Pathology Flashcards

1
Q

What is the definition of hyperplasia, hypertrophy, atrophy and metaplasia?

A

Hyperplasia - increase in the number of cells
Hypertrophy - increase in the size of cells
Atrophy - decrease in the size of a cell that has at one time been of normal size
Metaplasia - change of epithelial cell type from one to another at a specific site or location (the epithelium is normal in appearance in an abnormal location)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

What are the two types of hyperplasia? Give an example for each

A

Physiological hyperplasia - occurs due to a normal stressor (breasts in pregnancy, liver growth after resection
Pathological hyperplasia - occurs due to an abnormal stressor (adrenal gland growth due to production of ACTH by a pituitary adenoma)

Hyperplasia only happens in cells that divide (not the heart or neuron)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What are the two types of hypertrophy? Give an example for each

A

Physiological hypertrophy - occurs due to a normal stressor (enlargement of skeletal muscle after exercise)
Pathological hypertrophy - occurs due to an abnormal stressor (increase in heart size due to aortic stenosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

What are the two types of atrophy? Give an example for each

A

Physiological atrophy - occurs due to a natural stressor (decrease in size of uterus post-pregnancy)
Pathological atrophy - occurs due to an abnormal stressor (usually due to a loss of stilumus to that organ - blood supply, innervation, decreased workload, aging)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What is the mechanism of metaplasia? Give an example

A

The epithelium normally present at a site cannot handle the new environment so it converts to a type of epithelium that can adapt (Barrett’s oesophagus - squamous to glandular)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What are some causes of cell injury?

A
  • Trauma
  • Thermal injury, hot or cold
  • Poisons
  • Infectious organisms
  • Ionising radiation
  • Drugs
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What are the six mechanisms of cellular injury? Give examples for each

A

Mechanical disruption - trauma, osmotic pressure
Failure of membrane function integrity - damage to ion pumps, complement-mediated cytolysis, specific blockage of ion channels, alteration of membrane lipids
Membrane damage - free radicals
Deficiency of metabolites - oxygen, glucose, hormones
Blockage of metabolic pathways - interruption of protein synthesis, respiratory poisins
DNA damage or loss - ioinising radiation, chemotherapy, free radicals

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

What are the two ways in which cell damage can occur?

A

Reversibly and irreversibly

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What are the four things that the effect of cellular injury on a tissue depend on?

A
  • the duration of injury
  • the nature of the injurious agent
  • the proportion and type of cells that are affected
  • the ability of tissues to regenerate
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What is the definition of ischaemia and infarction?

A

Ischaemia is the pathological process resulting from a lack of oxygen due to impaired blood supply
Infarction is the specific process of necrosis resulting from lack of blood supply

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What is reperfusion injury?

A

Under some conditions it is possible for the onset of cell death to be delayed until blood flow has been restored.
It may be due to the generation of reactive oxygen species, free radicals and damage to calcium pups.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

In reperfusion injury, are cells damaged via apoptosis or necrosis

A

Cells damaged in this way probably go through apoptosis rather than necrosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What is a free radical?

A

Atoms or groups of atoms with an unpaired electron, as such they may enter into chemical bond formation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What are some of the clinicopathological events involving free radicals?

A
  • toxicity of some poisons
  • oxygen toxicity
  • tissue damage in inflammation
  • intracellular killing of bacteria
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What is necrosis and what causes it?

A

Death of tissues, causes include ichaemia, metabolic and truma. It is a pathological process

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

What are the six different types of necrosis? Give examples of each

A
  • Coagulative necrosis in most tissues, firm pale area with ghost outlines on microscopy (most common)
  • Colliquative necrosis is seen in the brain, the dead area is liquefied
  • Causeous necrosis is seen in TB, there is a pale yellow, semi-solid material
  • Gangrene is necrosis with putrefaction: it follows vascular occlusion or certain infections and is black
  • Fibrinoid necrosis is a microscopic feature in arterioles in malignant hypertension
  • Fat necrosis may follow trauma and cause a mass (often in the breast), or may follow pancreatitis visible as multiple white spots
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

What are the different types of gangrene?

A
  • Wet - in the bowel (incarcarated hernia) or appendicitis
  • Dry - usually seen in the toes (gradual arterial or small vessel obstruction in diabetes)
  • Gas - result of infection by Clostridium perfringens
  • Synergistic - infection by a combination of organisms
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

What are the two types of cell appearances following injury? Describe them

A
  • Hydropic change - cells when the cytoplasm becomes pale and swollen due to the accumulation of fluid (hypoxia or chemical poisoning) - often reversible
  • Fatty change - vacuolation of cells often due to the accumulation of lipid droplets as a result of a disturbance to ribosomal function and uncoupling of lipid from protein metabolism. (often in the liver) - moderate changes are reversible but severe fatty change is irreversible
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

What is the process of coagulative necrosis?

A

Following devitalisation, the cells retain their outline as their proteins coagulate and metabolic activity ceases.
Later, it may become soft as a result of digestion by the macrophages

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q
A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

What is the morphology of caseous necrosis?

A

A pattern of necrosis in which the dead tissue lacks any structure

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What is the process of fibrinoid necrosis?

A

Arterioles are under such pressure that there is necrosis of the smooth muscle wall. This allows seepage of plasma into the media with consequent deposition of fibrin.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

How does trauma cause fat necrosis?

A

The release of intra-cellular fat elicits a brisk inflammatory response, the polymorphs and macrophages phagocytosing the fat, proceeding eventually to fibrosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

What is the process of fat necrosis in pancreatitis?

A

There is release of pancreatic lipase. As a result, fat cells have their stored fat split into fatty acids, which then combine with calcium to precipitate out as white soaps

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

What is apoptosis?

A

Individual cell deletion in physiological growth control and in disease

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

What does reduced apoptosis cause?

A

Cell accumulation (neoplasia)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

What does increased apoptosis cause?

A

Extensive cell loss (atrophy)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

What are some of the roles of apoptosis?

A
  • Continuining control of organ size
  • Unwanted or defective cells undergo apoptosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

What factors control apoptosis?

A

Inhibitors include growth factors, cell matrix, sex steroids and some viral proteins
Inducers include growth factor withdrawal, loss of matrix attachment, glucocorticoids, some viruses, free radicals, ioinising radiation, DNA damage

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

How do apoptosis inducers and inhibitors work?

A

They act via the bcl-2 protein family, inhibiting or activating the death pathway, resulting in activation of caspases
OR
Activation of plasma membrane receptor Fas (CD95) by its ligand bypasses bcl-2 to activate other caspases

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

What is the process of apoptosis?

A

Degradation of the cytoskeletal framework
Fragmentation of DNA and loss of mitochondrial function
Nucleus shrinks (pykonosis) and fragments (karyorrhexis)
Cell shrinks, retaining an intact plasma membrane
Alteration of this membrane rapidly induces phagocytosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

What happens to dead cells not phagocytosed?

A

They break into smaller membrane-bound fragments called apoptotic bodies.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

What gene checks the integrity of the genome before mitosis? What happens next?

A

p-53 gene
Defective cells are swtiched to apoptosis instead

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

What diseases are associated with increased apoptosis?

A

AIDs (activate CD4, inducing apoptosis
Neurodegenerative disorders

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

What is the difference in induction of apoptosis vs necrosis?

A

Apoptosis - may be induced by physiological or pathological stimuli
Necrosis - invariably due to pathological injury

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

What is the difference in biochemical events of apoptosis vs necrosis?

A

Apoptosis - energy-dependent fragmentation of DNA by endogenous endonucleases - lysosomes intact
Necrosis - impairment or cessation of ion homeostasis - lysosomes leak lytic enzymes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

What is the difference in extent of apoptosis vs necrosis?

A

Apoptosis - single cell
Necrosis - cell groups

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

What is the difference in cell membrane integrity of apoptosis vs necrosis?

A

Apoptosis - maintained

Necrosis - lost

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

What is the difference in morphology of apoptosis vs necrosis?

A

Apoptosis - cell shrinkage and fragmentation to form apoptotic bodies with dense chromatin
Necrosis - cell swelling and lysis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

What is the difference in inflammatory response of apoptosis vs necrosis?

A

Apoptosis - none
Necrosis - usual

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

What is the difference in fate of dead cells of apoptosis vs necrosis?

A

Apoptosis - ingested (phagocytosed) by neighbouring cells
Necrosis - ingested (phagocytosed) by neutrophil polymorphs and macrophages

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

What is the definition of inflammation?

A

A response of vascularised tissues to infection and damaged tissues that brings cells and molecules of host defence from the circulation to the sites where they are needed, in order to eliminate the offending agents

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

In a broad sense

What are the key mediators of inflammation?

A

Phagocytic leukocytes, antibodies and complement proteins

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

What are some examples of key components of the innate immune system?

A

Natural killer cells, dendritic cells, and epithelial cells, as well as soluble factors such as the proteins of the complement system.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

What are the steps of the typical inflammatory reaction?

A
  • The offending agent, often in extrasvascular tissues, is recognised by host cells and molecules
  • Leukocytes and plasma proteins are recruited from the circulation to the site where the offending agents are located
  • The leukocytes and proteins are activated and work together to destroy and eliminate the offending substance
  • The reaction is controlled and terminated
  • The damaged tissue is repaired
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

How do blood vessels help in inflammation?

A
  • The blood vessels dilate to slow down blood flow, and by increasing their permeability, they enable certain selected proteins to enter the site of infection of tissue damage
  • The blood vessel epithelium characteristics change, such that circulating leukocytes first stop then enter the site of infection
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

What do leukocytes do once they are recruited?

A

They are activated and acquire the ability to ingest and destroy microbes and dead cells, as well as foreign bodies

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

What are some of the harmful effects of inflammation?

A
  • Acute temporary things such as pain and functional impairment
  • There are many diseases in which the inflammatory reaction is misdirected (against self tissues in autoimmune diseases), occurs against normally harmless substances (allergies) or is inadequately controlled
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

What cells are involved in these acute diseases:
1. ARDS
2. Asthma
3. Glomerulonephritis
4. Septic shock

A
  1. ARDS - neutrophils
  2. Asthma - IgE antibodies; eosinophils
  3. Glomerulonephritis - antibodies and complement, neutrophils, monocytes
  4. Septic shock - cytokines
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

What is the role of inflammatory mediators such as plasma proteins?

A

They initiate and amplify the inflammatory response and determine it’s pattern, severity, and clinical and pathological manifestations

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

Tell me about acute inflammation e.g. duration, mechanisms, and resolution

A
  • Typically develops within minutes or hours and is of short duration, lasting for several hours or a few days
  • Main characteristics are the exudation of fluid and plasma proteins (oedema), and the emmigration of leukocytes, particularly neutrophils,
  • When it reaches its desired goal, the reaction subsides, but if the response fails to clear the stimulus, the reaction can develop into chronic inflammation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

Tell me about chronic inflammation

A
  • It is of longer duration and is associated with more tissue destruction
  • Associated with the presence of lymphocytes and macrophages, the proliferation of blood vessels, and the deposition of connective tissue
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

What is the onset of acute vs chronic inflammation?

A

Acute - minutes to hours
Chronic - days

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

What are the cellular infiltrates in acute vs chronic inflammation?

A

Acute - neutrophils
Chronic - lymphocytes and macrophages

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

How are local and systemic signs present in acute vs chronic inflammation?

A

Acute - prominent
Chronic - less

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

How does the termination of inflammation occur?

A

It is terminated when the offending agent is eliminated
The reaction resolved because the mediators are broken down and dissipated, the leukocytes have a short life span in tissues
In addition, anti-inflammatory mechanisms are activated, serving to control the host and prevent it from causing excessive damage

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

What are some examples of things that trigger inflammation?

A
  • Infections - bacterial, viral, fungal, parasitic
  • Tissue necrosis elicits inflammation regardless of the cause of cell death, which may include ischaema, trauma, physical and chemical injury
  • Foreign bodies - splinters, dirt, sutures - or endogenous molecules depositing in tissues e.g. urate in gout, cholesterol in atherosclerosis
  • Immune reactions - autoimmune diseases, allergies
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

What are some cells and receptors that recognise microbes and damaged cells?

A
  • Cellular receptors for microbes in the plasma membrane (extracellular), the endosomes (ingested), and the cytosol (intracellular) - e.g. Toll-like receptors TLRs
  • Sensors of cell damage - all cells have cytosolic receptors that recognise a diverse set of molecules that are liberted of altered as a consequence of cell damage
  • Other cellular receptors involved in inflammation - many leukocytes express receptors for the Fc tails of antibodies and for complement proteins
  • Circulating proteins - the complement system reacts against microbes and produces inflammatory mediators, mannose-binding lectin recognises microbial sugars and promotes ingestion of the microbes and activation of the complement system
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

Where are TLRs expressed?
What do they do?

A

They are expressed on many cell types, including epithelial cells, dendritic cells, macrophages and other leukocytes
Engagement of these receptors triggers production of molecules involved in inflammation including adhesion molecules on endothelial cells, cytokines and other mediators

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

What are some examples of cytosolic sensors of cell damage?

A
  • Uric acid - a product of DNA breakdown
  • ATP - released from damaged mitochondria
  • Reduced intracellular K+ concentrations - reflecting loss of ions because of membrane injury
  • DNA in the cytoplasm because it should normally be in the nucleus
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

What do cytosolic sensors of cell damage do?

A
  • They activate a multiprotein cytosolic complex called inflammasome, which induces the production of IL-1
  • IL-1 recruits leukocytes and thus induces inflammation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

What do the leukocyte receptors for the Fc tails do?

A

They recognise microbes coated with antibodies and complement (opsonisation) and promote ingestion and destruction of the microbes as well as triggering inflammation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

What are the three major components of acute inflammation?

A
  1. Dilation of small vessels, leading to an increased blood flow
  2. Increased permability of the microvasculature enabling plasma proteins and leukocytes to leave the circulation
  3. Emigration of the leukocytes from the microcirculation, their accumulation in the focus on injury, and their activation to eliminate the offending agent
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

What is an exudate vs a transudate?

A

Exudate is an extravascular fluid that has a high protein concentration and contains cellular debris. Its presence implies an ongoing inflammatory response
Transudate is a fluid with low protein content, little or no cellular material, and low specific gravity. It is essentially an ultrafiltrate of blood plasma that is produced as a result of imbalance between hydrostatic and osmotic pressure without an increase in vascular permability

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

What is edema?

A

An excess of fluid in the interstital tissue or serous cavities, it can be either exudate or transudate

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
66
Q

What is pus?

A

A purulent inflammatory exudate rich in leukocytes (mostly neutrophils), the debris of dead cells, and in many cases, microbes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
67
Q

What are the changes in vascular flow and caliber after injury?

A
  • Vasodilation is induced by the action of many mediators, notably histamine, on vascular smooth muscle. First the arterioles then capillaries, causing increased blood flow and erythema
  • Increased permeability of the microvasculature, with the outpouring of protein-rich fluid into extravascular tissues
  • These changes lead to engorgement of small vessels, which slowly moving red cells, a condition termed stasis (again causing erythema)
  • As stasis develops, blood leukocytes, principally neutrophils, accumulate along the vascular endothelium, simultaneously, endothelial cells are activated by inflammatory mediators and express increased levels of adhesion molecules, adhering to leukocytes, which then migrate through the vascular wall
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
68
Q

What are the different mechanisms of increased vascular permability in short? (we’ll go into more detail after this)

A
  • Contraction of endothelial cells resulting in increased endothelial spaces is the most common mechanism of vascular leakage
  • Endothelial injury, resulting in endothelial cell necrosis and detachment
  • Increased transport of fluid and proteins called transcytosis, through the endothelial cell
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

What chemical mediators trigger endothelial cell contraction?

A

Histamine, bradykinin, leukotrienes, and other chemical mediators

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
70
Q

When does endothelial cell contraction happen and how long does it last for?

A

It is called the immediate transient response because it happens occurs rapidly after exposure to the mediator and is often short-lived (15 to 30 minutes)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

What is delayed prolonged leakage?

A
  • In some forms of mild injury (e.g. sunburn), vascular leakage begins after a delay of 2-12 hours and lasts for several hours or even days
  • It may be caused by contraction of endothelial cells or mild endothelial damage
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
72
Q

What is the mechanism behind endothelial injury causing increased vascular permeability?

A

Direct damage to the endothelium is encountered in severe injuries and is induced by the action of microbes and microbial toxins that target endothelial cells

Neutrophils that adhere to the endothelium during inflammation may also injure the endothelial cells and thus amplify the reaction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
73
Q

What does leakage occur due to endothelial injury, necrosis and detatchment after injury?

A

In most instances, leakage starts immediately after injury and is sustained for several hours until the damaged vessels are thrombosed or repaired

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
74
Q

How do lymphatic vessels respond to inflammation?

A

In inflammation, lymphatic flow increases and helps drain oedema fluid. In addition to fluid, leukocytes and cell debris, as well as microbes, may find their way into the lymph.

Lymphatic vessels proliferate during inflammatory reactions to handle the increased load.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
75
Q

What are the most important leukocytes in inflammation?

A

Neutrophils and macrophages

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q

What is the function of leukoytes in the inflammatory response?

A
  • Ingest and destroy bacteria and other microbes, as well as necrotic tissue and foreign substances
  • Produce growth factors that aid in repair
  • When strongly activated, they may induce tissue damage and prolong inflammation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
77
Q

The journey of leukocytes from the vessel lumen to the tissue is a multi-step process that is mediated and controlled by what?

A

Adhesion molecules and cytokines called chemokines

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
78
Q

What are the three phases of leukocyte migration?

A
  1. In the lumen: margination, rolling, and adhesion to the endothelium
  2. Migration across the endothelium and vessel wall
  3. Migration in the tissue toward a chemotactic stimulus
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
79
Q

How does margination occur?

A

In normal flowing venules, red cells are confined to a central axial column, displacing the leukocytes towards the wall of the vessel

Because of stasis (blood flow slowing) in early inflammation, haemodynamic conditions change, and more white cells assume a peripheral position along the enothelial surface

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
80
Q

At what stage of the inflammatory process do neutrophils predominate?

A

6 - 24 hours

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
81
Q

At what stage of the inflammatory process do monocytes predominate?

A

24 - 48 hours

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
82
Q

What are the mediators that trigger the following:
1. Leukocyte rolling on endothelium
2. Firm attachment to the endothelium

A
  1. Selectins (P-selectin (platelets), L-selectin (leukocytes), E-selectin (endothelium)) trigger leukocyte rolling
  2. Integrins trigger firm attachment to the endothelium
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

Which cytokines promote expression of selectins and integrins on endothelium?

A

TNF
IL-1

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
84
Q

Which cytokines increases the avidity of integrins for their ligands and promote directional migration of the leukocytes?

A

Chemokines

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
85
Q

What is transmigration or diapedesis?

A

It is migration of the leukocytes through the endothelium.
It often happens mainly in the postcapillary venules

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
86
Q

After exiting the circulation, the leukocytes move in the tissues toward the site of injury. What is this process called?

A

Chemotaxis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
87
Q

What are some endogenous chemoattractants? (things released in an injury that attract leukocytes in inflammation)

A
  • Cytokines, particularly those of the chemokine family e.g. IL-8
  • Components of the complement system (mainly C5a)
  • Arachidonic acid (AA) metabolites, mainly leukotriene B4
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
88
Q

What are the three steps of phagocytosis?

A
  1. Recognition and attachment of the particle to be ingested by the leukocyte
  2. Engulfment, with subsequent formation of a phagocytic vacuole
  3. Killing or degradation of the ingested molecule
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
89
Q

What phagocytic receptors bind and ingest molecules?

A
  • Mannose receptors - a lectin that binds to terminal mannose and fucose (which are only on microbes) therefore recognises microbes and not host cells
  • Scavenger receptors - bind to LDLs
  • Receptors for various opsonins - main ones are IgG antibodies, C3b breakdown product and certain lectins (mannose-binding lectins)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
90
Q

How does engulfment take place in phagocytosis?

A
  • After a particle is bound to the phagocyte receptors, extensions of the cytoplasm (psuedopods) flow around it
  • The plasma membrane pinches off to form a vesicle (phagosome) that engulfs the particle
  • The phagosome fuses with a lysosome granule, resulting in release of the granules contents into the phagosome
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
91
Q

What molecules are used to perform intracellular destruction of microbes and debris once in the phagosome and how?

A
  • Reactive oxygen series are produced by the oxidisation of NADPH by NADPH oxidase - it accompanies phagocytosis - produced by lysosome - may be released extracellularly, causing tissue damage
  • Reactive nitrogen series
  • Lysosomal enzymes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
92
Q

What are neutrophil extracellular traps?

A

Extracellular fibrillar networks that provide a high concentration of antimicrobial substances at sites of infection and prevent the spread of the microbes by trapping them in the fibrils

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
93
Q

What are mediators of inflammation?

A

Substances that initiate and regulate inflammatory reactions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
94
Q

What are the most important mediators of acute inflammation?

A

Vasoactive amines
Lipid products (prostaglandins and leukotrienes)
Cytokines (including chemokines)
Products of complement activation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
95
Q

How are inflammatory mediators usually created?

A

They are either secreted by cells or generated by plasma proteins.
Cell-derived mediators are usually sequestered by intracellular granules and can be rapidly secreted by granule exocytosis (e.g. histamine in mast cell granules) or are synthesised de novo (e.g. prostagladins, leukotrienes, cytokines) in response to a stimulus

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
96
Q

What are the main cells that create inflammatory mediators?

A

The major cell types that produce mediators of inflammation are the sentinels that detect invaders and damage in tissues, such as macrophages, dendritic cells, and mast cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
97
Q

How long do inflammatory mediators last for?

A

They are usually short-lived.

They quickly decay or are inactivated by enzymes, or they are otherwise scavenged or inhibited.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
98
Q

Can one inflammatory mediator stimulate another one?

A

Yes!

For instance, products of complement activation can stimulate the release of histamine. the cytokine TNF acts on endothelial cells to stimulate the production of another cytokine IL-1.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
99
Q

Where is histamine created and what does it do?

A

It is created in mast cells, basophils and platelets.
It causes vasodilation, increased vascular permeability and endothelial activation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
100
Q

Where are prostaglandins created and what do they do?

A

It is created in mast cells, macrophages, endothelial cells and leukocytes
It causes vasodilation, pain and fever

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
101
Q

Where are leukotrienes created and what do they do?

A

They are created in mast cells and leukocytes.
They cause increased vascular permability, chemotaxis, leukocyte adhesion, and activation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
102
Q

Where are cytokines (TNF, IL-1 and IL-6) created and what do they do?

A

They are created in macrophages, endothelial cells and mast cells.
Locally, they cause endothelial activation (expression of adhesion molecules).
Systemically, they cause fever, metabolic abnormalities, hypotension (shock)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
103
Q

Where is complement created and what does it do?

A

It is in the plasma, created by the liver.
It causes leukocyte chemotaxis and activation, direct target killing (membrane attack complex), vasodilation (mast cell stimulation)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
104
Q

Where are kinins created and what do they do?

A

They are found in the plasma, created in the liver.
They cause increase vascular permeability, smooth muscle contraction, vasodilation and pain

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
105
Q

How does histamine increase vascular permeability?

A

It binds to H1 receptors on microvascular endothelial cells, creating interendothelial gaps in venules and causing endothelial contraction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
106
Q

What things stimulate the release of histamine?

A
  1. physical injury, such as trauma, cold or heat
  2. binding of antibodies to mast cells (immediate hypersensitivity reactions)
  3. products of complement called anaphylatoxins (C3a and C5a)
  4. Neuropeptides (substance P) and cytokines (IL-1 and IL-8) may also trigger histamine release
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
107
Q

What are some examples of arachidonic acid metabolites?

A

The lipid mediators prostaglandins and leukotrienes are produced from arachidonic acid (AA) present in membrane phospholipids, and stimulate vascular and cellular reactions in acute inflammation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
108
Q

Arachidonic acid (AA) is usually found esterified in the membrane phospholipids. How is it released?

A

Through the action of cellular phospholipases (mainly phospholipase A2).
An increase in cytoplasmic Ca2+ and activation of various kinases in response to external stimuli, activates phospholipase A2

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
109
Q

What is created when AA is released from the membrane phospholipids?

A

From the AA, two major enzymes: cyclooxygenases and lipoxygenases synthesise many AA-derived inflammatory mediators - called eicosanoids
Cyclooxygenases create prostaglandins
Lipooxygenases create leukotrienes and lipoxins

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
110
Q

How are prostaglandins generated?

A

By the actions of cyclooxygenase 1 and 2.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
111
Q

What is the difference between COX-1 and COX-2?

A

COX-1 is produced in response to an inflammatory stimuli and is also constitutively expressed in most tissues, where it may serve a homeostatic function
COX-2 is induced by inflammatory stimuli and thus generates prostaglandins, but it is low or absent in normal tissues

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
112
Q

What is the function of prostacyclin?

A

It is a vasodilator and a potent inhibitor of platelet aggregation, and also markedly potentiates the permeability-increasing and chemotactic effects of other mediators

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
113
Q

What are the most important cytokines in inflammation? What do they do?

A

TNF & IL-1 serve critical roles in leukocyte recruitment by promoting adhesion of leukocytes to endothelium and their migration through vessels.
* They increase expression of adhesion molecules (E- and P- selectin)
* They increase the production of other cytokines, growth factors and eicosanoids
* They produce fever and can cause sepsis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
114
Q

What are chemokines and what is their role?

A

A family of small proteins that act as chemoattractants for specific types of leukocytes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
115
Q

What are the main morphological features of acute inflammation?

A

Dilation of small bloods vessels, accumulation of leukocytes and fluid in the extravascular tissue

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
116
Q

What are the different morphological types of acute inflammation?

A
  • Serous inflammation is marked by exudation of cell-poor fluid into spaces created by cell injury or into body cavities lined by the peritoneum, pleura or pericardium
  • Fibrinous inflammation is where a fibrinous exudate develops when the vascular leaks are large or there is a local procoagulant stimulus
  • Purulent (suppurative) inflammation is characterised by the production of pus, an exudate containing neutrophils, the liquefied debris of necrotic cells and edema fluid
  • Ulcers are a local defect, or excavation, of the surface of an organ or tissue that is produced by the sloughing of inflamed necrotic tissue
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
117
Q

What is (or isn’t) in the fluid of serous inflammation?

A

Typically, the fluid is not infected by destructive organisms and does not contain large numbers of leukocytes.
The fluid may be derived from plasma or from the secretions of mesothial cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
118
Q

What is the mechanism of fibrinous inflammation?

A

With greater increase in vascular permeability, large molecules such as fibrinogen pass out of the blood, and fibrin is formed and deposited in the extracellular space.
A fibrous exudate is characteristic of inflammation of the body cavities, such as the meninges, pleura and pericardium.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
119
Q

How does fibrinous inflammation present histologically?

A

Fibrin appears as an eosinophilic meshwork of threads, or sometimes as an amorphuos coagulum

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
120
Q

How does fibrinous inflammation resolve?

A

Fibrinous exudates may be dissolved by fibrinolysis and cleared by macrophages.

If the fibrin is not removed, over time it may stimulate the growth of fibroblasts and blood vessels and thus lead to scarring.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
121
Q

What are abscesses?

A

They are localised collections of purulent inflammatory tissue caused by a suppuration buried in a tissue, an organ or a confined space

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
122
Q

What components make up an abscess?

A

Abscesses have a central region that appears as a mass of nectrotic leukocytes and tissue cells. There is usually a zone of preserved neutrophils around this necrotic focus, and outside this region, there may be vascular dilation and parenchymal and fibroblastic proliferation, indicating chronic inflammation and repair.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
123
Q

How does an ulcer develop?

A

During the acute stage, there is intense polymorphonuclear infiltration and vascular dilation in the margins of the defect.
With chronicity, the margins and the base of the ucler develop fibroblastic proliferation, scarring and the accumulation of lymphocytes, macrophages and plasma cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
124
Q

What are the outcomes of acute inflammation?

A
  • Complete resolution - the injury is short-lived, there is little tissue destruction and the damaged parenchymal cells can regenerate
  • Healing by connective tissue replacement (scarring or fibrosis) - there is substantial tissue destruction, abundant fibrin exudation that can’t be cleared, connective tissue grows into the area of damage, converting it into a mass of fibrous tissue, a process called organisation
  • Chronic inflammation occurs when the acute inflammatory response cannot be resolved, either due to the microbe or an inadequate immune response
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
125
Q

What is chronic inflammation?

A

A response of prolonged duration (weeks to months) in which inflammation, tissue injury and attempts of repair co-exist, in varying combinations

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
126
Q

What are the causes of chronic inflammation?

A
  • Persistent infections by infections that are difficult to eradicate (e.g.mycobacteria, funghi, parasites) - these may be a delayed hypersensitivity reaction or granulomatous reactions
  • Hypersensitivity diseases such as autoimmune diseases or allergic diseases
  • Prolonged exposure to potentially toxic agents, either endogenous or exogenous for example silicosis (exogenous) or atherosclerosis (endogenous)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
127
Q

What are the morphological characteristics of chronic inflammation?

A
  • Infiltration with mononuclear cells, which include macrophages, lymphocytes and plasma cells
  • Tissue destruction, induced by the persistent offending agent, or by the inflammatory cells
  • Attempts at healing by connective tissue replacement of damaged tissue, accomplished by angiogenesis and fibrosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
128
Q

What is the most dominant cell in chronic inflammation and what does it do?

A

Macrophages contribute to the reaction by secreting cytokines and growth factors that act on various cells, by destryoing foreign invaders and tissues, and by activating other cells, notably T lymphocytes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
129
Q

What are the stages in the life cycle of a macrophage?

A
  • They are tissue cells derived from haematopoietic stem cells in the bone marrow and from progenitors in the embryonic yolk sac and fetal liver during early development. They circulate as monocytes
  • Monocytes enter the blood, migrate into various tissues and differentiate into macrophages
  • The life span of a monocyte in 1 day, whereas a tissue macrophage can last for several months or years
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
130
Q

What are the names of macrophages in the following organs:
1. Liver
2. Spleen and lymph nodes
3. Central nervous system
4. Lungs

What do they form together?

A
  1. Liver - Kuppfer cells
  2. Spleen and lymph nodes - Sinus histiocytes
  3. CNS - microglial cells
  4. Lungs - alveolar macrophages

Together they form the mononuclear phagocyte system

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
131
Q

There are two major pathways of macrophage activation. What are they?

A
  • Classical macrophage activation may be induced by microbial products such as endotoxins, which engage TLRs and other cells by T cell-drived signals (cytokine IFN-gamma). Classically activate macrophages (M1) produce NO and ROS and upregulate lysosomal enzymes. These things help them kill ingested organisms, trigger inflammation via cytokines.
  • Alternate macrophage activation is induced by cytokines other than IFN-gamma (IL-4 and IL-13), produced by T lymphocytes and other cells. The function of alternately activated macrophages (M2) is tissue repair. They secrete GF that stimulates angiogenesis, activate fibroblasts and stimulate collagen synthesis.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
132
Q

What are the main functions of macrophages in chronic inflammation?

A
  • Ingest and eliminate microbes and dead tissue
  • Initiate the process of tissue repair
  • Secrete mediators of inflammation, such as cytokines (TNF, IL-1, chemokines) and eicosanoids
  • Display antigens to T-lymphocytes and respond to signals from T cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
133
Q

What are the roles of lymphocytes in chronic inflammation?

A

Microbes and other environmental antigens activate T and B lymphocytes, which amplify and propogate chronic inflammation.
Lymphocytes may be the dominant population in the chronic inflammation seen in autoimmune and other hypersensitivity diseases

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
134
Q

There are three subsets of CD4+ T cells that secrete different cytokines and ellicit different types of inflammation. Tell me them…

A
  1. TH1 cells produce the cytokine IFN-gamma, which activates macrophages via the classical pathway
  2. TH2 cells secrete IL-4, IL-5 and IL-13, which recruit and activate eosinophils and are reponsible for the alternative pathway of macrophage activation.
  3. TH17 cells secrete IL-17 and other cytokines, which induce the secretion of chemokines, reponsible for the recruitment of neutrophils (and monocytes) to the reaction
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
135
Q

Lymphocytes and macrophages interact in a bidirectional way, and these interactions play an important role in propogating chronic inflammation. Tell me about them…

A
  • Macrophages display antigens to T cells, express membrane molecules (called costimulators), and produce cytokines (IL-12) that stimulate T cell responses.
  • Activated T cells produce cytokines (IFN-gamma), which recruit and activate macrophages, promoting more antigen presentation and cytokine secretion
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
136
Q

What is a tertiary lymphoid organ? What is the term used for it’s formation?

A

In some chronic inflammatory reactions, the accumulated lymphocytes, antigen-presenting cells, and plasma cells cluster together to form lymphoid tissues resembling - this is a tertiary lymphoid organ.

It’s formation is called lymphoid organogenesis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
137
Q

Apart from lymphocytes and macrophages, what other cells are involved in chronic inflammation?

A
  • Eosinophils are abundant in immune reactions mediated by IgE and parasitic infections. Their recruitment is similar to neutrophils, via the chemokine eotaxin. They contain major basic protein, which is toxic to parasites but also mammalian epithelial cells
  • Mast cells are present in chronic inflammatory reactions, and because they secrete a plethora of cytokines, they may promote inflammatory reactions in different situations.
  • Although neutrophils are characteristic of acute inflammation, many forms of chronic inflammation still show large numbers of neutrophils
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
138
Q

What is granulomatous inflammation?

A

A form of chronic inflammation characterised by collections of activated macrophages, often with T lymphocytes, and sometimes associated with central necrosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
139
Q

There are two types of granulomatous inflammation, which differ in pathogenesis. What are they and what is their pathogeneses?

A
  • Foreign body granulomas are incited by relatively inert foreign bodies, in the abscense of T cell-mediated immune responses. Typically occur around talc (IVDU) or sutures, things large enough to not be phagocytosed. Epitheliod cells and giant cells are opposed to the foreign body surface.
  • Immune granulomas are caused by agents capable of inducing a persistent T-cell mediated immune response. The agent is often difficult to erradicate. In such responses, macrophages keep activatign T cells and T cells keep activating macrophages until giant cells epitheliod cells form.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
140
Q

What are epithelioid and giant cells?

A
  • The activated macrophages may develop abundant cytoplasm and begin to resemble epithelial cells, these are called epithelioid cells
  • Some activated macrophages may fuse, forming multinucleate giant cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
141
Q

What are the two types of reaction that could occur in repair of damaged tissue?

A
  • Regeneration by proliferation of residual (uninjured) cells and maturation of tissue stem cells
  • The deposition of scar tissue to form a scar
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
142
Q

How does regeneration occur in tissue repair?

A

Some tissues are able to replace the damaged components and essentially return to a normal state - this is called regeneration
It occurs by proliferation of cells that survive the injury and retain the capacity to proliferate

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
143
Q

In what organs does regeneration often occur in tissue repair?

A

The rapidly dividing epithelia of the skin and intestines, and in some parenchymal organs, notably the liver.
In other cases, tissue stem cells may contribute to the restoration of the damaged tissues

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
144
Q

When would scar tissue form in tissue healing?

A

If the injured tissues are incapable of complete restitution, or if the supporting structures of the tissue are severely damaged, repair occurs by the laying down of connective (fibrous) tissue.
Although the fibrous scar is not normal, it provides enough structural stability that the injured tissue can continue to function

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
145
Q

What is fibrosis? and how it this linked with organisation?

A

The term fibrosis is often used to describe the extensive depostion of collagen that occurs in the lungs, liver, kidney and other organs as a consequence of chronic inflammation or in the myocardium after extensive ischaemic necrosis.
If fibrosis occurs in a tissue space occupied by an inflammatory exudate, it is called an organisation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
146
Q

The regeneration of injured cells and tissues involves cell proliferation. What is the driven by and dependent on?

A

It is driven by growth factors and is critically dependent on the integrity of the extracellular matrix, and by the development of mature cells from the stem cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
147
Q

Several cell types proliferate during tissue repair. What are they and why do they proliferate?

A
  • Remnants of the injured tissue - which attempt to restore normal structure
  • Vascular endothelial cells - to create new vessels that provide the nutrients required for tissue repair
  • Fibroblasts - the source of the fibrous tissue that forms the scar to fill defects that cannot be corrected by regeneration
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
148
Q

The ability of tissues to repair themselves is determined, in part, by their intrinsic proliferative capacity. Based on this, tissues are divided into three groups. What are the groups and tell me about them?

Not examples yet, that will be next …

A
  • Labile (continuously dividing) tissues - cells of these tissues are constantly being lost and replaced by maturation from tissue stem cells and by proliferation of mature cells. They can readily regenerate after injury, as long as the stem cells aren’t damaged
  • Stable tissues - cells of these tissues are quiescent (in the G0 stage of the cell cycle) and have only minimal proliferative activity in their normal state. However, they are capable of dividing in response to injury but have a limited capacity for regeneration
  • Permanent tissues - The cells of these tissues are considered to be terminally differentiated and nonproliferative in post-natal life. Repair is normally dominated by scar formation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
149
Q

Give me some examples of labile, stable and permanent tissues…

A
  • Labile - haematopoeitic cells in the bone marrow, stratified squamous epithelium of the skin, oral cavity, vagina, and cervix ; the cuboidal epithelia of the ducts draining exocrine organs (salivary glands, pancreas, biliary tract); columnar epithelium of GI tract, uterus, fallopian tubes and the transitional epithelium of the urinary tract
  • Stable - the parenchyma of most solid tissues, such as liver, kidney and pancreas. They also include endothelial cells, fibroblasts, smooth muscle cells.
  • Permanent - cardiac myocytes and neurons
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
150
Q

Where and by what cells are growth factors released that drive tissue repair?

A
  • They are typically produced by cells near the site of damage.
  • The most important sources are macrophages activated by the tissue injury, but epithlial cells and stromal cells also produce some of them.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
151
Q

How do growth factors drive tissue repair?

A
  • Several growth factors bind to ECM proteins and are displayed at high concentrations
  • All growth factors activate signaling pathways that ultimately induce the production of proteins that are involved in driving cells through the cell cycle and other proteins that release blocks on the cell cycle.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
152
Q

In addition to growth factors, what else can stimulate cell proliferation in tissue repair?

A

Cells use integrins to bind to ECM proteins, and signals from the integrins can stimulate cell proliferation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
153
Q

What are the most important stem cells for regeneration after injury? Where are they and what do they do?

A

Tissue stem cells live in specialised niches, and it is believed that injury triggers signals in these niches that activate quiescent stem cells to proliferate and differentiate into mature cells that repopulate the injured tissue

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
154
Q

How is the loss of blood cells corrected and what triggers this?

A

Loss of blood cells is corrected by the proliferation of haematopoietic stem cells in the bone marrow and other tissues, driven by growth factors called colony-stimulating factors, which are produced as the response to the reduced number of blood cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
155
Q

What are the steps in scar formation that follow tissue injury and the inflammation response?

A
  1. Angiogenesis to supply nutrients and oxygen needed to support the repair process. Newly formed vessels are leaky because of incomplete interendothelial junctions and because VEGF, the growth factor that drives angiogenesis, increases vasular permeability
  2. Formulation of granulation tissue - migration and proliferation of fibroblasts and the deposition of loose connective tissue, together with the vessels and interspersed leukocytes, form granulation tissue. It progressively invades the site of injury
  3. Remodelling of the connective tissue - maturation and reorganising of the connective tissue produces the stable fibrous scar. The amount of connective tissue increases in the granulation tissue, eventually resulting in a scar
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
156
Q

What is the histological appearance of granulation tissue?

A

Proliferation of fibroblasts and new thin-walled delicate capillaries (angiogenesis) in a loose extracellular matrix, often with admixed inflammatory cells, especially macrophages

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
157
Q

How soon after injury do the steps of tissue repair take place?

A
  • Repair begins within 24 hours of injury by the emmigration of fibroblasts and the induction of fibroblast and endothelial cell proliferation.
  • By 3-5 days, the specialised granulation tissue that is characteristic of healing is apparent.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
158
Q

What is angiogenesis and when does it happen?

A

Angiogenesis is the process of new blood vessel development from existing vessels
It is critical:
* in healing at sites of injury
* in the development of collateral circulations at sites of ischaemia
* in allowing tumours to increase in size beyond the constraints of their original blood supply

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
159
Q

What are the steps of angiogenesis?

A
  1. Vasodilation in response to NO and increased permeability induced by VEGF
  2. Seperation of pericytes from the abluminal surface and breakdown of the basement membrane to allow formation of a vessel sprout
  3. Migration of endothelial cells towards the site of tissue injury
  4. Proliferation of endothelial cells just behind the leading front (‘tip’) of migrating cells
  5. Remodelling into capillary tubes
  6. Recruitment of periendothelial cells (pericytes for small vessels, smooth muscle for large) to form the mature vessel
  7. Suppression of endothelial proliferation and migration and deposition of the basement membrane
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
160
Q

The process of angiogenesis involves several signalling pathways, cell-cell interactions, ECM proteins and tissue enzymes.
What growth factors are required (name 5) and what do they do?

A
  • Vascular endothelial growth factors (VEGF), mainly VEGF-A, stimulates both migration and proliferation of endothelial cells, initiating the process of capillary sprouting. It promotes vasodilation by stimulating the production of NO and contributes to the formation of the vascular lumen
  • Fibroblast growth factors, mainly FGF-2, stimulates the proliferation of endothelial cells. It also promotes the migration of macrophages and fibroblasts to the area, and stimulates epithelial cell migration to cover epidermal wounds
  • Angiopoietins 1 and 2 (Ang 1 & 2) are growth factors that play a role in angiogenesis and the structural maturation of new vessels. Ang1 interacts with an endothelial tyrosine kinase receptor called Tie1
  • PGDF recruits smooth muscle cells
  • TGF-beta suppresses endothelial cell proliferation and migration, and enhances the production of ECM proteins
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
161
Q

What is Notch signalling?

A

Through ‘cross-talking’ with VEGF, the Notch signalling pathway regulates the sprouting and branching of new vessels and thus ensures that the new vessels that are formed have the proper spacing to effectively supply the healing tissue with blood.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
162
Q

The process of angiogenesis involves several signalling pathways, cell-cell interactions, ECM proteins and tissue enzymes.
What is the function of ECM proteins?

A

ECM proteins participate in the process of vessel sprouting, largely with interactions with integrin receptors in endothelial cells and providing the scaffold for vessel growth

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
163
Q

The process of angiogenesis involves several signalling pathways, cell-cell interactions, ECM proteins and tissue enzymes.
What is the function of the tissue enzymes?

A

Enzymes in the ECM, notably the matrix metalloproteinases (MMPs), degrade the ECM to permit remodelling and extension of the vascular tube

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
164
Q

What are the two steps in the laying down of connective tissue in tissue repair?

A
  1. Migration and proliferation of fibroblasts to the site of injury
  2. The deposition of ECM proteins produced by these cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
165
Q

The process of laying down connective tissue is orchestrated from what? and where are these produced?

A

The proceses are orchestrated by locally produced cytokines and growth factors, including PDGF, FGF-2 and TGF-beta.

The major sources for these are inflammatory cells, particularly macrophages activated by the alternative pathway (M2). Mast cells and lymphocytes can also release them

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
166
Q
A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
167
Q

What is the most important cytokine for the synthesis and deposition of the connective tissue proteins?

A

Transforming growth factor - beta
TGF-beta

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
168
Q

Where is TGF-beta produced and what does it do?

A
  • It is produced by most of the granulation tissue, including alternatively activated macrophages.
  • It stimulates fibroblast migration and proliferation, increased synthesis of collagen and fibronectin, and decreased degradation of the ECM due to inhibition of metalloproteinases
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
169
Q

What factors regulate TGF-beta production?

A

The levels in tissues are primary regulated not by the transcription of the gene but by the posttranscriptional activation of latent TGF-beta, the rate of secretion of the active molecule, and factors in the ECM (notably integrins), that enhance or diminish TGF-beta activity

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
170
Q

What are the processes in which TGF-beta is involved in?

A
  • Scar formation after injury
  • Development in fibrosis in the lung, liver and kidney that follow chronic inflammation
  • It serves as an anti-inflammatory cytokine that limits and terminates the inflammatory response
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
171
Q

How does TGF-beta limit and terminate the inflammatory response?

A

It inhibits leukocyte production and the activity of other leukocytes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
172
Q

How do fibroblasts change as healing progresses and what is the outcome of this?

A

As healing progresses, the number of proliferating fibroblasts and new vessels decreases; however, the fibroblasts progressively assume a more synthetic phenotype, and hence there is an increased deposition of ECM.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
173
Q

When does collagen synthesis by fibroblasts begin after injury and how long does it last for?

A

It begins early in wound healing (3-5 days) and continues for several weeks, depending on the size of the wound.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
174
Q

What is a scar tissue composed of?

A

Largely inactive, spindle-shaped fibroblasts, dense collagen, fragments of elastic tissue and other ECM components

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
175
Q

What happens to the scar as it matures?

A

There is progressive vascular regression, which eventually transforms the highly vascularised granulation tissue into a pale, largely asvascular scar.
Some of the fibroblasts also acquire features of smooth muscle cells, including the presence of actin filaments, and are called myofibroblasts.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
176
Q

How do myofibrils change a scar over time?

A

The contribute to the contraction of a scar over time

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
177
Q

What affects the outcome of the repair process?

A

It is influenced by a balance between the synthesis and degradation of the ECM proteins

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
178
Q

What family degrades the ECM proteins in scar formation?

A

The degradation of collagens and other ECM components is accomplished by a family of matrix metalloproteinases (MMPs), so called because they are dependent on metal ions (zinc) for their activity.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
179
Q

What are some examples of some matrix metalloproteinases (MMPs)?

A
  • Interstitial collagenases which cleave fibrillar collagen (MMP-1, -2 and -3)
  • Gelatinases which degrade amorphous collagen and firbonectin (MMP-2 and -9)
  • Stromelysins which degrade a variety of ECM constituents, including proteoglycans, laminin, fibronectin and amorphous collagen (MMP -3, -10 and -11)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
180
Q

What cells produce MMPs and what regulates them?

A
  • MMPs are produced by a variety of cell types (fibroblasts, macrophages, neutrophils, synovial cells, and some endotheliali cells)
  • Their synthesis and secretion is regulated by growth factors, cytokines and other agents
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
181
Q

MMPs have to be tightly controlled. What processes control this?

A
  • They are produced as inactive precursors (zymogens) that must first be activated by proteases (e.g.plasmin) likely to be present only at the site of injury
  • In addition, activated collagenases can be rapidly inhibited by specific tissue inhibitors of metalloproteinases (TIMPs), produced by most mesenchymal cells.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
182
Q

What are the factors that affect tissue repair?

A
  • Infection prolongs inflammation
  • Diabetes prolongs wound healing
  • Nutritional status particularly protein deficiency and vitamin C deficiency
  • Glucocorticoids (steroids) inhibit TGF-beta production and diminish fibrosis
  • Mechanical factors such as increased local pressure or torsion
  • Poor perfusion due to either atherosclerosis and diabetes or to obstructed venous damage
  • Foreign bodies
  • The type and extent of tissue injury - the type of cells, as explained earlier (you know this gurl)
  • The location of the injury depends on the type of acute inflammation that happens and some of those repair better than others
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
183
Q

What is healing by first intention vs second intention?

A
  • First intention is when the injury only involves the epithelial layer, the principal mechanism of repair is epithelial regeneration
  • Second intention is when cell of tissue loss is more extensive, such as in large wounds, abscesses, ulceration and ischaemic necrosis in parenchymal organs, the repair process involves a combination of regeneration and scarring
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
184
Q

What are the steps of first intention healing?

A
  1. Inflammation
  2. Proliferation of epithelial and other cells
  3. Maturation of the connective tissue scar
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
185
Q

What happens immediately during healing of first intention?

A
  • Wounding causes the rapid activation of coagulation pathways, which results in the formation of a blood clot on the wound surface.
  • In addition to trapped red cells, the clot contains fibrin, fibronectin, and complement proteins.
  • The clot serves to stop bleeding and act as a scaffold for migrating cells, which are attracted by growth factors, cytokines and chemokines.
  • Release of VEGF leads to increased vascular permeability and oedema.
  • As dehydration occurs at the external surface of the clot, a scab covering the wound is formed.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
186
Q

What happens within 24 hours of healing with first intention?

A
  • Neutrophils are seen at the incision margin, migrating towards the fibrin clot. They release proteolytic enzymes that begin to clear the debris.
  • Basal cells at the cut edge of the epidermis begin to show increased mitotic activity.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
187
Q

What happens between 24-48 hours of healing with first intention?

A
  • Epithelial cells from both edges have begun to migrate and proliferate along the dermis, depositing basement membrane components as they progress.
  • The cells meet in the midline beneath the surface scab, yielding a thin but continuous epithelial layer that closes the wound
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
188
Q

What happens by day 3 of healing with first intention?

A
  • Neutrophils have been largely replaced by macrophages, and granulation tissue progressively invades the incision space.
  • As you know, macrophages clear extracellular debris, fibrin and other foreign material, promoting angiogenesis and ECM deposition.
  • Collagen fibres are now evident at the incision margins.
  • Epithelial cells continue to proliferate, forming a covering approaching the thickness of the epidermis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
189
Q

What happens by day 5 of healing with first intention?

A
  • Neovascularisation reaches its peak as granulation tissue fills the incisional space. These new vessels are leaky, allowing the passage of plasma proteins and fluid into the extravascular space. Thus, new granulation tissue is often oedematous.
  • Migration of fibroblasts to the site on injury is driven by chemokines, TNF, PDGF, TGF-beta and FGF. Their subsequent proliferation is triggered by multiple growths factors, including PDGF, EGF, TGF-beta and FGF, and the cytokines IL-1 and TNF.
  • The fibroblasts produce ECM proteins, and collagen fibrils become more abundant and begin to bridge the incision.
  • The epidermis recovers it’s normal thickness as differentiation of surface cells yields a mature epiderminal architechture with surface keratinisation.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
190
Q

What happens during the second week of healing with first intention?

A
  • There is continued collagen accumulation and fibroblast proliferation.
  • The leukocyte infiltrate, oedema, and increased vascularity are substantially diminished.
  • The process of ‘blanching’ begings, accomplished by increasing collagen deposition within the incisional scar and the regression of the vascular channels
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
191
Q

What happens by the end of the first month of healing with first intention?

A
  • The scar comprises a cellular connective tissue largely devoid from inflammatory cells and covered by an essentially normally epidermis
  • However, the dermal appendages destroyed in the line of incision are permanently destroyed.
  • The tensile strength of the wound increases with time
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
192
Q

How does secondary intention differ from primary intention?

A
  • The fibrin clot is larger, there is more exudate and necrotic debris, therefore the inflammatory response is larger and may cause inflammation-mediated injury
  • Much larger amounts of granulation tissue, which generally results in a greater mass of scar tissue
  • At first, a provisional matrix containing fibrin, plasma fibronectin, and type 3 collagen is formed, but in about 2 weeks this is replaced by a matrix composed primarily by type 1 collagen
  • Wound contraction generally plays a larger role in secondary intention by myofibrils. Within 6 weeks, large skin defects may be reduced by 5-10% of their original size
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
193
Q

What are the phases of cell cycle?

A
  • G1 - presynthetic growth
  • S - DNA synthesis
  • G2 - premitotic growth
  • M - mitotic
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
194
Q

What are quiescent cells? What phase of the cycle are they in?

A

Cells that are not actively cycling. They are said to be in the G0 phase.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
195
Q

Where can cells enter G1 from?

A

Either from G0 or after completing a round of mitosis, as for continuously replicating cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
196
Q

What regulates the cell cycle?

A

It is regulated by activators and inhibitors.

  • Cell cycle progression is driven by proteins called cyclins and cyclin-associated enzymes called cyclic-dependant kinases (CDKs)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
197
Q

How are cyclins and cyclic-dependant kinases linked?

A
  • CDKs acquire the ability to phosphorylate protein substrates by forming complexes with the relevant cyclins.
  • Transiently increased synthesis of a particular cyclin leads to increased kinase activity of the appropriate CDK binding partner.
  • As the CDK completes its round of phosphorylation, the associated cyclin is degraded and the CDK activity abades.
  • Thus, as cyclin levels rise and fall, the activity of associates CDKs likewise wax and wane.
  • The different CDKs happen sequentially, like a relay race
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
198
Q

Where are the cell cycle checkpoints and what do they check for?

A
  • The G1-S checkpoint monitors the integrity of the DNA before irreversibly commiting cellular resources to DNA replication.
  • The G2-M restriction point ensures that there has been accurate gene replication before the cell actually divides.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
199
Q

What happens if the checkpoints in the cell cycle detect an error in DNA replication?

A
  • When cells do have DNA irregularities, checkpoint activation delays cell cycle progression and triggers DNA repair mechanisms
  • If the genetic derangement is too severe to be repaired, the cells will undergo apoptosis.
  • Alternatively, they may enter a non-replicative state called senescence - primarily throught the p53-dependant mechanisms
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
200
Q

What enzyme ensures the checkpoints in the cell cycle do their job? How does it do that?

A

It is the job of the CDK inhibitors: they accomplish this by modulating the CDK-cyclin complex activity.

Some examples are p21, p27, and p57

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
201
Q

What is the Warburg effect? Why is it important?

A
  • It is the effect to ensure that there are enough resources to perform DNA replication
  • It involves increased cellular uptake of glucose and glutamine, increased glycolysis, and (counter-intuitively) decreased oxidate phosphorylation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
202
Q

What are stem cells?

A

During development, stem cells give rise to all the various differentiated tissues.
In the adult organism, stem cells replace damaged cells and maintain tissue populations as individual cells within them undergo replicative senescence due to attirition of telemeres.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
203
Q

What are two important characteristics of stem cells?

A
  • Self-renewal, which permits stem cells to maintain their numbers
  • Asymmetric division, in which one daughter cell enters a differentiation pathway and gives rise to mature cells, while the other remains undifferentiated and retains its self-renewal capacity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
204
Q

What are the two varieties of stem cells?

A
  • Embryonic stem cells are the most undifferentiated. They are present in the inner cell mass of the blastocyst, have virtually limitless cell renewal capacity, and can give rise to every cell in the body. They are totipotent.
  • Tissue stem cells are found in intimate association with differential cells a given tissue. They are normally protected within specialised tissue microenvironments called stem cell niches. They have a limited repertoire of differentiated cells they can generate.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
205
Q

Where are mesenchymal stem cells found and what can they differentiate to?

A

Mesenchymal stem cells are found in bone marrow.
They can differentiate into a variety of stromal cells including chondrocytes (cartilage), osteocytes, adipocytes, and myocytes.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
206
Q

What is a bone fracture?

A

A fracture is defined as the loss of bone integrity due to mechanical injury and/or diminished bone strength

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
207
Q

How can you describe fractures?

A
  • Simple - the overlying skin is intact
  • Compound - the bone communicates with the skin surface
  • Comminuted - the bone is fragmented
  • Displaced - the ends of the bone at the fracture site are not aligned
  • Stress - a slowly developing fracture that follows a period of increased physical activity in which the bone is subjected to repetitive loads
  • “Greenstick” - extending only partially through the bone, common in infants, where the bone is soft
  • Pathological - involving bone weakened by an underlying disease process, such as tumour
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
208
Q

What happens in the bone immediately after a fracture?

A
  • Rupture of the blood vessels results in a haematoma, which fills the fracture gap and surround the area of bone injury.
  • The clotted blood provides a fibrin mesh, sealing off the fracture site and at the same time creates a framework for the influx of inflammatory cells and ingrowth of fibroblasts and new capillaries.
  • Simultaneously, degranulated platelets and migrating inflammatory cells release PDGF, TGF-beta, FGF and other factors, which activate osteoprogenitor cells in the periosteum, medullary cavity, and surrounding soft tissues and stimulate osteoclastic and osteoblastic activity.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
209
Q

What happens in the bone by the end of the first week after a fracture?

A
  • Organisation of the haematoma, matrix production in adjacent tissues, and remodeling of the fractured ends of the bone
  • This fusiform and predominantly uncalcified tissue - called soft tissue callus - provides some anchorage between the ends of the fractured bones but not structural rigidity for weight bearing
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
210
Q

What happens in the bone by two weeks after a fracture?

A
  • The soft tissue callus is transformed into a bony callus.
  • The activated osteoprogenitor cells deposit subperiosteal trabeculae of woven bone that are orientated perpendicular to the cortical axis and within the medullary cortex.
  • In some cases, the activated mesenchymal cells also form chondrocytes that make fibrocartilage and hyaline cartilage.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
211
Q

When does the bony cartilage after a bone fracture reach it’s maximal girth?

A

At the end of the second or third week. It helps stabilise the fracture site.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
212
Q

What happens to the newly formed cartilage after a bone fracture?

A

The newly formed cartilage along the fracture line undergoes endochondral ossification, forming a contigunous network of bone with newly deposited bone trabeculae in the medulla and beneath the periosteum.

In this fashion, the fracture ends are bridged, and as it mineralises, the stiffness and strength of the callus increases to the point that controlled weight bearing is tolerated.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
213
Q

How does the bony callus disappear in healed bone?

A

As the callus matures and is subject to weight-bearing forces, the callus portions that are not physically stressed are resorbed.
In this manner, the callus is reduced in size and the shape and the outline of the fractured bone are re-established as lamellar bone

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
214
Q

The sequence of events in the healing of a fracture can be easily impeded or blocked. Give some examples of what by…

A
  • Inadequate immobilisation - causes nonunion
  • Infection
  • Malnutrition and skeletal dysplasia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
215
Q

What is pathological calcification?

A

The abnormal tissue deposition of calcium salts, together with smaller amounts of iron, magnesium and other mineral salts

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
216
Q

What are the two types of calcification?

A
  • Dystrophic calcification occurs despite normal serum levels of calcium and in the absence of derangements of calcium metabolism
  • Metastatic calcification - the deposition of calcium in otherwise normal tissues, almost always results from hypercalcaemia secondary to some disturbance in calcium metabolism
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
217
Q

In what conditions would dystrophic calcification take place?

A

It is encountered in areas of necrosis, whether they are of coagulative, caseous or liquefactive type, and in foci of enzymatic necrosis of fat.
Commonly happens in atherosclerosis and heart valves

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
218
Q

How does dystrophic calcification present macroscopically and histologically?

A
  • Macroscopically, calcium salts appear as fine, white granules or clumps, often felt as gritty deposits
  • Histologically, they have a basophilic, amorphous, granular, sometimes clumped appearance. You can see psammoma bodies, which are mineral deposits around necrotic cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
219
Q
A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
220
Q

Are dystrophic and metastatic calcifications causes of organ dysfunction?

A
  • Dystrophic calcification is often a cause of organ dysfunction, such as in calcific valvular disease
  • Metastatic calcification if not often a cause of organ dysfunction, but on occasion massive involvement of the lungs produces some respiratory compromise
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
221
Q

What are some common causes of transudates?

A

Heart failure
Liver failure
Kidney failure
Severe nutritional deficiency

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
222
Q

What are some causes of increased hydrostatic pressure?

A

Increases in hydrostatic pressure are mainly caused by disorders that impair venous return. For example:
Congestive heart failure
Constrictive pericarditis
Ascites
Venous obstruction or compression (thrombosis, external pressures e.g. mass, lower extremity inactivity)
It is also sometimes caused by arteriolar dilation. For example in heat or neurohumoral dysregulation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
223
Q

What causes reduced plasma osmotic pressure?

A

Under normal circumstances albumin accounts for almost half of the total plasma protein; it follows that conditions leading to inadequate synthesis or increased loss of albumin from the circulation commonly cause reduced plasma oncotic pressure.
* Reduced synthesis occurs mainly in severe liver disease and protein malnutrition
* Increased loss occurs in nephrotic syndrome

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
224
Q

How does sodium and water retention occur in oedema?

A

Increased salt retention (with obligate retention of water) causes both increased hydrostatic pressure and diminshes osmotic pressure (due to dilution).
Salt retention occurs whenever renal function is compromised, such as in primary kidney diseases and in cardiovascular diseases that cause hypoperfusion, resulting in the activation of the renin-angiotension-aldosterone axis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
225
Q

How does lymphatic obstruction cause lymph-oedema?

A

Trauma, fibrosis, invasive tumours, and infection agents can all disrupt lymphatic vessels and impair the clearance of interstitial fluid, resulting in lymphoedema in the affected part of the body.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
226
Q

What are the different places you can get oedema?

A

Subcutaneous oedema
Pulmonary oedema
Pulmonary effusions
Peritoneal effusions (ascites)
Brain oedema

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
227
Q

How is oedema recognised microscopically?

A

It is appreciated as clearing and separation of the extracellular matrix and subtle cell swelling

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
228
Q

What is hyperemia vs congestion?

A

Hyperemia and congestion both stem from increased blood volumes within tissues.
Hyperemia is an active process in which arteriolar dilation (inflammation or exercising muscles) leads to increased blood flow. Affected tissues turn red (erythema) because of increased delivery of oxygenated blood
Congestion is a passive process resulting from reduced outflow of blood from a tissue. It can be systemic (heart failure) or localised (isolated venous obstruction)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
229
Q

What does chronic congestion lead to and why?

A

As a result of increased hydrostatic pressures, congestion commonly leads to edema.
The associated chronic hypoxia may lead to ischaemic tissue injury and scarring.
Capillary rupture can also produce small haemorrhagic foci; subsequent catabolism of extravasated red cells can leave clusters of haemosiderin-laden macrophages

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
230
Q

Microscopically, what will you see in acute pulmonary congestion vs chronic pulmonary congestion?

A
  • Acute pulmonary congestion exhibits engorged alveolar capillaries, alveolar septal emboli, and focal intraalveolar haemorrhage
  • In chronic pulmonary congestion (often caused by congested heart failure) the septa are thickened and fibrotic, and the alveoli often contain numerous haemosiderin-laden macrophages called heart failure cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
231
Q

Microscopically, what will you see in acute hepatic congestion vs chronic hepatic congestion?

A
  • In acute hepatic congestion, the central vein and sinusoids are distended. Because the centrilobular area is distal end of the hepatic blood supply, centrilobular hepatocytes may undergo ischaemic necrosis with the periportal hepatocytes may only develop fatty change (better oxygenated)
  • In chronic passive hepatic congestion, the centrilobular regions are grossly red-brown and slightly depressed and are accentuated against the surrounding zones of uncongested tan liver (nutmeg liver).
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
232
Q

What is haemostasis?

A

A precisely organised process involving platelets, clotting factors, and endothelium that occurs at the site of the vascular injury and culminates in the formation of a blood clot, which serves to prevent or limit the extent of bleeding

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
233
Q

What two groups are abnormal haemostasis divided into?

A
  • Haemorrhagic disorders - characterised by excessive bleeding, haemostatic mechanisms are either blunted or insufficient to prevent abnormal blood loss.
  • Thrombotic disorders - blood clots (thrombi) form within intact blood vessels or within the chambers of the heart.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
234
Q

Are thrombotic and haemorrhagic disorders always separate entities?

A

No, sometimes in generalised activation of clotting paradoxically produces bleeding due to the consumption of coagulation factors, as in DIC.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
235
Q

What are the four steps in haemostasis?

A

1) Arteriolar vasoconstriction
2) Primary haemostasis - the formation of the platelet plug
3) Secondary haemostasis - deposition of the fibrin
4) Clot stabilisation and resorption

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
236
Q

What happens in arteriolar vasoconstriction during haemostasis?

A
  • It occurs immediately and markedly reduced blood flow to the injured area.
  • It is mediated by reflex neurogenic mechanisms and augmented by the local secretion of factors such as endothelin, a potent endothelium-derived vasoconstrictor.
  • This effect is transient and bleeding would continue if it wasn’t for the next steps
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
237
Q

What happens in primary haemostasis - formation of the platelet plug?

A
  • Disruption of the endothelium exposes subendothelium vWF and collagen, which promote platelet adherence and activation.
  • Activation of platelets results in a dramatic shape change (from small rounded discs to flat plates with spiky protrusions that markedly increase surface area), as well as the release of secretory granules (ADP, TxA2)
  • Within minutes, the secreted products recruit additional platelets, which undergo aggregation to form a primary haemostatic plug
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
238
Q

What happens in secondary haemostasis - deposition of fibrin?

A
  • Tissue factor is exposed at the site of injury. Tissue factor is a membrane-bound procoagulant glycoprotein that is normally expressed by subendothelial cells in the vessel wall, such as smooth muscle cells or fibroblasts.
  • Tissue factor binds and activates factor VII, setting in motion a cascade of reactions that generates thrombin.
  • Thrombin cleaves circulating fibrinogen into insoluble fibrin, creating a fibrin mesh-work, and is also a potent activator of platelets, leading to additional platelet aggregation at the site of injury.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
239
Q

What happens during clot stabilisation and resorption during haemostasis?

A
  • Polymerised fibrin and platelet aggregates undergo contraction to form a solid, permanent plug that prevents further haemorrhage.
  • At this stage, counteregulatory mechanisms (e.g. tissue plasminogen factor t-PA) are set into motion that limit clotting to the side of injury and eventually lead to clot resorption and tissue repair.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
240
Q

What are platelets?

A

Disc-shaped anucleate cell fragments that are shed from megakaryocytes in the bone marrow into the bloodstream.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
241
Q

What do platelets need to function?

A

Their function depends on several glycoprotein receptors, a contractile cytoskeleton, and two types of cytoplasmic granules.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
242
Q

What cytoplasmic granules do platelets rely on to function?

A
  • α-granules have the adhesion molecule P-selectin on their membranes and contain proteins involved in coagulation, such as fibrinogen, coagulation factor V, and vWF, as well as protein factors that may be involved in wound healing, such as fibronectin, platelet factor 4 (a heperin-binding chemokine), platelet-derived growth factor (PDGF), and transforming growth factor-β.
  • Dense (or δ) granules contain ADP and ATP, ionised calcium, serotonin, and adrenaline
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
243
Q

After a traumatic vascular injury, platelets encounter consistuents of the subendothelial connective tissue, such as vWF and collagen. On contact with these proteins, platelets undergo a sequence of reactions the culminate the formation of a platelet plug. What are they?

A
  1. Platelet adhesion
  2. Platelets rapidly change shape
  3. Secretion of granules content
  4. Platelet aggregation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
244
Q

How does platelet adhesion take place in plug formation?

A
  • Platelet adhesion is mediated largely by interactions with vWF, which acts as a bridge between the platelet surface receptor glycoprotein 1b (Gp1B) and exposed collagen.
  • Notably, the genetic deficiencies of vWF or Gp1B (Bernard-Soulier syndrome) result in bleeding disorders.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
245
Q

What happens when the platelets change shape in plug formation?

A
  • The platelets change shape following adhesion, being converted from smooth discs to ‘sea urchins’ with greatly increased surface area.
  • This change is accompanied by alterations in glycoprotein IIb/IIIa that increase it’s affinity for fibrinogen, and by the translocation of the negatively charged phospholipids (particularly phosphatidylserine) to the platelet surface.
  • These phospholipids bind calcium and serve as nucleation sites for the assembly of coagulation factor complexes.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
246
Q

What is platelet activation in the plug formation? What triggers it?

A

Secretion of the granule contents occurs along with the changes in shape; these two events together are known as activation.

  • Platelet activation is triggered by a number of factors, including the coagulation factor thrombin and ADP.
  • Thrombin activates platelets through a special type of G-protein coupled receptor, protease-activated receptor (PAR), which is switched on by a proteolytic cleavage by thrombin.
  • ADP is a component of dense body granules; thus, platelet activation and ADP release begets additional rounds of platelet activation, a phenomenon known as recruitment.
  • Activated platelets also produce the prostaglandin thromboxane A2 (TxA2), a potent inducer of platelet aggregation.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
247
Q

What happens during platelet aggregation?

A
  • Platelet aggregation follows their activation.
  • The conformational change in glycoprotein 2b/3a that occurs with platelet activation allows a binding of fibrinogen, a large bivalent plasma polypeptide that forms bridges between adjacent platelets, leading to their aggregation
  • The inital wave of aggregation is reversible, but concurrent activation of thrombin stabilises the platelet plug by causing further platelet activation and aggregation, and by promoting irreversible platelet contraction
  • Platelet contraction is dependent of the cytoskeleton and consolidates the aggregated platelets.
  • In parallel, thrombin also converts fibrinogen into insoluble fibrin, cementing platelets in place and creating the definitely secondary hemostatic plug.
  • Entrapped red cells and leukocytes are also found in haemostatic plugs, in part due to adherence of leukocytes to P-selectin expressed on activated platelets
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
248
Q

What is the coagulation cascade?

A

It is a series of amplifying enzymatic reactions that leads to the deposition of an insoluble fibrin clot.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
249
Q

What does each step in the coagulation cascade have in common? Where do they happen?

A

Each reaction step involves an enzyme (an activated coagulation factor), a substrate (an inactive proenzyme form of a coagulaton factor), and a cofactor (a reaction accelerator).
These components are assembled on a negatively charged phospholipid surface, provided by activated platelets.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
250
Q

Which ion and vitamin are important in the coagulation cascade?

A
  • Calcium ions - assembly of reaction complexes depend on calcium, which binds to γ-carboxylated glutamic acid residues that are present in factors
  • The enzymatic reactions that produce γ-carboxylated glutamic acid use Vitamin K as a cofounder and are antagonised by many drugs.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
251
Q

What are the two pathways in the coagulation cascade? What do we use to assess them?

A

Extrinsic and Intrinsic Pathways
* The prothrombin time (PT) assay assess the function of the proteins in the extrinsic pathway (factors VII, X, V, II and fibrinogen). In brief, tissue factor, phospholipids, and calcium are added to plasma and the time for a fibrin clot to form is recorded
* The partial thromboplastin time (PTT) assay screens the function of the proteins in the intrinsic pathway (factors XII, XI, IX, VIII, X, V, II, and fibrinogen). In this assay, clotting of plasma is initiated by addition of negative-charged particles (e.g. ground glass) that activate factor XII (Hageman factor) together with the phospholipids and calcium, and the time to fibrin clot formation is recorded

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
252
Q

What do the deficiencys in the different clottin factors lead to? What does this show?

A
  • Deficiencies in factors V, VII, VIII, IX and X are associated with moderate to severe bleeding disorders
  • Prothrombin deficiency is incompatible with life
  • Factor XI deficiency is only associated with mild bleeding
  • Individuals with factor XII deficiency do not bleed and are in fact susceptible to thrombosis

This shows that the process in vivo and in the lab are different!

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
253
Q

Which is the most important coagulation factor?

A

Thrombin, because its various enzymatic activities control divers aspects of haemostasis and link clotting to inflammation and repair

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
254
Q

What are thrombin’s most important roles?

A
  • Conversion of fibrinogen into crosslinked fibres - Thrombin directly converts soluble fibrinogen into fibrin monomers that polymerise into an insoluble clot, and also amplifies the coagulation process, not only by activating factor XI, but also by activating factors V and VIII.
  • Stabilises the secondary haemostatic plug by activating factor XIII, which covalently cross-links fibrin.
  • Platelet activation - thrombin in a potent inducer of platelet activation, and aggregation through its ability to activate PARs, therefore linking platelet function to coagulation
  • Pro-inflammatory effects - PARs are also expressed on inflammatory cells, endothelium, and other cell types, and activation of these receptors by thrombin is believed to mediate proinflammatory effects that contribute to tissue repair and angiogenesis
  • Anticoagulant effects - upon encountering normal endothelium, thrombin changes from a procoagulant to an anticoagulant. This reversal in function prevents clotting from extending beyond the site of vascular injury
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
255
Q

Once initiated, coagulation must be restricted to the site of vascular injury. What are the factors that limit coagulation?

A
  • Simple dilution - blood flowing past the site of injury washes out activated coagulation factors, which are rapidly removed by the liver
  • The requirement for negatively charged phospholipids - which are mainly provided by platelets that have been activated by contact with the subendothelial matrix at sites of vascular injury.
  • Factors that are expressed by intact endothelium adjacent to the injury
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
256
Q

Activation of the coagulation cascade also sets into motion the fibrinolytic cascade that limits the size of the clot and contributes to it’s later dissolution. Tell me about this process…

A
  • Fibrinolysis is largely accomplished through the enzymatic activity of plasmin, which breaks down fibrin and interferes with its polymerisation.
  • There is an elevated level of breakdown products of fibrinogen (often called fibrin split products) e.g. d-dimer.
  • Plasmin is generated by enzymatic catabolism of the inactive circulating precursor plasminogen, either by a factor XII-derived pathway or by plasminogen activators e.g. tPA.
  • Once activated, plasmin is tightly controlled by counterregulatory factors such as α2-plasmin inhibitor, a plasma protein that binds and rapidly inhibits plasmin.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
257
Q

Normal endothelial cells express a multitude of factors that inhibit the procoagulant activities of platelets and coagulation factors and that augment fibrinolysis. What are the different antithrombotic properties of endothelium?

A
  • Platelet inhibitory effects - Intact epithelium acts as a barrier that shields platelets from subendothelial vWF and collagen. It also releases NO, prostacyclin (PGI2), and ADP which inhibit platelet activation and aggregation. They also bind and alter the activity of thrombin, which is a potent activators of platelets.
  • Anticoagulant effects - Intact epithelium shields coagulation factors from tissue factor. It also expresses multiple factors that actively oppose coagulation, such as thrombomodulin, endothelial protein C receptor, heparin-like molecules, and tissue factor pathway inhibitor.
  • Fibrinolytic effects - normal endothelial cells synthesise t-PA, a key component of the fibrinolytic pathway
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
258
Q

How do thrombomodulin and endothelial protein C receptor have anticoagulant effects?

A

They bind thrombin and protein C, respectively, in a complex on the endothelial cell surface. When bound in this complex, thrombin loses it’s ability to activate coagulation factors and platelets, and instead cleaves and activated protein C, a vitamin K-dependent protease that requires a cofactor, protein S. Activated protein C/protein S complex is a potent inhibitor of coagulation factors Va and VIIIa

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
259
Q

How do Heparin-like molecules have anticoagulant properties?

A

Heparin-like molecules on the surface of endothelium bind and activate anti-thrombin III, which then inhibits thrombin and factors IXa, Xa, XIa, and XIIa.
The clinical utility of heparin and related drugs is based on their ability to stimulate antithrombin III activity.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
260
Q

How does tissue factor pathway inhibitor have anticoagulant properties?

A

TFPI, like Protein C, requires Protein S as a cofactor, and binds and inhibits tissue factor/factor VIIa complexes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
261
Q

What are the very broad causes of haemorrhagic disorders?

A
  • Primary or secondary defects in vessel walls e.g. aortic dissection or arterial ruptures
  • Platelet disorders
  • Coagulation factor disorders e.g. haemophilia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
262
Q

How do defects of primary haemostasis present? Give an example

A
  • Platelet defects or von Willebrand disease
  • Often present with small bleeds in skin or mucosal membranes, which take the form of petechiae or purpura
  • The capillaries of the mucosa and skin are more prone to rupture following minor trauma and under normal circumstances platelets seal these defects virtually immediately.
  • Mucosal bleeding associated with disorders of primary haemostasis may present with epistaxis, GI bleeding, or menorrhagia. A fatal complication is intracerebral haemorrhage
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
263
Q

What are petechiae, purpura and ecchymoses?

A
  • Petechiae - minute 1-2mm haemorrhages
  • Purpura - slightly larger (>3mm) haemorrhages
  • Ecchymoses - haemorrhages of 1-2 cm in size
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
264
Q

How do defects of secondary haemostasis present? Give an example

A
  • Coagulation factor defects e.g. haemophilia
  • Often present with bleeding into soft tissue (muscles) and joints
  • Haemarthrosis after minor trauma is particularly characteristic of haemophilia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
265
Q

What does the clinical significance of haemorrhage depend on?

A
  • Volume of the bleed
  • Rate at which it occurs
  • Location
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
266
Q

What percentage of blood loss may lead to haemorrhagic shock?

A

Over 20%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
267
Q

What are the primary abnormalities that lead to thrombosis? (Virchow’s triad)

A
  • Endothelial injury
  • Stasis or turbulent blood flow
  • Hypercoagulability of the blood
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
268
Q

Where in the body is endothelial injury the most common cause of thrombosis and why?

A

Endothlial injury leading to platelet activation almost inevitably underlies thrombus formation in the heart and the arterial circulation, where the high rates of blood flow impede clot formation.
These clots are typically rich in platelets, which is why aspirin is good.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
269
Q

What is endothelial activation or dysfunction?

A

Severe endothelial injury may trigger thrombosis by exposing vWF and tissue factor. However, inflammation and other noxious stimuli also promote thrombosis by shifting the pattern of gene expression in endothelium to one that is ‘prothrombotic’. This is called endothelial activation or dysfunction.
It can be caused by physical injury, infection, abnormal blood flow, inflammatory mediators, metabolic abnormalities and toxins

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
270
Q

What are the main endothelial changes that occur during endothelial activation?

A
  • Procoagulant changes - endothelial cells activated by cytokines downregulate the expression of thrombomodulin, which results is sustained action of thrombin, which stimulates platelets and augments inflammation through PARs expressed on platelets and inflammatory cells. Endothelial cells also downregulate the expression of other anticoagulants, such as protein C and tissue factor pathway inhibitor
  • Antifibrinolytic effects - activated endothelial cells secrete plasminogen activator inhibitors (PAIs), which limit fibrinolysis, and downregulate the expession of t-PA
The opposite of this diagram!!
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
271
Q

What is laminar flow?

A

Normal blood flow is laminar such that the platelets (and other blood cellular elements) flow centrally in the vessel lumen, separated from endothelium by a slower moving layer of plasma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
272
Q

How do stasis and turbulent flow cause endothelial injury and thrombosis?

A
  • Promote endothelial activation, enhancing procoagulant activity and leukocyte adhesion, in part through flow-induced changes in the expression of adhesion molecules and pro-inflammatory factors
  • Disrupt laminar flow and bring platelets into contact with the endothelium
  • Prevent washout and dilution of activated clotting factors by fresh flowing blood and the inflow of clotting factor inhibitors
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
273
Q

What are some of the causes of stasis and turbulent blood flow in vessels?

A
  • Ulcerated atherosclerotic plaques not only expose subendothelial vWF and tissue factor but also cause turbulence
  • Aortic and arterial dilatations (aneurysms) result in local stasis
  • Acute MIs are associated with areas of noncontractile myocardium and cardiac aneurysms, both are associated with stasis and flow abnormalities
  • Hyperviscosity such as in polycythaemia vera causes increased resistance to flow and stasis
  • Deformed red cells in sickle cell anaemia impede blood flow through small vessels, resulting in stasis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
274
Q

What is hypercoagulability? What groups can it be split into

A

Also called thrombophilia, hypercoagulability can be loosely defined as any disorder of the blood that predisposes to thrombosis.
It can be divided into primary (genetic) and secondary (acquired) disorders

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
275
Q

What are some primary causes of hypercoagulability?

A

Common
* Factor V mutation (Arg to Glu substitution in amino acid residue 506 leading to resistance to factor C; Factor V Leiden
* Prothrombin mutation (G20210A noncoding sequence variant leading to increased prothrombin levels)
* Increased levels of factors VIII, IX, XI or fibrinogen

Rare
* Antithrombin III deficiency
* Protein C deficiency
* Protein S deficiency

Very rare
* Fibrinolysis defects
* Homozygous homocystinuria

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
276
Q

How do the following secondary causes of hypercoagulability cause it?
* oral contraceptive
* disseminated cancers
* old age

A
  • Oral contraceptive - increased hepatic synthesis of coagulation factors and reduced anticoagulant synthesis
  • Disseminated cancers - release of various pro-coagulants from tumours
  • Older age - reduced endothelial PGI2 production
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
277
Q

What are some secondary causes of hypercoagulability?

A

High risk for thrombosis
* prolonged bed rest or immobilisation
* MI
* AF
* Tissue injury (fracture, burn, surgery)
* Cancer
* Prosthetic cardiac valves
* DIC
* Heparin-induced thrombocytopaenia
* Antiphospholipid syndrome

Lower risk for thrombosis
* Cardiomyopathy
* Nephrotic syndrome
* Hyperestrogenic states (pregnancy, oral contraceptive)
* Sickle cell anaemia
* Smoking

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
278
Q

What is Heparin-Induced Thrombocytopaenic Syndrome?

A
  • HIT occurs following the administration of unfractionated heparin, which may induce the appearance of antibodies that recognise complexes of heparin and platelet factor 4 on the surface of platelets, as well as complexes of heparin-like molecules and platelet factor 4-like molecules on endothelial cells.
  • Binding of these antibodies to platelets results in their activation, aggregation and consumption. This produces a pro-thrombotic state.
  • It happens less in LMWH
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
279
Q

How may antiphospholipid antibody syndrome present?

A
  • Recurrent thromboses
  • repeated miscarriages
  • cardiac valve vegetations
  • thrombocytopaenia
  • pulmonary hypertension from recurrent subclinical PEs
  • stroke
  • bowel infarction
  • renovascular hypertension
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
280
Q

What are the two types of antiphospholipid antibody syndrome?

A
  • Secondary antiphospholipid antibody syndrome is when it occurs with individual with a well-defined autoimmune disease, such as SLE
  • In primary antiphospholipid antibody syndrome, patients exhibit only the manifestations of a hypercoagulable state and lack evidence of other autoimmune disorders; occasionally it comes following drugs or an infection
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
281
Q

Which way to arterial and venous thrombi occur and which ways do they grow?

A
  • Arterial or cardiac thrombi usually begin at sites of turbulence and tend to grow retrograde
  • Venous thrombi often begin at sites of stasis and extend in the direction of blood flow
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
282
Q

What is a line of Zahn? Where does it happen and what does it mean?

A
  • Thrombi often have grossly and microscopically apparent laminations called lines of Zahn, which are pale platelet and fibrin deposits alternating with darker red cell-rich layers
  • They signify that a thrombus formed in flowing blood; they can therefore distinguish post-mortem to ante-mortem clots
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
283
Q

What are mural thrombi?

A

Thrombi that form in heart chambers or in the aortic lumen.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
284
Q

What is different in the content or arterial vs venous clots?

A

Arterial thrombi often consist of a friable meshwork of platelets, fibrin, red cells, and degenerating leukocytes
Venous thrombi contain more enmeshed red cells and relatively few platelets - they are therefore known as red or stasis thrombi

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
285
Q

If a patient survive the initial thrombus, in the ensuing days to weeks, thrombi can have one of four processes happen. What are they?

A
  • Propagation - thrombi accumulate additional platelets and fibrin
  • Embolisation - thrombi dislodge and travel to other sites in the vasculature
  • Dissolution - the result of fibrinolysis, which can lead to shrinkage and total disappearance of the recent thrombi - only happens in the first few hours
  • Organisation and recanalisation - older thrombi become organised by the ingrowth of endothelial cells, smooth muscle cells and fibroblasts. Capillary channels eventually form that reestablish the continuity of the original lumen, albiet to a variable degree. Eventually, with remodeling and contraction of the mesenchymal elements, only a fibrous lump may remain
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
286
Q

What veins do superficial and deep venous thrombosis affect?

A

Superficial - in the saphenous veins - rarely embolise but same symptoms as DVT
Deep - in the popliteal, femoral or iliac veins - often embolise

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
287
Q

What happens in DIC?

A

Widespread formation of thrombi in the microcirculation. These microvascular thrombi can cause diffuse circulatory insufficiency and organ dysfunction, particularly of the brain, lungs, heart and kidneys.
The runaway thrombosis ‘uses up’ platelets and coagulation factors and often activates fibrinolytic mechanisms . Thus, symptoms initially related to thrombosis can evolve into a bleeding catastrophe, such as haemorrhagic stroke or hypovolaemic shock.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
288
Q

What is an embolism?

A

An embolus is a detached intravascular solid, liquid, or gaseous mass that is carried by the blood from it’s point of origin to a distant site, where it often causes tissue dysfunction or infarction.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
289
Q

What is the most common thromboembolic disease?

A

Pulmonary emboli from the DVT

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
290
Q

Where do pulmonary emboli travel?

A

Fragmented thrombi from DVTs are carried through progressively larger veins and the right side of the heart before slamming into the pulmonary arterial vasculature.
Depending on the size of the embolus, it can occlude the main pulmonary artery, straddle the pulmonary artery bifurcation (staddle embolus), or pass out into the smaller branching arteries.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
291
Q

Once you have one PE, what happens to your risk?

A

It increases significantly.
Frequently there are multiple emboli, occuring either sequentially or simultaneously as a shower of small emboli from a single large mass

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
292
Q

What is a paradoxical embolism?

A

Rarely, a venous embolus passes through an interatrial or interventricular defect and gains access to the systemic arterial circulation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
293
Q

At what percentage occlusion does a PE cause sudden death or Cor Pulmonale?

A

When emboli obstruct 60% or more of the pulmonary circulation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
294
Q

Why does embolic obstruction of medium-sized pulmonary arteries with subsequent vascular rupture not often cause an infarct? Why is this different in small end-arteriolar pulmonary branches

A

Because the lung is supplied by both the pulmonary arteries and the bronchial arteries, and an intact bronchial circulation is usually sufficient to perfuse the attached area.
This is not the case in small end-arteriolar pulmonary branches, so they often produce haemorrhage or infarction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
295
Q

Where do most systemic thromboembolisms come from?

A

80% arise from intracardiac mural thrombi, two thirds of which are associated with left ventricular wall infarcts and one quarter with left atrial dilatation and fibrillation.

The remainder originates from aortic aneurysms, atherosclerotic plaques, valvular vegetations, or venous thrombi

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
296
Q
A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
297
Q

Where do systemic thromboembolisms often end up?

A

75% in the lower extremities
10% in the brain
May also involve the intestines, kidneys, spleen, and upper extremities

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
298
Q

How do fat and marrow emboli occur?

A

Microscopic fat globules - sometimes associated with marrow- can by found in the pulmonary vasculature after fractures of long bones, soft tissue trauma and burns
Presumably these injuries rupture vascular sinusoids in the marrow or small venules, allowing marrow and adipose tissue to herniate into the vascular space and travel to the lung.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
299
Q

Do fat embolisms often give symptoms?

A

Fat embolism occurs in 90% of invidiuals with severe skeletal injuries, but less than 10% have any clinical findings.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
300
Q

What is fat embolism syndrome?

A

The term applied to the minority of patients who become symptomatic.
Characterised by pulmonary insufficiency, neurologic symptoms, anaemia and thrombocytopaenia, and is fatal in about 5 to 15% of cases

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
301
Q

What are the symptoms of fat embolism syndrome?

A

Typically, 1 to 3 days after injury, there is a sudden onset of tachypnoea, dyspnoea, and tachycardia; irritability and restlessness can progress into delirium or come

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
302
Q

Why does thrombocytopaenia occur in fat embolism syndrome?

A

It is attributed to platelet adhesion to fat globules and subsequent aggregation or splenic sequestration; anaemia can result from similar red cell aggregation and/or haemolysis.
A diffuse petechial rash is related to rapid onset of thrombocytopaenia and can be a useful diagnostic feature.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
303
Q

What is the pathogenesis of fat embolism syndrome?

A
  • Fat microemboli and associated red cell and platelet aggregates can occlude the pulmonary and cerebral microvasculature.
  • Release of fatty acids from the fat globules exacerbated the situation by causing local toxic injury to endothelium, and platelet activation and granulocyte recruitment (with free radical, protease, and eicosanoid release) complete the vascular assault.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
304
Q

What is an air embolism?

A

Gas bubbles within the circulation can coalesce to form frothy masses that obstruct vascular flow and cause distal ischaemic injury.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
305
Q

What volume of air is required to have an effect on the pulmonary circulation?

A

Generally more than 100cc

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
306
Q

What is decompression sickness? Why does it happen?

A

It occurs when an individual experiences sudden decrease in atmospheric pressure.
When air is breathed at high pressure, increased amounts of gas (particularly nitrogen) are dissolved in the blood and tissues. If the diver then ascends too rapidly, the nitrogen comes out of the solution in the tissues and blood

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
307
Q

What happens if you get an air embolus in your lungs?

A

Gas bubbles in the vasculature cause oedema, haemorrhage and focal atelectasis or emphysema, leading to a form of respiratory distress called the chokes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
308
Q

What happens in caisson disease? Who does it happen to?

A

Persistence of gas emboli in the skeletal system causes multiple foci of ischaemic necrosis; common in the femoral head, tibia and humeri
Happens in chronic decompression sickness, like workers in pressurised vehicles

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
309
Q

What is an amniotic fluid embolis? What is it’s mortality?

A

It is the infusion of amniotic fluid or fetal tissue into the maternal circulation via a tear in the placental membranes or rupture of uterine veins.
It accounts for roughly 10% of maternal deaths in the US, the mortality rate is up to 80% and 85% of survivors are left with permanent neurological damage

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
310
Q

What are the symptoms of an amniotic fluid embolus?

A

The onset is characterised by sudden severe dyspnoea, cyanosis and shock, followed by neurological impairment ranging from headache to seizures.
If the patient survives the initial crisis, pulmonary oedema typically develops, frequently accompanied by DIC.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
311
Q

How does an amniotic fluid embolus vary from a PE?

A

Much of the morbidity and mortality of an amniotic fluid embolism may stem from the biochemical activation of coagulation factors and components of the innate immune system by substances in the amniotic fluid, rather than the mechanical obstruction of a PE

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
312
Q

What is an infarct?

A

An infarct is an area of ischaemic necrosis caused by occlusion or either the arterial supply or venous drainage.
Arterial thrombosis or arterial embolism underlies the vast majority of infarctions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
313
Q

How can infarcts be classified? Tell me about both groups

A
  • Red infarcts occur
    1) with venous occlusions (e.g. testicular torsion)
    2) in loose, spongy tissues (e.g. lung), where blood can collect in the infarcted zone
    3) in tissues with dual circulations (e.g. lung and intestine) that allow to blood to flow from an unobstructed parallel supply in a necrotic zone
    4) in tissues previously congested by sluggish venous outflow
    5) when flow is reestablished to a site of previous arterial occlusion and necrosis
  • White infarcts occur with arterial occlusions in solid organs with end-arterial circulation (e.g. heart, spleen and kidney), and where tissue density limits the seepage of blood from adjoining capillary beds into the necrotic area.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
314
Q

What shape are infarcts?

A

Infarcts tend to be wedge-shaped, with the occluded vessel at the apex and the periphery of the organ forming the base.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
315
Q

How does the morphology differ over the first few days and based on the location of infarcts?

A
  • When the base is a serousal surface, there may be an overlying fibrinous exudate resulting from an acute inflammatory response to mediators release from injured and necrotic cells
  • Fresh infarcts are poorly defined and slightly haemorrhagic, but over a few days their margins become better defined by a narrow rim of congestion attributable to inflammation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
316
Q

Why are some infarcts red and some white?

A
  • Over time, infarcts resulting from arterial occlusion without a dual blood supply typically become progressively paler and more sharply defined - white infarcts
  • Extravasated red cells in haemorrhagic infarcts are phagocytosed by macrophages, which convert heme iron to haemosiderin, extensive haemorrhage can leave a firm, brown, haemosiderin-rich residuum.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
317
Q

What are septic infarctions?

A

They occur when infected cardiac valve vegetations embolise or when microbes seed necrotic tissue.
In these cases, the infarct is converted into an abscess, with a corresponding greater inflammatory response

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
318
Q

What are the factors that influence development of an infarct?

A
  • Anatomy of the vascular supply - is there an alternative blood supply?
  • Rate of occlusion - slowly developing occlusions are less likely to cause infarction, because they provide time for collateral pathways of perfusion to form.
  • Tissue vulnerability to hypoxia - neurons undergo irreversible damage within 3-4 minutes, myocardial cells last 20-30 minutes, fibroblasts within myocardium can last hours
  • Hypoxaemia - the lower the oxygen, the more likely the infarct
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
319
Q

What is shock?

A

Shock is a state in which diminished cardiac output or reduced effective circulating blood volume impairs tissue perfusion and leads to cellular hypoxia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
320
Q

What are the three main types of shock? Why do they happen? What are the other types of shock?

A

1) Cardiogenic shock results from low cardiac output due to myocardial pump failure. This can be due to intrinsic myocardial damage (infarction), ventricular arrythmias, extrinsic compression (tamponade), or outflow obstruction (PE)
2) Hypovolaemic shock results from low cardiac output due to low blood volume, such as can occur with massive haemorrhage or fluid loss from severe burns
3) Shock associated with systemic inflammation may be triggered by microbial infections, burns, trauma or pancreatitis. The common pathogenic feature is a massive outpouring of inflammatory mediators from innate and adaptive immune cells that produce vasodilation, vascular leaking and venous blood pooling. These result in tissue hypoperfusion, cellular hypoxia and metabolic derangements
4) Less commonly, shock can be neurogenic or anaphylactic, where acute vasodilation leads to hypotension and tissue hypoperfusion

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
321
Q

What are most common triggers for septic shock?

A

Gram-positive bacterial infections, followed by gram-negative bacterial infections and then funghi

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
322
Q

In broad terms, what are the factors believed to play a major role in the pathophysiology of septic shock?

A
  • Inflammatory and counter-inflammatory responses
  • Endothelial activation and injury
  • Induction of a procoagulant state
  • Metabolic abnormalities
  • Organ dysfunction
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
323
Q

What inflammatory responses have a play in the pathophysiology of septic shock?

A
  • Microbial cell wall consituents engage receptors on innate immune cells.
  • Likely initiators of inflammation are signalling pathways that lie downsteam of TLRs (which recognise PAMPs), and G-protein coupled receptors that detect bacterial pathogens.
  • Upon activation, innate immune cells produce TNF,IL-1, IFN-γ, IL-12 and IL-18
  • ROS, platelet activating factor and prostaglandins are also elaborated
  • These effector molecules induce endothelial cell activation to upregulate adhesion molecule expression and further stimulate chemokines and cytokines production.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
324
Q

How is the complement cascade involved in the pathophysiology of septic shock?

A
  • The complement cascade is also activated by microbial components, directly and indirectly by the proteolytic activity of plasmin
  • This results in the production of anaphylotoxins (C3a, C5a), chemotactic fragments (C5a) and opsonins (C3b), all of which contribute to the pro-inflammatory state
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
325
Q

How does the hyperinflammatory state initiated by sepsis also activate counter-regulatory immunosuppressive mechanisms? What does this mean?

A
  • Septic patients may oscillate between hyperinflammatory and immunosuppressed states during their clinical course.
  • Proposed mechanisms for the immune suppression include a shift from pro-inflammatory TH1 cells to anti-inflammatory TH2 cells, production of anti-inflammatory mediators (e.g. soluble TNF receptor, IL-1 receptor antagonist, and IL-10), lymphocyte apoptosis, the immunosuppressive act of apoptotic cells, and the induction of cellular energy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
326
Q

How does endothelial activation and injury contribute to the pathophysiology of septic shock?

A
  • It leads to widespread vascular leakage and tissue oedema, which have deleterious effects on both nutrient delivery and waste removal.
  • The oedema created impedes tissue perfusion
  • Activated endothelium also upregulates production of NO and other vasoactive inflammatory mediators (e.g. C3a, C5a, and PAF), which may contribute to vascular smooth muscle relaxation and systemic hypotension
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
327
Q

How does the induction of a procoagulant state contribute to the pathophysiology of septic shock?

A
  • Pro-inflammatory cytokines increase tissue factor production by monocytes and possibly endothelial cells as well, and decrease the production of endothelial anti-coagulant factors (e.g. TFPI, thrombomodulin and protein C)
  • They also dampen fibrinolysis by increasing plasminogen activator inhibitor 1 expression.
  • The vascular leak and vasodilation causes stasis.
  • All of these things together lead to systemic activation of thrombin and the deposition of fibrin-rich thrombi in small vessels throughout the body, often compromising tissue perfusion
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
328
Q

How do metabolic abnormalities contribute to the pathophysiology of septic shock?

A
  • Septic patients exhibit insulin resistance and hyperglycaemia.
  • Cytokines such as TNF and IL-1, and stress hormones (glucagon, GH, and glucocorticoids) and catecholamines all drive gluconeogenesis.
  • Pro-inflammatory cytokines suppress insulin release whilst promoting insulin resistance in the liver and other tissues, likely by impairing the expression of GLUT-4.
  • Hyperglycaemia reduced neutrophil function - and causes increased endothelial cell adhesion molecule expression
  • Cellular hypoxia and diminished oxidative phosphorylation leads to increased lactate production and lactic acidosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
329
Q

How does organ dysfuction contribute to the pathophysiology of septic shock?

A
  • Systemic hypotension, interstitial oedema and small vessel thrombosis all decrease the delivery of oxygen and nutrients to the tissue, which fail to properly utilise those nutrients that are delivered due to cellular hypoxia.
  • High levels of cytokines and secondary mediators diminish myocardial contractility and cardiac output, and increased vascular permeability and endothelial injury can lead to the acute respiratory distress syndrome.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
330
Q

What affects the outcomes and severity of septic shock?

A
  • The extent and virulence of the infection
  • the immune status of the host
  • the presence of other co-morbid conditions
  • the pattern and level of mediation production
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
331
Q

What are the three general phases of hypovolaemic and cardiogenic shock?

A
  • An initial nonprogressive phase during which reflex compensatory mechanisms are activated and perfusion of vital organs is maintained
  • A progressive stage characterised by tissue hypoperfusion and onset of worsening circulatory and metabolic imbalances, including a lactic acidosis
  • An irreversible stage that sets in after the body has incurred cellular and tissue injury so severe that even if the haemodynamic defects are corrected, survival is not possible
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
332
Q

What are the neurohumeral mechanisms that occur in the early non-progressive stage of shock that help to maintain cardiac output and blood pressure?

A
  • Baroreceptor reflexes, catecholamine release, activation of the renin-angiotensin axis, ADH release, and generalised sympathetic stimulation.
  • The net effect is tachycardia, peripheral vasoconstriction, and renal conservation of fluid
  • Coronary and cerebral vessels are less sensitive to the sympathetic response and therefore maintain relatively normal caliber, blood flow and oxygen delivery
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
333
Q

What happens during the progressive phase of shock?

A
  • There is widespread tissue hypoxia
  • In the setting of persistent oxygen deficit, intracellular aerobic respiration is replaced by anaerobic glycolysis with excessive production of lactic acid
  • The lactic acidosis lowers the tissue pH and blunts the vasomotor response, arterioles dilate, and blood begins to pool in the microcirculation
  • Peripheral pooling not only worsens the cardiac output, but also put the endothelial cells at risk of developing anoxic injury with subsequent DIC.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
334
Q

What happens during the irreversible stage of shock?

A
  • Widespread cell injury is reflected in lysosomal enzyme leakage, further aggrevating the shock state.
  • If ischaemic bowel allows intestinal flora into the circulation, bacterial septic shock may be superimposed
  • At this point, the patient may develop anuria as a result of acute tubular necrosis and acute renal failure
  • They may develop a coagulopathy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
335
Q

What are the morphological adrenal changes during shock?

A

There is cortical cell depletion. This does not reflect adrenal exhaustion but rather the conversion of the relatively inactive vacuolated cells to metabolically active cells that utilise stored lipids for the synthesis of steroids

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
336
Q

Immunity is protection from infectious pathogens. The mechanisms of defense against microbes fall into two broad categories. What are they?

A
  • Innate immunity refers to the mechanisms that are ready to react to infections even before the occur, and that have evolved to specifically recognise and combat microbes
  • Adaptive immunity consists of mechanisms that are stimulated by microbes and are capable of recognising microbial and non-microbial substances.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
337
Q

When do innate immunity and adaptive immunity take place?

A
  • Innate immunity is the first line of defence. It is mediated by cells and molecules that recognise products of microbes and dead cells and induce rapid protective host reactions.
  • Adaptive immunity develops later, after exposure to microbes and other foreign substances, and is even more powerful that innate immunity in combating infections
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
338
Q

What are the stages of innate immunity?

A

Innate immunity is always present, ready to provide defense against microbes and to eliminate damaged cells.
The three stages are: recognition of microbes and damaged cells, activation of various mechanisms, and elimination of unwanted substances

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
339
Q

What are the major components of innate immunity?

A

Epithelial barriers that block entry of microbes, phagocytic cells (mainly neutrophils and macrophages), dendritic cells, natural killer cells, and several plasma proteins, including the proteins of the complement system.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
340
Q

What are the epithelial barriers in innate immunity?

A
  • Epithelia of the skin and GI and resp tracts provide mechanical barriers to the entry of microbes from the external environment
  • Epithalial cells also produce antimicrobial molecules, such as defensins, and lymphocytes located in the epithelia combat microbes at these sites
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
341
Q

How do monocytes and neutrophils contribute towards innate immunity?

A

They are phagocytes in the blood that can rapidly be recruited to any site of infection; monocytes nter the tissues and mature to macrophages.
All tissues contain resident macrophages, which not only sense the presence of microbes and other offeding agents, but also ingest (phagocytose) these invaders and destroy them

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
342
Q

How do dendritic cells contribute to innate immunity?

A
  • They are a specialised cell population present in epithelia, lymphoid organs and most tissues.
  • They capture protein antigens and display peptides for recognition by T lymphocytes.
  • They are endowed with a rich collection of receptors that sense microbes and cell damage and stimulate the secretion of cytokines, mediators that play critical roles in inflammation and anti-viral defense
  • Thus, dendritic cells are involved in the initiation of innate immune responses, but not destruction of microbes and other offending agents.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
343
Q

What are the roles of natural killer cells in innate immunity?

A

They provide early protection against many viruses and intracellular bacteria

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
344
Q

What are pathogen-associated molecular patterns (PAMPs)?

A

Cells that participate in innate immunity are capable of recognising certain microbial components that are shared among related microbes and are often essential for infectivity (and thus cannot be mutated). These are called PAMPs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
345
Q

What are damage-associated molecular patterns?

A

Leukocytes recognise molecules released by injured and necrotic cells, which are called damage-associated molecular patterns (DAMPs).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
346
Q

What and where are pattern recognition receptors?

A

The cellular receptors that recognise PAMPs and DAMPs are often called pattern recognition receptors. They are located in all the cellular compartments where microbes may be present: plasma membrane receptors detect extracelluls microbes, endosomal receptors detect ingested microbes, and cytosolic receptors detect microbes in the cytoplasm

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
347
Q

What are the different classes of pattern recognition receptors?

A
  • Toll-like receptors
  • NOD-like receptors
  • C-type lectin receptors
  • RIG-like receptors
  • G protein-coupled recetprs
  • Mannose receptors
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
348
Q

Where are Toll-like Receptors (TLRs) and what do they do?

A

They are present in the plasma membrane and endosomal vesicles.
All these receptors signal by a common pathway that culminates in the activation of two sets of transcription factors:
1) NF-κB, which stimulates the synthesis and secretion of cytokines and the expression of adhesion molecules
2) Interferon regulatory factors (IRFs), which stimulate the production of antiviral cytokines, type I interferons

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
349
Q

Where are NOD-like receptors (NLRs) and what do they recognise?

A

They are cytosolic receptors that recognise a wide variety of substances, including products of necrotic cells (uric acid and released ATP), ion disturbances (loss of K+), and some microbial products.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
350
Q

What is the inflammasome and how is it related to NLRs? What does it do?

A

Several of the NLRs signal via a cytosolic multiprotein complex - the inflammasome, which activates an enzyme (caspase-1) that cleaves a precursor form of IL-1 to generate the biologically active form.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
351
Q

Where are C-type lectin receptors (CLRs)? What do they do?

A
  • They are expressed on the plasma membrane of macrophages and dentritic cells.
  • They detect fungal glycans and ellicit inflammatory reactions to funghi.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
352
Q

Where are RIG-like receptors (RLRs)? What do they do?

A
  • They are located in the cytosol of most cell types
  • They detect nucliec acids of viruses that replicate in the cytoplasm of infected cells.
  • They stimulate the production of antiviral cytokines
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
353
Q

Where are G protein-coupled receptors? What do they do?

A
  • They are on neutrophils, macrophages and most other types of leukocytes
  • They recognise short bacterial peptides containing N-formylmethionyl residues, which is produced by all bacteral proteins and few mammalian proteins, which enables neutrophils to detect bacterial proteins and stimulate chemotactic responses of the cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
354
Q

What do mannose receptors do?

A

They recognise microbial sugars and induce phagocytosis of the microbes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
355
Q

The innate immune system provides host defense by two main reactions. What are they?

A
  • Inflammation - cytokines and products of complement activation are produced during innate immune reactions and trigger the vascular and cellular components of inflammation. The recruited leukocytes destroy microbes and ingest and eliminate damaged cells
  • Antiviral defense - type I inferferons produced in response to viruses act on infected and uninfected cells and activate enzymes that degrade viral nucleic acids and inhibit viral replication.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
356
Q

How many different receptors does the innate immune system use compared to the adaptive immune system and why?

A

Innate immunity does not have memory or fine antigen specificity. It is estimated that innate immunity uses about 100 different receptors to recognise 1,000 molecular patterns.
In contrast, adaptive immunity uses two types of receptors (antibodies and T-cell receptors), each with millions or variations

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
357
Q

What does the adaptive immune system consist of?

A

Lymphocytes and their products, inlduing antibodies.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
358
Q

There are two types of adaptive immunity. What are they and what are they mediated by?

A
  • Humoral immunity, which protects against extracellular microbes and their toxins, is mediated by B lymphocytes and their secreted products, antibodies (AKA immunoglobulins).
  • Cell-mediated immunity, which is responsible for defence against intracellular microbes, is mediated by T-lymphocytes.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
359
Q

Where are lymphocytes and why are they there?

A

They are not fixed in particular tissues but constantly circulate among lymphoid and other tissues via the vlood and the lymphatic circulation.
This promotes immune surveillance by allowing lymphocytes to home to any site of infection.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
360
Q

What are the different types of lymphocytes (5) and what do they do?

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
361
Q

What are naive lymphocytes?

A

Mature lymphocytes that have not encountered the antigen for which they are specific are said to be naive (immunologically inexperienced).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
362
Q

After lymphocytes are activated by recognition of antigens, they differentiate into what? What do these cells do?

A
  • Effector cells, which perform the function of eliminated microbes
  • Memory cells, which live in a state of heightened awareness and are able to react rapidly and strongly to combat the microbe in case it returns
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
363
Q

What is clonal selection?

A

Lymphocytes specific for a large number of antigens exist before exposure to antigen, and when an antigen enters, it selectively activates the antigen-specific cells. This fundamental concept is called clonal selection.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
364
Q

Antigen receptor diversity is generated by somatic recombination of the genes that encode the receptor proteins. How does this process happen?

A

All cells of the body contain antigen receptors genes in the germline configuration, in which the genes encoding these receptors consist of spatially separated segments that cannot be expressed as proteins. During lymphocyte maturation, these gene segments recombine in random sets and variations are introduced at the sites of recombination, forming many different geners that can be transcribed and translated into functional antigen receptors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
365
Q

What is the enzyme in developing lymphocytes that mediates recombination of the gene segments?

A

It is the product of RAG-1 and RAG-2 (recombination activating genes)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
366
Q

What cells contain recombined antigen receptor genes? How does this influence tumour identification?

A

Germline receptor genes are present in all cells in the body, but only T and B cells contain recombined antigen receptor genes (T-cell receptors in T cells and immunoglobulin in B cells).
Hence the presence of recombined T-cell receptors or Ig genes, is a marker of T- or B-lineage cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
367
Q

There are three major populations of T cells, which serve distinct functions. What are they and what do they do?

A
  • Helper T lymphocytes stimulate B lymphocytes to make antibodies and activate other leukocytes (e.g. phagocytes) to destroy microbes
  • Cytotoxic T lymphocytes kill infected cells
  • Regulatory T lymphocytes limit immune responses and prevent reactions against self-antigens
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
368
Q

Where do T cells develop and from what?

A

They develop in the thymus from precursors that arise from haematopoetic stem cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
369
Q

Where are mature T cells found and what percentage of lymphocytes do that constistute?

A

They are found in the blood, where they constitute 60-70% of lymphocytes, and in T-cell zones of peripheral lymphoid organs.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
370
Q

How do T cells recognise antigens and what is this thing made up of?

A

Each T cell recognises a specific cell-bound antigen by means of an antigen-specific TCR.
In approximately 95% of T cells, the TCR consists of a disulfide-linked heterodimer made up of an α and a β polypeptide chain, each having a variable (antigen-binding) region and a constant region.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
371
Q

The αβ TCR recognises peptide antigens that presented by what?

A

Major histocompatibility complex (MHC) molecules on the surfaces of antigen-presenting cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
372
Q

What is MHC restriction? What is its purpose?

A

By limiting the specificity of T cells for peptides displayed by cell surface MHC molecules, called MHC restriction, the immune system ensures that T cells see only cell-associated antigens (e.g. those derived from microbes in cells or from proteins ingested by cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
373
Q

What forms the TCR complex?

A

Each TCR is noncovelently linked to six polypeptide chains, which form the CD3 complex and the ζ chain dimer. The CD3 and ζ proteins are invarient (identical) in all T cells. They are involved in the transduction of signals into the T cell that are triggered by binding of antigen to the TCR. Together, with the TCR, these proteins form the TCR complexes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
374
Q

In addition to CD3 and ζ proteins, T cells express several other proteins that assist the TCR complex in functional responses. These include CD4, CD8, CD28 and integrins. CD4 and CD8 are expressed on two mutually exclusive subsets of αβ T cells. What percentage are CD4 and what percentage are CD8. What do they do?

A
  • Approximately 60% of mature T cells are CD4+ and about 30% are CD8+.
  • Most CD4+ T cells function as cytokine-secreting helper cells that assist macrophages and B lymphocytes to combat infections.
  • Most CD8+cells function as cytotoxic (killer) T lymphocytes to destroy host cells harboring microbes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
375
Q

What MHC molecules do CD4+ and CD8+ bind to? What happens after that?

A
  • During antigen recognition, CD4 molecules bind to class II MCH molecules that are displaying antigen.
  • CD8 molecules bind to class I MHC molecules
  • The CD4 or CD8 coreceptor initiates signals that are necessary for activation of the T cells.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
376
Q

What cells are the only cells in the body capable of producing antibody molecules?

A

B lymphocytes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
377
Q

Where are B lymphocytes produced and where are they found?

A

B lymphocytes develop from precursors in the bone marrow.
Mature B cells consistute 10% to 20% of the circulating peripheral lymphocyte population and are also present in peripheral lymphoid tissues such as lymph nodes, spleen and mucose-associated lymphoid tissues.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
378
Q

How do B cells recognise antigens? How do they bind to them?

A
  • B cells recognise antigen via the B-cell antigen receptor complex.
  • Membrane bound antibodies of the IgM and IgD isotypes, present on the surface of all mature, naive B cells, are the antigen-binding component of the B-cell receptor complex.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
379
Q

What happens in B cells after they have bound to an antigen?

A

After stimulation by antigen and other signals, B cells develop into plasma cells, vertiable protein factories for antibodies.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
380
Q

How many antibody molecules can plasma cells secrete per second?

A

A single plasma cell can secrete hundreds to thousands of antibody molecules per second.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
381
Q

What are plasmablasts?

A

Antibody-secreting cells also detected in human peripheral blood.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
382
Q

What does the B-cell antigen receptor complex consist of?

A

It contains a heterodimer of Igα (CD79a) and Igβ (CD79b), which are essential for signal transduction through the antigen receptor

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
383
Q

Apart from CD79a and CD79b, what are some other molecules that B cells also express that are essential for their reponses and what do they do?

A
  • The type 2 complement receptor (CD2 or CD21), which recognises complement products generated during innate immune responses to microbes
  • CD40, which receives signals from helper T cells
  • CD21 is also used by the EBV as a receptor to enter and infect B cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
384
Q

What is the purpose of dendritic cells?

A

They are the most important antigen-presenting cells for initiating T-cell responses against protein antigens.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
385
Q

Several features of dendritic cells account for their key role in antigen presentation. What are they?

A
  • They are located at the right place to capture antigens - under epithelia, the common site of entry of microbes and foreign antigens, and in the interstitia of all tissues, where antigens may be produced
  • They express many receptors for capturing and responsing to microbes (and other antigens), including TLRs and lectins
  • In response to microbes, dendritic cells are recruited to the T-cell zones of lymphoid organs, where they are ideally located to present antigens to T cells.
  • Dendritic cells express high levels of MHC and other molecules needed for presenting antigens to and activating T cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
386
Q

What is a follicular dendritic cell? What do they do?

A

A second type of cell with dendritic morphologi present in the germinal centers of lypmhoid follicles in the spleen and lymph nodes and is called the follicular dendritic cell.

These cells bear Fc receptors for IgG and receptors for C3b and can trap antigen bound to antibodies or complement proteins. Such cells play a role in humoral immune responses by presenting antigens to B cells and selecting the B cells that have the highest affinity for the antigen, thus improving the quality of the antibody produced.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
387
Q

What is the function of natural killer cells?

A

The function of NK cells is to destory irreversibly stressed and abnormal cells, such as virus-infected cells and tumour cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
388
Q

What is the morphology of natural killer cells?

A

They are somewhat larger than small lymphocytes, and they contain abundant azurophilic granules

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
389
Q

What ability of natural killer cells makes then an early line of defense against viral infections and, perhaps, some tumours?

A

Natural killer cells are endowed with the ability to kill a variety of virus-infected cells and tumour cells, without prior exposure to or activation by these microbes or tumours.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
390
Q

What are the two cell surface molecules on natural killer cells? What do they do?

A

Two cell surface molecules, CD16 and CD56, are commonly used to identify NK cells.
CD16 is an Fc receptor for IgG, and it confers on NK cells the ability to lyse IgG-coated target cells. This process is known as antibody-dependent cell-mediated cytotoxicity.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
391
Q

How is the functional activity of NK cells regulated?

A
  • By a balance between signals from activating and inhibitory receptors.
  • There are many types of activating receptors, of which the NKG2D family is the best characterised - they recognise surface molecules that are induced by various kinds of stress, such as infections and DNA damage.
  • NK cell inhibitory receptors recognise self class I MHC molecules, which are expressed on all healthy cells. The inhibitory receptors prevent NK cells from killing normal cells.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
392
Q

How does viral infection or neoplastic transformation trigger natural killer cells?

A

It often enhances expression of ligands for activating receptors and at the same time reduces the expression of class I MHC molecules.
As a result, the balance is tilted towards activation, and the infected or tumour cell is killed.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
393
Q

What cytokines are associated with natural killer cells?

Which ones do they secrete? Which ones stimulate proliferation and activate killing?

A
  • Natural killer cells secrete cytokines such as interferon-γ, which activates macrophages to destroy ingested microbes, and thus NK cells provide early defense against intracellular microbial infections.
  • The activity of NK cells is regulated by many cytokines, including the interleukins (IL-2, IL-15, and IL-12)
  • IL-2 and IL-15 stimulate proliferation of NK cells
  • IL-12 activated killing and secretion of IFN-γ
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
394
Q

What are the peripheral lymphoid organs and what do they do?

A
  • Lymph nodes - antigen-presenting cells in the nodes are able to sample the antigens of microbes that may enter through epithelia into tissues and are carried in the lymph. Dendritic cells transport antigens of microbes from epithelia and tissues via lymph vessels to the nodes. Thus, the antigens become concentrated in lymph nodes
  • Spleen - Blood entering the spleen flows through a network of sinusoids. Bloodborne antigens are trapped by dendritic cells and macrophages in the spleen
  • The cutaneous and mucosal lymphoid systems are located under the epithelia of the skin and the GI and resp tracts, respectively. Pharyngeal tonsils and Peyer’s patches of the intestine are two anatomically defined mucosal lymphoid tissues
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
395
Q

Within the peripheral lymphoid organs, T lymphocytes and B lymphocytes are segregated into different regions. What are they?

A
  • In lymph nodes the B cells are concentrated in discrete structures, called follicles, located around the periphery, or cortex, of each node. If the B cells in a follicle have recently responded to an antigen, this follicle may contain a central region called a germinal center.
  • The T lymphoctyes are concentrated in the paracortex, adjacent to the follicles.
  • The follicles contain the follicular dendritic cells, involved in B cell activation, and the paracortex contains the dendritic cells that present antigens to T lymphocytes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
396
Q

What is lymphocyte recirculation and why is it important?

A

Lymphocytes constantly recirculate between tissues and home to particular sites; naive lymphocytes traverse the peripher lympoid organs where immune responses are initiated and antigens are concentrated and effector T cells migrate to sites of infection and inflammation, to locate and eliminate microbes at any site of infection.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
397
Q

What is the purpose of MHC molecules?

A

To display peptide fragments of protein antigens for recognition by antigen-specific T cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
398
Q

What are human leukocyte antigens (HLA)?

A

In humans, the MHC molecules are called HLA because they were initially detected on leukocytes by the binding of antibodies.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
399
Q

On the basis of their structure, cellular distibution and function, MHC gene products are classified into two major classes. What are these classes and where are they found?

A
  • Class I MHC molecules are expressed on all nucleated cells and platelets. They are heterodimers made of a polymorphic α heavy chain linked convalently to a smaller non-polymorphic β2-microglobulin.
  • Class II MHC molecules are mainly expressed on cells that present ingested antigens and respond to T-cell help (macrophages, B lymphocytes, and dendritic cells).
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
400
Q

What is the role of Class I and Class II MHC molecules?

A
  • Class I MHC molecules display peptides that are derived from proteins, such as viral and tumour antigens, that are located in the cytoplasm and usually produced in the cell, and class I-associated peptides are recognised by CD8+ T lymphocytes
  • Class II MHC molecules present antigens that are internalised into vesicles, and are typically derived from extracellular microbes and soluble proteins, class II-peptide complex is recognised by CD4+ T cells, which function as helper cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
401
Q

MHC molecules play several key roles in regulating T cell-mediated immune reponses. What are they?

A

1) Because diferent antigenic peptides bind to different MHC molecules, it follows that an individual mounts an immune response against a protein antigen only if they inherit the genes for those MHC molecules that can bind peptides derived from the antigen and present it to T cells
2) By segregating cytoplasmic and internalised antigens, MHC molecules ensure that the correct immune reponse is mounted against different microbes - cytotoxic lymphocyte mediated killing of cells harboring cytoplasmic microbes, and helper T cell-mediated antibody and macrophage activation to combat extracellular microbes.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
402
Q

Cytokines contribute to different types of immune responses. How do they contribute to innate vs adaptive immune responses? Give examples for each type

A
  • In innate immune responses, cytokines are produced rapidly after encounter with microbes and other stimuli, and function to include infammaltion and inhibit virus replication - TNF, IL-1, IL-2, type 1 IFNs, IFN-γ, and chemokines, produced mainly by macrophages, dendritic cells and natural killer cells
  • In adaptive immune responses, cytokines are produced principally by CD4+ T lymphocytes activated by antigen and other signals, and function to promote lymphocyte proliferation and differentiation and to activate effector cells. - IL-2, IL-4, IL-5, IL-17, IFN-γ.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
403
Q

What cells secrete IL-2 and what does it do?

A

One of the earliest responses of CD4+ helper T cells is secretion of the cytokine IL-2 and expression of high-affinity receptors for IL-2. IL-2 is a growth factor that acts on these T lymphocytes and stimulates their proliferation, leading to an increase in the number of antigen-specific lymphocytes.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
404
Q

Some of the activated CD4+ T cells differentiate into effector cells that secrete different sets of cytokine and perform different functions. What are these sets and their functions?

A
  • T helper 1 cells secrete the cytokine IFN-γ, which is a potent macrophage activator. The combination of CD40- ad IFN-γ mediated activation results in “classical” macrophage activation, leading to the induction of microbicidal substances in macrophages and the destruction of ingested microbes
  • T helper 2 cells produce IL-4, which stimulates B cells to differntiate into IgE-secreting plasma cells, and IL-5, which activates eosinophils. Eosinophils and mast cells bind to IgE-coated microbes such as helminthic parasites, and function to eliminate helminths. They also induce the ‘alternative’ activation pathway of macrophages
  • T helper 17 cells, which produce IL-17, recruit neutrophils and monocytes, which destroy some extracellular bacteria and funghi and are involved in some inflammatory diseases
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
405
Q

What do activated CD8+ T lymphocytes differetiate into? What do they do?

A

They differentiate into cytotoxic T lymphocytes that kill cells harboring microbes in the cytoplasm. By destroying the infected cells, CTLs eliminate the reservoirs of infection

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
406
Q

Antibody responses to most protein antigens require T cell help and are said to be T-dependent. In what way is this the case?

A
  • B cells ingest protein antigens into vesicles, degrade them, and display peptides bound to class II MHC molecules for recognition by helper T cells.
  • The helper T cells are activated and express CD40L and secrete cytokines, which work together to stimulate the B cells.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
407
Q

What antigens are T-independent? What happens here?

A

Many polysaccharide and lipid antigens cannot be recognised by T cells but have multiple identical antigenic determinants (epitopes) that are able to engage many antigen receptor molecules on each B cell and initate the process of B-cell activation; these responses are said to be T-independent

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
408
Q

Each plasma cell is derived from an antigen-stimulated B cell and secretes antibodies that recognise the same antigen that initiated the reponse. What type of antibodies do different molecules stimulate the secretion of?

A
  • Polysaccarides and lipids stimulate secretion mainly of IgM antibody
  • Protein antigens, by virtue of CD40L- and cytokine-mediated helper T-cell actons, induce the production of antibodies of different classes, or isotypes (IgG, IgA, IgE)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
409
Q

What is affinity maturation and isotype switching? Where do they occur and what stimulates them?

A
  • Isotype switching is induced by cytokines including IFN-γ and IL-4.
  • Helper T cells also stimulate the production of antibodies with high affinities for the antigen. This process, affinity maturation, improved the quality of the humoral immune response
  • They occur mainly in germinal centers, which are formed by proliferating B cells, especially in helper T cell-dependent response to protein antigens
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
410
Q

What are follicular helper T cells? What do they do?

A

Some activated B cells migrate into follicles and form germinal centers, which are the major sites of isotype switching and affinity maturation. The helper T-cells that stimulate these process in B lymphocytes migrate to and reside in the germinal centers and are called follicular helper T cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
411
Q

The humoral immune response combates microbes in many ways. What are they?

A
  • Antibodies bind to microbes and prevent then from infecting cells, thus neutralising the microbes
  • IgG antibodies coat (opsonise) microbes and target them for phagocytosis as phagocytes express receptors for the Fc tails of IgG.
  • IgG and IgM activate the complement system by the classical pathway, and complement products promote phagocytosis and destruction of microbes
  • IgA is secreted from mucoseal epithelia and neutralises microbes inthe lumens of the respiratory and GI tracts
  • IgG is actively transported across the placenta and protects the newborn until the immune system becomes mature
  • IgE and eosinophils cooperate to kill parasites, mainly by release of eosinphil granules contents that are toxic to the worms.
  • TH2 cytokines stimulate the production of IgE and activeate eosinophils, and thus the response to helminths is orchestrated by TH2 cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
412
Q

There are several important general features of hypersensitivity disorders. What are they?

A
  • Hypersensitivity reactions can be eilicited by exogenous environmental antigens (microbial and non-microbial) or endogenous self antigens
  • Hypersensitivity usually results from an imbalance between the effector mechanisms of immune responses and the control mechanisms that serve to normally limit such responses
  • The development of hypersensitivity diseases is often associated with the inheritance of a particular susceptibility genes (HLA genes)
  • The mechanisms of tissue injury in hypersensitivity reactions are the same as the effector mechanisms of defense against infectious pathogens
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
413
Q

What are the different types of hypersensitivity reactions?

A

1) Immediate hypersensitivity - caused by TH2 cells, IgE antibodies, and mast cells and other leukocytes. Mast cells release mediators that act on vessels and smooth msucle and proinflammatory cytokiens that recruit inflammatory cells
2) Antibody-mediated disorders - secreted IgG and IgM antibodies injure cells by promoting their phagocytosis or lysis and injure tissues by inducing inflammation
3) Immune complex-mediated disorders - IgG and IgM antibodies bind antigens usually in the circulation, and the antigen-antibody complexes deposit in tissues and induce inflammation. The leukocytes that are recruited produce tissue damage by release of lysosomal enzymes and generation of toxic free radicals
4) Cell-mediated immune disorders - sensitised T lymphocytes (Th1 and Th17 cells and CTLs) are the cause of the tissue injury. Th2 cells indue lesions that are part of immediate hypersensitivity reactions and are not considered a form of type IV hypersentivity

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
414
Q

How is an immediate or type 1 hypersensitivity reaction triggered?

A

It is a rapid immunological reaction occurring in a previously sensitised individual that is triggered by the binding of an antigen to IgE antibody on the surface of mast cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
415
Q

Local reactions are diverse and vary depending on the portal of entry of the allergen in type 1 hypersensitivity reaction?

A

Localised cutaneous rash or blisters (shin allergy, hives), nasal and conjunctival discharge (allergic rhinitis and conjunctivitis), hay fever, bronchial asthma, or allergic gastroenteritis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
416
Q

Many local type 1 hypersensitivity reactions have two well-defined phases. What are they and what happens during them?

A

1) The immediate reaction is characterised by vasodilation, vascular leakage, and depending on the location, smooth muscle spasm or glandular secretions. These changes usually become evident within minutes after exposure to an allergen and tend to subside in a few hours.
2) A second, late-phase reaction sets in 2-24 hours later without additiona exposure to antigen and may last for several days. This late-phase reaction is characterised by infiltration of tissues with eosinophils, neutrophils, basophils, monocytes and CD4+ T cells, as well as tissue destruction, typically in the form of mucosal epithelial cell damage

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
417
Q

Most immediate hypersensitivity disorders are caused by excessive responses from what cells? What do they do?

A

Most are caused by excessive Th2 responses and these cells play a central role by stimulating IgE production and promoting inflammation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
418
Q

The first step in the generation of Th2 cells is the presentation of the antigen (likely by dendritic cells) to naive CD4+ helper T cells. How do naive CD4+ helper cells respond to this?

A
  • In response to antigen and other stimuli (IL-4), the T cells differentiate into Th2 cells.
  • The newly minted Th2 cells produce a number of cytokines upon consequence encounter with the antigen (IL-4, IL-5, IL-13).
  • IL-4 acts on B cells to stimulate class swtiching to IgE and promotes the development of Th2 cells
  • IL-5 is involved in the development and activation of eosinophils.
  • IL-13 enhances IgE production and acts on epithelial cells to stimualte mucus secretion
  • In addition, Th2 cells produce chemokines that attract more Th2 cells, as well as other leukocytes to the reaction site
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
419
Q

Where are mast cells? What do they contain?

A
  • They are bone marrow-derived cells that are widely distributed in the tissues, being abundant near blood vessels and nerves and in subepithelial tissues
  • Mast cells have cytoplasmic membrane-bound granules that contain a variety of biologically active mediators
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
420
Q

How are mast cells activated in type 1 hypersensitivity reactions?

A
  • They are activated by the cross-linking of high-affinity IgE Fc receptors
  • They may also be triggered by complement components C5a and C3a (anaphylatoxins), both of which act by binding to receptors on the mast cell membrane.
  • Other mast cell secretagogues include some chemokines (e.g. IL-8), drugs such as codeine, morphine, adenosine, bee stings and physical stimuli (heat, cold, sunlight)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
421
Q

How are basophils and mast cells similar and what makes them different?

A

They are similar in many ways, including the presence of cell surface IgE Fc receptors as well as cytoplasmic granules.
In contrast to mast cells, however, basophuls are not normally present in tissues but rather circulate in the blood in extremely small numbers.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
422
Q

Mast cells and basophils express a high-affinity receptor, called FcεRI. What is this specific for and what does this mean?

A

FcεRI is specific for Fc portion of IgE and therefore avidly binds IgE antibodies.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
423
Q

When are masts cells sensitised?

A

IgE-coated mast cells are said to eb sensitised, because they are sensitive to a subsequent encounter with the specific antigen.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
424
Q

When a sensitised mast cell is exposed to the same antigen, what happens?

A

The cell is activated, leading eventually to the release of an arsenal of powerful mediators responsible for the clinical features of immediate hypersensitivity reactions.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
425
Q

What are the steps of mast cell activation?

A
  • The antigen binds to the IgE antibodies precious attached to the amst cells
  • Multivalent antigens bind to and cross-link adjacent IgE antibodies.
  • The underlying Fcε receptors are brought together, and this activates signal transudction pathways from the cytoplasmic portion of the receptors
  • These signals lead to the production of mediators that are responsible for the initial, sometimes explosive, symptoms of immediate hypersensitivity, and they also set into motion the events that lead to the late-phase reaction
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
426
Q

Mast cell activation leads to degranulation, with the discharge of preformed (primary) mediators that are stored in the granules, and de novo synthesis and release of secondary mediators, including lipid products and cytokines. Name some preformed mediators and what they do?

A
  • Vasoactive amines - most important is histamine, causing smooth muscle contraction, increased vascular permeabilityk and increased mucus secretion
  • Enzymes - contained in the granule matrix, include neutral proteases (chymase, tryptase) and several acid hydrolases. The enzymes cause tissue damage and lead to the generation of kinins and activated components of complement (C3a) by acting on their precursor proteins
  • Proteoglycans - these include heparin and chondroitin sulfate, they seve to packes and store the amines in the granules
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
427
Q

Mast cell activation leads to degranulation, with the discharge of preformed (primary) mediators that are stored in the granules, and de novo synthesis and release of secondary mediators, including lipid products and cytokines. Name some lipid mediators and what they do?

A

The major lipid mediators are arachidonic acid-derived products. Reactions in the mast cell membranes lead to activation of phospholipase A2.
* Leukotrienes C4 and D4 are the most potent vasoactive and spasmogenic agents known, they are several thousant times more active than histamine in increaseing vascular permeability and causing bronchial smooth muscle contraction. Leukotriene B4 is highly chemotactic for neutrophils, eosinophils and monocyes
* Prostaglandin D2 is the most abundant mediator produced in mast cells by the COX pathway, it causes intense bronchospasm and increased mucus secretion
* Platelet-activating factor is a lipid mediated produced by some mast cell populations but is not derived from AA. It causes platelet aggregation, release of histmine, bronchospasm, increased vascular permeability, and vasodilation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
428
Q

Mast cell activation leads to degranulation, with the discharge of preformed (primary) mediators that are stored in the granules, and de novo synthesis and release of secondary mediators, including lipid products and cytokines. Name some cytokines and their roles.

A
  • TNF, IL-1 and chemokines, which promote leukocytes recruitment
  • IL-4, which amplified the Th2 response
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
429
Q

On a morphological level, what happens during the late-phase reaction during type 1 hypersensitivity reactions?

A

In the late-phase reaction, leukocytes are recruited that amplify and sustain the inflammatory response without additional exposure to the tiggering antigen.
* Eosinophils are often an abundant leukocyte population in these reactions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
430
Q

How are eosinophils recruited to sites of immediate hypersensitivity?

A

By chemokines, such as eotaxin, and others that may be produced by epithelial cells, Th2 cells, and mast cells
The Th2 cytokine IL-5 is the most potent eosinophil-activating cytokine known.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
431
Q

Upon activation in late-phase immediate hypersensitivity reactions, what do eosinophils do?

A

They liberate proteolytic enzymes, as well as two unique proteins called major basic protein and eosinophil cationic protein, which damage tissues.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
432
Q

An increase propensity to develop immediate hypersensitivity reactions is called atopy. What are the morphological changes that atopic individuals tend to have?

A

They tend to have higher serum IgE levels and more IL-4 producing Th2 cells than does the general population.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
433
Q

Studies in patients with asthma reveal linkage to polymorphisms in several genes. What chromosome are these genes encoded, what are they and what do they do?

A
  • Some of these genes are located in the chromosome 5q31 region
  • These include genes encoding the cytokines IL-3, IL-4, IL-5, IL-9, IL-13 and GM-CSF.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
434
Q

What is nonatopic allergy?

A

It is estimated that 20-30% of immediate hypersensitivity reactions are triggered by non-antigenic stimuli, such as temperature extremes and exercsie, and do not invovle Th2 cells or IgE; such reactions are sometimes called nonatopic allergy

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
435
Q

Give me an overview of what happens during antibody-mediated (type II) hypersensitivity reactions

A

Anibodies that react with antigens present on cell surfaces or in the extracellular matrix cause disease by destroying these cells, triggering inflammation, or interfering with normal functions.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
436
Q

What are the three steps that occur during antibody mediated (type II hypersensitivity) reactions?

A

1) Opsonisation and phagocytosis
2) Complement and Fc receptor-mediated inflammation
3) Antibody-mediated cellular dysfunction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
437
Q

What happens during opsonisation and phagocytosis in antibody mediated (type 2) hypersensitivity reaction?

A
  • Cells opsonised by IgG antibodies are recognised by phagocyte Fc receptors.
  • When IgM or IgG antibodies are deposited on the surfaces of cells, they may activated the complement system and C3b and C4b are deposited on the surfaces of the cells and recognised by phagocytes that express receptors for these proteins.

The net result is phagocytsis of the opsonised cells and their destruction.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
438
Q

Clinically, in what situations does antibody-mediated cell destruction and phagocytosis occur?

A

1) Transfusion reactions, in which cells from an incompatible donor react with and are opsonised by preformed antibody in the host
2) Heamolytic disease of the newborn, in which there is an antigenic difference between the mother and the fetus, and IgG antierythrocyte antibodies from the mother cross the placenta and cause destruction of fetal red cells
3) Autoimmune haemolytic anaemia, agranulocytosis, and thrombocytopaenia, in which individuals produce antibodies to their own blood cells, which are then destroyed
4) Certain drug reactions, in which a drug acts as a “hapten”, by attaching to plasma membrane proteins of red cells and antibodies are produced against the drug protein complex

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
439
Q

What happens during the inflammation stage of antibody mediated (type II) hypersensitivity?

A
  • When antibodies deposit in fixed tissues, such as basement membranes and extracellular matrix, the resultant injury is due to inflammation.
  • The deposited antibodies activate complement, generating by-products, including chemotactic agents (C5a), which direct the migration of polymorphonuclear leukocytes and monocytes and anaphylatoxins (C3a and C5a), which increase vascular permeability
  • The leukocytes are activated by engagement of their C3b and Fc receptors.
  • This results in the production of other substances, that damage tissues, such as lysosomal enzymes, including proteases capable of digesting basement membrane, collagen, elastin, and cartilage, and ROS
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
440
Q

Clinically, in what situations does antibody-mediated inflammation occur as the mechanism responsible for tissue injury?

A

In some forms of glomerulonephritis, vascular rejection in organ grafts

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
441
Q

How does cellular dysfunction happen during antibody-mediated (type II) hypersensitivity reactions? Give examples

A

In some cases, antibodies directed against cell surface receptors impair or dysregulate function without causing cell injury or inflammation.
* In myasthenia gravis, antibodies reactive with acetylcholine receptors in the motor end plates of skeletal muscles block neuromuscular transmission and therefore cause muscle weakness
* In Graves disease, antibodies against the thyroid-stimulating hormone receptor on thyroid epithelial cells stimulate the cells, resulting in hyperthyroidism

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
442
Q

Give a general overview of what happens in immune complex-mediated (type III) hypersensitivity?

A

Antigen-antibody complexes produce tissue damage mainly by eliciting inflammation at the sites of deposition

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
443
Q

How is the pathological reaction usually initiated in immune complex-mediated (type III) hypersensitivity?

A
  • It is usually initiated when antigen combines with antibody in the cirulation, creating immune complexes that typically deposit in vessel walls.
  • Less frequently, the complexes may be formed at sites where antigen has been “planted” previously (called in situ immune complexes).
  • The antigens that form immune complexes may be exogenous or endogenous
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
444
Q

What organs do immune complex-mediated (type III) hypersensitivity reactions often take place in?

A

They tend to be systemic, but often preferentially involve the kidney (glomerulonephritis), joints (arthritis), and small blood vessels (vasculitis), all of which are common sites of immune complex deposition

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
445
Q

The pathogenesis of systemic immune complex disease can be divided into three phases. What are they and what happens in each one?

A

1) Formation of immune complexes - the introduction of a protein antigen triggers an immune response that results in the formation of antibodies, typcically about a week after injection. These antibodies are secreted into the blood, where they react with the antigen still present and form antigen-antibody complexes
2) Deposition of immune complexes - the circulating antigen-antibody complexes are deposited in various tissues, often in the glomeruli and joints
3) Inflammation and tissue injury - once the complexes are deposited, they initiate an acute inflammatory reaction. The resultant inflammatory lesion is termed vasculitis if it occurs in blood vesels, glomerulonephritis if it occurs in renal glomeruli, arthritis if it occurs in the joints

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
446
Q

What are the symptoms of immune complex-mediated (type III) reactions? When do thy happen?

A

During the inflammation and tissue injury phae (approx 10 days after antigen administration), clinical features such as fever, urticaria, joint pains, lymph node enlargement and proteinuria appear.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
447
Q

What antibodies are involved in immune complex-mediated (type III) hypersensitivity?

A

Complement-fixing antibodies (IgG and IgM) and antibodies that bind to leukocyte Fc receptors (some subclasses of IgG) induce the pathological lesions of immune complex disorders.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
448
Q

Give some examples of immune complex-mediated diseases

A

SLE
Post-strep glomerulonephritis
Polyarteritis nodosa
Reactive arthritis
Serum sickness

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
449
Q

What is an Arthus reaction?

A

The Arthus reaction is localised area of tissue necrosis resulting from acute immune complex vasculitis, usually elicited in the skin.
These complexes precipitate in the vessel walls and cause fibrinoid necrosis, and superimposed thrombosis worsens the ischaemic injury.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
450
Q

Give a general overview of the process of T Cell-mediated (type IV) hypersensitivity?

A

The cell mediated type of hypersensitivity is caused by inflammation resulting from cytokines produced by CD4+ T cells and cell killing by CD8+ T cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
451
Q

Give a general overview of what happens in CD4+ T cell-mediated hypersensitivity

A

In CD4+ T cell-mediated hypersensitivity reactions, cytokines produced by the T cells induce inflammation that may be chronic and destrucive.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
452
Q

What is the prototype of T cell-mediated inflammation? What happens during this?
Give an example

A

Delayed-type hypersensitivity, a tissue reaction to antigens give to immune individuals. An antigen administered into the skin of a previously immunised individual results in a detectable cutaneous reaction within 24-48 hours

An example of this is the tuberculin reaction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
453
Q

The inflammatory reactions stimulated by CD4+ T cells can be divided into sequential stages. What are these?

A

1) Activation of CD4+ T cells - naive CD4+ T cells recognise peptides displayed by dendritic cells and secrete IL-2, which functions as an autocrine growth factor to stimulate proliferation of the antigen-responsive T cells. The T cells differentiate into either Th1 (due to IFN-γ) or Th17 (via IL-1, IL-6 or IL-12).
2) Responses of differentiated effector T cells - upon repeat exposure to an antigen, Th1 cells secrete IFN-γ, which activates macrophages and they do their thing; if the activation is sustained, continued inflammation and tissue injury result

This produces granulomatous inflammation, which you know from before

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
454
Q

Give some examples of T cell-mediated (type IV) diseases…

A

RA
MS
T1DM
TBD
Psoriasis
Contact sensitivity

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
455
Q

What happens during CD8+ T Cell-mediated cytoxocity in T cell-mediated (type IV) hypersensitivity?

A

CD8+ CTLs kill antigen-expressing target cells. Tissue destruction by CTLs is important in T cell-mediated diseases, such as type 1 diabetes. The killing of infected cells leads to the elimination of the infection, but in some cases it is responsible for cell damage that accompanies the infection.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
456
Q

What is the principal mechanism of T cell-mediated killing of targets involves what?

A

Perforins and granzymes, preformed mediators contained in the lysosome-like granules of CTLs.
* CTLs that recognise the target cells secrete a complex consisting of perforin, granzymes, and other proteins which enters target cells by endocytosis.
* In the target cell cytoplasm, perforin facilitates the release of the granzymes from the complex. Granzymes are proteases that cleave and activate caspases, which induce apoptosis of teh target cells
* Activated CTLs also express Fas ligand, which can bind to Fas expressed on target cells and trigger apoptosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
457
Q

What is the complement system?

A

It is a collection of soluble proteins and membrane receptors that function mainly in host defence against microbes and in pathological inflammatory reactions.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
458
Q

What does the complement system consist of?

A

More than 20 proteins, some of which are numbered C1 through C9

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
459
Q

When does the complement system function?

A

In both innate and adative immunity for defense against microbial pathogens.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
460
Q

Broadly, what are the effects of complement activation?

A

Severeal cleavage products of complement proteins are elaborated that cause increased vascular permeability, chemotaxis, and opsonisation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
461
Q

Complement proteins are present in inactive forms in the plasma, what is the critical step in complement activation?

A

The proteolysis of the third (and most abundant) component, C3.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
462
Q

Cleavage of C3 in the complement pathway can occur by one of three pathways. What are these pathways?

A
  • The classical pathway, which is triggered by fixation of C1 to antibody (IgM or IgG) that has combined with antigen
  • The alternative pathway, which can be triggered by microbial surface molecules (e.g. endotoxin or LPS), complex polysaccharides, cobra venom, and other substances, in the absence of antibody
  • The lectin pathway, in which plasma mannose-binding lectin binds to carbohydrates on microbes and directly activates C1
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
463
Q

All three complement pathways converge lead to the same outcome. What is this and what happens next?

A
  • They all lead to the formation of an active enzyme called the C3 convertase, which splits C3 into two functionally distinct fragments, C3a and C3b.
  • C3a is released, and C3b becomes convalently attached to the cell or molecules where complement is being activated.
  • More C3b then binds to the previously generated fragments to form C5 convertase, which cleaves C5 to release C5a and leave C5b attached to the cell surface.
  • C5b binds the late components (C6-C9), culminating in the formation of the membrane attack complex (MAC)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
464
Q

The complement system has three main functions. What are they and how do they occur?

A
  • Inflammation - C3a, C5a (anaphylatoxins) and, to a lesser extent, C4a stimulate histamine release from mast cells and thereby increase vascular permeability and cause vasodilation. C5a is also a chemotactic agent and activates the lipooxygenase pathway of AA, releasing leukotrienes.
  • Opsonisation and phagocytosis - C3b and iC3b (inactive), when fixed to a microbial cell wall, act as opsonins and promote phagocytosis
  • Cell lysis - the deposition of the MAC on cells makes these cells permaeblt ot water and ions and results in lysis of the cells. - this is particularly importnat in microbes with thin walls such as Neissaria bacteria
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
465
Q

The activation of complement is tightly controlled by cell-associated and circulatin regulatory proteins. What are these and what do they do?

A
  • C1 inhibitor (C1 INH) blocks the activation of C1
  • Decay accelerating factor (DAF) and CD59 are two proteins that are linked to plasma memrbanes by a glycophosphatidyl anchor. DAF presents formation of C3 convertases and CD59 inhibits formation of the MAC
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
466
Q

What are oncogenes and proto-oncogenes?

A

Proto-oncogenes - normal cellular genes whose products promote cell proliferation
Oncogenes - mutated or overexpressed versions of proto-oncogenes that function autonomously, having lost dependence on normal growth promoting signals

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
467
Q

How are oncogenes created?

A

By mutations in proto-oncogenes and encode proteins called oncoproteins that have the ability to promote cell growth in the absence of normal growth-producing signals

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
468
Q

What are the roles of proto-oncogenes?

A

They have multiple roles, but all participate at some level in signaling pathways that drive proliferation. Thus, pro-growth proto-oncogenes may encode growth factors, growth factor receptors, signal transducers, transcription factors or cell cycle components.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
469
Q

When discussing tumours, what does differentiation and anaplasia mean?

A

Differentiation refers to the extent to which neoplastic parenchymal cells resemble the corresponding normal parenchymal cells, both morphologically and functionally
Lack of differentiation is called anaplasia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
470
Q

Are benign or malignant cells well differentiated or not?

A
  • In general, benign tumours are well differentiated, mitoses are usually rare and of normal configuration.
  • Malignant neoplasms exhibit a wide range of parenchymal cell differentiation, most exhibit morphological alterations that betray their malignant nature.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
471
Q

Malignant neoplasms that are composed of poorly differentiated cells are said to be anaplastic. This is often associated with many other morphological changes. What are these?

A
  • Pleomorphism - variety in size and shape, cells within the same tumour are not uniform
  • Abnormal nuclear morphology - the nuclei are often disproportionately large for the cell
  • Mitoses - many cells are in mitosis, reflecting the high proliferative activity of the parenchymal cells.
  • Loss of polarity - the orientation of anaplastic cells is markedly disturbed. Sheets or large amsses of tumour cells grow in an anarchic, disorganised fashion
  • Large central area of ischaemic necrosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
472
Q

What are the ranges of cells that occur with malignant pleomorphisms?

A

Cells within the same tumour are not uniform, they range from small cells with an undifferentiated appearance, to tumour giant cells many time larger than their neighbours.
Some tumor giant cells possess only a single huge polymorphic nucleus, whilst others may have two or more large hyperchromatic nuclei.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
473
Q

What is abnormal about the nuclear morphology of malignant cells?

A

They have a nuclear-to-cytoplasm ratio that may approach 1:1 instead of the normal 1:4 or 1:6.
The nuclear shape is variable and often irregular, and the chromatin is often coarsely clumped and distributed along the nuclear membrane, or more darkly stained than normal (hyperchromatic)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
474
Q

The better the differentiation of the transformed cell, the more completely it retains the functional capabilities of its normal counterpart. What does this mean for endocrine tumours?

A

Benign neoplasms and well-differentiated carcinomas of endocrine glands frequently secrete hormones characteristic of their origin. Increased levels of these hormones in the blood are used clinically to detect and follow such tumours.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
475
Q

In some instances of highly anaplastic undifferentiated cells, new and unanticipated functions emerge. What does this mean? Give some examples

A

Some tumours express fetal proteins that are not produced by comparable cells in the adult, while others express proteins that are normally only found in other types of adult cells. e.g. bronchogenic carcinomas may produce corticotropin, PTH, insulin, glucagon and other hormones, giving rise to paraneoplastic syndromes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
476
Q

What is dysplasia?

A

It is a term that literally means ‘disordered growth’. It is encountered principally in epithelia and is characterised by a constellation of changes that include the loss of uniformity of the individual cells as well as loss in their architectural orientation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
477
Q

Morphologically, what are some characteristics of dysplastic cells?

A

They may exhibit considerable pleomorphism and often contain large hyperchromatic nuclei with a high nuclear-to-cytoplasmic ratio.
In addition, mitotic figures are more abundant that in the normal tissue and rather than being confined to the basal layer may instead be seen at all levels, including surface cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
478
Q

What is a carcinoma in situ?

A

When dysplastic changes are marked and involve the full thickness of the epithelium, but the lesion does not penetrate the basement membrane, it is considered a pre-invasive neoplasm and is referred to as carcinoma in situ

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
479
Q

What is an invasive tumour?

A

Once the tumor cells breach the basement membrane, the tumour is said to be invasive

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
480
Q

Does dysplasia always progress to cancer?

A

Dysplastic changes are often found adjacent to foci of invasive carcinoma, and in some situations, frequently antedates the appearance of cancer.
Some mutations associated with cancers may be present in even ‘mild’ dysplasia.
Therefore, although dysplasia may be a pre-cursor to malignant transformation, it does not always progress to cancer.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
481
Q

How does the process of growth vary between malignant and benign tumours?

A
  • The growth of cancers is accompanied by progressive infiltration, invasion, and destruction of the surrounding tissue
  • Nearly all benign tumours grow and cohesive expansile masses that remain localised to their site of origin and lack the capacity to infiltrate, invade or metastasize to distant sites
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
482
Q

What is a capsule associated with benign tumours and what does it consist of?

A

Because benign tumours grow and expand slowly, they usually develop a rim of compressed fibrous tissue called a capsule that separates them from the host tissue.
This capsule consists largely of extracellular matrix deposited by stromal cells such as fibroblasts, which are activated by hypoxic damage resulting from the pressure of the expanding tumour

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
483
Q

How does the capsule of a benign tumour affect how it feels clinically?

A

Such encapsulation does not prevent tumour growth, but it creates a tissue plane that makes the tumour discrete, readily palpable, moveable (non-fixed), and easily excisable by surgical enucleation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
484
Q

How do malignant tumors vary from benign tumours re: the capsule and demarcation from normal tissue?

A

Malignant tumours are, in general, poorly demarcated from the surrounding normal tissue, and a well-defined cleavage plane in lacking.
Slowly explanding malignant tumours, however, may develop an apparently enclosing fibrous capsule and may push along a broad fron into adjacent normal structures.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
485
Q

Histologically, what would you see in ‘pseudoencapsulated’ malignant masses?

A

They almost always show rows of cells penetrating the margin and infiltrating the adjacent structures, a crablike pattern of growth that consitutes the popular image of cancer.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
486
Q

Next to the development of metastases, invasiveness is the most reliable feature that differentiates cancers from benign tumours. How?

A

Most malignant tumours do not recognise normal anatomic boundaries and can be expected to penetrate the wall of the colon or uterus for example.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
487
Q

Some cancers seem to evolve from a preinvasive stage - carcinoma in situ. Name some examples.

A

Carcinomas of the skin, breatst and uterine cervix

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
488
Q

What is metastasis?

A

Metastasis is defined by the spread of a tumour to sites that are physically discontinuours with the primary tumour, and unequivocally marks a tumour as malignant, as by definition noeplasms do not metastasise.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
489
Q

In general, the likelihood of a primary tumour to metastasise is what?

A

It correlates with lack of differentiation, aggressive local invasion, rapid growth, and large size.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
490
Q

What percentage of newly diagnosed solid tumours present with metastases?

A

30%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
491
Q

Dissemination of cancers may occur through one of three pathways. What are these pathways?

A

1) Direct seeding of body cavities or surfaces
2) Lymphatic spread
3) Haematogenous spread

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
492
Q

Where does seeding of body cavity and surfaces take place?

A

Seeding of body cavities and surfaces may occur whenever a malignant neoplasm penetrated into a natural ‘open field’ lacking physical barriers.
Most often involved is the peritoneal cavity, but any other cavity - pleural, pericardial, subarachnoid, and joint spaces - may be affected.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
493
Q

What is pseudomyxoma peritonei?

A

Sometimes mucus-secreting appendiceal carcinomas or ovarian carcinomas fill the peritoneal cavity with a gelatinous neoplastic mass referred to as pseudomyxoma peritonei

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
494
Q

What is the most common pathway for the initiall dissemination of carcinomas?

A

Lymphatic spread

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
495
Q

How do tumours spread through lymphatics?

A

Tumours do not contain functional lymphatics, but lymphatic vessels located at the tumor margins are apparently sufficien for the lymphatic spread of tumour cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
496
Q

What cancers often spread through seeding of body cavities?

A

It is particularly characteristic of carcinomas arising in the ovaries, which, not infrequently, spread to peritoneal surfaces, which become coated with a heavy cancerous glaze.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
497
Q

What cancers often spread via lymphatic spread? and via which nodes?

A
  • Because carcinomas of the breast usually arise in the upper outer quadrants, they generally disseminate first to the axillary lymph nodes. Cancers of the inner quadrants drain to the nodes along the internal mammary arteries. Thus, the infraclavicular and supraclavicular nodes may become involved
  • Carcinomas of the lyng arising in the major respiratory passages metastasis first to the perihilar tracheobronchial and mediastinal nodes.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
498
Q

What are skip metastases regarding lymph nodes? Why do they happen?

A

Local lymph nodes, however, may be bypassed - so called skip metastasis - because of venous-lymphatic anastomoses or because inflammation or radiation has obliterated lymphatic channels

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
499
Q

What is a sentinal lymph node?

A

A senitel lymph node is defined as ‘the first node in a regional lymphatic basin that receives lymph flow from the primary tumour’

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
500
Q

Does nodal enlargement in proximity to a cancer always equate with dissemination of the primary lesion?

A

Drainage of tumour cell debris or tumour antigens, or both, also induces reactive changes within nodes.
Thus, enlargement of nodes may be caused by the spread and growth of cancer cells or reactive hyperplasia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
501
Q

Are arteries or veins more or less readily penetrated by haematogenous spread of metastases?

A

Arteries, with their thicker walls, are less readily penetrated than the veins.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
502
Q

In what situation does arterial spread of metastases occur?

A

Arterial spread may occur, however when tumour cells pass through the pulmonary capillary beds or pulmonary arteriovenous shunts or when pulmonary metastases themselves give rise to additional tumour emboli.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
503
Q

In what situation does venous invasion of metastases occur?

A

With venous invasion, the bloodborne cells follow the venous flow draining the site of the neoplasm, and the tumour cells often come to rest in the first capillary bed they encounter. Understandably, the liver and lungs ar the most frequently involved in such haematogenous dissemination.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
504
Q

Certain cancers have a propensity for invasion of veins. What are they and where do they spread?

A
  • Renal cell carcinoma often invades the branches of the renal vein and then the renal vein itself, from where it may grow in a snakelike fashion up the inferior vena cava, sometimes reaching the right side of the heart
  • HCCs often penetrate portal and hepatic radicles to grow within them into the main venous channels.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
505
Q

What are the different modes of activation in tumours that proto-oncogenes can do?

A

Overexpression
Amplification
Point mutation
Translocation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
506
Q

Why is angiogenesis essentional in tumour growth?

A

Even if a solid tumour possesses all of the genetic aberrations that are required for malignant transofrmation, it cannot enlarge beyond 1 to 2mm in diameter unless it has the capacity to induce angiogenesis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
507
Q

What happens due to neovascularisation?

A

Perfusion supplied needed nutrients and oxygen, and newly formed endothelial cells stimulate the growth of adjacent tumour cells by secreting growth factors, such as insulin-like growth factos (IGFs) and PDGF.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
508
Q

Is tumour neovascularisation normal?

A

No, it is effective at delivering nutrients and removing wastes but it is not entirely normal; the vessels and leaky and dilated, and have a haphazard pattern of connection, features that can be appreciated on angiograms.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
509
Q

What is an angiogenic switch?

A

Early in their development, most human tumours do not induce angiogenesis. Starved of nutrients, these tumours remain small or in situ, possible for years, until an angiogenic switch terminates the stage of vascular quiescence.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
510
Q

What is the molecular basis of the angiogenic switch?

A

It involves increased production of angiogenic factors and/or loss of angiogenic inhibitors. These factors may be produced by the tumour cells themselves or by inflammatory cells or other stromal cells associated with the tumours.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
511
Q

What are the two phases of the metastatic cascade?

A

1) invasion of the extracellular matrix
2) vascular dissemination, homing of tumour cells and colonisation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
512
Q

Tumour cells must interact with the extracellular matrix at several stages in the metastatic cascade. What are they?

A
  • A carcinoma must first breach the underlying basement membrane, then traverse the interstitial connective tissue, and ultimately gain access to the circulation by penetrating the vascular basement membrane.
  • This process is repeated in revers when tumour cell emboli extravasate to a distant site.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
513
Q

Invastion of the ECM initiates the metastatic cascade and is an active process that can be resolved into several steps. What are they?

A
  • “Loosening up” of tumour cell-tumour interactions
  • Degradation of ECM
  • Attachment to novel ECM components
  • Migration and invasion of tumour cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
514
Q

Dissociation of cancer cells from one another is often the result of alterations in intercellular adhesion molecules and is the first step in the process of invasion. How is the function of E-cadherins related to this?

A

Normal epithelial cells are tightly glued to each other and the ECM by a variety of adhesion molecules. E-cadherins mediate the homotypic adhesion of epithelial cells, serving to both hold the cells toegterh and to relay signals between the cells.
In several epithelial tumours, including adenocarcinomas of the colon, stomach and breast, E-cadherin function is lost.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
515
Q

Degradation of the basement membrane and interstitial connective tissue is the second step in invasion. How does it occur?

A

Tumour cells may accomplish this by either secreting proteolytic enzymes themselves or by inducing stromal cells (e.g. fibroblasts and inflammatory cells) to elaborate proteases.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
516
Q

Many different families of proteases have been implicated in tumour cell invasion. Name some and explain what they do?

A
  • Metalloproteinases (MMPs) regulate tumour invasion not only by remodeling insoluble components of the basement membrane and interstitial matrix but also by releasing ECM-sequestered growth factors.
  • Cathepsin D
  • Urokinase plasminogen activator
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
517
Q

The third step of tumour invasion involves changes in attachment of tumour cells to ECM proteins. How does this happen?

A
  • Loss of adhesion in normal cells leads to induction of apoptosis, but tumour cells are resistant to this form of cell death.
  • Additionally, the matrix itself is modified in ways that promote invasion and metastasis - e.g. cleavage of the basement membrane proteins collagen IV and laminin by MMP2 or MMP9 generates novel sites that bind to receptors on tumour cells and stimulate migration.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
518
Q

What is measles?

A

Measles is an acute viral infection that affects multiple organs and causes a wide range of disease, from mild, self-limiting infections to severe systemic manifestations

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
519
Q

What type of virus is the measles virus?

A

It is a single-stranded RNA virus of the paramyxovirus family, which includes mumps, RSV, parainfluenze and human metapneumovirus.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
520
Q

How many serotypes of the measles virus are there? How is it transmitted?

A

There is only one serotype of measles virus.
Measles virus is transmitted by respiratory droplets.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
521
Q

What are three cell-surface receptors that have been identified for measles? Where are they and what do they bind to?

A
  • CD46 (a complement-regulartory protein that inactivates C3 convertases) is expressed on all nucleated cells
  • Signaling lypmhocytic activation molecule (SLAM, a molecule involved in T-cell activation) is expressed on cells of the immune system
  • Nectin 4 (adherens junction protein) is expressed on epithelial cells.

All of these receptors bind the viral haemgglutinin protein.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
522
Q

How does the measles virus replicate?

A
  • It can replicate in a variety of cell types, including epithelial cells and leukocytes.
  • The virus initially multiplies within the respiratory tract and then spreads to local lymphoid tissues
  • Replication of the virus in lymphatic tissue is followed by viremia and systemic dissemination to many tissues, including the conjunctiva, skin, respiratory tract, urinary tract, small blood vessles, lymphatic system, and CNS
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
523
Q

How do children develop immunity to measles?

A
  • Most children develop T-cell-mediated immunity to measles virus that helps control the viral infection and produces the measle rash.
  • Antibody-mediated immunity to measles virus protects against reinfection.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
524
Q

What does measles cause, that is responsible for much of measles-related morbidity and mortality? How does this happen?

A
  • It can cause transient but profound immunosuppression, resulting in secondary bacterial and viral infections, which are responsible for much of measles-related morbidity and mortality.
  • Alterations of both innate and adaptive immune responses occur following measles infection, including defects in dendritic cell and lymphocyte function.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
525
Q

What are some rare late complications of measles?

A

Subacute sclerosing panencephalitis and measles inclusion body encephalitis (in immunocompromised individuals).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
526
Q

What is the morphology of measles virus?

A
  • The blotchy, reddish brown rash of measles virus infection on the face, trunk and proximal extremities is produced by dilated skin vessels, oedema, and a mononuclear perivascular infiltrate.
  • Ulcerated mucosal lesions in the oral cavity near the opening of the Stensen ducts (the pathognomic Koplic spots) are marked by necrosis, neutrophilic exudate, and neovascularisation.
  • The lymphoid organs typically have marked follicular hyperplasia, large germinal centers, and randomly distributed multinucleate giant cells, called Warthin-Finkeldey cells, which have eosinophilic nuclear and cytoplasmic inclusion bodies, found in the lung and sputum
Koplic spots
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
527
Q

What is mumps?

A

Mumps is an acute systemic viral infection usually associated with pain and swelling of the salivary glands

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
528
Q

What type of virus is mumps?

A

Like measles, mumps is a member of paramyxovirus family.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
529
Q

What are the two types of surface glycoproteins that the mumps virus has?

A
  • One with hemagglutinin and neuraminidase activities
  • The other with cell fusion and cytolytic activities
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
530
Q

How do mumps viruses replicate and effect cells?

A
  • They enter the upper respiratory tract through inhalation of respiratory droplets, spread to draining lymph nodes where they replicate in lymphocytes (preferentially in activated T cells), and then spread through the blood to the salivary and other glands
  • Mumps virus infects salivary gland ductal epithalial cells, resulting in desquamation of involved cells, oedema, and inflammation that leads to the classic salivary gland pain and swelling.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
531
Q

Where can mumps spread to?

A

Mumps can spread to other sites, including the CNS, testis, ovary and pancreas.
Aseptic meningitis is the most common extrasalivary gland complication of mumps infection, occurring in up to 15% of cases.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
532
Q

What can be used to get a definitive diagnosis of mumps?

A

Serology, viral culture, or PCR assays.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
533
Q

What is the morphology of mumps parotitis?

A
  • Mumps parotitis is bilateral in 70% of cases
  • The affected glands are enlarged, have a doughy consistency, and are moist, glistening, and reddish-brown on cross-section.
  • On microscopic examination the gland interstitium is oedematous and diffusely infiltrated by macrophages, lymphocytes, and plasma cells, which compress acini and ducts.
  • Neutrophils and nectrotic debris may fill the duct lumen and cause focal damage to the lining epithelium
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
534
Q

What is the morphology of mumps orchitis?

A
  • In mumps orchitis testicular swelling may be marked, caused by oedema, mononuclear cell infiltration, and focal haemorrhages.
  • Because the testis is tightly contained within the tunica albuginea, parenchymal swelling may compromise the blood supply and cause areas of infarction.
  • The testicular damage can lead to scarring, atrophy, and, if severe, sterility.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
535
Q

How does mumps affect the pancreas and the CNS?

A
  • Infection and damage of acinar cells in the pancreas may release digestive enzymes, causing parenchymal and fat necrosis and neutrophil-rich inflammation.
  • Mumps encephalitis causes perivenous demyelination and perivascular mononuclear cuffing.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
536
Q

What is poliovirus infection?

A

Poliovirus causes an acute systemic viral infection, leading to a wide range of manifestations, from mild, slef-limited infections to paralysis of limb muscles and respiratory muscles.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
537
Q

What type of virus is polio?

A

It is a spherical, unencapsulated ENA virus of the enterovirus genus.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
538
Q

What type of vaccine is polio?

A
  • Salk formalin-fixed (killed) vaccine
  • Sabin oral, attenuated (live) vaccine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
539
Q

How is poliovirus transmitted and how does it replicate?

A
  • Polio, like other enteroviruses, is transmitted by the fecal-oral route.
  • The virus infects human cells by binding to CD155, an epithelial adhesion molecule.
  • The virus is ingested and replicates in the mucosa of the pharynx and gut, including tonsils and Peyer patches in the ilerum.
  • It then spreads through lymphatics to lymph nodes and eventually the blood, producing transient viremia and fever.
  • Although most polio infections are symptomatic, in 1% of people it invades the CNS and replicates in motor neurons of the spinal cord (spinal poliomyelitis) or brain stem (bulbar poliomyelitis).
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
540
Q

What is Viral haemorrhagic fever?

A

It is a severe life-threatening multisystem syndrome in which there is vascular dysregulation and damage, leading to shock.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
541
Q

What causes viral haemorrhagic fever?

A

It is caused by enveloped RNA viruses belonging to four different genera: Arenaviridae, Filoviridae, Bunyaviridae, and Flaviviridae.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
542
Q

What are the symptoms of viral haemorrhagic fever?

A

It can produce a spectrum of illnesses, ranging from a mild acute disease, characterised by fever, headache, myalgia, rash, neutropaenia, and thrombocytopaenia to severe, life-threatening disease in which there is sudden haemodynamic deterioration and shock.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
543
Q

How is viral haemorrhagic fever transmitted?

A
  • All the viruses pass through an animal or insect host during their life cycles and therefore their ranges are restricted to areas in which their hosts reside.
  • Humans are incidental hosts who are infected when they come into contact with infected hosts (typically rodents) or insect vectors (mosquitos adn ticks). Some viruses that cause haemorrhagic fever (Ebola) also can spread from person to person
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
544
Q

What is the pathogenesis of viral haemorrhagic fever?

A
  • The pathogenesis of the infection and its complications vary among the different viruses but there are some common features
  • Damage to blood vessels is often prominent. It may be caused by direct or indirect infection of and damage to endothelial cells, or infection of macrophages and dendritic cells leading to production of inflammatory cytokines.
  • There may be haemorrhagic manifestations, including petechiae, caused by a combinations of thrombocytopaenia or platelet dysfunction, endothelial injury, cytokine-induced DIC, and deficiency of clotting factors because of hepatic injury.
  • Necrosis of tissues secondary to the vascular lesions and haemorrages may be seen and varies from mild and focal to massive, but the attendant inflammatory response is usually minimal.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
545
Q

What is a latent infection?

A

Latency is defined as the persistence of viral genomes in cells that do not produce infectious virus.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
546
Q

What happens when a latent infection is reactivated?

A

Dissemination of the infection and tissue injury stem from reactivation of the latent virus.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
547
Q

What are the viruses that most frequently establish latent infections in humans?

A

Herpesviruses

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
548
Q

What type of virus are herpesviruses?

A

They are large encapsulated viruses with double-stranded DNA genomes that encode approximately 70 proteins.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
549
Q

What are the patterns of infection caused by herpesvirus?

A

Herpesviruses cause acute infection followed by latent infection in which the viruses persist in a noninfectious form with periodic reactivation and shedding of infectious virus.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
550
Q

There are eight types of human herpesviruses, belonging to three subgroups that are defined by the type of cell most frequently infected and the site of latency. What are the groups, give some examples and what cells do they infect?

A
  • α-group viruses, including HSV-1, HSV-2 and VZV, which infect epithelial cells and produce latent infection in neurons
  • Lymphotropic β-group viruses, including CMV, human herpesvirus-6 (which causes exanthem subitum, also known as roseola infantum and sixth disease, a benign rash of infants), and human herpesvirus-7, which infect and produce latent infection in a variety of cell types
  • γ-group viruses EBV and KSHV/HHV-8, the cause of Kaposi sarcoma, which produce latent infection mainly in lymphoid cells.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
551
Q

What are the similarities of HSV-1 and HSV-2?

A

HSV-1 and HSV-2 differ serologically but are closely related genetically and cause a similar set of primary and recurrent infections

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
552
Q

How do HSV-1 and HSV-2 replicate and infect?

A
  • Both viruses replicate in the skin and the mucous membranes at the site of entry of the virus (usually oropharynx or genitals), where they produce infectious virions and cause vesicular lesions of the epidermis.
  • The viruses spread to sensory neurons that innervate these primary sites of replication.
  • Viral nucleocapsids are transported along axons to the neuronal cell bodies, where the viruses establish latent infection
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
553
Q

In immunocompetent hosts, how long does it take for primary HSV infection to resolve?

A

A few weeks, although the virus remains latent in nerve cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
554
Q

What happens for the HSV during latency?

A
  • During latency the viral DNA remains within the nucleus of the neuron, and only latency-associated viral RNA transcripts (LATs) are synthesised.
  • No viral proteins appear to be produced during latency.
  • LATs may contribute to latency by conferring resistance to apoptosis, silencing lytic gene expression through heterochromatin formation, and serving as precursors for microRNAs that downregulate expression of critical HSV lytic genes.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
555
Q

How does reactivation occur?

A
  • Reactivation of HSV-1 and HSV-2 may occur repeatedly with or without symptoms, and results in the spread of virus from the neurons to the skin or to mucous membranes.
  • Reactivation can occur in the presence of host immunity, because HSVs have developed ways to avoid immune recognition.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
556
Q

What ways have HSV developed to avoid immune recognition?

A
  • HSVs can evade antiviral CTLs by inhibiting the MHC class I recognition pathway
  • They can elude humoral immune defenses by producing receptors for teh Fc domain of immunoglobulin and inhibitors of complement.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
557
Q

HSV-1 is the major infectious cause of corneal blindness in the US. How does it cause it?

A
  • Corneal epithelial disease is thought to be due to direct viral damage, whilst corneal stromal disease appears to be immune-mediated.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
558
Q

HSV-1 is the major cause of fatal sporadic encephalitis in the US. Where in the brain does it involve?

A

When the infection spreads to the brain, it usually involves the temporal lobes and orbital gyri of the frontal lobes.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
559
Q

How does HSV-2 infection effect HIV transmission?

A

HSV-2 infection increases the risk of HIV transmission by four-fold and increases the risk of HIV acquisition by two-to-three-fold.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
560
Q

What is the morphology of HSV-infected cells?

A

They contain large, pink to purple intranuclear inclusions (Cowdry type A) that consist of viral replication proteins and virions at various stage of assembly that push the host cell chromatin out to the edges of the nucleus. Due to cell fusion, HSVs also produc inclusion-bearing multinucleated syncytia.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
561
Q

Where are HSV-1 and HSV-2 lesions?

A
  • They cause lesions ranging from self-limited cold sores and gingivostomatitis to life-threatening disseminated visceral infection and encephalitis.
  • Fever blisters or cold sores favor the facial skin around mucosal orifices (lips, nose), where their distribution is frequently bilateral and independent of skin dermatomes.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
562
Q

What is gingivostomatitis? Is it HSV-1 or -2?

A
  • It is usually encountered in children, and is caused by HSV-1
  • It is a vesicular eruption extending from the tongue to the retropharynx and causing cervical lymphadenopathy.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
563
Q

What is herpetic whitlow in HSV? Who does it happen to?

A

Swollen, erythematous HSV lesions of the fingers of palm (herpetic whitlow) occur in infants and, occasionally, in health care workers

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
564
Q

What are genital herpes? Is it caused by HSV-1 or -2?

A
  • It is more often caused by HSV-2 than by HSV-1.
  • It is characterised by vesicles on the genital mucous membranes as well as on the external genitalia that are rapidly converted into superficial ulcerations, rimmed by an inflammatory infiltrate
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
565
Q

How is HSV transmitted to neonates? Is it usually HSV-1 or HSV-2? How does it affect them?

A
  • Usually HSV-2 can be transmitted to neonates during passage through the birth canal of infected mothers.
  • Although HSV-2 infection in the neonate may be more mild, more commonly it is fulminating with generalised lymphadenopathy, splenomegaly and necrotic foci throughout the lungs, liver, adrenals and CNS
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
566
Q

Two forms of corneal lesions are caused by HSV. What are they? How do they present?

A

1) Herpes epithelial keratitis shows typical virus-induced cytolysis of the superficial epithelium
2) Herpes stromal keratitis is characterised by infiltrates of mononuclear cells around keratinocytes and endothelial cells, leading to neovascularisation, scarring, opacification of the cornea, and eventual blindness. Here the damage is caused by an immunologic reaction to the HSV infection, rather than the cytopathic effects of the virus itself

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
567
Q

What is Varicella-Zoster Virus?

A

Acute infection with VZV causes chickenpox and reactivation of latent VZV causes shingles.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
568
Q

What is the pathological process of VZV?

A
  • Like, HSV, VZV infects mucous membranes, skin, and neurons and causes a self-limited primary infection in immunocompetent individuals.
  • Also like HSV, VZV evades immune responses and establishes a latent infection in sensory ganglia.
  • In contrast to HSV, VZV is transmitted by respiratory aerosols, disseminates haemtogenously, and causes widespread vesicular skin lesions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
569
Q

Where is VZV infection seen? How is it reactivated?

A
  • Latent VZV infection is seen in neurons and/or satellite cells around neurons in the dorsal root ganglia.
  • Reactivation and clinical recurrences causing shingles are uncommon but may occur many years after the primary infection
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
570
Q

Where is localised recurrence of VZV most frequent and painful?

A

In dermatomes innervated by the trigeminal ganglia, where the virus is more likely to be latent.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
571
Q

How is VZV infection diagnosed?

A

By viral culture, or detection of viral antigens in cells scraped from superficial lesions.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
572
Q

How long after a respiratory infection does the chickenpox rash present? How does it present?

A
  • The chickenpox rash occurs approx 2 weeks after respiratory infection. Lesions appear in multiple waves centrifugally from the torso to the head and extremities.
  • Each lesion progresses rapidly from a macule to a vesicle, which resembles a dewdrop on a rose petal.
  • After a few days most chickenpox vesicles rupture, crust over, and heal by regeneration, leaving no scars. However, bacterial superinfection of vesicles that are ruptured by trauma may lead to destruction of the basal epidermal layer and residual scarring.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
573
Q

How does chickenpox appear histologically?

A

Chickenppox lesions show intraepithelial vesicles with intranuclear inclusions in epithelial cells at the base of the vesicles.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
574
Q

When does shingles occur?

A

When VZV that has long remained latent in dorsal root ganglia after a previous chickenpox infection is reactivated and infects sensory nerves that carry it to one or more dermatomes.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
575
Q

What cells does VZV infect in shingles and how does it present?

A

The virus infects keratinocytes and causes vesciular lesions, which, unlike chickpox, are often associated with intense itching, burning, or sharp pain because of concomitant radiculoneuritis. This pain is especially severe when the trigeminal nerves are involved

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
576
Q
A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
577
Q

What is Ramsay Hunt syndrome?

A

Rarely, the geniculate nucleus is involved in a shingles infection, causing facial paralysis (Ramsay Hunt syndrome).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
578
Q

Histologically, what do the sensory ganglia appear like in shingles?

A

They contain a dense, predominantly mononuclear infiltrate, with herpetic intranuclear inclusions within neurons and their supporting cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
579
Q

What is cytomegalovirus?

A

CMV, a β-group herpesvirus, can produce a variety of disease manifestations, depending on the age of the host, and, more importantly, on the host’s immune status.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
580
Q

How does CMV infection start?

A

CMV latently infects monocytes and their bone marrow progenitors and can be reactivated when cellular immunity is depressed

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
581
Q

How does CMV vary due to the host’s immune system?

A

CMV cause an asymptomatic or mononucleosis-like infection in healthy individuals but devastating systemic infections in neonates and in immunocompromised people, in whom the virus may infect many different cell types and tissues.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
582
Q

How do CMV-infected cells appear?

A

CMV-infected cells exhibit gigantism of both the entire cell and its nucleus, which typically contains a large inclusion surrounded by a clear halo “owl’s eye)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
583
Q

Transmission of CMV can occur be several mechanisms, depending on the age group affected. What are these?

A
  • Transplacental transmission, from a newly acquired or primary infection in a mother who does not have protective antibodies (congenital CMV)
  • Neonatal transmission, through cervical or vaginal secretions at birth, or later throough breat milk from a mother who has an active infection (perinatal CMV)
  • Transmission through saliva during preschool years, especially in day care centers. Toddlers so infected readily transmit the virus to their parents
  • Tramission by the genital route is the dominant mode after about 15 years of age. Spread may also occur via respiratory secreation and the fecel-oral route.
  • Iatrogenic transmission, at any age through organ transplants or blood transfusions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
584
Q

How does acute CMV infection affect the immune system?

A

It induces transient but severe immunosuppression. CMV can infect dendritic cells and impair antigen processing and the ability of dentritic cells to stimulate T lymphocuyes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
585
Q

How does CMV evade the immune system?

A
  • By downmodulating MHC class I and II molecules and by producing homologues of TNF receptor, IL-10, and MHC class I molecules.
  • Interestingly, CMV can evade NK cells by producing ligands that block activating receptors and class I-like proteins that engage inhibitory receptors.
  • thus, CMV both hides from and actively suppresses immune responses.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
586
Q

How do MCV infected cells appear?

A
  • They are strikingly enlarged, often to a diameter of 40μ, and show cellular and nuclear pleomorphism.
  • Prominent intranuclear basophilic inclusions spanning half the nuclear diameter are usually set off from the nuclear membrane by a clear halo.
  • Within the cytoplasm of infected cells, smaller basophilic inclusions can also be seen.
  • In the glandular organs, the parenchymal epithalial cells are infected; in the brain, the neurons; in the lungs, the alveolar macrophages and epithelial and endothelial cells; and in the kidneys, the tubular epithelial and glomerular endothelial cells.
  • Disseminated CMV causes focal necrosis with minimal inflammation in virtually any organ.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
587
Q

How does congential CMV present?

A
  • Infection acquired in utero may take many forms.
  • Inapproximately 95% of cases it is asymptomatic.
  • However, sometimes when the virus is acquired from a mother with primary infection (who does not have protective antibodies), classic cytomegalic inclusion disease develops.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
588
Q

How does cytomegalic inclusion disease present in utero? How is diagnosis of neonatal CMV made?

A
  • It resembles erythroblastosis fetalis.
  • Affected infants may suffer IUGR, and present with jaundice, hepatosplenomegaly, anaemia, bleeding due to thrombocytopaenia, and encephalitis.
  • In fatal cases, the brain is often smaller than normal (microcephaly) and may show foci of calcification
  • Diagnosis of neonatal CMV is made by viral culture or PCR amplification of viral DNA in urine or saliva
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
589
Q

What are the effects for an infant who had cytomegalic inclusion disease?

A
  • The infants who survive usually have permanent deficits, including intellectual disability, hearing loss, and other neurologic impairments.
  • The congenital infection is not always devastating, however, and may take the form of interstitial pneumonitis, hepatitis, or a haematologic disoder
  • Most infants with this milder form recover, although a few develop intelectual disability later
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
590
Q

How does perinatal infection of CMV usually present?

A
  • Infection acquired during passage through the birth canal or from breast milk is usually asymptomatic due to protective maternal anti-CMV antibodies, which are transmitted to the fetus across the placenta.
  • Despite the lack of symptoms, many of these infants continue to excrete CMV in their urine or saliva for months to years.
  • Subtle effects on hearing and intelligence later in life have been reported in some studies
  • Much less commonly, infected infant develop interstitial pneumonitis, failure to thrive, rash or hepatitis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
591
Q

What is the most common clinical manifestation of CMV infection in immunocompetent hosts beyond the neonatal period?

A

In healthy young children and adults the disease is nearly always asymptomatic, 50-100% of adults have had previous exposure. If there are symptoms, most commonly they are an infectious mononucleosis-like illness, with fever, atypical lymphocytosis, lymphadenopathy and hepatitis, marked by hepatomegaly and abnormal liver function tests.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
592
Q

What organs does CMV primarily affect in immunosuppressed patients? What happens in these organs

A
  • The lungs (pneumonitis) and GI (colitis)
  • In the pulmonary infection an interstitial mononuclear infiltrate with foci of necrosis develops, accompanied by the typical enlarged cells with inclusions. The pneumonitis can progress to full-blown ARDS.
  • Intestinal necrosis and ulceration can develop and be extensive, leading to the formation of pseudomembranes and debilitating diarrhoea.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
593
Q

What are transforming viral infections? Name some

A
  • Some viruses can transform infected cells into benign or malignant tumour cells.
  • Oncogenic viruses can stimulate cell growth and survival by a variety of mechanisms.
  • Examples include EBV, HPV, HBV.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
594
Q

What does EBV cause?

A

EBV causes infectious mononucleosis, a benign, self-limited lymphoproliferative disorder, and is associated with the pahtogenesis of several human tumours, most commonly certain lymphomas and nasopharyngeal carcinomas.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
595
Q

What are the symptoms of infectious mononucleiosis?

A

It is characterised by fever, sore throat, generalised lymphadenopathy, splenomegaly, and the appearace in the blood of atypical activated T lymphocytes (mononucleiosis cells).
Some people develop hepatitis, meningoencephalitis, and pneumonitis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
596
Q

In what age group does EBV usually occur?

A

Infectious mononucleiosis occurs principally in late adolescents or young adults among upper socioeconomic classes in developed nations.
In the rest of the wold, primary infection with EBV occurs in childhood and is usually asymptomatic.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
597
Q

How is EBV transmitted?

A

By close human contact, frequently through the saliva during kissing.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
598
Q

How does EBV infect the body?

A
  • EBV infects B cells and possibly epithelial cells of the oropharynx.
  • An EBV envelope glycoprotein binds CD21 (CR2), the receptor for C3d component of complement, which is present on B cells.
  • In a minority of B cells, infection is lytic, leading to viral replication and eventual cell lysis accompanied by release of virions, which may infect other B cells.
  • In most B cells however, EBV establishes latent infection, during which the virus persists as an extrachromosomal episome.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
599
Q

A small number of EBV-encoded proteins are believed to be particularly important in the establishment of latency. What are they and what do they do?

A
  • Epstein-Barr nucleur antigen 1 (EBNA1) binds the EBV genome to host cell chromosomes during mitosis, thereby ensuring that viral episomes are partitioned evenly to daughter cells when infected cells divide
  • Latent membrane protein 1(LMP1) drives B-cell activation and proliferation. LMP1 does so by mimicking a constitutively active form of CD40, a B cell surface receptor. Like activating CD40, LMP1 binds to TNF receptor-assocated factors, adaptor molecules that trigger downstream events that activate NF-κB and the JAK/STAT signaling pathway. In addition, LMP1 prevents apoptosis by activating Bcl-2.
  • EBNA2 also promotes B-cell activation and replication. It turns on the transcription of several host cell genes, inclduing genes that encode proteins that drive cell cycle antry, such as cyclin D.
  • EBV produces a homologue of IL-10 (vIL-10), which inhibits its amcrophages and dendritic cells and suppresses anti-viral T cell responses.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
600
Q

What happens as a result of the actions of the EBV proteins?

A
  • B cells that are latently infected with EBV are activated and begin to proliferate and to disseminate.
  • This uncontrolled, expanding polyclonal population of EBV-infected B cells secretes antibodies with many specificities, inclduing antibodies that recognise sheep or horse red cells.
  • These so-called heterophile antibodies are detected in diagnostic tests for mononucleosis.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
601
Q

Where in the body is EBV shed?

A
  • In the saliva.
  • It is not known whether the source of the virus is B cells, oropharyngeal epithelial cells, or both
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
602
Q

The symptoms of infectious mononucleosis appear uopn initiation of the host immune response. What happens during this response on a cellular level?

A
  • Cellular immunity mediated by EBV-specific CD8+ cytotoxic T cells and CD16+ NK cells is the most important component of this response.
  • The reactive proliferation of T cells is largely centered in lymphoid tissues, which accounts for the lymphadenopathy and splenomegaly.
  • Early in the course of the infection, IgM antibodies are formed against viral capsid antigens; later IgG antibodies are formed that persist for life.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
603
Q

How does the response to EBV vary from healthy to immunosupressed people?

A
  • In healthy people, the fully developed humoral and cellular responses to EBV act as brakes on viral shedding, resulting in the elimination of B cells expressing the full complement of EBV latency-associated genes.
  • In hosts with acquired defects in cellular immunity, reactivation of EBV can lead to B-cell proliferation, which can progress through a multistep process to EBV-associated B-cell lymphomas.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
604
Q

EBV contributes to the development of which lymphoma? How?

A

It contributes to the development of some cases of Burkitt lymphoma, in which a chromosomal translocation (most commonly an 8:14 translocation) involving the MYC oncogene is the critical ongongenic event.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
605
Q

What are the morphological changes in EBV?

A
  • The peripheral blood shows absolute lymphocytosis. Many of these are large, atypical lymphocytes, characterised by an abundant cytoplasm containing multiple clear vacuolations, an oval, indented, or folded nucleus, and scattered cytoplasmic azurophilic granules.
  • The lymph nodes, histologically, have expansion of paracortical areas due to activation of T cells (immunoblasts). A minor population of EBV-infected B cells can also be detected in the paracortex.
  • EBV-infected B cells resembling Reed-Sternberg cells may be found.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
606
Q

What are the physical and histological changes in the spleen in EBV?

A
  • The spleen in enlarged in most cases. It is usually soft and fleshy, which a hyperemic cut surface
  • The histological changes are an expansion of white pulp follicles and red pulp sinusoids due to the presence of numerous activated T cells.
  • These spleen are especially vulnerable to rupture, possibly in part because the rapid increase in size produces a tense, fragile splenic capsule.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
607
Q

What are the physical and histological changes to the liver in EBV?

A
  • Heptamegaly is at most moderate.
  • On histological examination, atypical lymphocytes are seen in the portal areas and sinusoids, and scattered, isolated cells or foci of parenchymal necrosis may be present.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
608
Q

What are the clinical features of EBV?

A
  • In young children is classically presents with fever, sore throat, lymphadenitis, and other features mentioned earlier.
  • However, malaise, fatigue and lymphadenopathy are the common presentation in young adults with infectious mononucleiosis and can raise the specter of leukaemia or lymphoma
  • EBV can also prsent as a fever of unknown origin without significant lymphadenopathy or other localised findings, hepatitis resembling one of the hepatotropic viral syndromes, or a febrile rash resembling rubella.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
609
Q

What does the diagnosis of infectious mononucleiosis involve?

A

The diagnosis depends on:
1) lymphocytosis with the characteristic atypical lymphocytes in the peripheral blood
2) a positive heterophile antibody reaction (Monospot test)
3) a rising titer of specific antibodies for EBV antigens (viral capsid antigens, ealry antigens, or Epstein-Barr nuclear angiten)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
610
Q

In most patients, infectious mononucleosis resolves within 4-6 weeks, but sometimes the fatigue lasts longer. One of more complications ocassionally supervene. What are these?

A
  • Most common is marked hepatic dysfunction with jaundice, elevated hepatic enzyme levels, disturbed appetite, and rarely, even liver failure
  • Other complications involve the nervous system, kidneys, bone marrow, lungs, eyes, heart and spleen.
  • Splenic rupture can occur even with minor trauma, leading to haemorrhage that may be fatal.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
611
Q

What are the common conditions caused by staphylococcal aureus?

A
  • Skin lesions (boils, carbuncles, impetigo, and scalded-skin syndrome)
  • Abscesses
  • Sepsis
  • Osteomyelitis
  • Pneumonia
  • Endocarditis
  • Food poisoning
  • Toxic shock syndrome
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
612
Q

What type of bacteria are staph aureus?

A
  • Pyogenic, gram-positive cocci that form clusters resembling bunches of grapes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
613
Q

What type of infections does staph. epidermidis cause?

A

Opportunistic infections in:
* catheterised patients
* patients with prosthetic cardiac valves
* drug addicts

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
614
Q

What is the pathogenesis of staph aureus infections?

A
  • It produces a mulitude of virulence factors, which include surface proteins involved in adherence and evasion of the host immune response, secreted by enzymes that degrade host structures, secreted toxins that damage host cells, and proteins that cause antibiotics resistance.
  • S. aureus expresses surface receptors for fibrinogen (called clumping factor), fibronectin, and vitronectin and use these molecules to find to host endothelial cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
615
Q

How do staphylococci infecting prosthetic valves and catheters do that?

A

They have a polysaccharide capsule that allows them to attach to artificial materials and resist host cell phagocytosis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
616
Q

Staphylococco have their surface protein A. What does this do?

A

It binds the Fc portion of immunoglobulins, allowing the organism to escape antibody-mediated killing.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
617
Q

S. aureus produces multiple membrane-damaging (haemolytic) toxins. What are they and what do they do?

A
  • α-toxin, a protein that intercalates into the plasma membrane of host cells, forming pores that allow toxic levels of calcium to leak into cells
  • β-toxin, a sphingomyelinase
  • δ-toxin, which is a detergent-like peptide.
  • Staphylococcal γ-toxin and leukocidin lyse red cells and phagocytes, respectively
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
618
Q

How do the exfoliative A and B toxins produced by S.aureus work?

A
  • They are serine proteases that cleave the desmosomal protein desmoglein 1, which holds epidermal cells tightly together.
  • This causes keratinocytes to detach from one another and from the underlying basement membrane, resulting in a loss of barrier function that often leads to secondary skin infections
  • Exfoliation may occur locally at the site of infection (bullous impetigo) or may result in widespread loss of superficial epidermis (staphycoccal scalded-skin syndrome).
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
619
Q

Superantigens produced by S.aureus may cause what?

A

Food poisoning and toxic shock syndrome.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
620
Q

What is the pathophysiology of toxic shock syndrome?

A
  • It is often caused by hyperabsorbent tampons, which become colonised with S.aureus during use. It can also be caused by strep. pyogenes.
  • The most common sites are the vagina and infected surgical sites.
  • Bacterial superantigens cause polyclonal T cell proliferation by binding to conserved portions of MHC molecules and to relatively conserved portions of T-cell receptor β chains.
  • In this manner, superantigens may stimulate up to 20% of T lymphocytes, leading to release of TNF and IL-1 in such large amounts that they trigger the inflmmatory response.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
621
Q

What are the clinical features of toxic shock syndrome?

A
  • It is characterised by hypotension, renal failure, coagulopathy, liver disease, respiratory distress, a generalised erythematous rash, and soft tissue necrosis at the site of infection.
  • If not treated promptly, it can be fatal.
  • Superantigens produced by S.aureus also cause vomiting, presumably by affecting the CNS or enteric nervous system
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
622
Q

What is the morphology of S.aureus infection?

A

Where the lesion is located in the skin, lungs, bones or heart valves, S.aureus causes pyogenic inflammation that is distinctive for its local destruction of host tissue.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
623
Q

Where do staphylococcal skin infections begin?

A

Excluding impetigo, which is restricted to the superficial epidermis, staphylococcal skin infections are centered around the hair follicles where they begin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
624
Q

What is a furuncle or boil in association with S.aureus? Where and how do they happen?

A
  • A focal suppurative inflammation of the skin and subcutaneous tissue.
  • They may be soliatry or multiple or recur in successive crops.
  • Furuncles are most frequent is moist, hairy areas, such as the face, axillae, groin, legs and submammary folds.
  • Beginning in a single hair follicle, a boil develops into a growing and deepening abscess that eventually ‘comes to a head’ by thinning and rupturing the overlying skin.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
625
Q

What is a carbuncle associated with S.aureus? Where and how do they happen?

A
  • A carbuncle is a deeper suppurative infection that spreads laterally beneath the deep subcutaneous fascia and then burrow superficially to erupt in multiple adjacent skin sinuses
  • Carbuncles typically appear beneath the skin of the upper back and posterior neck, where fascial planes favor their spread
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
626
Q

What is hidradenitis?

A

It is a chronic suppurative infection of apocrine glands, most often in the axilla. Caused by S.aureus

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
627
Q

What is the morphology of S.aureus lung infections? What normally causes them

A
  • They have a polymorphonuclear infiltrate similar to that of S.pneumoniae infections, but cause much more tissue destruction.
  • They usually arise from a haematogenous source, such as an infected thrombus, or in the setting of a predisposing condition such as infleunza.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
628
Q

What is staphylococcal scalded-skin syndrome? Who does it happen in and how does it happen?

A
  • It occurs most frequently in children with S.aureus infection of the nasopharynx or skin.
  • There is a sunburn-like rash that spreads over the entire body and evolves into fragile bullae that lead to partial or total skin loss.
  • The desquamation of the epidermis in staphylococcal scalded-skin syndrome occurs at the level of the granulosa layer, distinguishing it from the toxic epidermal necrolysis, or Lyell disease, which is secondary to drug hypersensitivity and causes desquamation at the level of the epidermal-dermal junction.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
629
Q

What are some species that cause respiratory diseases and what diseases do the often cause?

A
  • Streptococcus pyogenes - pharyngitis
  • Corynebacterium diphtheria - diphtheria
  • Bordatella pertussis - pertussis
  • Streptococcus pnuemonia - lobar pneumonia
  • Mycobacterium tuberculosis - TB
  • Legionella pneumophila - Legionnaire disease
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
630
Q

What are some species that cause gastrointestinal diseases and what diseases do the often cause?

A
  • Helicobacter pylori - peptic ulcers
  • Vibrio cholerae, enterotoxigenic E.coli - non-inflammatory gastroenteritis
  • Shigella, Salmonella, Campylobacter, enterohaemorrhagic E.coli - Inflammatory gastroenteritis
  • Salmonella typhi - enteric (typhoid) fever
  • Clostridium difficile - pseudomembranous colitis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
631
Q

What are some species that cause nervous system diseases and what diseases do the often cause?

A
  • Neisseria meningitidis, Streptococcus pneumonia, Haemophilus influenza, Listeria monocytogenes - acute meningitis
  • Clostridium tetani, Clostridium botulinum - paralytic intoxications, tetanus and botulism
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
632
Q

What are some species that cause urogenital diseases and what diseases do the often cause?

A
  • Escherichia coli, Pseudomonas aeruginosa, Entercoccus species - UTIs
  • Neisseria gonorrhoeae - gonorrhea
  • Chlamydia trachomatis - chlamydia
  • Treponema pallidum - syphilis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
633
Q

What are some species that cause skin and adjacent soft tissue diseases and what diseases do the often cause?

A
  • Staphylococcus aureus - abscess, cellulitis
  • Streptococcus pyogenes - impetigo, erysipela, necrotising fasciitis
  • Clostridium perfringens - gas gangrene
  • Bacillus anthracis - cutaneous anthrax
  • Pseudomonas aeruginosa - burn infections
  • Mycobacterium leprae - leprosy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
634
Q

What are some species that cause disseminated infections and what diseases do the often cause?

A
  • Yersinia pestis - plague
  • Borrelia burgdorferi - lyme disease
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
635
Q

What are some species that cause disseminated neonatal infection and what diseases do the often cause?

A
  • Streptococcus agalactiae, Listeria monocytogenes - neonatal bacteraemia, meningitis
  • Treponema pallidum - congenital syphilis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
636
Q

What infections do Streptococci cause?

A
  • Suppurative infections of the skin, oropharynx, lungs and heart valves
  • Postinfectious syndromes, including rheumatic fever, immune complex glomerulonephritis, and erythema nodosum.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
637
Q

What type of bacteria are Streptococci?

A

They are gram-positive cocci that grow in pairs or chains.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
638
Q

β-haemolytic streptococci are typed according to their surface carbohydrate (Lancefield) antigens. What are the different types of β-haemolytic streptococci and what do they cause?

A
  • Strep. pyogenes (group A) causes pharyngitis, scarlet fever, erysipelas, impetigo, rheumatic fever, toxic shock syndrome, and glomerulonephritis.
  • Strep. agalactiae (group B) colonises the female genital tract and causes sepsis and meningitis in neonates and chorioamnionitis in pregnancy.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
639
Q

What is the most important α-haemolytic stretococcus? What does it cause?

A

Strep. pneumoniae is a common cause of CAP in older adults and meningitis in children and adults.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
640
Q
A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
641
Q

What does Strep. mutans cause?

A

Dental caries

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
642
Q

What type of bacteria are enterococci? What do they cause

A
  • Enterococci are gram-positive cocci that grow in chains.
  • They are often resistant to commonly used antibiotics
  • They are a significant cause of endocarditis and urinary tract infections.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
643
Q

What is the pathogenesis of Strep. pyogenes?

A
  • Expresses M protein, a surface protein that prevents bacteria from being phagocytosed
  • Expresses a complement 5a peptidase that degrades degrades this chemotactic peptide
  • It secretes a phage-encoded pyrogenic exotoxin that causes fever and rash in scarlet fever
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
644
Q

What do S.pyogenes, S.agalactiae, and S.pneumoniae all have that allows them to resist phagocytosis?

A

They have capsules that resist phagocytosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
645
Q

How is post-streptococcal acute rheumatic fever cause?

A

By antistreptococcal M protein antibodies and T cells that cross-react with cardiac proteins

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
646
Q

Although the antiphagocytic capsule is the most important virulence factor of S.pneumoniae, what other virulence factor does it produce?

A

Pneumolysin, a toxin that inserts into host cell membranes and lyses cells, greatly increasing tissue damage

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
647
Q

How does S.mutans produce caries?

A

By metabolising sucrose to lactic acid (which causes demineralisation of tooth enamel) and by secreting high-molecular-weight glucans that promote aggregation of bacteria and plaque formation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
648
Q

Enterococci are low-virulence bacteria, although they do have an antiphagocytic capsule and produce enzymes that injur host cells, what is the primary cause of the emergence of enterococci?

A

Their resistance to antibiotics

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
649
Q

What is the morphology of streptococcal infections?

A

They are characterised by diffuse interstitial neutrophilic infiltrates with minimal destruction of host tissues.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
650
Q

What causes Erysipelas? How does it present both clincically and histologically?

A
  • It is caused by exotoxins from superficial infection with S.pyogenes
  • Characterised by rapidly spreading erythematous cutaneous swelling that may begin on the face, or less frequently, on the body or an extremity. The rash has a sharp, well-demarcated, serpiginous border and may form a ‘butterfly’ distribution on the face.
  • Histologically, there is a diffuse, oedematous, neutrophilic inflammatory reaction in the dermis and epidermis extending into the subcutaneous tissues. Microabscesses may be formed, but tissue necrosis is usually minor.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
651
Q

What are the symptoms of streptococcal pharyngitis?

A
  • Oedema, epiglottic swelling, and punctate abscesses of the tonsillar crypts, sometimes accompanied by cervical lymphadenopathy.
  • Swelling associated with severe pharyngeal infection may encroach on the airways, especially if there is peritonsillar or retropharyngeal abscess formation.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
652
Q

What bacteria causes scarlet fever? How does it present?

A
  • Associated with pharyngitis cause by S.pyogenes.
  • Most common between the ages of 3 and 15 years
  • Manifested y a punctate erythematous rash that is most prominent over the trunk and inner aspects of the arms and legs.
  • The face is also involved, but usually a small area about the mouth remains relatively unaffected, producing circumoral pallor.
  • They skin usually becomes hyperkeratotic and scaly during defervescence.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
653
Q

What is the bacteria that causes Diphtheria? How is it spread?

A

It is caused by Corynebacterium diphtheriae, a slender gram-positive rod with clubbed ends that spreads from person to person in respiratory droplets or skin exudate.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
654
Q

What symptoms does diphtheria infection cause?

A
  • Respiratory diphtheria causes pharyngeal, or less often, nasal or laryngeal infection.
  • There is toxin-mediated formation of a gray pharyngeal membrane, and damage to the heart, nerves, and other organs.
  • Cutaneous diphtheria causes chronic ulcers with a dirty gray membrane, but does not cause systemic damage.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
655
Q

What is the pathogenesis of diphtheria?

A
  • C. diphtheriae produces a phage-encoded A-B toxin that blocks host cell protein synthesis.
  • The A fragment does this by catalysing the covalent transfer of ADP–ribose to elongation factor-2 (EF-2).
  • This inhibits EF-2 function, which is required for the translation of mRNA into protein.
  • A single molecule of diphtheria toxin can kill a cell by ADP-ribosylating, and thereby inactivating, more than a million EF-2 molecules.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
656
Q

How does immunisation with diphtheria toxoid work?

A

Immunisation with diphtheria toxoid (formalin-fixed toxin) stimualtes production of toxin-neutralising antibodies that protect people from the lethal effects of the toxin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
657
Q

What is the morphology of C.diphtheriae?

A
  • Inhaled C.diphtheriae carried in respiratory droplets proliferate at the side of attachment on the mucosa of the nasopharynx, oropharynx, larynx, or trachea. The bacteria also form satellite lesions in the oesophagus or lower airways.
  • Release of exotoxin causes necrosis of the epithelium, accompanied by an outpouring of a dense fibrinosuppurative exudate. The coagulaton of this exudate on the ulcerated necrotic surface created a tough, dirty gray to black, superficial membrane, sometimes called pseudo-membrane because it is not formed by viable tissue.
  • There is an intense neutrophilic infiltration with marked vascular congestion, interstitial edema and fribin exudation.
  • When the membrane sloughs off its inflamed and vascularised bed, bleeding and asphyxiation may occur.
  • With control of the infection, the membrane is coughed up or removed by enzymatic digestion, and the inflammatory reaction subsides.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
658
Q

Although the bacterial invasion of diphtheriae remains localised, what happens with entry of exotoxin into the blood adn its systemic distribution?

A

There may be a fatty change in the myocardium with isolated myofiber necrosis, polyneuritis with degeneration of the myelin sheaths and axis cylinders, and (less commonly) fatty change and focal necroses of parenchymal cells in the liver, kidneys and adrenals.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
659
Q

What type of bacteria causes cholera?

A

Vibrio cholerae are comma-shaped, gram-negative bacteria, that cause cholera, a disease that has been endemic in the Ganges Valley of India and Bangladesh for almost all of recorded history

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
660
Q

There is a marked seasonal variation in the incidence of cholera in most climates. Why?

A

Due to radpi growth of Vibrio bacteria at warm temperatures.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
661
Q

Where do people get cholera from?

A

While the bacteria can be present in food, the infection is primarily transmitted by contaminated drinking water.
Thus, cholera can become rampant in areas devastated by natural or man-made disasters, such as earthquakes or war, that threaten sewage systems and drinking water supplied.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
662
Q

What is the pathogenesis of cholera?

A
  • Cholera toxin is composed of five B subunits and a single A subunit.
  • The B subunit binds GM1 ganglioside on the surface of intestinal epithelial cells, and is carried by endocytosis to the endoplasmic reticulum, a process called retrograde transport.
  • Here, the A subunit is reduced by protein disulfide isomerase, and a fragment of the A subunit is unfolded and released.
  • This peptide fragment is transported into the cytosol using host cell machinery that moves misfolded proteins from the endoplasmic reticulum to the cytosol. The A subnit refolds to void degradation.
  • The refolded A subunit peptide then interacts with cytosolic ADP ribosylation factors (ARFs) to ribosylate and activate the stimulatory G protein Gsα, which stimulates adenylate cyclase and the resulting cAMP opens the cystic fibrosis transmembrane conductance regulator, CFTR, which releases chloride ions into the lumen.
  • Chloride and sodium absoprtion are also inhibited by cAMP.
  • The resulting accumulation accumulation of chloride, bicarbonate, and sodium within the lumen creates an osmotic driving force that draws water into the lumen and causes massive diarrhoea
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
663
Q

What are the clinical features of cholera?

A
  • Most individuals exposed are asymptomatic or develop only mild diarrhoea.
  • In those with severe disease there is an abrupt onset of watery diarrhoea and vomiting following an incubation period of 1 to 5 days.
  • The voluminous stools resemble rice water and are sometimes described as having a fishy odor.
  • The rate of diarrhoea may reach 1L per hour, leading to dehydration, hypotension, muscular cramping, anuria, shock, LOC and death.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
664
Q

What is the most common bacterial enteric pathogen in developed countries? What do you get it from?

A

Campylobacteri jejuni is an important cause of traveler’s diarrhoea
* Most infections are associated with ingestion of improperly cooked chicken, but outbreaks can also be caused by unpasteurised mild or contaminated water

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
665
Q

The pathogenesis of Campylobacter infection remains poorly defined, but what are the four major properties that contribute to virulence?

A
  • Motility - flagella allow campylobacter to be motile
  • Adherence - the motility facilitates adherence and colonisation, which are necessary for mucosal invasion.
  • Cytotoxins that cause epithelial damage and a cholera toxin-like enterotoxin are also released by some C.jejuni isolates.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
666
Q

With Campylobacter infection, when does dysentry and enteric fever occur?

A
  • Dysentry, i.e. blood diarrhoea, is generally associated with invasion and only occurs with a small minority of Campylobacter strains.
  • Enteric fever occurs when bacteria proliferate within the lamina propria and mesenteric lymph nodes.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
667
Q

What are some extra-intestinal complications of Campylobacter?

A
  • Campylobacter infection can result in reactive arthritis, primarily in patients with HLA-B27.
  • Erythema nodosum and Guillain-Barre syndome.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
668
Q

What is the morphology of Campylobacter and how is it diagnosed?

A
  • Diagnosis is primarily by stool culture, since biopsy findings are nonspecific, and reveal acute self-limited colitis with features ocmmon to many forms of infectious colitis.
  • Mucosal and intraepithelial neutrophil infiltrates are prominent, particularly within the superficial mucosa; cryptitis (neutrophil infiltration of the crypts) and crypt abscesses may also be present.
  • Importantly, crypt architecture is preserved, although this can be difficult to assess in cases with severe mucosal damage
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
669
Q

What are the clinical features and inbucation period of campylobacter?

A
  • Incubation period is up to 8 days
  • Watery diarrhoea, either acute of following an influenza-like prodrome, is the primary symptom, but dysentery develops in 15% of adults and more than 50% of children.
  • Patients may shed bacteria for 1 month or more after clinical resolution.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
670
Q

What type of bacteria are shigella?

A

Shigella are gram-negative unencapsulated, nonmotile, facultative anaerobes that belong to teh enterobacteriaceae family and are closely related to enteroinvasive E.coli.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
671
Q

How is shigella transmitted? What populations are most affected?

A
  • By the faecal-oral route or via contaminated water and food.
  • In the US and Europe, children in daycare, migrant workers, travelers, and those in nursing homes are more commonly affected.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
672
Q

What is the pathogenesis of Shigella?

A
  • Shigella are resistant to the harsh acidic environment of the stomach, thereby explaining teh extremely low infective dose
  • Once in the intestine, organisms are taken up by M, or microfold cells. There are epithelial cells, which are specialised for sampling and presentation of luminal antigens.
  • Shigella proliferate intracellularly, escape into the lamina propria, and are phagocytosed by macrophages, in which they induce apoptosis.
  • The ensuing inflammatory response damages surface epithelia and allows shigella within the intestinal lumen to gain access to the basolateral membranes of colonic epithelial cells, which is the preferred domain for invasion.
  • All Shigella species carry virulence plasmids, some of which encode a type III secretion system capable of directly injecting bacterial proteins into the host cytoplasm.
  • Shigella. dysenteriae serotype 1 also release the Shiga toxin Stx, which inhibits eukaryotic protein synthesis, resulting in host cell damage and death.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
673
Q

Where in the colon are Shigella infections most prominent and why?

A

In the left colon, but the ileum may also be involved, perhaps reflecting the abundance of M cells in the dome epithelium over the Peyer patches.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
674
Q

What is the histology of Shigella?

A

The mucose is haemorrhagic and ulcerated, and pseudomembranes may be present
* The histology of early cases is similar to other acute self-limiting colitides, such as Campylobacter, but because of the tropism for M cells, apthous-appearing ulcers similar to those seen in Crohn disease may occur

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
675
Q

What are the clinical features of Shigella, including the incubation period?

A
  • After an incubation period of up to 1 week, Shigella causes self-limiting disease characterised by about 1 week fo diarrhoea, fever and abdominal pain
  • The initially watery diarrhoea progresses to a dysenteric phase in approx 50% of patients, and constitutional symptoms can persist for as long as 1 month.
  • The subacute presentation that develops in a minority of patients is characterised by several weeks of waxing and waning diarrhoea that can mimic new-onset ulcerative colitis.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
676
Q

What are the extra-intestinal complications of shigella?

A
  • They are uncommon and include a triad of sterilr reactive arthritis, urethritis and conjunctivitis that preferentially affects HLA-B27 positive men between 20 and 40 years of age.
  • Haemolytic-uraemic syndrome, may also occur after infection with serotype 1 infection that secretes Shiga toxin.
  • Toxic megacolon and intestinal obstruction are uncommon complications
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
677
Q

How do antibiotics affect Shigella and Campylobacter?

A
  • No effect in Campylobacter
  • Shortens the clinical course and reduces the duration of organism shedding in stools in Shigella
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
678
Q

What type of bacteria is Salmonella? How are they divided

A

Salmonella, which are classified within then Entero-bacteriaceae family of gram-negative bacilli, are divided into Salmonella typhi, the causative agent of typhoid fever and the nontyphoid Salmonella, of which S.enteritidis is the most common.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
679
Q

What causes Salmonella?

A

Ingestion of contaminated food, particularly raw or udnercooked meat, poutlry, eggs, and milk.
Centralised food processing can lead to large outbreaks

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
680
Q

Can you get a vaccine for Salmonella?

A

Yes, and so can farm animals e.g. egg-laying hens

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
681
Q

What increases your risk of Salmonella?

A

Very few viable Salmonella are necessary to cause infection, and the absence of gastric acid, in individuals with atrophic gastritis or those on acid-suppressive therapy, further reduces the required inoculum.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
682
Q

What is the pathogenesis of Salmonella?

A
  • Salmonella possess virulence genes that encode a type III secretion system capable of transferring bacterial proteins into M cells and enterocytes.
  • The transferred proteins activate host Rho GTPases, thereby triggering actin rearrangement and bacterial endocytosis which, in turn, allows bacterial growth within endosomes.
  • In addition, flagellin, the core protein of bacterial flagellae, activates TLR5 on host cells and icnreases the local inflammatory response.
  • Similarly, bacterial lipopolysaccharide activates TLR4, although some Salmonella strains express a virulence factor that prevents TLR4 activation.
  • Salmonella also secrete a molecule that induces epithelial cell release of the eicosanoid hepoxilin A3, thereby drawing neutrophils into the intestinal lumen and potentiaing mucosal damage
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
683
Q

What are the clinical features on Salmonella?

A
  • They are clinically indistinguishable from other enteric pathogens, and symptoms range from loose stools to cholera-like profuse diarrhoea to dysentery.
  • Fever often resolves within 2 days, but diarrhoea can persist for a week and organisms can be shed in the stool for several weeks after resolution.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
684
Q

Are antibiotics recommended in Salmonella?

A

Antibiotics therapy is not recommended in uncomplicated cases because it can prolong the carrier state or even cause relapse and does not typically shorten the duration of diarrhoea.
Most salmonella infections are self-limited, but deaths do occur.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
685
Q

What bacteria causes typhoid fever?

A
  • It is caused by Salmonella enterica and its two subtypes, typhi and paratyphi.
  • The majority of cases in endemic countries are due to S.typhi, while infection by S.paratyphi is more common among travelers, perhaps because travelers tend to be vaccinated against S.typhi.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
686
Q

What countries are associated with typhoid?

A

India, Mexico, the Philippins, Pakistan, El Salvador, and Haiti.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
687
Q

How is S.typhi and S.paratyphi transmitted?

A

Humans are the sole reservoir for S.typhi and S. paratyphi and transmission occurs from person to person or via food or contaminated water.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
688
Q

What is the pathogenesis of S.typhi?

A
  • S.typhi are able to survive in gastric acid and, once in the small intestine, they are taken up by and invade M cells.
  • Bacteria are then engulfed by mononuclear cells in the underlying lymphoid tissue.
  • Unlike S.enteritidis, S.typhi can then disseminate via lymphatic and blood vessels.
  • This causes reactive hyperplasia of phagocytes and lymphoid tissues throughout the body.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
689
Q

What is the morphology of typhoid fever?

A
  • Infection causes Peyer patches in the terminal ileum to enlarge into sharp, delineated, plateau-like elevations up to 8cm in diameter.
  • Draining mesenteric lymph noces are also enlarged.
  • Neutrophils accumulate within the superficial lamina propria, and macrophages containing bacteria, red cells, and nuclear debris mix with lymphocytes and plasma cells in the lamina propria.
  • Mucosal damage creates oval ulcers, oriented along the axis of the ileum, that may perforate.
  • The draining lymph nodes also harbor organisms and are enlarged due to phagocyte accumulation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
690
Q

How does typhoid affect the liver and spleen morphologically?

A
  • The spleen is enlarged and soft, with uniformly pale red pulp, obliterated follicular markings, and prominent phagocyte hyperplasia.
  • The liver shows small, randomly scattered foci of parenchmal necrosis in which hepatocytes are replaced by macrophage aggregates, called typhoid nodules; such nodules may also develop in the bone marrow and lymph nodes.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
691
Q

What are the clinical features of typhoid?

A
  • Patients experience anorexia, abdominal pain, bloating, nausea, vomiting, and blood diarrhoea followed by a short asymptomatic phase that gives way to bacteraemia and fever with flu-like symptoms.
  • Patients have sustained high fevers and abdominal tenderness, that may mimic appendicitis
  • Rose spots, small erythematous maculopapular lesions, are seen on the chest and abdomen.
  • Symptoms abate after several weeks, although relapse can occur
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
692
Q

Do antibiotics help with typhoid?

A
  • Blood cultures are positive in more than 90% of affected individuals during the febrile phase.
  • Antibiotic treatment can prevent further disease progression.
  • In patients who do not receive antibiotics, the initial febrile phase continues for up to 2 weeks.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
693
Q

What is AIDS?

A

Acquired immunodeficiency syndrome is a disease causes by the retrovirus human immunodeficiency virus (HIV) and characterised by profound immunosuppression that leads to opportunistic infections, secondary neoplasms, and neurologic manifestations.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
694
Q

Epidemiological studies in the US have identified five groups of adults at high risk for developing AIDS. What are they?

A
  • Homosexual or bisexual men constitute the largest group, accounting for more than 50% of the reported cases.
  • IVDUs with no previous history of homosexuality are the next largest group, representing about 20% of infected indiviuals
  • Haemophiliacs, especially those who receive large amounts of factor VIII or factor IX concentrated before 1985
  • Recepients of blood and blood components who are not haemophiliacs but who received transufusions of HIV-infected whole blood or components account for 1% of patients
  • Heterosexual contacts of members of other high-risk groups (chiefly IVDUs), globally heterosexual transmission is most common mode by which HIV is spread - mainly female partners of male IVDUs.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
695
Q

The transmission of HIV occus under conditions that facilitate exchange of blood or bodily fluids containing the virus or virus-infected cells. What are the three major routes and tell me a bit about them?

A
  • Sexual transmission is clearly the dominant mode of infection worldwide, accounting for >75% of cases. Viral tramission occurs in two ways 1) direct inoculation into the blood vessels breached by trauma and 2) infection of dendritic cells or CD4+ cells within the mucosa.
  • Parenteral transmission occurs by sharing of needles and syringes with HIV-containing blood.
  • Mother-to-infant transmission is the major cause of paediatric AIDS. It can occur by three routes: 1) in utero by transplacental spread. 2) during delivery through an infected birth canal, and 3) after birth by ingestion of breast milk. Transmission during birth and in the immediate period after is the most common.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
696
Q

How do STIs affect HIV transmission? Why?

A

Sexual transmission of HIV is enhanced by coexisting STDs, especially those associated with genital ulceration - syphillips, herpes. Other STDs are also co-factors for transmission, perhaps because in these genital inflammatory states there is a greater concentration of the virus and virus-containing cells in genital fluids, as a result of increased numbers of inflammatory cells in the semen

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
697
Q

Transmission of HIV by transfusion of blood of blood products has been virtually eliminated by three public health measures. What are they?

A

1) Screening of donated blood and plasma for antibody to HIV
2) Stringent purity criteria for factor VIII and factor IX preperations
3) Screening of donors on the basis of history

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
698
Q

What type of virus is HIV?

A

A non-transforming human retrovirus belonging to the lentivirus family.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
699
Q

What is the structure of HIV?

A
  • The HIV-1 virion is spherical and contains an electron-dense, cone-shaped core surrounded by a lipid envelope derived from the host cell membrane
  • The virus core contains 1) the major capsid protein 24; 2) nucleocapsid protein p7/p9; 3) two copies of viral genomic RNA; and 4) the three viral enzymes (protease, reverse transcriptase, and integrase).
  • The viral core is surrounded by a matrix protein called p17, which lies underneath the virion envelope.
  • Studding the viral envelope are two viral glycoproteins, gp120 and gp41, which are critical for HIV infection of cells.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
700
Q

What does the HIV-1 RNA genoma contain?

A
  • The gag, pol and env genes, which are typical of retroviruses.
  • The products of the gag and pol genes are large precursor proteins that are cleaved by the viral protease to yield the mature proteins.
  • HIV contains several other accessory genes, including tat, rev, vif, nef, vpr and vpu, which regulate the synthesis and assembly of infectious viral particles and the pathogenicity of the virus
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
701
Q

Two genetically different but related forms of HIV have been isolated from patients with AIDS. What are these and where are they most common?

A
  • HIV-1 is the most common type associated with AIDS in the US, Europe and Central Africa
  • HIV-2 causes a similar disease principalle in West Africa and India
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
702
Q

HIV-1 can be divided into three subgroups. What are they? Tell me more about the most common one

A
  • Group M (major) viruses are the most common worldwide, and they are further divided into several subtypes designated A through K. Subtype B is the most common in Western Europe and the US.
  • Group O (outlier)
  • Group N (neither M nor O)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
703
Q

What is the broad pathogenesis of HIV and AIDs?

A
  • The two major targets of HIV infection are the immune system and the CNS.
  • Profound immune deficiency results chiefly from infection and subsequent loss of CD4+ T cells as well as the impairment in the function of surviving helper T cells. Macrophages and dendritic cells are also targets of HIV infection.
  • HIV enters thebody through mucosal tissues and blood and first infects T cells as well as dendritic cells and macrophages
  • The infection becomes established in lymphoid tissues, where the virus may remain latent for long periods.
  • Active viral replication is associated with more infection of cells and profression to AIDS.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
704
Q

What does the life cycle of HIV consist of?

A

Infection of cells, integration of the provirus into the host cell genome, activation of viral replication, and production and release of infectious virus.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
705
Q

How does the infection of cells by HIV occur?

A
  • HIV infects cells by using the CD4 molecule as a receptor and various chemokine receptors as corectpors.
  • The initial step of infection is the binding of gp120 envelope glycoprotein to CD4 molecules, which leads to a conformation change that results in the formation of a new recognition site on gp120 for the coreceptors CCR5 and CXCR4.
  • The conformational changes in gp41 result in the exposure of a hydrophobic region called the fusion peptide at the tip of gp41.
  • This peptide inserts into the cells membrane of the target cells, leading to fusion of the virus with the host cell.
  • After fusion, the virus core containing the HIV genome enters the cytoplasm of the cell
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
706
Q

How do chemokines effect HIV infection of cells in culture?

A

They sterically hinder it by occupying their receptors, and therefore, the level of chemokines in the tissues may influence the efficiency of viral infection in vivo

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
707
Q

Once HIV is internalised into the cells, the RNA genome of the virus undergoes reverse trascription, leading to the synthesis of double-stranded complementary DNA (cDNA; proviral DNA). What are the options of what can happen next?

A
  • In quiescent T cells, HIV cDNA may remain in the cytoplasm in a linear episomal form.
  • In dividing T cells, the cDNA circularises, enters the nucleus, and is then integrated into the host genome.
  • After this integration, the provirus may be silent for months of years, a form a latent infection.
  • Alternatively, proviral DNA may be transcribed, with the formation of complete viral particles that bud from the cell membrane. Such productive infection, when associated with extensive viral budding, leads to death of infected cells.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
708
Q

What type of T cells does HIV infect?

A

HIV infects memory and activated T cells but is ineffecient at productively infecting naive (unactivatived T cells).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
709
Q

Why can HIV not productively infect naive T cells? How does this change in activated T cells?

A
  • Naive T cells contain an active form of an enzyme that introduces mutations in the HIV genome.
  • This enzyme is called APOBEC3G. It is a cytidine deaminase that introduces cytosine-to-uracil mutations in the viral DNA that is produced by reverse transcription.
  • These mutations inhiti further DNA replication.
  • Activation of T cells converts cellular APOBEC3G into an inative, high-molecular-mass complex, which explains why the virus can replicate in previously activated (memory) T cells and T-cell lines.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
710
Q

How is HIV linked to NF-κB?

A
  • Activation of T cells upregulates several transcription factors, including NF-κB, which stimulate transcription of genes encoding cytokines such as IL-1
  • The long-terminal-repeat sequences that flank the HIV genome also contain NF-κB-binding sites that can be triggered by the same transcription factors.
  • Induction of NF-κB-in latently infected CD4+ cells activates the transcription of HIV proviral DNA and leads ultimately to the production of virions and to cell lysis.
  • It seems that HIV thrives when the host T cells and macrophages are physiologically activates, an act that can best be described as ‘subversion from within’
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
711
Q

How does infection effect HIV infection?

A
  • HIV-infected people are at increased risk for recurrent exposure to other infections, which lead to increased lymphocyte activation and production of proinflammatory cytokines.
  • These, in turn, stimulate more HIV production, loss of additional CD4+ T cells, and more infection.
  • A vicious cycle may be set up that cluminates in inexorable destruction of the immune system.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
712
Q

What is the main mechanism of T-cell depletion in HIV infection?

A
  • Loss of CD4+ T cells is mainly because of infection of the cells and direct cytopathic effects of the replicating virus.
  • Many infected cells may be in mucosal and other peripheral lymphoid organs and death of these cells is a major cause of the relentless, and eventually profound, cell loss
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
713
Q

In addition to firect killing of cells by the virus, other mechanisms may contribute to the loss of T cells. What are they?

A
  • Chronic activation of uninfected cells, responding to HIV itself or to infections that are common in individuals with AIDS, leads to apoptosis of these cells by the process of activation-induced cell death.
  • Non-cytopathic (abortive) HIV infection activates the inflammasome pathway and leads to a form of cell death called pyroptosis - inflammatory cytokines and cellular contents are released.
  • HIV infects cells in lymphoid organs (spleen, lymph nodes and tonsils) and may cause progressive destruction of the architecture and cellular composition of lymphoid tissues.
  • Loss of immature precursors of CD4+ T cells, either by direct infection of thymus progenitor cells or by infection of accessory cells that secrete cytokines.
  • Fusion of infected and uninfected cells with formation of giant cells
  • Defects in Th1-type responses relative to the Th2 type, defects in intracellular signaling.
  • Selective loss of memory CD4+ helper T cells.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
714
Q

In addition to infection and loss of CD4+ T cells, infection of macrophages and dendritic cells is also important in the pathogenesis of HIV infection. What are some aspects of HIV infection of macrophages?

A
  • HIV-1 can infect and multiply in terminally differentiated nondividing macrophages. THe Vpr protein allows nuclear targeting of the HIV preintegration complex through the nuclear pore.
  • Infected macrophages bud relatively small amounts of virus from the cell surface, but these cells contain large numbers of virus particles often located in intracellular vacuoles.
  • Macrophages are quire resistant to the cytopathic effects of HIV, so in late stages, when CD4+ T cell numbers are low, macrophages are an important site of continued viral replication.
  • They have functional defects such as impaired microbicidal activity, decreased chemotaxis, decreased secretion of IL-1, inappropriate secretion of TNF, and poor capacity to present antigens.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
715
Q

In addition to macrophages, two types of dendritic cells are also important targets for the initiation and maintenance of HIV infection. What are they and what do they do?

A
  • Mucosal dendritic cells are infected by the virus and may transport it to regional lymph nodes, where the virus is transmitted to CD4+ T cells
  • Dentritic cells also express a lectin-like receptor that specifically binds HIV and displays it in an intact, infectious form to T cells, thus promoting infection of T cells.
  • Follicular dendritic cells in the germinal centers of lymph nodes are potential reservoirs of HIV.They have receptors for the Fc portion of immunoglobulins and hence they trap HIV virions coated with anti-HIV antibodies. These virions retain their ability to infect CD4+ T cells as they traverse the intricate meshwork formed by the dendritic processes of the follicular dendritic cells.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
716
Q

How is the function of B cells affected by HIV infection?

A
  • There is polyclonal activation of B cells, resulting in germinal center B-cell hyperplasia, bone marrow plasmacytosis, hypergammaglobulinaemia, and formation of circulating immune complexes.
  • This activation may result from: reactivation of or reinfection with CMV and/or EBV, both of which are polyclonal B-cell activators; gp41 itself can promote B-cells growth and differentiation; HIV-infected macorphages produce increased amounts of IL-6, which stimulates proliferation of B cells.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
717
Q

What is the pathogenesis of CNS involvement in HIV infection?

A
  • Macrophages and microglia are the predominant cell types in the brain that are infected with HIV
  • The neurological deficit is likely caused indirectly by viral products and by soluble factors produced by infected microglia, e.g. IL-1, TNF and IL-6
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
718
Q

What is the natural history of HIV infection?

A
  • Virus typically enters through mucosal epithelia
  • An acute retroviral syndrome
  • A middle, chronic phase, in which most individuals are asymptomatic
  • Clinical AIDS
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
719
Q

What happens during the acute infection of HIV?

A
  • Acute infection is characterised by infection of memory CD4+ T cells (which express CCR5) in mucosal lymphoid tissues, and death of many infected cells.
  • Mucosal infection is often associated with damage to the epithelium, defects in mucosal barrier functions, and translocation of microbes across the epithelium.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
720
Q

Mucosal infection is followed by dissemination of the virus and the development of host immune responses. How does this happen?

A
  • Dendritic cells in epithelia at sites of virus entry capture the virus and then migrate into the lymph nodes.
  • Once in lymphoid tissue, dentritic cells may pass HIV on to CD4+ T cells through direct cell-t-cell contact.
  • Within days after the first exposure to HIV, viral replication can be detected in the lymph nodes
  • This replication leads to viremia. The virus disseminates throughout the body and infects helper T cells, macrophages, and dendritic cells in peripheral lymphoid tissues.
  • As the infection. spreads, the individual mounts antiviral humoral and cell-mediated immune responses evidenced by seroconversion and by the development of virus-specific CD8+ cytotoxic T cells.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
721
Q

What immune responses to HIV infection occur at what time?

A
  • A cytotoxic T lymphocyte response to HIV is detectable by 2-3 weeks after the initial ifnection, and it peaks by 9-12 weeks.
  • Marked expansion of virus-specific CD8+ T cell clones occurs during this time, and up to 1-% of patients CTLs may be specific at 12 weeks.
  • The humoral immune response to HIV peaks at about 12 weeks.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
722
Q

What is the acute retroviral syndrome in HIV infection?

A
  • It is the clinical presentation of the initial spread of the virus and the host response.
  • 40-90% if individuals who acquire a primary infection develop this syndrome
  • It typically occurs 3-6 weeks after infection, and resolves spontaneously in 2-4 weeks.
  • Clinically, this phase is associated with a self-limited acute illness with nonspecific symptoms, including sore throat, myalgias, fever, weight loss and fatigue, resembling a flulike syndrome.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
723
Q

What does the viral set point in HIV mean and what does it predict?

A
  • The viral load at the end of the acute phase reflects the equilibrium reached between the virus and the host response, and in a given patient it may remain fairly stable for several years.
  • The level, called the viral set point, is a predictor of the rate of decline of CD4+ T cells, and, therefore, profression of HIV disease.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
724
Q

Because the loss of HIV immune containment is associated with declining CD4+ T cell counts, the CDC classification of HIV infection stratifies three categories on the basis of CD4+ cell counts. What are they and what levels are there?

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
725
Q

What happens during the chronic phase of HIV?

A
  • Lymph nodes and the spleen are sites of continuous HIV replication and cell destruction
  • During this period, few or no clinical manifestations of the HIV infection are present.
  • Although the majority of peripheral blood T cells do not harbor the virus, destruction of CD4+ T cells within lymphoid tissues continues and the numer of circulating blood CD4+ T cells steadily declines.
  • Host defenses begin to wane, and the proportion of the surviving CD4+ cells infected with HIV increases, as does the viral burden per CD4+ cell.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
726
Q

The final phase of HIV infection is AIDS. What is this characterised by?

A
  • A breakdown of host defence, a dramatic increase in plasma virus, and severe, life-threatening clinical disease.
  • Long-lasting fever (>1 month), fatigue, weight loss and diarrhoea.
  • After a variable period, serious opportunistic infections, secondary neoplasms, or clinical neurologic disease emerge, and the patient is said to have developed AIDS.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
727
Q

In the absence of treatment, most patients with HIV infection progress to AIDS after how long?

A
  • After a chronic phase lasting 7-10 years
  • Exceptions to this typical course are exemplified by rapid progressors and long-tern nonprogressors
  • In rapid progressors the midd,e chronic phase in telescoped to 2-3 years after primary infection.
  • About 5-15% of infected individuals are long-term nonprogressors, defined as untreated HIV-1-infected individuals who remain asymptomatic for 10 years or more, with stable CD4+ T-cell counts and low levels of plasma viremia.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
728
Q

What are the clinical features of AIDS?

A
  • They range from a mild acte illness ro severe disease.
  • In the US, the typical adult patient presents with fever, weight loss, diarrhoea, generalised lymphadenopathy, multiple opportunistic infections, neurologic disease, and , in many cases, secondary neoplasms
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
729
Q

What are the AIDS-defining opportunistic infections?

A

Protozoal and Helminthic infections
Cryptosporidiosis or isosporidiosis (enteritis)
Pneumocystis jireoveci (pneumonia or disseminated infection)
Toxoplasmosis (pneumonia or CNS infection)
Fungal infections
Candidiasis (oesophageal, tracheal, or pulmonary)
Cryptococcosis (CNS infection)
Coccidiodomycosis (disseminated)
Histoplasmosis (disseminated)
Bacterial infections
Mycobacteriosis tuberculosis and mycobacterium avium-intracellulare
Nocardiosis (pneumonia, meningitis, disseminated)
Salmonella infections, disseminated
Viral infections
CMV (pulmonary, intestinal, retinitis, or CNS infections)
HSV (localised or disseminated infection)
VZV (localised or disseminated infection)
Progressive multifocal leukoencephalopathy

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
730
Q

What are some AIDS-defining neoplasms?

A

Kaposi sarcoma
Primary lymphoma of brain
Invasive cancer of uterine cervix - likely secondary to HPV

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
731
Q

What is Kaposi sarcoma?

A
  • A vascular tumour
  • The lesions of KS are characterised by the proliferation of spindle-shaped cells that express markers of both endothelial cells (vascular or lymphatic) and smooth muscle cells.
  • The tumour is usually widespread in AIDS, affecting the skin, mucous membranes, GI tract, lymph nodes and lungs.
  • There is also a profusion of slitlike vascular spaces, suggesting that the lesions may arise from primitive mesenchymal precursors of vascular channels.
  • In addition, KS lesions display chronic inflammatory cell infiltrates.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
732
Q

What is the pathogenesis of KS?

A
  • The spindle cells produce proinflammatory and angiogenic factors, which recruit the inflammatory and neovascular components of the lesion, and the latter components supply signals that aid in spindle cell survival and growth
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
733
Q

What causes Kaposi sarcoma? How?

A
  • It is caused by the KS herpesvirus, also called human herpesvirus 8 (HHV8).
  • HHV8 establishes latent infection, during which several proteins are produced with potential roles in stimulating spindle cell proliferation and preventing apoptosis. These include a viral homologue of cyclin D and several inhibitors of p53
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
734
Q

Why lymphoma is linked with Kaposi Sarcoma? why?

A
  • KSHV infection is not restricted to endothelial cells.
  • This virus is related phylogenetically to the lymphotropic subfamily of herpesvirus (γ-herpesvirus). Its genome is found in B cells of infected subjects
  • It is linked to rare B-cell lymphomas in AIDS patients (primary effusion lymphoma) and to multicentric Castleman disease, a B-cell lymphoproliferative disorder
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
735
Q

What percentage of people with AIDS develop lymphoma?

A

Roughly 5% present with lymphoma, and approx another 5% develop lymphoma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
736
Q

What are the mechanisms of lymphoma development in AIDS?

A
  • Unchecked proliferation of B cells infected with oncogenic herpesvirus in the setting of profound T cell depletion (AIDS). T cell immunity is required to restrain the proliferation of B cells infected with oncogenic viruses such as VBD and KSHV. AIDS patients are at high risk of developing aggressive B cell lymphomas composed of tumour cells infected by oncogenic viruses, particularly EBV.
  • Germinal center B-cell hyperplasia in the setting of early HIV infection. In germinal centers, B cells diversify their immunoglobulin genes via lesions introduced into their DNA by the enzyme activation-induced deaminase (AID). AID can cause mutations in oncogenes implcated in B-cell lymphomagenesis.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
737
Q

How does EBV in HIV cause lymphomas?

A
  • Once EBV immunity is established, it persists as a latent infection in memory B cells.
  • Activation of such cells, by antigen or by cytokines, reawakens an EBV-encoded program of gene expression that drives B-cell proliferation.
  • Patients with AIDS have high levels of several cytokines, some of which, including IL-6 are growth factors for B cells.
  • In the absence of T-cell immunit, these activated, EBV infected clones proliferate and eventually acquire additional somatic mutations, leading to their outgrowth as full-blown EBV positive B-cell lymphomas.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
738
Q

Apart from B-cell lymphoma, what are some other EBV-related proliferations that are common in AIDS?

A
  • Hodgkin lymphoma - all instances of HIV-associated Hodgkin lymphoma, the characteristic tumour cells (Reed-Sternberg cell) are infected with EBV.
  • Oral hair leukoplakia (white projections on the tongue), which results from EBV-driven sqaumour cell proliferation of the oral mucosa.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
739
Q

What are some CNS diseases in AIDS?

A
  • Self-limited meningo-encephalitis occuring at the time of seroconversion
  • Aseptic meningitis
  • Vaculoar myelpathy
  • Peripheral neuropathies
  • Most commonly, a progressive encephalopathy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
740
Q

In very broad terms, what do the antiretroviral drugs do for HIV?

A
  • They target the viral reverse transcriptase, protease, and integrase.
  • They are given in combination to reduce the emergence of mutants that develop resistance to any one
  • Treatment regimens are commonly called highly active antiretroviral therapy (HAART) or combination antiretrovoral therapy.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
741
Q

What is the immune reconstitution inflammatory syndrome that may occur with antiretrovirals? Why does it happen?

A
  • Some patients with advanced disease who are given antiretroviral therapy develop a paradoxical clinical deterioration during the period of recovery of the immune system. This occurs despite increasing CD4+ T cell counts and decreasing viral load.
  • It is postulated to be a poorly regulated host response to the high antigenic burden of persistent microbes.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
742
Q

What are the adverse effects of HAART?

A
  • Lipoatrophy (loss of facial fat)
  • Lipoaccumulation (excess fat deposition centrally)
  • Elevated lipids
  • Insulin resistance
  • Peripheral neuropathy
  • Premature cardiovascular kidney and liver disease
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
743
Q

What is the morphology of early stage HIV infected lymph nodes?

A
  • A marked hyperplasia of B cell follicles. The follicles are enlarged and often take on unusual, serpiginous shapes
  • The mantle zones that surroun the follicles are attenuated, and teh germinal centers impinge on interfollicular T cell areas.
  • This hyperplasia of B cells is the morphologic reflection of the polyclonal C-cell activation and hypergammaglobulinaemia seen in HIV-infected individuals.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
744
Q

How does the morphology of lymph nodes change throughout HIV infection?

A
  • With disease progression, the frenzy of B-cell proliferation subsides and gives way to a pattern of severe lymphoid involution.
  • The lymph nodes are depleted of lymphocytes, and the organised network of follicular dendritic cells is disrupted. The germinal centers may even become hyalinised.
  • These ‘burn-out’ lymph nodes are atrophic and small and may harbor numerous opportunistic pathogens, often with macrophages.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
745
Q

What are some modifiable and non-modifiable risk factors for atherosclerosis?

A

NON-MODIFIABLE
* Genetics - familial hypercholesterolaemia, diabetes risk
* Age - between ages 40-60, the incidence of MI increases five-fold. Death rates from IHD rise with each decade
* Gender - very low risk in premenopausal women. After menopause however, the incidence increases in women and at older ages actually exceeds that of men

MODIFIABLE
* Hyperlipidaemia - specificially LDL levels
* HTN - both systolic and diastolic levels - chronic HTN is the most common cause of LV hypertrophy
* Smoking
* Diabetes mellitus induces hypercholesterolemaeia and markedly increases the risk of atherosclerosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
746
Q

What are some additional risk factors for atherosclerosis?

A
  • Inflammation is present during all stages of atherogenesis and is intimately linked with atherosclerotic plaque formation and rupture. CRP correlates with IHD risk.
  • Hyperhomocystinemia is associated with premature vascular disease. Although low folate and vit G12 levels can ingrease homocystiene, supplements don’t affect the CVD risk
  • Metabolic syndrome associated with central obesity, is characterised by insulin resistance, HTN, dyslipidaemia, hypercoagulability, and a proinflammatory state
  • Lipoprotein a is an altered form of LDL and is associated with coronary and cerebrovascular disease risk, independent of total cholesterol or LDL levels.
  • Factors affecting hemostasis (plasminogen activator inhibitor 1, thrombin)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
747
Q

What is CRP? How does it work?

A

CRP is an acute phase reactant synthesisted primarily by the liver.
Its expression is increased by a number of inflammatory mediators, particularly IL-6, and it augmetns the innate immune response by binding to bacteria and activating the classical complement cascade.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
748
Q

What is the pathogenesis pathway of atherosclerosis?

A
  • Endothelial injury and dysfunction, causing increased vascular permeability, leukocyte adhesion and thrombosis
  • Accumulation of lipoproteins (mainly LDL and its oxidised forms) in the vessel wall
  • Monocyte adhesion to the endothelium, followed by migration into the intima and transformation into macrophages and foam cells
  • Platelet adhesion
  • Factor release from activated platelets, macrophages and vascular wall cells, inducing smooth muscle cell recruitment, either from the media or from circulating precursors
  • Smooth muscle cell proliferation, extrcellular matrix production and recruitment of T cells
  • Lipid accumulation both extracellularly and within cells (macrophages and smooth muscle cells)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
749
Q

How does endothelial injury precipitate atherosclerosis?

A
  • Early human lesions begin at sites of morphologically intact endothelium. Thus, endothelial dysfunction underlies most atherosclerosis.
  • The intact but dysfunctional endothelial cells exhibit increased endothelial permeability, enhanced leukocyte adhesion, and altered gene expression.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
750
Q

Endothelial injury can be caused by toxins from cigarette smoke, infectious agent and inflammatory cytokines. But what are the two most important causes of endothelial dysfunction?

A
  • Haemodynamic disturbances such as stasis or flow turbulence
  • Hypercholesterolaemia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
751
Q

What are the different types of hypercholesterolaemia?

A
  • Lipids are transported in the bloodstream bound to specific apoproteins (forming lipoprotein complexes)
  • Dyslipoproteinemias include 1) increased LDL cholesterol levels, 2) decreased HDL cholesterol levels, and 3) increased levels of the abnormal lipoprotein (a)
  • Familial hypercholesterolaemia, caused by defective LDL receptors and inadequate hepatic LDL uptake, can precipitate MIs before age 20.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
752
Q

What are the mechanisms by which hyperlipidaemia contributes to atherogenesis?

A
  • Chronic hyperlipidaemia can directly impair endothelial cell function by increasing local reactive oxygen species production; besides causing membrane and mitochondrial damage, oxygen free radicals accelerate nitric oxide decay, dampening its vasodilator activity.
  • Lipoproteins accumualte within the intima, where they may aggregate or become oxidised by free radicals produced by inflammatory cells. Such modified LFL is then accumulated by macrophages. Because the modified lipoproteins cannot be completely degraded, chronic ingestion leads to the formation of lipid-filled macrophages ‘foam cells; smooth muscle cells can similarly transform into lipid-laden foam cells by ingesting modified lipids through LDL-receptor related proteins.
  • Not only are the modified lipoproteins toxic to endothelial cells, smooth muscle cells, and macrophages, but their binding and uptake also stimulates the release of growth factors, cytokines, and chemokines that create a vicious cycle of monocyte recruitment and activation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
753
Q

How does inflammation cause atherosclerosis?

A
  • It causes endothelial damage (you know how).
  • Activated macrophages produce ROS that enhance LDL oxidation, and elaborate growth factors that drive smooth muscle cell proliferation.
  • Activated T cells in the growing intimal lesions elaborate inflammatory cytokines, e.g. interferon-γ, which , in turn, can activate macrophages as well as endothelial cells and smooth muscle cells
  • These leukocytes and vascular wall cells release growth factors that promote smooth muscle cell prolfieration and syntehsis of extracellular matrix proteins. Thus, many of the lesions of atherosclerosis are atrributable to the chronic inflammatory reaction in the vessel wall.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
754
Q

How does smooth muscle proliferation and matrix synthesis take place in atherosclerosis?

A
  • Intimal smooth muscle cell proliferation and extracellular matrix deposition convert a fatty streak into a mature atheroma and contribute to the progressive growth of atherosclerotic lesions.
  • Intimal smooth muscle cells have a proliferative and synthetic phenotype distinct from the underlying medial smooth muscle cells.
  • Several growth factors are implicated in smooth muscle cell proliferation, including platelet-derived growth factor (PDGF), FGF and TGF-α. These factors also stimulate smooth muscle cells to syntehsis extracellular matrix (notably collagen), which stabilised atherosclerotic plaques.
  • In contract, activated inflammatory cells in atheromas may increase breakdown of extracellular matrix components, resulting in unstabel plaques
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
755
Q

What are atheromas?

A

They are dynamic lesions consisting of dysfunctional endothelial cells, proliferating smooth muscle cells, and admixed T lymphocytes and macrophages.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
756
Q

How does the make-up of atheromas change over time?

A
  • At early stages, intimal plaques are little more than aggregates of smooth muscle cells, macrophages, and foam cells; death of these cells releases lipids and necrotic debris
  • With progression, the atheroma is modified by extracellular matrix synthesised by smooth muscle cells; connective tissue is particularly prominent on the intimal aspect forming a fibrous cap, although lesions typically retain a central core of lipid-laden cells and fatty debris that can become calcified.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
757
Q

What is the outcome of an atheroma?

A

The intimal plaque may progressively encroach on the vessel lumen, or may compress the underlying media, leading to its degeneration this in turn may expose thrombogenic factors, resulting in thrombus formation and acute vascular occlusion

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
758
Q

What are fatty streaks in atherosclerosis?

A
  • Fatty streaks are composed of lipid-filled foamy macrophages.
  • Beginning as multiple minute flat yellow spots, they eventially coalesce into elongated streaks 1cm long or longer
  • These lesions are no sufficiently raised to cause significant flow disturbances.
  • Although fatty streaks can evolve into plaques, not all are destined to become advanced lesions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
759
Q

What are the key processes in atherosclerosis?

A

Intimal thickening and lipid accumulation, which together form plaques

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
760
Q

How do atherosclerotic plaques and lesions present morphologically?

A
  • Atheromatous plaques are white-yellow and encroach on the lumen of the artery; superimposed thrombu over ulcerated plaques is red-brown.
  • Atherosclerotic lesions are patchy, usually involving only a portion of any given arterial wall adn are rarely circumferential; on cross-section, the lesions therefore appear “eccentric”. The focality of atherosclerotic lesions is attributable to the vagaries of vascular haemodynamics.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
761
Q

What are the most common vessels prone to atherosclerosis?

A

The most extensively involved vessels are the lower abdominal aorta, the coronary arteries, the popliteal arteries, the internal carotid arteries, and the vessels of the circle of Willis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
762
Q

Atherosclerotic plaques have three principal components. What are they?

A

1) Smooth muscle cells, macrophages and T cells
2) Extracellular matrix, including collagen, elastic fibers, and proteoglycans
3) Intracellular and extracellular lipids

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
763
Q

How are the components of an atherosclerotic plaque lay out?

A
  • Typically, there is a superficial fibrous cap composed of smooth muscle cells and relatively dense collagen
  • Beneath and to the side of the cap (the “shoulder”) is a more cellular area containing macrophages, T cells, and smooth muscle cells.
  • Deep to the fibrous cap is a necrotic core, containing lipid (primarily cholesterol and cholesterol esters), debris from dead cells, foam cells (lipid-laden macrophages and smooth muscle cells), fibrin, variably organised thrombus, and other plasma proteins; the cholesterol content is frequently present as crystalline aggregates that are washed out during routine tissue processin and leave behind only empty “clefts”.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
764
Q

Atherosclerotic plaques generally continue to change and progress over time, what are the processes that allow this?

A

Through cell death and degradation, synthesis and degradation (remodeling) of ECM, and organisation of any superoimproved thrombus.
Moreover, atheromas often undergo calcification.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
765
Q

Atherosclerotic plaques are susceptible to which clinically important pathologic changes?

A
  • Rupture, ulceration, or erosion of the surface of atheromatous plaques exposes highly thrombogenic substances and leads to thrombosis, which may partially or completely occlude the vessel lumen.
  • Haemorrhage into a plaque - rupture of the overlying fibrous cap, or of the thin-walled vessels in the areas of neovascularisation, can cause intraplaque haemorrhage; a contained haemtoma may expand the plaque or induce plaque rupture
  • Atheroembolism - plaque rupture can discharge atherosclerotic debris into the blood stream, producing microemboli
  • Aneurysm formation - atherosclerosis-induced pressure or ischaemic atrophy of the underlying media, with loss of elastic tissue, causes weakness and potential rupture
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
766
Q

What is atherosclerotic stenosis?

A
  • In small arteries, the atherosclerotic plaques can gradually occlude vessel lumina, compromising blood flow and causing ischaemic injury.
  • At early stages of stenosis, outward remodeling of the vessel media tends to preserve the size of the lumen. However, there are limits of the extent of remodeling, and eventually the expanding atheroma impinges on the lumen to such a dgeree that blood flow is compromised.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
767
Q

What is critical atherosclerotic stenosis?

A
  • The stage at which the occlusion is sufficiently severe to produce tissue ischaemia.
  • In the coronary circulations, this typically occurs at when the occlusion produces a 70% decrease in luminal cross-scetional area.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
768
Q

In what conditions does a plaque rupture?

A

When they are unable to withstand mechanical stresses generated by vascular shear forces.
The events that trigger abrupt changes in plaques and subsequent thrombosis are complex and include both intrinsic factors (e.g. plaque structure and composition) and extrinsic elements (e.g. blood pressure, platelet reactivity, vessel spasm)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
769
Q

The composition of atherosclerotic plaques is dynamic and can contribute to risk of rupture. What compositions increase the risk of rupture?

A

Plaques that contain large areas of foam cells and extracellular lipid, and those in which the fibrous caps are thin or contain few smooth muscle cells of have clusters of inflammatory cells, are more likely to rupture; there are referred to as “vulnerable plaques”

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
770
Q

What is the major structural component of the fibrous cap in atherosclerotic caps? What affects its integrity?

A

Collagen accounts for its mechnical strength and stability.
Thus, the balance of collagen synthesis versus degradation affects cap integrity.
Collagen in atherosclerotic plaque is produced primarily by smooth muscle cells so that loss of these cells results in a less sturdy cap

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
771
Q

What controls collagen turnover in atherosclerotic plaques?

A
  • Collagen turnover is controlled by metalloproteinases (MMPs), enzymes elaborated largely by macrophages and smooth muscle cells within the atheromatous plaque
  • Conversely, tissue inhibitors of metalloproteinases (TIMPs) produced by endothelial cells, smooth muscle cells and macrophages modulate MMP activity.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
772
Q

How does cholesterol deposits affect atherosclerotic plaques?

A
  • In general, plaque inflammation results in a net increase in collagen degradation and reduced collagen synthesis, thereby destabilising the mechanical integrity of the fibrous cap.
  • The inflammation induced by cholesterol deposits themselves may contribute to plaque destabilisation.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
773
Q

Influencs extrinsic to plaques also contribute to acute plaque changes. Like what?

A
  • Adrenergic simulation can increase systemic BP or induce local vasoconstriction, thereby increasing the physical stresses on a given plaque.
  • The adrenergic stimulation associated with wakening can cause BP spikes that have causally inked to the pronounced circadian periodicity for onset of acute MI.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
774
Q

Vasoconstriction compromises lumen size, and, by increasing the local mechanical forces, can potentiate plaque disruption. What may stimualte vasoconstriction at sites of atheroma?

A
  • Circulating adrenergic agonist
  • Locally released platelet contents
  • Endothelial cell dysfunction with impaired secretion of endothelial-derived relaxing factors (NO) relative to contracting factors (endothelin)
  • Mediators released from peri-vascular inflammatory cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
775
Q

What is an aneurysm?

A

A localised abnormal dilation of a blood vessle or the heart that may be congenital or acquried

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
776
Q

What are the different types of aneurysm? Give examples

A
  • A ‘true’ aneurysm is when it involves an attenuated but intact arterial wall or thinned ventricular wall of the heart - atheroscleroric, syphilitic and congenital vascular aneurysms, ventricular aneurysms that follow MIs
  • A false aneurysm is a defect in the vascular wall leading to extravascular haematoma that freely communicates with the intravascular space “pulsating haematoma” - ventricular rupture after MI or a leak at the sutured junction of a vascular graft with a natural artery
  • An arterial dissection arises when blood enters a defect in the arterial wall and tunnels between its layers. Dissections are often but always aneurysmal.

All of these can rupture, often with catastrophic consequences

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
777
Q

Descriptively, aneurysms are classified by macroscopic shape and size. How would you do this?

A
  • Saccular aneurysms are spherical outpouchings involving only a portion of the vessel wall; they may vary from 5 to 20 cm in diameter and often contain thrombus
  • Fusiform aneurysms are diffuse, circumferential dilations of a long vascular segment; they vary in diameter (up to 20cm) and in length and can involve extensive portions of the aortic arch, abdominal aorta or even the iliacs.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
778
Q

When do aneurysms occur?

A
  • To maintain their structural and functional integrity, arterial walls constantly remodel by synthesising, degrading, and repairing damage to their ECM constituents.
  • Aneurysms can occur when the structure or function of the connective tissue within the vascular wall is compromised.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
779
Q

What are some inherited defects in connective tissues that increase the risk of aneurysm and dissection? How?

A
  • In Marfan syndrome, defective synthesis of the scaffolding protein fibrillin leads to aberrant TGF-β activity and weakening of elastic tissue; in the aorta, this may result in progressive dilation.
  • In Loeys-Dietz syndrome mutations in TGF-β receptors lead to defective synthesis of elastin and collagens I and III. Aneurysms in such individuals can rupture fairly easily (even at small size) and are thus considered to follow an “aggresive” course
  • Weak vessel walls due to defective type III collagen synthesis are also a hallmark of the vascular forms of Ehlers-Danlos syndrome
  • Altered collagen cross-linking associated with vitamin C deficiency (scurvy) is an example of a nutritional basis for aneurysm formation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
780
Q

How is aneurysm formation linked to collagen synthesis and degradation? What enzymes are also related?

A
  • Increased matrix metalloprotease (MMP) expression, particularly by macrophages in atherosclerotic plaque or vasculitis, likely contributes to aneurysm development; these enzymes have the capacity to degrade virtually all components of the ECM in the arterial wall (collagens, elastin, proteoglycans, laminin, fibronectin).
  • Decreased expression of tissue inhibitors of metalloproteases (TIMPs) can also contribute to the ECM degradation.
  • The risk of aneurysm formation in the setting of inflammatory lesions (atherosclerosis) may be associated with MMP and/or TIMP polymorphisms
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
781
Q

In development of aneurysms, the vascular wall is weakened through loss of smooth muscle cells or the synthesis of non-collagenous or nonelastic extracellular matrix. How does this happen?

A
  • Ischaemia of the inner media occurs when there is atherosclerotic thickening of the intima, which increases the distance that oxygen and nutrients must diffuse.
  • Systemic hypertension can also cause significant narrowing of arterioles of the vasa vasorum (e.g. in the aorta), which causes outer medial ischaemia
  • Medial ischaemia may lead to “degenerative changes” of the aorta, whereby smooth muscle cell loss - or change in the synthetic phenotype - leads to scarring (and loss of elastic fibers), inadequate extracellular matrix synthesis, and production of increasing amoutns of amorphous ground substance (glycosaminoglycan).
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
782
Q

Histologically, what is cystic medial degeneration?

A

It is smooth muscle cell loss leading to scarring (and loss of elastic fibers), inadequate ECM synthesis, and production of increasing amounts of amorphous ground substance (glycosaminoglycan) that occurs in Marfan syndrome and scurvy

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
783
Q

How does tertiary syphilis cause aortic aneurysms>

A

The obliterative endarteritis characteristic of late-stage syphilis shows a predilection for small vessels, including those of the vasa vasorum of the thoracic aorta.
This leads to ischaemic injury of the aortic media and aneurysmal dilation, which sometimes involves the aortic valve annulus.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
784
Q

The two most important causes or aortic aneurysms are atheroscleosis and HTN. Where do they often affect the aorta?

A

Atherosclerosis is a greater factor in AAAs, while HTN is the most common etiology associated with ascending aortic aneurysms.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
785
Q

Other factors that weaken vessel walls and lead to aneurysms include trauma, vasculitis, congenital defects (fibromuscular dysplasia and berry aneurysms) and infections such as mycotic aneurysms. Where can mycotic aneurysms originate?

A

1) from embolisation of a septic embolus, usually as a complication of infective endocarditis
2) as an extension of an adjacent suppurative process
3) by circulating organisms directly infecting the arterial wall

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
786
Q

In what populations do AAAs occur more frequently?

A
  • Men
  • Smokers
  • Age >50
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
787
Q

Where in the aorta is an AAA? How big can they be?

A

Usually positioned below the renal arteries and above the bifurcation of the aorta. They can occasionally affect the renal and superior or inferior mesenteric arteries, either by direct extensior of by occluding vessel ostia with mural thrombi.
AAA can be saccular or fusiform, up to 15cm in diameter and up to 25cm in length

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
788
Q

What is the morphology of a AAA?

A

There is severe complicated atherosclerosis and destruction and thinning of the underlying aortic media; the aneurysm frequently contains a bland, laminated, poorly organised mural thrombus.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
789
Q

Although they are less common than the usual atherosclerotic anuerysm, what are three AAA variants thet merit special mention because of their unusual features? tell me about them

A
  • Inflammatory AAA account for 5-10% of all AAA; they typically occur in younger patients, who often present with back pain and elevated inflammatory markers (elevated CRP).
  • A subset of inflammatory AAA may be a vascular manifestation of an entity called immunoglobulin G4 (IgG4)-related disease. A disorder marked by high plasma levels of IgG4 and tissue fibrosis associated with frequent infiltrating IgG4-expressing plasma cells
  • Mycotic AAA are lesions that have become infected by the lodging of circulating microorganisms in the wall. In such cases, suppuration further destroys the media, potentiating rapid dilation and rupture
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
790
Q

How are inflammatory AAA characterised morphologically? What causes it?

A
  • Inflamatory aneurysms are characterised by abundant lymphoplasmacytic inflammation with many macrophages (and even giant cells) associated with dense periaortic scarring that can extend into the anterior retroperioneum.
  • The cause is a presumed localised immune response to the abdominal aortic wall; remarkably, most cases are not associated with inflammation of other arteries.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
791
Q

What are the clinical features of AAA?

A

Most cases of AAA are asymptomatic and are discovered incidentally on physical exam as an abdominal mass (often palpably pulsating) that may mimic a tumour. The other clinical manifestations of AAA include:
* rupture into the periotoneal cavity of retroperitoneal tissues with massive, potentially fatal haemorrhage
* obstruction of a vessel branching off from the aorta, resulting in ischaemic injury to the supplied tissue; for example, iliac, renal, mesenteric or vertebral arteries
* embolism from atheroma or mural thrombus
* impingement on an adjacent structure, for example, compression of a ureter or erosion of vertebrae

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
792
Q

The risk of rupture is directly related to the size of the aneurysm. Tell me about the risk related to the size:

A
  • 4cm or less - no risk
  • 4-5cm - 1% per year
  • 5-6 cm - 11% per year
  • > 6cm - 25% per year
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
793
Q

How quickly do aneurysms expand?

A

Most expand at a rate of 0.2 to 0.3 cm/year but 20% expand more rapidly.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
794
Q

How are AAA managed?

A

In general, aneurysms 5cm or larger are managed aggresively, usually by surgical bypass with prosthetic grafts, although treatment via endoluminal approaches using stent grafts rather than surgery is now available for selected patients

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
795
Q

Timely surgery for AAA is critical. Why?

A

Operative mortality for unruptured aneurysms is approx 5%, whereas emergency surgery after rupture carries a mortality rate of >50%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
796
Q

What are the main associations for thoracic aortic aneurysms?

A

They are most commonly associated with HTN, although other causes such as Marfan syndrome and Loeys-Dietz syndrome are increasingly recognised.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
797
Q

What are the clinical features of thoracic aortic aneurysm?

A

1) respiratory difficulties due to encroachment on the lungs and airways
2) difficulty in swallowing due to compression of the oesophagus
3) persistent cough due to compression of the recurrent laryngeal nerves
4) pain caused by erosion of bone (i.e. ribs and vertebral bodies)
5) cardiac disease as the aortic aneurysm leads to aortic valve dilation with valvular insufficiency or narrowing of the coronary ostia causing MI
6) rupture

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
798
Q

What is an aortic dissection?

A

Aortic dissection occurs when blood separates the laminar planes of the media to form a blood-filled channel within the aortic wall; this can be catastrophic if the dissection then ruptures through the adventitia and haemorrages into adjacent spaces.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
799
Q

Aortic dissection occurs principally in two groups of patients. What are they? What are the rarer groups of populations

A

1) men aged 40-60 years with antecedent HTN (>90% of cases)
2) younger adults with systemic or localised abnormalities of connective tissue affecting the aorta (e.g. Marfan syndrome)

It can also be iatrogenic e.g. following arterial cannulations during coronary catheterisation or bypass.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
800
Q

Rarely pregnancy is associated with aortic dissection. What trimester and why?

A
  • This typically occurs during or after third trimester
  • It may be related to hormone-induced vascular remodeling and the haemodynamic stresses of the perinatal period.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
801
Q

What is the pathogenesis of aortic dissection?

A
  • HTN is the major risk factor for aortic dissection
  • Aortas of hypertensive patients have medial hypertrophy of vasa vasorum associated with degenerative changes such as loss of medial smooth muscle cells and disorganised ECM, suggesting that ischaemic injury (due to diminished flow through the vasa vasorum, possibly exacerbated by high wall pressures) is contributory.
  • The trigger for intimal tear and initial intramural aortic haemorrhage is not known in most cases.
  • Once a tear has occurred, blood flow under systemic pressure dissects through the media, leading to progression of the haematoma.
  • Accordingly, aggressive pressure-reducing therapy may be effective in liminiting an evolving dissection.
  • In some cases, disruption of penetrating vessels of the vasa vasorum can give rise to an intramural haematoma without an intimal tear
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
802
Q
A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
803
Q

Morphologically, what changes occur prior to/at the beginning of an aortic dissection?

A
  • The most frequent preexisting histologically detectable lesion is cystic medial degeneration, and inflammation is characteristically absent.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
804
Q

An aortic dissection usually initiates with an intimal tear. Where is the location that the vast majority of spontaneous dissections occur? What shape are they? Where do they go?

A
  • The tear often occurs in the ascending aorta, usually within 10cm of the aortic valve
  • Such tears are typically transverse with sharp, jagged edges up to 1-5cm in length.
  • The dissection can extend retrograde towards the heart as well as distally, sometimes into the iliac and femoral arteries
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
805
Q

Which layers of the aorta does a dissection often affect?

A

The dissecting haematoma spreads characteristally along the laminor planes of the aorta, usually between the middle and outer thirds.
It can rupture through the adventitia causing massive haemorrhage (e.g. into the thoracic or abdominal cavities) or cardiac tamponade (haemorrhage into the pericardial sac)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
806
Q

What is a double-barreled aorta? What happens to this over time?

A

In some instances, the dissecting haematoma reenters the lumen of the aorta through a second distal intimal tear, creating a new false vascular channel - double-barreled aorta.
This averts a fatal extraaortic haemorrhage, and over time, such false channels can be endothelialised to become recognisable chronic dissections

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
807
Q

What are the classifications of aortic dissections?

A
  • The more common (and dangerous) proximal lesions (called type A dissections), involving either both the ascending and descending aorta (type 1 Debakey) or just the ascending aorta only (type II DeBakey)
  • Distal lesions not involving the ascending part and usually beginning distal to subclavian artery (called type B dissections or DeBakey type III)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
808
Q

What are the clinical features of an aortic dissection?

A

The classic clinical symptoms or aortic dissection are the sudden onset of excruciating pain, usually beginning in the anterior chest, radiating to the back between the scapulae, and moving downward as the dissection progresses; the pain can be confused with that of an MI.
Common clinical manifestations also include cardiac tamponade and aortic insufficiency

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
809
Q

What is the most common cause of death from aortic dissection?

A

Rupture of the dissection into the pericardial, pleural, or peritoneal cavities. Retrograde dissection into the aortic root can also disrupt the aortic valve annulus.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
810
Q

Where can aortic dissections extend into? What does it cause?

A

They can extend into the great arteries of the neck or into the coronary, renal, mesenteric, or iliac arteries, causing vascular obstruction and ischaemic consequences such as MI; involvement of spinal arteries can cause transverse myelitis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
811
Q

What is the treatment for type A dissections?

A

Rapid diagnosis and institution of intensive antihypertensive therapy coupled with surgical plication of the aortic intimal tear can save 65-85% of patients.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
812
Q

What is the prognosis of type A dissections?

A

Mortality approaches 70% in those who present with haemorrhage or symptoms related to distal ischaemia, and the overall 10-year survival is only 40-60%.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
813
Q

What is the prognosis and treatment of type B dissections?

A

Most type B dissections can be managed conservatively; patients have a 75% survival rate whether they are treated with surgery or antihypertensive medication only/

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
814
Q

What is ischaemic heart disease

A

IHD represents a group of physiologically related syndromes resulting from myocardial ischaemia - an imbalance between myocardial supply (perfusion) and cardiac demand for oxygenated blood.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
815
Q

What is myocardial ischaemia vs hypoxia?

A

Ischaemia not only limits tissue oxygenation (and thus ATP generation), but also reduced the availability of nutrients and the removal of metabolic wastes.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
816
Q

What are the main causes of myocardial ischaemia?

A

> 90% cases of myocardial ischaemia results from reduced blood flow due to obstructive atherosclerotic lesions in teh epicardial coronary arteries.
Consequently, IHD is frequently referred to as coronary artery disease

But it can also be caused by coronary emboli, myocardial vessel inflammation, or vascular spasm.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
817
Q

Does IHD have a long or short period of progression?

A

In most cases there is a long period (up to decades) of silent, slow progression of coronary lesions before the sudden onset of symptoms.
Thus, IHD is often the late manifestatoin of coronary atherosclerosis that began during childhood or adolsecence.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
818
Q

How can IHD present?

A

As one or more of the following clinical syndromes:
* Myocardial infarction, where ischaemia causes frank cardiac necrosis
* Angina pectoris, where ischaemia is not severe enough to cause infarction, but the symptoms nevertheless portend infarction risk
* Chronic IHD with heart failure
* Sudden cardiac death

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
819
Q

The number of deaths from IHD has significantly improved. Why?

A
  • Prevention - modifying important risk factors, such as smoking, blood cholesterol, and HTN, weight loss and increased exercise
  • Diagnostic and therapeutic advances such as statins, CCUs, thrombolysis, PCI, stents, CABG, implantable defibrillators
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
820
Q

More than 90% of patients with IHD have atherosclerosis involving one or more coronary arteries. What percentage surface area obstruction means significant coronary artery disease?

A

A fiex lesion obstructing greater than 75% of vascular cross-sectional area defines sigificant coronary artery disease; this is generally the threshold for symptomatic ischaemia precipitated by exercise

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
821
Q

Obstruction of what % of cross-sectional area of the coronary lumen can lead to inadequate coronary blood flow even at rest?

A

90%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
822
Q

Progressive myocardial ischaemia induced by slowly developing occlusions may lead to what?

A

It may stimulate the formation of collateral vessels over time, which can often protect against myocardial ischaemia and infarction and mitigate the effects of high-grade stenoses.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
823
Q

Clinically significant plaques can be located anywhere along the course of the coronary vessels. Where most commonly?

A

Particularly the RCA, although they tend to predominate within the first several centimeters of the LAD and LCX. Somtimes the major epicardial bracnhes are also involved (i.e. LAD diagonal bracnhes, LCX obtuse marginal branches, or posterior descending branch of the RCA), but the atherosclerosis of the intramural (penetrating) branches is rare.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
824
Q

The risk of an individual developing clinically important IHD depends on what?

A

In part on the number, distribution, sturcture, and degree of obstruction of atheromatous plaques. Also the acute change of a plaque

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
825
Q

Acute coronary syndromes are typically initiated by what?

A

An unpredictable and abrupt conversion of a stable atherosclerotic plaque to an unstable and potentially life-threatening atherothrombotic lesion through rupture, superficial erosion, ulceration, fissuring, or deep haemorrhage.
In most instances, plaque changes - typically associated with intralesional inflammation - may precipitate the formation of a superimposed thrombus that partially or completely occludes the artery

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
826
Q

What are the consequences of myocardial ischaemia?

A
  • Stable angina results from increases in myocardial oxygen demand that outstrip the ability of stenosed coronary arteries to increase oxygen delivery; it is usually not associated with plaque disruption
  • Unstable angina is caused by plaque disruption that results in thrombosis and vasoconstriction, and leads to severe but transient reductions in coronary blood flow.
  • Myocardial infarction is often the result of acute plaque change that induces an abrupt thrombotic occlusion, resulting in myocardial necrosis
  • Sudden cardiac death may be caused by regional myocardial ischaemia a fatal ventricular arrhythmia.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
827
Q

What is angina pectoris?

A

It is characterised by payxoysmal and usually recurrent attacks of substernal or precordial chest discomfort caused by transient (15 seconds to 15 minutes) myocardial ischaemia that is insufficient to induce myocyte necrosis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
828
Q

What causes the pain of angina?

A

The pain itself is likely a consequence of the ischaemia-induced release of adenosine, bradykinin, and other molecules that stimulate sympathetic and vagal afferent nerves.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
829
Q

There are three overlapping patterns of angina pectoris caused by varying combinations of decreased perfusion, increased demand, and coronary arterial pathology. What are they and in what situations do they happen?

A
  • Stable angina is the most common form of angina; caused by an imbalance in coronary perfusion (due to chronic stenosing coronary atherosclerosis) relative to myocardial demand, such as physical activity, emotion or stress.
  • Prinzmetal variant angina is an uncommon form of episodic myocardial ischaemia; caused by coronary artery spasm.
  • Unstable or crescendo angina refers to a pattern of increasing frequent, prolonged (>20 mins), or severe angina or chest discomfort described as frank pain. In most patients, is it caused by the disruption of an atherosclerotic plaque with superimposed partial thrombosis and possibly embolisation or vasospasm (or both)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
830
Q

How is stable angina often described and what usually relieves it?

A

As a deep, poorly localised pressure, squeezing, or burning sensation (like indigestion), but unusually as pain, and is usually relieved by rest (decreasing demand) or administering vasodilators, such as nitroglycerin and calcium channel blockers (increasing perfusion)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
831
Q

What does Prinzmetal angina usually respond well to?

A

Generally responds promptly to vasodilators

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
832
Q

What does increases the incidence and risk factors of MI?

A
  • Age - Mi can occur at virtually any age; nearly 10% occur in people <40. 45% occur in people <65 years old.
  • Race Black and white people are equally affected
  • Through middle age male gender incrases the relative risk of MI
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
833
Q

What is the pathogenesis of coronary arterial occlusion?

A
  • A coronary artery atheromatous plaque undergoes an acute change consisting of intraplaque hemorrhage, erosion or ulceration, or rupture or fissuring
  • When exposed to subendothelial collagen and necrotic plaque contents, platelets adhere, become activated, release their granule contents, and aggregate to form microthrombi
  • Vasospasm is stimulated by mediators released from platelets
  • Tissue factor activates the coagulation pathway, adding to the bulk of the thromus
  • Within minutes, the thrombus can expand to completely occlude the vessel lumen
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
834
Q

In approximately 10% of cases, transmural MI occurs in the absence of the typical coronary atherothrombosis. In such situations, what happens?

A
  • Vasospasm with or without coronary atherosclerosis, perhaps in association with platelet aggregation or due to drug ingestion (e.g. cocaine or ephedrine)
  • Emboli from the left atrium in association with AF, a left-sided mural thrombus, vegetations of infective endocarditis, intracardiac prosthetic material or paradoxical emboli from the right side of the heart or the peripheral veins, traversing a patent foramen ovale and into the coronary rteries
  • Ischaemia without detectable or significant coronary atherosclerosis and thrombosis may be caused by disorders of small intramural coronary vessels (vasculitis), haematological abnormalities (sickle cell disease), amyloid deposition in vasular walls, vascular dissection, marked hypertrophy (AS), shock
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
835
Q

What is the myocardial response to coronary arterial obstruction?

A

Coronary arterial obstruction diminishes blood flow to a region of myocardiam, causing ischaemia, rapid myocardial dysfunction, and eventually - with prolonged vascular compromise - myocyte death.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
836
Q

What does the outcome of myocardial infarction depend on?

A

Predominantly on the severity and duration of flow deprivation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
837
Q

What are the early biological consequences of MI? How does this affect myocardial function?

A
  • The cessation of aerobic metabolism within seconds, leading to inadequate production of high energy phosphates (e.g. creatine phosphate and ATP) and accumulation of potentially noxious metabolites (e.g. lactic acid)
  • Because of the exquisite dependence of myocardial function on oxygen and nutrients, myocardial contractility ceases within a minute or so of the onset of severe ischaemia. Such loss of function actually precipitates heart failure long before myocyte death occurs
838
Q

What are the ultrastructural changes that occur in MI? How long do they take? are they irreversible?

A
  • Ultrastructural changes - myofibrillar relaxation, glycogen depletion, cell and mitochondrial swelling - develop within a few minutes of the onset of ischemia
  • There early manifestations of ischaemic injury are potentially reversible
  • Only severe ischemia (blood flow <10% of normal) lasting 20-30 minutes or longer leads to irreversible damage (necrosis) of cardiac myocytes.
839
Q

What is the earliest detectable feature of myocyte necrosis? What are these used for?

A
  • The disruption of the integrity of the sacrolemmal membrane, allowing intracellular macromolecules to leak out of necrotic cells into the cardiac interstitium and ultimately into the microvasculature and lymphatics
  • This escape of intracellular myocardial proteins into the circulation forms the basis for blood tests that can sensitively detect irreversible myocyte damage, and important for managing MI.
840
Q

What are approximate time of onset of key events in ischaemic cardiac myocytes?

A
  • Onset of ATP depletion - seconds
  • Loss of contractility - under 2 minutes
  • ATP reduced to 50% of normal - 10 minutes
  • ATP reduced to 10% of normal - 40 minutes
  • Irreversible cell injury - 20-40 minutes
  • Microvascular injury - over 1 hour
841
Q

Where is ischaemia most pronounced in the heart?

A
  • Due to the myocardial perfusion pattern from epicardium to endocardium, ischaemia is most pronounced in the subendocardium; thus, irreversibly injury of ischaemic myocytes occurs first in the subendocardial zone.
  • With more extended ischaemia, a wavefront of cell death moves through the myocardium to encompass progressively more of the transmural thickness and breadth of the ischaemic zone.
842
Q

The precise location, size, and specific morphologic features of an acute MI depend on what?

A
  • The location, severity, and rate of development of coronary obstructions due to atherosclerosis and thromboses
  • The size of the vascular bed perfused by the obstructed vessels
  • The duration of the occlusion
  • The metabolic and oxygen needs of the myocardium at risk
  • The extent of collateral blood vessels
  • The presence, site and severity of coronary arterial spasm
  • Other factors, such as heart rate, cardiac rhythm and blood oxygenation
843
Q

How long does it take for necrosis to involve half of the thickness of the myocardium and the full way through? What may change this?

A
  • Necrosis involves approximately half of the thickeness of the myocardium in 2-3 hours of the onset of severe MI, and is usually transmural within 6 hours.
  • However, in instances where chronic sublethal ischaemia has induced a more well-developed coronary collateral circulation, the progression of necrosis may follow a more protracted course (12 hours or longer)
844
Q

What does the LAD branch of the coronary artery supply?

A

Most of the apex of the heart, the anterior wall of the left ventricle, and the anterior two thirds of the ventricular septum.

845
Q

What is the “dominant” coronary artery?

A

Either RCA or LCX - that perfuses the posterior third of the septum is called ‘dominant’ even though the LAD and PCX perfuse the majority of the left ventricular myocardium

846
Q

What proportion of the population are right dominant circulation and what does this mean?

A
  • Present in 80% of individuals
  • The RCA supplies the entire right ventricular free wall, the posterobasal wall of the left ventricle, and the posterior third of the ventricular septum, while the LCX generally perfuses only the lateral wall of the left ventricle
  • Thus, RCA occlusions can potentially lead to left ventricular damage.
847
Q

How do coronary collaterals affect coronary blood supply?

A
  • Although most hearts have numerous intercoronary anastomoses, relatively little blood normally courses through these.
  • However, when a coronary artery is progressively narrowed over time, blood flows via the collaterals from the high- to low-pressure circulating causing the channels to enlarge.
  • Through such progressive dilation and growth of collaterals, stimulated by ischaemia, blood flow is provided to areas of myocardium that would otherwise be deprived of adequate perfusion.
  • Indeed, in the setting of extensive collateralisation, the normal epicardial perfusion terroties may be so expanded that subsequent occlusion leads to infarction in paradoxical distributions.
848
Q

The distribution of myocardial necrosis correlates with the location and cause of the decreased perfusion. How does the location affect myocardial necrosis?

A
  • Transmural infarction. MI caused by an epicardial vessel (in the absence of any therapeutic intervention) are typically transmural - the necrosis involves virtually the full thickness of the ventricular wall in the distribution of the affected coronary.
  • Subendocardial (nontransmural) infarction. As the subendocardial zone is normally the least perfused region of myocardium, this area is most vulnerable to any reduction in coronary flow. It typically involves roughly the inner third of the ventricular wall.
  • Multifocal microinfarction pattern is seen when there is pathology involving only smaller intramural vessels.
849
Q

What usually causes the different patterns of myocardial infarction?

A
  • Transmural infarction is usually associated with a combination of chronic coronary atherosclerosis, acute plaque change, and superimposed thrombosis
  • Subendocardial (nontransmural) infarction can occur as a result of plaque disruption followed by a coronary thrombus that becomes lysed (therapeutically or spontaneously) before myocardial necrosis extends across the full thicckness of the wall. They can also result from prolonged, severe reduction in systemic blood pressure, as in shock on superimpsoed on chronic, otherwise noncritical, coronary stenoses
  • Multifocal infarction may occur in the setting of microembolisation, vasculitis, or vascular spasm, for example, due to endogenous catechols (adrenaline) or drugs (cocaine)
850
Q

In the subendocardial infarcts that occur as a result of global hypotension rather than an acute plaque, how is the pattern of infarction different?

A

The myocardial damage is usually circumferential, rather than being limited to the distribution of a singele major coronary artery

851
Q

How do elevated levels of catechols (adrenaline, cocaine) cause multifocal microinfarctions? What is the outcome?

A

They increase heart rate and myocardial contractility, exacerbating ischaemia caused by the vasospasm.
The utcome of such vasospasm can be sudden cardiac death (usually caused by a fatal arrhythmia) or an ischaemic dilated cardiomyopathy, so-called takotsubo cardiomyopathy

852
Q

How do STEMIs and NSTEMIs relate to the pattern of infarction?

A

A transmural infarct is referred to as a STEMI and a subendocardial infarct as an NSTEMI

853
Q

What proportion of the artery perfusion zone do transmural myocardial infarcts effect?

A

Nearly all transmural infarcts involve at least a portion of the left ventricle (comprising the free wall and ventricular septum) and encompass nearly the entire perfusion zone of the occluded coronary artery save for a narrow rim (approx 0.1mm) of preserved subendocardial myocardium that is preserved by diffusion of oxygen and nutrients from the ventricular lumen.

854
Q

Of MIs caused by a right coronary obstruction, what proportion affect which parts of the heart?

A
  • 15-30% extend from the posterior free wall of the septal portion of the left ventricle into the adjacent right ventricular wall.
  • Isolated infarction of the right ventricle is unusual (only 1-3% of cases), as is infarction of the atria.
855
Q

What are the frequencies of involvement of each of the three main arterial trunks and the correspondint sites of myocardial lesions resulting in infarction (in the typical right dominant heart)?

A
  • Left anterior descending coronary artery (40-50%): infacrts involving the anterior wall of left ventricle near the apex, the anterior portion of the ventricular septum; and the apex circumferentially
  • Right coronary artery (30-40%): infarcts involving the inferior/posterior wall of left ventricle; posterior portion of ventricular septum; and the inferior/posterior right ventricular free wall in some cases
  • Left circumflex coronary artery (15-20%): infarcts involving the lateral wall of left ventricle except at the apex
856
Q

What are the gross morphological changes in MI over time?

A

0-4 hours - none
4-24 hours - dark mottling
1-3 days - mottling with yellow-tan infarct center
3-7 days - hyperemic border; central yellow-tan softening
7-10 days - maximally yellow-tan and soft, with depressed red-tan margins
10-14 days - red-gray depressed infarct borders
2-8 weeks - gray-white scar, progressive from border towards core of infarct
>2 months - scarring complete

857
Q

What are the light microscope morphological changes over time following an MI?

A

0-4 hours - usually none; variable waviness of fibers at border
4-12 hours - early coagulation necrosis; edema; haemorrhage
12-24 hours - ongoing coagulation ecrosis; pyknosis of nucli; myocyte hypereosinophilia; marginal contraction band necrosis; early neutrophilic infiltrate
1-3 days - coagulation necrosis, with loss of nuclei and striations; brisk intersititial infiltrate of neutrophils
3-7 days - beginning disintegration of dead myofibers, with dying neutrophils; early phagocytosis of dead cells by macrophages at infarct border
7-10 days - well-developed phagocytosis of dead cells; granulation tissue at margins
10-14 days - well-established granulation tissue with enw blood vessels and collagen deposition
2-8 weeks - increased collagen deposition, with decreased cellularity
>2 months - dense collagenous scar

858
Q

What are the morphological changes seen on an electron microscope after an MI?

A

0-30 minutes - relaxation of myofibrils; glycogen loss; mitochondrial swelling
30 minutes - 4 hours - sarcolemmal disurption; mitochondrial amorphous densities

859
Q

What is an overview of the progressive sequence of morphological changes following an MI?

A

They involve typical ischaemic coagulative necrosis (although apoptosis may also occur), followed by inflammation and repair that closely parallels responses to injury in other tissues

860
Q

Early morphological recognition of acute MI can be difficult. But if the infarct preceded death by 2-3 hours, how is it possible to highlight the area of necrosis? What does this show?

A

By immersion of tissue slices in a solution of triphenyl-tetrazolium chloride. This stain imparts a brick-red colour to intact, noninfarcted myocardium where LDH activity is preserved. Because dehydrogenases leak out through the damaged membranes of dead cells, an infarct appears as an unstained pale zone

861
Q

Once an MI lesion is completely healed, can you determine its age?

A

No, the dense fibrous scar of 8-week-old and 10-year-old infarcts look virtually identical

862
Q

What is cardiac reperfusion?

A

Reperfusion is the restoration of blood flow to ischaemic myocardium threatened by infarction; the goal is to slavage cardiac muscle at risk and limit infarct size

863
Q

How can prompt cardiac reperfusion be accomplished after an MI?

A

By a host of coronary interventions, that is , thrombolysis, angioplasty, stent placement, or CABG surgery. The foal is to dissolve, mechanically alter or bypass the lesion that precipitated the acute infarction.

864
Q

What do the benefits of reperfusion following an MI correlate with?

A

1) the rapidity of re-establishing coronary blood (the first 3-4 hours following obstruction are critical)
2) the extent of restoration of blood flow and correction of the underlying causeal lesion

865
Q

What are the mechanisms of reperfusion of thrombolysis, PCI and CABG? How do they differ?

A
  • Thrombolysis can remove a thrombus, but does not alter the underlying atherosclerotic plaque that initiated it
  • PCI with stent placement not only eliminated a thrombotic occlusion but also relieves some of the original obstruction and instability caused by the underlying disrupted plaque
  • CABG provides a new conduit for flow bypassing the area of vessel blockage
866
Q

Reperfused MIs are usually hemorrhagic. Why?

A

Because the vascular is injured during ischaemia and there is bleeding after flow is restored.

867
Q

What does microscopic examination of reperfused cardiac muscle reveal?

A
  • Irreversibly injured myocytes exhibit contraction bands, intensely eosinophilic intracellular “stripe” composed of closely packed sarcomeres
  • These result from the exaggerated contraction of sarcomeres when perfusion is re-established, at which time the interior of cells with damaged membranes is exposed to a high concentration of calcium ions from the plasma
  • Thus, **reperfusion not only salvages reversibly injured cells but also alters the morphology of lethally injured cells.
868
Q

Although clearly beneficial, reperfusion can trigger deleterious complications such as what?

A

Arrhythmias, as well as damage superimposed on the original ischaemia, so-called reperfusion injury.

869
Q

What is reperfusion injury?

A

This term encompasses various forms of damage that can occur after restoration of flow to “vulnerable” myocardium that is ischaemic but not yet irreversibly damaged

870
Q

What causes reperfusion injury and what are the results of it?

A
  • Reperfusion injury may be mediated by oxidative stress, calcium overload, and inflammatory cells recruited after tissue reperfusion
  • Reperfusion-induced microvascular injury not only results in haemorrhage but can also cause endothelial swelling that occludes capillaries and may limit the reperfusion of criticall injured myocardium (called no-reflow)
871
Q

What is a stunned myocardium? How long does it last?

A

Biochemical abnormalities (and their functional consequences) may also persist for days to weeks in reperfused myocytes. Such changes are thought to underlie a phenomenon referred to as stunned myocardium, a state of prolonged cardiac failure induced by short-term ischaemia that usually recovers after several days

872
Q

What is hibernating myocardium? How do you get rid of it?

A

Myocardium that is subjected to chronic sublethal ischaemia may also enter into a state of lowered metabolism and function called hibernation.
Subsequent revascularisation (e.g. by CABG surgery, angiopasty or stenting) often restores normal function to such hibernating myocardium.

873
Q

What are the classic clinical features of an MI?

A
  • Patients with MI characteristically present with prolonged (>30 minutes) chest pain described as crushing, stabbing, or squeezing, associated with a rapid, weak pulse; profuse sweating (diaphoresis), and nausea and vomiting are common, and can suggest involvement of the posterior-inferior ventricle with secondary vagal stimulation.
  • Dyspnoea due to impaired contractility of the myocardium and the resultant pulmonary congestion and edema is a frequent symptom.
874
Q

The laboratory evaluation of MI is based on measuring the proteins that leak out of irreversibly damaged myocytes. What are these?

A
  • The most useful are cardiac specific troponins T and I, and the MB fraction of CK.
  • The diagnosis of MI is established when blood levels of these cardiac biomarkers are elevated.
875
Q

What does the rate of appearance of cardiac markers in the peripheral circulation following MI depend on?

A
  • their intracellular location and molecular weight
  • the blood flow and lymphatic drainage in the area of the infarct
  • the rate of elimination of the marker from the blood
876
Q

Following MI, how does the troponin level change over time?

A
  • Troponins I and T are not normally detectable in the circulation.
  • Following an MI, levels of both begin to rise at 3-12 hours
  • cTnT levels peak somewhere between 12-46 hours
  • cTnI levels are maimal at 24 hours
877
Q

Creatinine kinase is an enzyme expressed in brain, myocardium and skeletal muscle. It is a dimer composed of two isoforms. What are they and where are they found?

A
  • It is a dimer composed of two isoforms designated “M” and “B”.
  • MM homodimers are found predominantly in cardiac and skeletal muscle
  • BB homodimers in brain, lung and may other dissues
  • MB heterodimers are principally localised to cardiac muscle. Thus, the MB form of CK (CK-MB) is sensitive but not specific, sincis can also be elevated after skeletal muscle injury
878
Q

When do CK-MB, cTnI and cTnT return to normal following an MI?

A
  • CK-MB returns to normal in 48-72 hours
  • cTnI in 5-10 days
  • cTnT in 5-14 days
879
Q

What are factors associated with a poorer prognosis of MI?

A
  • Advanced age
  • Female gender
  • Diabetes mellitus
  • Previous MI
880
Q

How long after an MI do most deaths occur? Why/

A
  • Half of the deaths associated with acute MI occur within 1 hour of onset
  • Most commonly due to a fatal arrhythmia; most of the individuals never reach the hospital
881
Q

Mi therapeutic interventions include what?

A
  • Morphine to relieve pain and improve dyspneic symptoms
  • Prompt reperfusion to salvage myocardium
  • Antiplatelet agents such as aspirin
  • Anticoagulant therapy with unfractionated heparin LMWH
  • Nitrates to induce vasodilation and reverse vasospasm
  • Beta blockers to decrease myocardial oxygen demand and to reduce risk of arrhythmias
  • Antiarrhythmics to manage arrhythmias
  • ACE inhibitors to limit ventricular dilation
  • Oxygen supplementation to improve blood oxygen saturation
882
Q

Despite interventions, many patients have one of more of what complications following an MI?

A
  • Contractile dysfunction - MIs produce abnormalities in left ventricular function roughly proportional to their size.
  • Arrhythmias - many patients have myocardial irritability and/or conduction disturbances following MI that lead to potentially fatal arrhythmias
  • Myocardial rupture - the various forms of cardiac rupture typically occur when there is transmural necrosis of a ventricle
  • Ventricular aneurysm - true aneurysms of the ventricular wall are bounded by myocardium that has become scarred
  • Pericarditis - a fibrinous or fibrinohaemorrhagic pericarditis can occur (Dressler syndrome)
  • Infarct expansion - as a result of the weakening of necrotic muscle, there may be disproportionate stretching, thinning, and dilation of the infarct region, which is often associated with mural thrombus
  • Mural thrombus - the combination of a local abnormality in contractility (causing stasis) and endocardial damage (pro-thrombotic) can cause thrombus
  • Papillary muscle dysfunction
  • Progressive late heart failure
883
Q

How often does cardiogenic shock follow an MI? and what type of infarcts? What is the prognosis of this?

A
  • Severe “pump failure” (cardiogenic shock) occurs in 10-15% of patients following acute MI, generally with large infarcts involving more than 40% of the LV
  • Cardiogenic shock has a nearly 70% mortality rate
  • Right ventricular infarcts can cause right-sided heart failure associated with pooling of blood in the venous circulation and systemic hypotension
884
Q

What are some MI-associated arrhythmias?

A
  • Sinus bradycardia
  • AF
  • Heart block
  • Tachycardia
  • Ventricular premature contractions
  • VT
  • VF
885
Q

What type of MIs are associated with arrhythmias?

A

Because of the location of portions of the AV conduction system (bundle of His) in the inferoseptal myocardium, infarcts involving this site can also be associated with heart block

886
Q

What are the different types of cardiac rupture that occur post-MI?

A
  • Rupture of the ventricular free wal (most common), with haemopericardium and cardiac tamponade (A)
  • Rupture of the ventricular septum (less common), leading to an acute VSD and left-to-right shunting (B)
  • Papillary muscle rupture (least common), resulting in the acute onset of severe mitral regurgitation (C)
887
Q

How long after an MI does free-wall rupture occur? Where in the heart is at the most risk?

A
  • Free-wall rupture occurs most frequently 2 to 4 days after MI, when coagulative necrosis, neutrophilic infiltration, and lysis of the myocardial connective tissue have appreciably weakened the infarcted myocardium.
  • The anterolateral wall at the mid-ventricular level is the most common site
888
Q

What are some risk factors for free wall rupture post-MI?

A

Age >60
First MI
Large, transmural and anterior MI
Absence of LVH
Pre-existing frequency

889
Q

Why is ventricular rupture less common after your first MI?

A

Because associated fibrotic scarring tends to inhibit myocardial tearing

890
Q

What is a false aneurysm after a myocardial rupture? What does it consist of?

A

While acute free-wall ruptures are usually rapidly fatal, a fortuitously lcoated pericardial adhesion can abort a rupture and result in a false aneurysm (localised haematoma communicating with the ventricular cavity). The wall of a false aneurysm consists only of epicardium and adherent parietal pericardium and thus may still ultimately rupture

891
Q

Aneurysms of the ventricular wall are a late complication of what type of infarct? How do they occur and what are their complications?

A
  • They are a late complication of large transmural infarcts that experience early expansion
  • The thin scar tissue wall of an aneurysm pardoxically bulges during systole.
  • Complications of ventricular aneurysms include mural thrombus, arrhythmias, and heart failure; rupture of the tough fibrotic wall does not usually occur
892
Q

When does pericarditis often occur after MI?

A

About the second or third day following a transmural infarct as a result of underlying myocardial inflammation (Dressler sydrome)

893
Q

Why does papillary muscle dysfunction occur following MI?

A

Although papillary muscle rupture after an MI may certainly result in precipitous onset of mitral (or tricuspid) valve incompetence, most post-infarct regurgitation results from ischaemic dysfunction of a papillary muscle (and underlying myocardium), or later from ventricular dilation or from papillary msucle fibrosis and shortening

894
Q

The risk of postinfarct complications and the prognosis of the patient depend primarily on the infarct size, location, and fraction of the wall thickness involved (subendocardial or transmural). What infarcts lead to which complications?

A
  • Large, transmural infarcts yield a higher probability of cardiogenic shock, arrhythmias and late CHF.
  • Patient with anterior transmural infarcts are at greatest risk for free-wall rupture, expanson, mural thrombi and aneurysm
  • Posterior transmural infarcts are more likely to be complicated by conduction blocks, right ventricular involvement, or both; when acute CSDs occur in this area, they are more difficult to manage
895
Q

What is ventricular remodelling post-MI?

A

In addition to the sequence of repair in the infarcted tissues, the noninfarcted segments of the ventricle undergo hypertrophy and dilation; collectively, these changes are termed ventricular remodelling

896
Q

Why does ventricular remodelling occur post-MI? What are the complications of it? what medication helps?

A
  • The compensatory hypertrophy of noninfarcted myocardium is initially haemodynamically beneficial.
  • However, this adaptive effect may be overwhelmed by ventricular dilation (with or without aneurysm) and increased oxygen demand, which can exacerbate ischaemia and depress cardiac function.
  • There may also be changes in ventricular shape and stiffening of the ventricle due to scar formation and hypertrophy that further diminish cardiac output.
  • Some of these deleterious effects appear to be reduced by ACE inhibitors, which lessen the ventricular remodeling that occurs after infarction.
897
Q

Long-term prognosis after MI depends on many factors, but what is the most important?

A

The residual left ventricular function and the extent of any vascular obstructions in vessels that peruse the remaining viable myocardium

898
Q

What is the prognosis of MIs? What have we been doing to improve that?

A
  • The overall total mortality within the first year can be as high as 30%; thereafter, each passing year is associated with an additional 3-4% mortality among survivors
  • Infarct prevention (through control of risk factors) in individuals who have never experienced MI (primary prevention) and prevention of reinfarction in MI survivors (secondary prevention) are important strategies that have received much attention and achieved considerable success.
899
Q

What is chronic IHD/ischaemia cardiomyopathy?

A

A progressive congestive heart failure as a consequence of accumulated ischaemic myocardial damage and/or inadequate compensatory responses.

900
Q

In what situations does chronic IHD occur?

A
  • In most instances, there has been prior MI and sometimes previous coronary arterial interventions and/or bypass surgery
  • Chronic IHD usually appears post-infarction due to the functional decompensation of hypertrophied noninfarcted myocardium. However, in other cases severe obstructive coronary artery disease may present as chronic congestive heart failure in the absence of prior infarction.
  • Patients with chronic IHD account for almost 50% of cardiac transplant receipients.
901
Q

What is the morphology of chronic heart failure?

A
  • Hearts from patients with chronic IHD have cardiomegaly, with left ventricular hypertrophy and dilation.
  • Invariably, there is some degree of stenotic coronary atherosclerosis.
  • Discrete scars representing healed infarcts are usually present.
  • the mural endocardium often has patchy fibrous thickenings (due to abnormal wall shear forces), and mural thrombi may be present.
  • Microscopic findings include myocardial hypertrophy, diffuse subendocardial vacuolisation, and fibrosis.
902
Q

What is infective endocarditis?

A

A microbial infection of the heart valves or the mural endocardium that leads to the formtion of vegetations composed of thrombotic debris and organisms, often associated with destruction of the underlying cardiac tissues.
The aorta, aneurysms, other blood vessels and prosthetic devices can also become infected.

903
Q

What usually causes infective endocarditis?

A

Although funghi and other classes of microorganisms can be responsible, most infections are bacterial (bacterial endocarditis)

904
Q

Traditionally, infective endocarditis has been classified on clinical grounds into acute and subacute forms. This subdivision relfects the range of the disease severity and tempo, which are determined in lareg part by the virulence of the infecting microorganism and whether underlying cardiac disease is present. Tell me about these types.

A
  • Acute infective endocarditis is typically caused by infection of a previously normal heart valve by a highly virulent organism (e.g. Staphylococcus aureus), that rapidly produces necrotising and destructive lesions. These infections may be difficult to cure with antibiotics alone, and usually require surgery. Despite appropriate treatment, death can ensue within days to weeks.
  • Subacute infective endocarditis is characterised by organisms with lower virulence (e.g. viridans streptococci) that cause insidious infections of deformed valves with overall less destruction. In such cases the disease may pursue a protracted course of weeks to months and can be achieved with antibiotics.
905
Q

Although highly virulent organisms can infect previously normal valves, a variety of cardiac and vascular abnormalities increase the risk of developing IE. What are they?

A

Rheumatic heart disease with valvular scarring has historically been the major antecedent disorder; as RHD becomes less common, it has been supplanted by mitral valve prolapse, degenerative calcific valvular stenosis, bicuspid aortic valve (whether calcified or not), artifical (prosthetic) valves, and unrepaired and repaired congenital defects.

906
Q

The causal organisms of infective endocarditis differ among the major high-risk groups. Tell me how…

A
  • Endocarditis of native but previously damaged or otherwise abnormal valves is caused most commonly (50-60% of cases) by Streptococcus viridans, a normal component of the oral cavity flora
  • Most virulent Staphylococcus aureus is the major offender in IE among IVDUs
  • Other bacterial causes include enterococci and the so-called HACEK group (Haemophilus, Actinobacillus, Cardiobacterium, Eikenella and Kingella), all commensals in the oral cavity.
  • Prosthetic valve endocarditis is cause most commonly by coagulase-negative staphylococci (e.g Staph epidermidis.
  • Other agents causing endocarditis include gram-negative bacilli and funghi
907
Q

In about 10% of all cases of infective endocarditis, no organism can be isolated from the blood (“culture-negative” endocarditis). What are the reasons for this?

A
  • Prior antibiotic therapy
  • Difficulties in isoalting the offending agent
  • Because deeply embedded organisms within the enlarging vegetation are not released into the blood
908
Q

Foremost among the factors predisposing to infective endocarditis are those that cause microorganism seeding into the blood stream (bacteraemia or fungaemia). What are some examples it this? How can it be managed?

A

The source may be an obvious infection elsewhere, a dental or srugical procedure, a contaminated needle shared by IVDUs, or seemingly trivial breaks in the epithelial barriers of the gut, oral cavity, or skin.
In patients with valve abnormalities, or with known bacteraemia, IE risk can be lowered by antibiotics prophylaxis.

909
Q

Vegetations on heart valves are the classic hallmark of IE. How do they present morphologically?

A

Vegetations on heart valves are the classic hallmark of IE; these are friable, bulky, potentially destructive lesions containing fibrin, inflammatory cells, and bacteria or other organisms.

910
Q

What valves are the most common sites of infective endocarditis? Is it only one valve they happen in?

A

The aortic and mitral valves are the most common sites of infection, although the valves of the right heart may also be involved, particularly in IVDUs.
Vegetations can be single or multiple and may involve more than one valve.

911
Q

What are some of the consequelae of infective endocarditis vegetations?

A

They can occasionally erode into the underlying myocardium and produce an abscess (ring abscess Fig B).
Vegetations are prone to embolisation; because the embolic fragments often contain virulent organisms, abscesses frequently develop where they lodge, leading to sequelae such as septic infarcts or mycotic aneurysms.

912
Q

How do the vegetations of subacute endocarditis differ from acute endocarditis? Both in terms of destruction and microscopically

A
  • The vegetations of subacute endocarditis are associated with less valvular destruction that those of acute endocarditis, although the distinction can be subtle.
  • Microscopically, the vegetations of subacute IE typically exhibit granulation tissue at their bases indicative of healing. With time, fibrosis, calcification, and a chronic inflammatory infiltrate can develop.
913
Q

What are the clinical features of infective endocarditis?

A
  • Acute endocarditis has a stormy onset with rapidly develpoing fever, chills, weakness, and lassitude. although fever is the most consistent sign of IE, it can be slight or absent, particularly in odler adults, and the only manifestations may be non-specific fatigue, loss of weight, and a flu-like syndrome.
  • Murmurs are present in 90% of patients with left-sided IE< either from a new valvular defect or from a pre-existing abnormality.
914
Q

What is the diagnostic criteria for infective endocarditis called and what are the points in it?

A

Duke’s criteria
Pathological criteria
* Microorganisms, demonstrated by culture or histological examination, in a vegetation, embolus from a vegetation, or intracardiac abscess
* Histological confirmation of active endocarditis in vegetation or intracardiac abscess

Clinical Criteria - major
* Positive blood cultures for a characteristic organism or persistently postive for an unusual organism
* Echo identification of a valve-related or implant-related mass or abscess, or partial separation of artificial valve
* New valvular regurgitation

Clinical Criteria - minor
* Predisposing heart lesion or IVDU
* Fever
* Vascular lesions, including arterial petechiae, subungual/splinter haemorrhages, emboli, septic infarcts, mycotic aneurysm, intracranial heamorrhage, Janeway lesions
* Immunologic phenomena including glomerulonephritis, Osler nodes, Roth spots, rheumatoid factor
* A single postitive culture for an unusual organism
* Echo findings consistent with but not diagnostic of endocarditis, including worsening or changing of a preexistent murmur

915
Q

What are cardiomyopathies?

A

Cardiomyopathies are a hetergenous group of diseases of the myocardium associated with mechanical and/or electrical dysfunction that usually (but not invaruably) exhibit inappropriate ventricular hypertrophy or dilatation and are due to a variety of causes that frequently are genetic.
Cardiomyopathies either are confined to the heart or are part of generalised systemic disorders, often leading to cardiovascular death of progressive heart failure-related disability

916
Q

How do cardiomyopathies manifest?

A

As failure of myocardial performance; this can be mechanical (eg., diastolic or systolic dysfunction) leading to CHF, or can culminate in life-threatening arrhythmias

917
Q

What are primary and secondary cardiomyopathies?

A
  • Primary cardiomyopathies can be genetic or acquired diseases of myocardium
  • Secondary cardiomyopathies have myocardial involvement as a component of a systemic or multiorgan disorder.
918
Q

What are the three pathological patterns of cardiomyopathy? and which ones are most/least common?

A
  • Dilated cardiomyopathy (including arrhythmogenic right ventricula cardiomyopathy) - 90% of cases
  • Hypertrophic cardiomyopathy
  • Restrictive cardiomyopathy - least common
919
Q

What is the left ventricular ejection fraction for the following cardiomyopathies:
* Dilated
* Hypertrophic
* Restrictive

A

Dilated < 40%
Hypertrophic - 50-80%
Restrictive - 45-90%

920
Q

What are the mechanisms of heart failure in the following cardiomyopathies?
* Dilated
* Hypertrophic
* Restrictive

A

Dilated - impairment of contractility (systolic dysfunction)
Hypertrophic - impairment of compliance (diastolic dysfunction)
Restrictive - impairment of compliance (diastolic dysfunction)

921
Q

What are the causes of the following cardiomyopathies:
* Dilated
* Hypertrophic
* Restrictive

A
  • Dilated - genetic, alcohol, peripartum, myocarditis; haemochromatosis; chronic anaemia; sarcoidosis; idiopathic
  • Hypertrophic - genetic ; Friedrich ataxia; storage disease; infants of diabetic mother
  • Restrictive - amyloidosis; radiation-induced fibrosis; idiopathic
922
Q

What are the some indirect myocardial dysfunctions associated with the following cardiomyopathies:
* Dilated
* Hypertrophic
* Restrictive

A
  • Dilated - IHD; valvular heart disease; hypertensive heart disease; congenital heart disease
  • Hypertrophic - hypertensive heart disease; aortic stenosis
  • Restrictive - pericardial constriction
923
Q

What are the characteristics of dilated cardiomypathy?

A

Dilated cardiomyopathy is characterised morphologically and functionally by progressive cardiac dilation and contractile (systolic) dysfunction, usually with concomitant hypertrophy.

924
Q

What percentage of dilated cardiomyopathy are familial? What genes are specific for this?

A
  • DCM is familial in at least 30-50% of cases, in which it is caused by mutations in a diverse group of more than 20 genes encoding porteins involved in the cytoskeleton, sarcolemma and nuclear envelope.
  • In particular, mutations in TTN, a gene that encodes titin, may account for approximately 20% of all cases of dilated cardiomyopathy
925
Q

What is the difference in presentationg ages between dilated cardiomyopathy caused by mitochondrial defects and those that are X-linked?

A
  • Mitochondrial defects typically mannifest in the paediatric population, while X-linked DCM typically presentes after puberty and into early adulthood.
926
Q

How does alcohol cause dilated cardiomyopathy?

A
  • Alcohol abuse is strongly associated with the development of dilated cardiomyopathy, raising the possibility that ethanol toxicity or a secondary nutritional disturbance can underlie myocardial injury.
  • Alcohol or its metabolites (especially acetaldehyde) have a direct toxic effect on the myocardium.
  • Moreover, chronic alcoholism may be associated with thiamine deficiency, which can lead to beriberi heart disease.
927
Q

A special form of dilated cardiomyopathy, termed peripartum cardiomyopathy can occur late in pregnancy or up to months postpartum. What is the mechanism of this?

A
  • The mechanism is poorly understood but is probably multi-factorial. Pregnancy-associated hypertension, volume overload, nutritional deficiency, other metabolic derangements, or immunological reactions have been proposed as causes.
  • The primary defect suggested is a microvascular angiogenic imbalance within the myocardium leading to functional ischaemic injury.
928
Q

How does iron overload cause dilated cardiomyopathy?

A
  • Dilated cardiomyopathy is the most common manifestation of iron overload from either haemochromastosis or from multiple transfusions..
  • It may be caused by interference with metal-dependent enzyme systems or to injury from iron-mediated production of reactive oxygen species.
929
Q
A
930
Q

What is takotsubo cardiomyopathy?

A
  • An entity characterised by left ventricular contractile dysfunction following extreme psychological stress
  • Affected myocardium may be stunned or show multi-focal contraction band necrosis.
  • The left ventricular apex is most often affected leading to “apical ballooning” that resembles a “takotsubo” - Japanese for “fishing pot for trapping octopus”
931
Q

What is the mechanism of carecholamine cardiotoxicity?

A
  • It likely relates either to direct myocyte toxicity due to calcium overload or to focal vasoconstriction in the coronary arterial macro- or microcirculation in the face of an increased heart rate.
932
Q

What is the gross morphology of dilated cardiomyopathy?

A
  • In dilated cardiomyopathy, the heart is usually enlarged, heavy (often weighing 2-3 times normal), and flabby, due to dilation of all chambers.
  • Mural thrombi are common and may be a source of thromboemboli
  • There are no primary valvular alterations; if mitral (or tricuspid) regurgitation is present, it results from left (or right) ventricular chamber dilation (functional regurgitation)
  • Either the coronary arteries are free of significant narrowing or the obstructions present are insufficient to explain the degree of cardiac dysfunction.
933
Q

What are the histological changes of dilated cardiomyopathy?

A
  • The histological abnormalities in DCM are nonspecific and usually do not point to a specific etiology
  • Most muscle cells are hypertrophied with enlarged nuclei, but some are attenuated, stretched, and irregular
  • Interstitial and endocardial fibrosis of variable degree is present, and small subendocardial scars may replace individual cells or groups of cells, probably reflecting healing of previous ischaemic necrosis of myocytes caused by hypertrophy-induced imbalance between perfusion and demand
  • Morever, the severity of morphological changes may not reflect either the degree of dysfunction or the patient’s prognosis
934
Q

At what age does dilated cardiomyopathy often occur?

A

It can occur at any age, including in childhood, but it mostly commonly affects individuals between the ages of 20 and 50.

935
Q

How does dilated cardiomyopathy tend to present?

A
  • It presents with slowly progressive signs and symptoms of chronic heart failure including dyspnoea, easy fatiguability, and poor exertional capacity
  • At the end stage, ejection fractions are typically less than 25%
  • Secondary mitral regurgitation and abnormal cardiac rhythms are common, and embolism from intracardiac thrombi can occur.
936
Q

How does death usually occur from dilated cardiomyopathy? How is it treated?

A
  • Death usually results from progressive cardiac failure or arrhythmia, and can occur suddenly
  • Although the annual mortality is high (10-50%), some severely affected patients respond well to pharmacologic therapy
  • Cardiac transplantation is also increasingly performed, and long-term ventricular assis can be beneficial.
937
Q

What is arrhythmogenic right ventricular cardiomyopathy? What does it cause?

A
  • Arrhythmogenic right ventricular cardiomyopathy is an inherited disease of myocardium causing right ventricular failure and rhythm distrubances (particularly ventricular tachycardia or fibrillation) with sudden death
  • Left sided involvement with left-sided heart failure may also occur.
938
Q

What is the morphology of arrhythmogenic right ventricular cardiomyopathy?

A
  • Morphologically, the right ventricular wall is severely thinned due to loss of myocytes, accompanied by extensive fatty infiltration and fibrosis.
  • Although myocardial inflammation may be present, ARVC is nor considered an inflammatory cardiomyopathy.
939
Q

What are the genetic links associated with arrhythmogenic right ventricular cardiomyopathy?

A
  • Classic ARVC has autosomal dominant inheritance with a variable penetrance.
  • The disease has been attributed to defective cell adhesion proteins in the desmosomes that link adjacent cardiac myocytes.
  • Naxos syndrome is a disorder characterised by arrhythmogenic right ventricular cardiomyopathy and hyperkeratosis of plantar palmar skin surfaces specifically associated with mutations in the gene encoding the desmosome-associated protein plakoglobin.
940
Q

What is hypertrophic cardiomyopathy? What is it’s incidence?

A

Hypertrophic cardiomyopathy (HCM) is a common (incidence, 1 in 500), clinically heterogenous, genetic disorder characterised by myocardial hypertrophy, poorly compliant left ventricular myocardium leading to abnormal diastolic filling, and (in about 1/3 of cases) intermittent ventricular outflow obstruction.

941
Q

What are the most common diseases that must be distinguished clinically from hypertrophic cardiomyopathy?

A
  • Depositiondiseases such as amyloidosis, Fabry disease
  • Hypertensive heart disease coupled with age-related subaortic septal hypertrophy.
  • Occasionally, valvular or congenital subvalvular aortic stenosis can also mimic HCM
942
Q

What are the genetic links associated with hypertrophic cardiomyopathy?

A
  • In most cases, the pattern of transmission is autosomal dominant with variable penetrance.
  • HCM is caused by mutations in any one of the several genes that encode sarcomeric proteins; there are more than 400 different known mutations in nine different genes, most being missense mutations
  • Mutations causing HCM are found most commonly in the gene encoding β-myosin heavy chains, following by genes coding for cardiac TnT, α-tropomyosin, and myosin-binding protein C.
  • The prognosis of HCM varies widely and correlates strongly with specific mutations
943
Q

What is the gross morphology of hypertrophic cardiomyopathy?

A
  • The essential feature of HCM is massive myocardial hypertrophy, usually without ventricular dilation.
  • The classic pattern involves disproportionate thickening of the ventricular septum relative to the left ventricle free wall (with a ratio of septum to free wall greater than 3:1), termed asymmetric septal hypertrophy. In about 10% of cases, the hypertrophy is concentric and symmetrical.
  • On longitudinal sectioning, the normally round-to-ovoid left ventricular cavity may be compressed into a “banana-like” configuration by bulging of the ventricular septum into the lumen.
  • The left ventricular outflow tract often exhibits a fibrous endocardial plaque associated with thickening of the anterior mitral leaflet.
944
Q

In hypertrophic cardiomyopathy, where is the hypertrophy?

A

Although marked hypertrophy can invovle the entire spetum, it is usually most prominent in the subaortic region.

945
Q

In hypertrophic cardiomyopathy, the left ventricular outflow tract often exhibits a fibrous endocardial plaque associated with thickening of the anterior mitral leaflet. Why? Will you see this on echo?

A

Both findings result from contact of the anterior mitral leaflet with the septum during ventricular systole; they coorelate with the echocardiographic “systolic anterior motion” of the anterior leaflet, with functional left ventricular outflow tract obstruction during mid-systole.

946
Q

What are the most important histological features of the myocardium in hypertrophic cardiomyopathy?

A

1) massive myocyte hypertrophy, with transverse myocyte diametes frequently greater than 40μm (normal, approx 15μm)
2) haphazard disarray of bundles of myocytes, individual myocytes, and contractile elements in sarcomeres within cells (termed myofiber disarray)
3) interstitial and replacement fibrosis

947
Q

What in the pathological changes that occur in hypertrophic cardiomyopathy? How does this present clinically?

A
  • The central abnormalities in HCM is reduced stroke volume due to impaired diastolic filling.
  • This is a consequence of a reduced chamber size, as well as the reduced compliance of the massively hypertrophied left ventricle.
  • In addition, approximately 25% of patients with HCM have dynamic obsutrction to the left ventricular outflow
  • The compromsied cardiac output in conjunction with a secondary increase in pulmonary venous pressure explains the exertional dyspnoea seen in these patients.
948
Q

What do you hear on auscultation of hypertrophic cardiomyopathy? Why?

A

Auscultation discloses a harsh systolic ejection murmur, caused by the ventricular outflow obstruction as the anterior mitral leaflet moves toward the ventricular septum during systole.

949
Q

Why does focal myocardial ischaemia commonly occur in hypertrophic cardiomyopathy, even in the absence of concomitant coronary artery disease?

A

Because of the massive hypertrophy, high left ventricular chamber pressure, and frequently thick-walled intramural arteries.

950
Q

What are the major clinical problems in hypertrophic cardiomyopathy?

A

AF, mural thrombus formation leading to embolisation and possible stroke, intractable cardiac failure, ventricular arrhythmias, and not infrequently, sudden death, especially with certain specific mutations.

951
Q

What is the course of progression of hypertrophic cardiomyopathy? How is it treated?

A
  • The natural history of HCM is highly variable.
  • Most patients can be helped by pharmacological intervention (eg, β-blockers) to decrease heart rate and contractility.
  • Some benefit can also be gained by reducing the septal myocardial mass, thus relieving the outflow tract obstruction. This can be achieved either by surgical excision of muscle or by carefully controlled septal infarction through a catheter-based infusion of alcohol
952
Q

What are the characteristics of restrictive cardiomyopathy? What can it be confused with?

A
  • It is characterised by a primary decrease in ventricular compliance, resulting in impaired ventricular filling during diastole.
  • Because the contractile (systolic) function of the left ventricle is usually unaffected, the function abnormality can be confused with that of constrictive pericarditis or HCM.
953
Q

What diseases in restrictive cardiomyopathy associated with?

A

It can be idiopathic or associated with distinct diseases or processes that affect the myocardium, pricipally radiation fibrosis, amyloidosis, sarcoidosis, metastatic tumours, or the deposition of metabolites that accumulate due to inborn errors of metabolism.

954
Q

What are the gross morphological features of restrictive cardiomyopathy?

A
  • The morphological features are not distinctive
  • The ventricles are of approximately normal size or slightly enlarged, the cavities are not dilated, and the myocardium is firm and non-compliant.
  • Biatrial dilation is commonly observed.
955
Q

What are the microscopic features of restrictive cardiomyopathy?

A

Microscopically, there may be only patchy or diffuse interstitial firbosis, which can vary from minimal to extensive.
Endomyocardial biopsy can often reveal a specific etiology.

956
Q

What are some important subgroups of restrictive cardiomyopathy?

A
  • Amyloidosis
  • Endomyocardial fibrosis
  • Loeffler endomyocarditis
  • Endocardial fibroelastosis
957
Q

What is endomyocardial fibrosis? Where is it common?

A

Endomyocardial fibrosis is principally a disease of children and young adults in Africa and other tropical areas, characterised by fibrosis of the ventricular endocardium and subendocardium that extends from the apex upward, often involving the tricuspid and mitral valves.

958
Q

What are the pathological outcomes of endomyocardial fibrosis?

A
  • The fibrous tissue markedly diminishes the volume and compliance of affected chambers and so causes a restrictive functional defect.
  • Ventricular mural thrombi sometimes develop, and indeed the endocardial fibrosis may result from thrombus organisation
959
Q

What is the pathological sequence of Loeffler endomyocarditis? How is it similar and different to endomyocardial fibrosis?

A
  • It results in endomycocardial fibrosis, typically with large mural thrombi, with an overall morphology similar to the tropical disease.
  • However, in addition to the cardiac changes, there is often a peripheral eosinophilia and eosinophilic infiltrates in multiple organs, including the heart.
  • The release of toxic products of eosinophils especially major basic protein, is postulated to initiate endomyocardial necrosis, followed by scarring of the necrotic area, layering of the endocardium by thrombus and finally organisation of the thrombus.
960
Q

What disease is associated with Loeffler endomyocarditis and how do we treat it? What happens if we dont?

A
  • Many patients with Loeffler endomyocarditis have a myeloproliferative disorder associated with chromosomal rearrangements involving either the platelet-derived growth factor receptors (PDGFR)-α or -β genes.
  • These rearrangmenets produce fusion genes that encode constitutively active PDGFR tyrosine kinases.
  • Treatment of such patients with the tyrosine kinase inhibitor imatinib has resulted in haematologic remissions associated with reversal of the endomyocarditis, which is otherwise often rapidly fatal.
961
Q

What is endocardial fibroelastosis? At what age is it most common and in what sub-type of the age?

A
  • It is an uncommon heart disease characterised by fibroelastic thickening that typically involved the left ventricular endocardium.
  • It is most common in the first 2 years of life; in a third of cases, it is accomopanied by aortic valve obstruction or other congenital cardiac anomalies?
962
Q

Endocardial fibroelastosis may actually represent a common morphological end-point of several different insults. What do these include?

A
  • Viral infections (eg, intrauterine exposure to mumps)
  • Mutations in the gene for tafazzin, which affected mitochondrial inner membrane integrity
  • Diffus involvement may be responsible for rapid and progressive cardiac decompensation and death
963
Q

What is myocarditis?

A

A diverse group of pathological entities in which infectious microorganisms and/or a primary inflammatory process causes myocardial injury.

964
Q

What are the major causes of myocarditis?

A
  • In the US, viral infections are the most common cause of myocarditis. Coxsackie viruses A/B and other enteroviruses account for most causes.
  • Other less common etiologic agents include CMV, HIV, influenza, chlamydia, rickets, meningococcus, candida
  • Immune-mediated reactions that cause it are post-viral, post-streptococcal (rheumatic fever), SLE, drug hypersensitivity and transplant rejections
  • Other cuases include sarcoidosis, giant cell myocarditis.
965
Q

How can a virus cause myocarditis?

A
  • Depending on the pathogen and the host, viruses can potentially cause myocardial injury either as a direct cytopathic effect, or by eliciting a destructive immune response.
  • Inflammatory cytokines produced in response to myocardial injury can also cause myocardial dysfunction that is out of proportion to the degree of actual myocyte damage.
966
Q

What is the gross morphology of myocarditis?

A
  • Grossly, the heart in myocarditis may appears normal or dilated; some hypertrophy may be present depending on disease duration.
  • In advanced stages, the ventricular myocardium is flabby and often mottled by either pale focci or minute haemorrhagic lesions.
  • Mural thrombi may be present
967
Q

What is the histological morphology of myocarditis?

A
  • Active myocarditis is characterised by an interstitial inflammatory infiltrate associated with focal myocyte necrosis.
  • A diffuse, mononuclear, predominantly lymphocytic infiltrate is most common.
  • Although endomyocardial biopsies are diagnostic in some causes, they can be spuriosly negative because inflmmatory involvement of the myocardium may be focal or patchy.
  • If the patient survives the acute phase, the inflammatory lesions either resolve, leaving no residual changes, or heal by progressive fibrosis
968
Q

Nonviral agents are also an important cause of infectious myocarditis, particularly the protozoan Typanosoma cruzi. What disease does this cause? Where? What is its prognosis?

A

Chagas disease is endemic in some regions of South America, with myocardial involvement in most infected individuals.
About 10% of patients die during an acute attack; others develop a chronic immune-mediated myocarditis that may progress to cardiac insufficiency in 10-20 years.

969
Q

What is the histological morphology of immune-mediated/hypersensitivity myocarditis?

A

Hypersensitivity myocarditis has interstitial infiltrates, principalle perivascular, composed of lymphocytes, macrophages, and a high proportion of eosinophils

970
Q

What is the histological morphology of giant-cell myocarditis? What is its prognosis?

A
  • It is characterised by a widespread inflammatory cellular infiltrate containing multinucleate giant cells (fused macrophages) interspersed with lymphocytes, eosinophils, plasma cells, and macrophages.
  • Focal to frequently extensive necrosis is present.
  • This variant likely represents the fulminant end of the myocarditis spectrum and carries a poor prognosis
971
Q

What is the histological morphology of Chagas disease myocarditis?

A

It is distinctive by virute of the parasitisation of scattered myofibers by trypanosomes accompanied by a mixed inflammatory infiltrate of neutrophils, lymphocytes, macrophages, and occasional eosinophils

972
Q

What are the clinical features of cardiomyopathy?

A
  • The clinical spectrum of myocarditis is broad.
  • At one end, the disease is entirely asymptomatic, and patients can expect a complete recovery without sequelae.
  • At the other extreme is the precitious onset of heart failure or arrhtyhmias, occasionally with sudden death,
  • Between these extremes are the many levels of involvement associated with symptoms such as fatigue, dyspnoea, palpitations, precordial discomfort and fever
  • Patients can develop dilated cardiomyopathy as a late complication of myocarditis.
973
Q

Which cardiotoxic cancer drugs can cause myocardial disease? What do they cause? At what levels is the most common one?

A
  • Cardiotoxicity has been associated with conventional chemotherapeutic agents, tyrosine kinase inhibitors, and certain forms of immunotherapy.
  • The anthracyclines doxorubicin and daunorubicin are the chemotherapeutic agents most often associated with toxic myocardial injury; they cause dilated cardiomyopathy with heart failure attributed primarily to peroxidation of lipids in myocyte membranes.
  • Anthra cycline toxicity is dose-dependent, with the cardiotoxicity risk increasing when cumuluative life-time doses exceed 500mg/m2
974
Q

Many therapeutic agents, including lithium, phenothiazine and chloroquine can idiosyncractically induce myocardial injury and sometimes death. What are some common histological findings in affected myocardium? How do you treat it?

A
  • Myofiber swelling, cytoplasma vacuolisation, and fatty change.
  • Discontinuining the offending agent often leads to prompt resolution, without apparent sequelae.
  • Occasionally, however, more extensive damage produces myocyte necrosis that can evolve to a dilated cardiomyopathy.
975
Q

What is pathogenesis of amyloidosis?

A

Amyloidosis results from the extracellular accumulation of protein fibrils that are prone to forming insoluble β-pleated sheets.

976
Q

How can cardiac amyloidosis occu?

A

It can appear as a consequence of systemic amyloidosis (eg, due to myeloma or inflammation-associated amyloid) or can be restricted to the heart, particularly in the elderly (senile cardiac amyloidosis)

977
Q

Who does senile cardiac amyloidosis occur in? What is the prognosis?

A

Senile cardiac amyloidosis characteristically occurs in individuals over 70 years old, and has a far better prognosis that systemic amyloidosis

978
Q

What is the pathogensis of senile cardiac amyloidosis?

A
  • Senile cardiac amyloid deposits are largely composed of transthyretin, a normal serum protein synthesised in the liver that transports thyroxine and retinol-binding protein.
  • Mutant formed of transthyretin can accelerate the cardiac (and associated systemic) amyloid deposition.
979
Q

What are the clinical features of cardiac amyloidosis?

A
  • Cardiac amyloidosis most frequently produces a restrictive cardiomyopathy, but it can also be asymptomatic, manifest as filation or arrhythmias, or mimic ischaemic or valvular disease.
  • The varied presentations depend on the predominant location of the deposits, for example, intersititium, conduction system, vasculature, or valves.
980
Q

What is the gross morphology of cardiac amyloidosis?

A

In cardiac amyloidosis the heart varies in consistency from normal to firm and rubbery.
The chambers are usually of normal size, but can be dilated and have thickced walls.
Small, semitranslucent nodules resembling drips of wax may be seen on the atrial endocardial surface, particularly on the left.

981
Q

What is the histology of cardiac amyloidosis?

A
  • Histologically, hyaline eosinophilic deposits of amyloid may be found in the interstitium, conduction tissues, valves, endocardium, pericardium, and small intramural coronary arteries
  • They can be distinguished from other deposits by special stains such as Congo red, which produces classic apple-green bifriendence when viewed under polarised light.
  • Intramural arteries and arterioles may have sufficient amyloid in their walls to compress and occlude their lumens, inclduing myocardial ischaemia
982
Q

What are the most important pericardial disorders?

A

Ones that cause fluid accumulation, inflammation, fibrous constriction or some combination of these processes, usually in association wiht other cardiac pathology or a systemic disease

983
Q

What are the different types of pericardial effusion? (Give me the normal volume of fluid in the pericardium).

A
  • Normally, the pericardial sac contains less than 50mL of thin, clear, straw-coloured fluid
  • Under various circumstances that parietal pericardium may be distended by serous fluid (pericardial effusion), blood (haemopericardium) or pus (purulent pericarditis)
984
Q

What is the difference between a chronic pericardial effusion and an acute one?

A
  • With long-standing cardiac enlargement or with slowly accumulating fluid, the pericardium has time to dilate. This permits a slowly accumulating pericardial effusion to become quite large without interfering with cardiac function. thus, with chronic effusions of less than 500mL, the only clinical significance is a characteristic globular enlargement of the heart shadow of chest radiographs.
  • In contract, rapidly developing fluid collections of as little as 200-300mL (eg, due to haemopericardium caused by a ruptured MI or aortix dissection) can produce clinically devastating compression of the thin-walled atria and venae cavae, or the ventricles themselves; cardiac filling is thereby restricted, producing potentially fatal cardiac tamponase
985
Q

Pericarditis can be split into primary and secondary. What are they and what causes them?

A
  • It can occur secondary to a variety of cardiac, thoracic, or systemic disorders, metastases from remote neoplasms, or cardiac surgical procedures
  • Primary percarditis is unusual and almost always of viral origin
986
Q

What are the different causes of percarditis?

A

Infectious agents
viruses
pyogenic bacteria
TB
funghi
Presumable immunologically mediated
rheumatic fever
SLE
scleroderma
postcardiotomy
post-MI (Dressler’s syndrome)
drug hypersensitivity reaction
Miscellanous
MI
uraemia
post-cardiac surgery
neoplasia
trauma
radiation

987
Q

What are the two types of acute percarditis?

A

Serous and fibrinous

988
Q

What causes serous pericarditis? How?

A
  • It is characteristically produced by non-infectious inflammatory diseases, including rheumatic fever, SLE, and scleroderma, as well as tumours or uraemia.
  • An infection in the tissues contiguous to the pericardium may incite sufficient irritation of the parietal pericardial serosa to cause a sterile serous effusion that can progress to serofibrinous pericarditis and ultimately to frank suppurative reaction.
  • In some instances a well-defined viral infection elsewhere - URTI, pneumonia, parotitis - antedates the percarditis and serves as the primary focus of infection
989
Q

In what age group does primary pericarditis often present?

A

Infrequently, usually in young adults, a viral pericarditis occurs as an apparent primary infection that may be accompanied by myocarditis

990
Q

How can tumours cause a serous pericarditis?

A

Tumours can cause a serous pericarditis by lymphatic invasion or direct contiguous extension into the pericardium.

991
Q

What is the histological morphology of acute serous pericarditis?

A

Histologically, serous pericarditis elicits a mild inflammatory infiltrate in the epipericardial fat consisting predominantly of lymphocytes; tumour-associated pericarditis may also exhibit neoplastic cells. Organisation into fibrous adhesions rarely occurs.

992
Q

What is fibrinous and serofibrinous pericarditis? Are they common? What causes them?

A
  • They are the most frequent types of pericarditis
  • They are composed of serous fluid variably admixed with a fibrinous exudate.
  • Common causes include acute MI, postinfarction (Dressler) syndrome, uraemia, chest radiation, rheumatic fever, SLE, and trauma. A fibrinous reaction also follows routine cardiac surgery
993
Q
A
994
Q

What is the morphology of fibrinous and serofibrinous pericarditis?

A
  • In fibrinous pericarditis the surface is dry, with a fine granular roughening.
  • In serofibrinous pericarditis a more intense inflammatory process inuced the accumulation of larger amounts of yellow to brown turbid fluid, conatining leukocytes, erythrocytes, and fibrin.
  • As this all inflammatory exudates, fibrin may be lysed with resolution of exudate, or can become organised.
995
Q

What are the symptoms of fibrinous pericarditis?

A
  • They characteristically include pain (sharp, pleuritic, and position dependent) and fever; congestive failure may also be present.
  • A loud pericardial friction rub is the most striking clinical finding.
996
Q

Purulent or suppurative pericarditis reflects an active infection caused by microbial invasion of the pericardial space. How can this occur?

A

Through:
* direct extension from neighbouring infections, such as empyema of the pleural cavity, lobar pneumonia, mediastinal infections, or extension of a ring abscess through the myocardium or aortic root
* seeding from the blood
* lymphatic extension
* direct introduction during cardiotomy.

997
Q

What are the gross morphological features or purulent pericarditis?

A
  • The exudate ranges from a thin cloudy fluid to frank pus up to 400-500mL in volume
  • The serosal surfaces are reddened, granular, and coated with the exudate.
998
Q

What are the microscopic features of purulent pericarditis?

A

Microscopically, there is an acute inflammatory reaction, which sometimes extends into surrounding structures to induce mediastinopericarditis.

999
Q

What are the clinical sequelae of purulent pericarditis?

A
  • Complete resolution is infreuent, and organised by scarring is the usual outcome
  • The intense inflammatory response and the subsequent scarring frequently produce constrictive pericarditis, a seious consequence
1000
Q

What is haemorrhagic pericarditis? What is it commonly caused by?

A
  • It has an exudate composed of blood mixed with a fibrinous or suppurative effusion
  • It is most commonly caused by the spread of a malignant neoplasm to the pericardial space. In such cases, cytologic examination of fluid removed through a pericardial tap often reveals neoplastic cells.
  • It can also be found in bacterial infections, in patients with an undering bleeding diathesis, and in TB.
  • It often follows cardiac surgery and os occasionally responsible for significant blood loss of even tamponade, requiring re-operation.
1001
Q

What is chronic pericarditis?

A
  • In some cases organisation merely produces plaque-like fibrous thickenings of the serosal membranes (“soldier’s plaque”) or thin, delicate adhesions that rarely cause impairment of cardiac function.
  • In other cases, fibrosis in the form of mesh-like stringy adhesions completely obliterates the pericardial sac. In most instances, this adhesive pericarditis has not effect on cardiac function.
1002
Q

What is adhesive mediastinopericarditis? When does it happen?

A
  • Adhesive mediastinopericarditis may follow infectious pericarditis, previous cardiac surgery, or mediastinal irradiation.
  • The pericardial sac is obliterated, and adherence of the external aspect of the parietal layer to surrounding structures strains cardiac function.
  • With each systolic contraction, the heart pulls not only against the parietal pericardium but also against the attached surrounding structures.
  • Systolic retraction of the rib cage, and diaphragm, pulsus paradoxus, and a vairety of other characteristic clinical findings may be observed.
  • The increased workload causes occasionally severe cardiac hypertrophy and dilation.
1003
Q

What is constrictive pericarditis?

A
  • The heart is encased in a dense, fibrous or fibrocalcific scar that limits diastolic expansion and cardiac output, features that mimic a restrictive cardiomyopathy.
  • A prior history of pericarditis may or may not be present.
1004
Q

How thick can the fibrous scar in constrictive pericarditis be? What does this mean for cardiac output?

A
  • The fibrous scar can be up to a centimeter in thickness, obliterating the pericardial space and sometimes calcifying; in extreme cases it can resemble a plaster mold (concretio cordis)
  • Because of the dense enclosing scar, cardiac hypertrophy and dilation cannot occur.
  • Cardiac output may be reduced at rest, but more importnatly the heart has little if any capacity to increase its output in reponse to increased systemic demands.
1005
Q

What are the clinical features and treatments of constrictive pericarditis?

A
  • Signs of pericarditis include distant or muffled heart sounds, elevated jugular venous pressure, and peripheral oedema
  • Treatment consists of surgical resection of the shell of constricting fibrous tissue (pericardectomy).
1006
Q

What is valvular stenosis and insufficency/regurgitation?

A
  • Stenosis is the failure of a valve to open completely, which impedes forward flow.
  • Insufficiency results from failure of a valve to close completely, thereby allowing reversed flow
1007
Q

What is function valve regurgitation?

A

It is used to descibre the incompetence of a vlave stemming from an abnormality in one of its support structures, as opposed to a primary valve defect.
For example, dilation of the right or left ventricle can pull the venticular papillary muscles down and outward, thereby preventing proper closure of otherwise normal mitral or tricuspid leaflets.

1008
Q

What are the clinical consequences of valve dysfunction?

A

They vary of the valve involved, the degree or impariemtn, the tempo of disease onset, and the rate and quality of compensatory mechanisms.
* For example, sudden destruction of an aortic valva cusp by infective endocarditis can cause acute, massive and rapidly fatal regurgitation.
* In contrast, rheumatic mitral stenosis typically develops indolently over yeats, and its clinical effects can be well tolerated for extended periods.

1009
Q

Valvular stenosis or insufficiency often produces secondary changes, both proximal and distal to the affected valve. What are these?

A
  • Generally, valvular stenosis leads to pressure overload cardiac hypertrophy, where mitral or aortic valvular insufficiency leads to volume overload.
  • Both situations can culminate in heart failure
  • In addition, the ejection of blood through narrowed stenotic valves can produce high speed “jets” of blood that injure the endocardium where they impact.
1010
Q

Valvular abnormalities can be congenital or acquired. Tell me about acquired valvular stenosis and acquired valvular insufficiency.

A
  • Acquired valvular stenosis has relative few causes; it is almost always a consequence of a remote or chronic injury of the valve cusps that declares itself clinically only after many years.
  • In contrast, acquired valvular insufficiency can result from intrinsic disease of the valve cuspss or damage to or distortion of the supporting structures (eg, the aorta, mitral annulus, tendinous cords, papillary muscles, ventricular free wall)
1011
Q

What are the most freuqent causes of the major functional valvular lesions?

A
  • Aortic stenosis: calcification and sclerosis of anatomically normal or congenitally bicuspid aortic valves
  • Aortic insuffiency: dilation of the ascending aorta, often secondary to HTN and/or aging
  • Miral stenosis: rheumatic heart disease
  • Miral insufficiency: myxomatous denegeration (mitral valve prolapse)
1012
Q

What is the most common of all valvular abnormalities? What is the usual cause?

A
  • Calcific aortic stenosis is usually the consequence of age-associated “wear and tear” of either anatomically normal valves or congenitally bicuspid valves (in approx 1% of the population).
  • It is likely a consequence of recurrent chronic injury due to hyperlipidaemia, HTN, inflammation, and other factors similar to those implicated in atherosclerosis.
1013
Q

What is the prevalence of aortic stenosis? Is it changing?

A

The prevalence of aortic stenosis is estimated at 2% and is increasing as the general population ages.

1014
Q

Aortic stenosis of previously normal valves usually comes to attention in the 7th-9th decades of life, whereas stenotic bicuspid valves tend to become clinically significant 1-2 decades earlier. Why?

A

Bicuspid valves incur greater mechanical stress than normal tricuspid valves, which may explain their accelerated stenosis.
The chronic progressive injury leads to valvular degeneration and incites the deposition of hydroxyapatite.

1015
Q

What is the gross morphology of aortic stenosis?

A

The gross morphologic hallfmark of non-rheumatic calcific aortic stenosis is mounded calcified masses within the aortic cusps that ultimately protrude through the outflow surfaces into the sinuses of Valvalva and prevent cuspal opening. The free edges of the cusps are usually not involved.

1016
Q

What is the microscopic morphology of calcific aortic stenosis?

A
  • Microscopically, the layered architecture of the valve is largely preserved.
  • The calcific process begins in the valvular fibrosa on the outflow surface of the valve, at the points of maximal cusp flexion (near the margins of attachment).
  • Inflammation is variable, and metplastic bone may be seen
1017
Q

What are the functional consequences of aortic stenosis?

Give me numbers please

A
  • In aortic stenosis, the obstruction to left ventricualr outflow leads to gradual narrowing of the valve orifice (valve area approx 0.5-1cm2 in severe aortic stenosis, normal approx 4cm2) and an increasing pressure gradient across the calcified valve, reaching 75 - 100 mmHg in severe cases.
  • Left ventricular pressures rise to 200 mmHg or more in such instances, producing concentric left ventricular (pressure overload) hypertrophy.
  • The hypertrophied myocardium tends to be ischaemic (as a result of diminished microcirculatory perfusion) and angine may occur.
  • Both systolic and diastolic myocardiacl function may be impaired; eventually, cardiac decompensation and CHF can ensue.
1018
Q

How does mitral involvement in rheumatic aortic stenosis and calcific aortic stenosis differ?

A

In calcific aortic stenosis, the mitral valve is generally normal,although some patient may have direct extension of aortic valve calcific deposits onto the anterior mitral leaflet.
In contrast, virtually all patients with rheumatic aortic stenosis also have concomitant and characteristic structural abnormalities of the mitral valve.

1019
Q

What is the prognosis of calcific aortic stenosis? How is it treated?

A
  • The onset of symptoms (angina, CHF, or syncope) in aortic stenosis heralds cardiac decompensation and carries an extremely poor prognosis.
  • If untreated, most patient with aortic stenosis will die within 5 years of developing angina, within 3 years of developing syncope, and within 2 years of CHF onset.
  • Treatment requires surgical valve replacement, as medical therapy is ineffective in severe symptomatic aortic stenosis.
1020
Q

What is the prevalence of a congenitally bicuspid aortic valve? Is it genetic? How so? What is it associated with?

A
  • Bicuspid aortic valve is a development of abnormality with prevalnce in teh population of approx 1%.
  • Some cases of BAV show familial clustering, often with associated aorta or left ventricular outflow tract malformations.
1021
Q

What is the pathology of a congenitally bicuspid aortic valve? What area is a major site for calcific deposits?

A
  • There are only two functional cusps, usually of unequal size, with the larger cusp having a midline raphe resulting from incomplete commissural separation during development; less frequently the cusps are of equal size and the raphe is absent.
  • The raphe is frequently a major site of calcific deposits.
1022
Q

Aortic valves can become bicuspid because of an acquired deformity. How? By what? and how can we tell?

A
  • Valves that become bicuspid because of an acquired deformity (e.g. rheumatic valve disease) have a fused commissure that produces a conjoined cusp that is generally twice the size of the non-conjoined cusp.
1023
Q

What are the clinical features of a bicuspid aortic valve?

A
  • Although BAV is usually asymptomatic early in life, late complications include aortic stenosis or regurgitation, infective endocarditis, and aortic dilation and/or dissection.
  • In particular, BAVs are predisposed to progressive calcification, similar to that occurring in aortic valves with initially normal anatomy.
1024
Q

Sturctural abnormalities of the aortic wall also commonly accompany bilateral aortic valves. What is the consequence of this?

A

It may potentiate aortic dilation and aortic dissection.

1025
Q

Where do calcific deposits usually deposit in the mitral valve? How do they appear?

A
  • As opposed to the predominantly cuspal involvement in aortic valve calcification, degenerative calcific deposits in the mitral valve typically develop in the fibrous annulus.
  • Grossly, these appear as irregular, stony hard, occasionally ulcerated nodules (2-5mm in thickness) at the base of the leaflets (fig C and D).
1026
Q

How does mitral annular calcification affect valvular function?

A

Mitral annular calcification usually does not affect valvular function. However, in exceptional cases it can lead to
* Regurgitation by interfering with physiologic contraction of the valve ring
* Stenosis by impairing opening of the mitral leaflets
* Arrhythmias and occasionally sudden death by penetration of calcium deposits to a depth sufficienc to impinge on the AV conduction system.

1027
Q

What are some of the clinical consequences of calcific mitral stenosis?

A

Because calcific nodules may provide a site for thrombus formation, patients with mitral annular calcification have an increased risk of embolic stroke, and the calcific nodules can become a nidus for infective endocarditis

1028
Q

What population does mitral annular calcification occur in?

A

Mitral annular calcification is most common in women older than 60 and individuals with mitral valve prolapse

1029
Q

What happens during a mitral valve prolapse? What is its prevalence and who does it effect?

A
  • One or both mitral valve leaflets are “floppy” and prolapse, or balloon back into the left atrium during systole.
  • It affects approximately 2-3% of adults in the US with an approximate 7:1 female:male ratio.
1030
Q

What is the pathogenesis of mitral valve prolapse?

A
  • The etiologic bases is unknown in most cases.
  • Uncommonly, MVP is associated with heritable disorders of connective tissue including Marfan syndrome, caused by fibrillin-1 mutations. Fibrillin-1 defects alter cell-matrix interactions and dysregulate TGF-β signaling.
1031
Q

What are the gross morphological changes in mitral valve prolapse?

A
  • The characteristic anatomic change in MVP in interchordal ballooning (hooding) of the mitral leaflets or portions thereof.
  • The affected leaflets are often enlarged, redundant, thick and rubbery.
  • The associated tendinous cords may be elongated, thinned, or even ruptured, and the annulus may be dilated.
  • The tricuspid, aortic or pulmonary valves may also be affected.
1032
Q

What are the key histological changes associated with mitral valve prolapse?

A
  • Marked thickening of the spongiosa layer with deposition of mucoid (myxomatous) material, called myxomatous degeneration
  • There is also attenuation of the collagenous fibrosa layer of the valve, on which the structural integrity of the leaflet depends.
1033
Q

In mitral valve prolapse, secondary changes reflect the stresses and tissue injury indicent to the billowing leaflets. What are these changes?

A

1) fibrous thickening of the valve leaflets, particularly where they rub against each other
2) linear fibrous thickening of the left ventricular endocardial surface where the abnormally long cords snap or rub against it
3) thickening of the mural endocardium of the left ventricle or atrium as a consequence of friction-induced injury induced by the prolapsing, hypermobile leaflets
4) thrombi on the atrial surfaces of the leaflets or the atrial walls (B)
5) focal calcifications at the base of the posterior mitral leaflet (C)

1034
Q

What are the clinical features of mitral valve prolapse?

A

Most individuals diagnosed with MVP are asymptomatic; in such cases the condition is discovered incidentally by auscultation of mid-systolic clicks, sometimes followed by a mid-late systolic murmur.
A minority of patients have chest pain mimicking angina (although not exertional in nature), and a subset have dyspnoea, presumably related to vascular insufficiency.

1035
Q

Although the great majority of patients with mitral valve prolapse have no untoward effects, approximately 3% develop on of four serious complications. What are they?

A

1) infective endocarditis
2) mitral insufficiency, sometimes with chordal rupture
3) stroke or other systemic infarct, resulting from embolism of leaflet thrombi
4) arrhythmias, both ventricular and atrial

Rarely, MVP is the only finding in sudden cardiac ceath

1036
Q

The risk of serious complications of mitral valve prolapse is high and low in which populations? What do we do to treat this?

A
  • The risk is very low in MVP discovered incidentally in young asymptomatic patients; the risk is higher for mel, older patients, and those with arrhythmias or mitral regurgitation.
  • Valve repair or replacement surgery can be done for symptomatic patients or those with increased risk for significant complications; indeed, in the US, MVP is the most common cause for mitral valve surgery
1037
Q

What is rheumatic fever?

A

It is an acute, immunologically mediated, multisystem inflammatory disease classically occuring a few weeks after an episode of group A strep pharyngitis, occasionally, it can follow strep infections at other sites, such as the skin.

1038
Q

What is acute rheumatic carditis?

A

It is a common manifestation of active rheumatic fever and may progress over time to chronic rheumatic heart disease (RHD), mainly manifesting as valvular abnormalities.

1039
Q

How is rheumatic fever characterised?

A

Principally by deforming fibrotic valvular disease, particularly involving the mitral valve; RHD is virtually the only cause of mitral stenosis.

1040
Q

What is the pathogenesis of rheumatic fever?

A
  • Acute rheumatic fever results from host immune responses to group A streptococcal antigens that cross-react with host proteins.
  • In particular, antibodies and CD4+ T cells directed against streptococcal M proteins can also in some cases recognise cardiac self-antigens.
  • Antibody binding can activate complement, as well as recruit Fc-receptor bearing cells (neutrophils and macrophages); cytokine production by the stimulated T cells leads to macrophage activation.
  • Damage to heart tissue may thus be caused by a combination of antibody- and T-cell-mediated reactions.
1041
Q

What are the key microscopic morphological features of rheumatic fever?

A
  • During acute RF, focal inflammatory lesions are found in various tissues.
  • Distinctive lesions occur in the heart, called Aschoff bodies, consisting of foci of T-lymphocytes, occasional plasma cells, and plump activated macrophages called Anitschkow cells (pathognomic for RF).
  • These macrophages have abudant cytoplasm and central round-to-ovoid nuclei (occasionally binucleate) in which the chromatin condenses into a central slender, wavy ribbon
1042
Q

What are some of the clinical consequences of rheumatic fever and how you they appear histologically?

A
  • During acute RF, diffus inflammation and Ashocc bodies may be found in any of the three layers of the heart, resulting in pericarditis, myocarditis, or endocarditis (pancarditis).
  • Inflammation of the endocardium and the left-sided valves typically results in fibroid necrosis within the cusps or tendinous cords. Overlying these necrotic foci and along the lines of closure are small (1-2mm) vegetations, called verrucae.
  • subendocardial lesions, prehaps exacerbated by regurgitant jets, can induce irregular thickenings called MacCallum plaques, usually in the left atrium
1043
Q

What are the cardinal anatomic changes of the mitral valve in chronic rheumatic heart disease?

A

Leaflet thickening, commissural fusion and shortening, and thickening and fusion of the tendinous cords (D).

1044
Q

What valves are normally effected in rheumatic heart disease?

A

In chronic disease, the mtiral valve is virtually always involved.
The mitral valve is affected in isolation roughly 2/3rds of RHD, and along with aortic valve in another 25% of cases.
Tricuspid valve involvement is infrequent, and the pulmonary valve is only rarely affected

1045
Q

How is the valve affected in chronic rheumatic mitral stenosis?

A
  • Calcification and fibrous bridging across the valvular ommissures create “fish mouth” or “buttonhole” stenoses.
  • With tight mitral stenosis, the left atrium progressively dilates and may harbor mural thrombi that can embolise.
1046
Q

What are the microscopic features of chronic rheumatic heart disease?

A

Valves show organised of the acute inflammation, with post-inflammatory neovascularisation and transmural fibrosis that obliterate the leaflet architecture.
Aschoff bodies are rarely seen in surgical specimens or autopsy tissue from patients with chronic RHD, as a result of the long intervals between the initial insult and the development of the chronic deformity

1047
Q

What are the clinical features of rheumatic fever? How do you diagnose it?

A

It is characterised by a constellation of findings:
1) migratory polyarthritis of the large joints
2) pancarditis
3) subcutaneous nodules
4) erythema marginatum of the skin
5) Sydenham chorea, a neurological disorder with involuntary rapid, purposeless movements.

The diagnosis is via the Jones criteria: evidence of a preceding group A strep infection, with the presence of two of the major manifestations above or one major and two minor manifestations (non-specific signs and symptoms that include fever, arthralgia, or elevated blood levels of acute-phase reactants)

1048
Q

When and in what age group does acute rheumatic fever often occur?

A

It typically appears 10 days to 6 weeks after a group A strep infection in about 3% of patients.
It occurs most often in children between ages 5 and 15, but first attacks can occur in middle to later life.

1049
Q

What antibodies are present in rheumatic fever and when?

A

Although pharyngeal cultures for strep are negative byt the time the illness begins, antibodies to one or more streptococcal enzymes, such as streptlysin O and DNase B can be detected in the serum of most patients with RF.

1050
Q

What are the predominant clinical manifestations of rheumatic fever in adults and children?

A

The predominant clinical manifestations are carditis and arthritis, the latter more common in adults than in children.
Arthritis typically begins with a polymigratory polyarthritis (accompanied by fever) in which one large joint after another becomes painful and swollen for a period of days and then subsides spontaneously, leaving no residual disability.

1051
Q

What are the clinical features related to acute carditis?

A

Pericardial friction rubs, tachycardia, and arrhythmias.
Myocarditis can cause cardiac dilation that may culminate in functional mitral valve insufficiency or even heart failure.
Approximately 1% of affected individuals die of fulminant RF involvement of the heart.

1052
Q

What happens in recurrent attacks of rheumatic fever?

A
  • After an initial attack there is increased vulnerability to reactivation of the disease with subseuquent pharyngeal infections, and the same manifestations are likely to appear with each recurrent attack.
  • Damage to the valves is cumulative. Turbulence induced by ongoing valvular deformities leads to additional fibrosis.
1053
Q

How do the lungs develop embryologically?

A
  • The respiratory system is an outgrowth from the ventral wall of the foregut.
  • The midline trachea develops two lateral outpocketings, the lung buds
  • The lung buds eventually divide into branches called lobar bronchi, three on the right and two on the left, thus giving rise to three lobes on the right and two on the left.
1054
Q

Developmental anomalies of the lung are rare. What are some of the more common ones?

A
  • Pulmonary hypoplasia is the defective development of both lungs, resulting in decreased weight, volume, and acini for body weight and gestational age
  • Foregut cysts arise from abnormal detachments of primitive foregut and are most often located in the hilum or middle mediastinum.
  • Pulmonary sequestration refers to a discrete area of lung tissue that 1) lacks any connection to the airway system and 2) has an abnormal blood supply arising from the aorta or its branches
1055
Q

What causes pulmonary hypoplasia?

A

It is caused by abnormalities that compress the lung or impede normal lung expansion in utero, such as CDH and oligohydramnios.

1056
Q

Foregut cysts are classfied depending on the wall structure. What are these classifications? What are the microscopic characteristics of the cyst?

A
  • They are classified as bronchogenic (most common), oesophageal, or enteric. A bronchogenic cyst is rarely connected to the tracheobronchial tree.
  • Microscopically, the cyst is lined by ciliated pseudostratified columnar epithelium. The wall contains bronchial glands, cartilage, and smooth muscle.
1057
Q

What are intralobar vs extralobar pulmonary sequestrations? When and why do they occur?

A
  • Extralobar sequestrations are external to lung and most commonly come attention in infants as mass lesions. They may be associated with other congenital anomalies.
  • Intralobar sequestrations occur within the lung. They usually present in odler childre, often due to recurrent localised infection or bronchiectasis.
1058
Q

What is atelectasis?

A

Atelectasis refers either to incomplete expansion of the lungs (neonatal atelectasis) or to the collapse of previously inflated lung, producing areas of relatively airless pulmonary parenchyma.

1059
Q

What are the main types of acquired atelectasis, which is encountered principally in adults? What causes each one each?

A
  • Resorption atelectasis stems from complete obstruction of an airway. It is most often caused by excessive secretions (mucus plugs) or exudates within smaller bronchi, as may occur in bronchial asthma, chronic bronchitis, bronchiectasis, and post-op states. Aspiration, foreign bodies and rarely tumours may also lead to airway obstruction
  • Compression atelectasis results whenever significant volumes of fluid (transudate, exudate, or blood), tumor or air accumulated within the pleural cavity.
  • Contraction atelectasis occurs when focal or generalised pulmonary or pleural fibrosis prevents full lung expansion
1060
Q

How do resorption and compression atelectasis effect the mediastinum?

A

In resorption atelectasis, the lung volume is diminshed and the mediastinum shifts toward the atelectatic lung
In compression atelectasis, the mediastinum is pushed away from the affected lung

1061
Q

What is the difference between obstructive and restrictive lung diseases?

A
  • Obstructive lung diseases are characterised by an increase in resistance to airflow due to partial or complete obstruction at any level from the trachea and larger bronchi to the terminal and respiratory bronchioles
  • Restrictive diseasess are characterised by reduced expansion of lung parenchyma and decreased total lung capacity
1062
Q

The distinction between obstructive and restrictive lung diseases is based primarily on lung function tests. How do these differ?

A
  • In individuals with diffuse obstructive disorders, pulmonary functions tests show decreased maximal airflow rates during forced expiration, usually expressed as a reduced FEV1/FVC ratio (less than 0.7)
  • Restrictive diseases are associated with proportionate decreases in both total lung capacity and FEV1, leading to normal FEV1/FVC ratio
1063
Q

Restrictive lung diseases occur in two broad kinds of conditions. What are they?

A

1) Chest wall disorders (e.g. severe obesity, pleural disease, hyphoscoliosis, and neuromuscular diseases such as poliomyelitis)
2) Chronic interstitial and infiltrative diseases, such as pneumoconioses and ILD.

1064
Q

What are some common obstructive lung diseases?

A

Emphysema
Chronic bronchitis
Asthma
Bronchiectasis

1065
Q

What is the anatomic site of:
* chronic bronchitis
* bronchiectasis
* asthma
* emphysema
* small airway disease, bronchiolitis

A

Chronic bronchitis - bronchus
Bronchiectasis - bronchus
Asthma - bronchus
Emphysema - acinus
Small airway disease, bronchiolitis - bronchiole

1066
Q

What are the main pathological changes of:
* chronic broncitis
* bronchiectasis
* asthma
* emphysema
* small airway disease, bronchiolitis

A

Chronic bronchitis - mucous gland hyperplasia, hypersecretion
Bronchiectasis - airway dilation and scarring
Asthma - smooth muscle hyperplasia, excess mucus, inflammation
Emphysema - airspace enlargement, wall destruction
Small airway disease, bronchiolitis - inflammatory scarring/obliteration

1067
Q

What is the etiology of:
* chronic bronchitis
* bronchiectasis
* asthma
* emphysema
* small airway disease, bronchiolitis

A

Chronic bronchitis - tobacco smoke, air pollutants
Bronchiectasis - persistent or severe infections
Asthma - immunologic or undefined causes
Emphysema - tobacco smoke
Small airway disease, bronchiolitis - tobacco smoke, air pollutants, miscellaneous

1068
Q

What are the signs/symptoms of:
* chronic bronchitis
* bronchiectasis
* asthma
* emphysema
* small airway disease, bronchiolitis

A

Chronic bronchitis - cough, sputum production
Bronchiectasis - cough, purulent sputum, fever
Asthma - episodic wheezing, cough, dyspnoea
Emphysema - dyspnea
Small airway disease, bronchiolitis - cough, dyspnoea

1069
Q

What does COPD consist of?

A

Emphysema and chronic bronchitis are often clinically grouped together and referred to as COPD, since most patients have features of both.;

1070
Q

What are the classic populations who get COPD?

A

There is a clear-cut association b etween heavy cigarette smoking and emphysema, and women and African Americans are more susceptible than other groups.

1071
Q

What is emphysema?

A

Emphysema is characterised by irreversible enlargement of the airspaces distal to the terminal bronchiole, accompannied by destruction of their walls without obvious fibrosis

1072
Q

Based on the segments of the respiratory units that involved emphysema is classified into four major types. What are they? Which ones cause significant obstruction? Which is most common?

A

1) Centriacinar
2) Panacinar
3) Paraseptal
4) Irregular

Of these, only the first two cause clinically significant airflow obstruction.
Centriacinar emphysema is the most common form, consistuting more than 95% of clinically significant cases.

1073
Q

What parts of the acini (the terminal respiratory units) does centriacinar emphysema effect? Where in the lung are the lesions commonly found?

A
  • The central or proximal parts of the acini, formed by respiratory bronchioles, are affected, whereas distal alveoli are spared.
  • Thus, both emphysematous and normal arispaces exist within the same acinus and lobule.
  • The lesions are more common and usually more severe in the upper lobes, particularly in the apical segements.
  • Inflammation around bronchi and bronchioles is common.
1074
Q

What parts of the acini are affected in panacinar emphysema? Where does it tend to happen?

A
  • The acini are uniformly enlarged from the level of the respiratory bronchiole to the terminal blind alveoli.
  • In contrast to centriacinar emphysema, panacinar emphysema tends to occur more commonly in the lower zone and in the anterior marghins of the lungs, and it is usually most severe at the bases
1075
Q

What part of the acini does distal acinar (paraseptal) emphysema usually happen? Where in the lungs does it happen?

A
  • The proximal portion of the acinus is normal, and the distal part is predominantly involved.
  • The emphysema is mroe striking adjacent to the pleura, along the lobular connective tissue septa, and at the margins of the lobules.
  • It occurs adjacent to areas of fibrosis, scarring or atelectasis and is usually more severe in the upper half of the lungs
1076
Q

What are the morphological characteristic findings in distal acinar (paraseptal) emphysema?

A

The characteristic findings are of multiple, continuous, enlarged airpsaces from less than 0.5cm to more than 2.0cm in diameter, sometimes forming cystlike structures.

1077
Q

Where and when is irregular emphysema found?

A
  • The acinus is irregularly involved, and is almost invariably associated with scarring.
  • In most instances it occurs in small foci and is clinically insignificant
1078
Q

What diseases are often associated with centriacinar, panacinar and distal acinar emphysema?

A
  • Centriacinar emphysema occurs predominantly in heavy smokers, often in association with chronic bronchitis (COPD)
  • Panacinar emphysema is associated with α1-antitrypsin deficiency
  • Distal acinar emphysema probably underlies many cases of spontaneous pneumothorax in young adults
1079
Q

In broad terms, what is the pathogenesis of emphysema?

A
  • Inhaled cigarette smoke and other noxious particles cause lung damage and inflammation
  • This result in parenchymal destruction (emphysema) and airway disease (bronchiolitis and chronic bronchitis).
1080
Q

What are the factors that influence the development of emphysema?

A
  • Inflammatory mediators and leukocytes including leukotriene B4, IL-8, TNF, are released by resident epithelial cells and macrophages and attract inflamatory cells, amplify the inflammatory process and induce structural changes
  • Protease-antiprotease imbalance - several proteases are released from the inflammatory cells and epithelial cells that break down connective tissue components
  • Oxidative stress - substances in tobacco smoke, alveolar damage, and ifnlammatory cells all produce oxidants, which may beget more tissue damage and inflammation
  • Infection may exacerbate the associated inflammation and chronic bronchitis
1081
Q

How do oxidants cause tissue damage in emphysema?

A
  • The gene NRF2 encodes a transcription factor that serves as a sensor for oxidants in alveolar epithelial cells and many other cell types
  • Intracellular oxidants activate MRF2, which upregulates the expression of multiple genes that protect cells from oxidant damage.
1082
Q

The idea that proteases are important in emphysema is based on the observation that patients with a deficientcy int eh antiprotease α1-antitrypsin have a markedley enhanced tendency to develop pulmonar emphysema. What is the pathogensis of α1-antitrypsin deficiency?

A

It is postulated that any injury (eg, that induced by smoking) that increases the activation and influx of neutrophils into the lung leads to local release of proteases, which in the absence of α1-antitrypsin activity result in excessive digestion of elastic tissue and emphysema.

1083
Q

Where is α1-antitrypsin usually found and what is its role?

A

α1-antitrypsin, normally present in serum, tissue fluids and macrophages, is a major inhibitor of proteases (particularly elastase) secreted by neutrophils during inflammation.

1084
Q

Where is the gene code from α1-antitrypsin?

A

α1-antitrypsin is encoded by teh proteinase inhibitor (Pi) locus on chromosome 14.

1085
Q

A number of factors contribute to airway obstruction in emphysema. What are they?

A
  • Small airways are normall held open by the elastic recoil of the lung parenchyma, and the loss of elastic tissue in the walls of alveoli that surround respiratory bronchioles reduces radial traction and thus causes the respiratory bronchioles to collapse during expiration.
  • This leads to functional airflow obstruction despite the absence of mechanical obstruction.
1086
Q

Even young smokers often have small airway inflammation associated with what changes?

A
  • Goblet cell hyperplasia, with mucus plugging of the lumen
  • Inflammatory infiltrates in bronchial walls consisting of neutrophils, macrophages, B cells (sometimes forming follicles) and T cells
  • Thickening of the bronchiolar wall due to smooth muscle hypertrophy and peribronchial fibrosis

Together these changes narrow the bronchiolar lumen and contribute to airway obstruction

1087
Q

What is the gross morphology of advanced emphysema?

A
  • Advanced emphysema produces voluminous lungs, often overlapping the heart and hiding it when the anterior chest wall is removed.
  • Generally, the upper two thirds of the lungs are more severely affected.
  • Large apical blebs or bullae are more characteristic of irregular emphysema secondary to scrring and ofdistal acinar emphysema
  • Large alveoli can easily be seen on the cut surface of fixed lungs
1088
Q

What is the microscopic morphology of emphysema?

A
  • Microscopically, abnormally large alveoli are separated by thin septa with only focal centriacinar fibrosis.
  • There is loss of attachments of the alveoli to the outer wall of small airways.
  • The pores of Kohn are so large that septa appear to be floating or protruding blindly into alveolar spaces with a club-shaped end
  • Prolonged vasoconstriction leads to changes of pulmonary arterial hypertension
  • As alveolar walls are destroyed, there is a decrease in the capillary bed area
  • With advanced disease, there are even larger abnormal airspaces and possibly bleds or bullae, which often deform and compress the respiratory bronchioles and vasculature of the lung
1089
Q

What proportion of the lung has to be damaged by emphysema before you get symptoms?

A

Symptoms do not appear until at least one third of the functioning piulmonary parenchyma is damaged

1090
Q

What symptoms occur in what order in emphysema?

A
  • Dyspnea usually appears first, beginning insidiously but progressing steadily
  • In some patients, cough or wheezing is the chief complaint, easily confused with asthma
  • Cough and expectoration are extremely variable and depend on the extent of the associated bronchitis
  • Weight loss is common and can be so severe as to suggest occult cancer.
1091
Q

Classically, how does a patient with severe emphysema appear?

A

Barrel-chested and dyspneic, with obviously prolonged expiration, sits forward in a hunched-over position, and breathes through pursed lips.

1092
Q

What are the symptoms and appearances of severe emphysema?

A

Cough is often slight, overdistention is severe, diffusion capacity is low, and blood gas values are relatively normal at rest.
Such patients may overventilate and remain well oxygenated, and therefore are somewhat ingloriously designated pink puffers.

1093
Q

How can emphysema effect the heart? How does this change prognosis?

A

Development of cor pulmonale and eventually congestive heart failure, related to secondary pulmonary HTN, is associated with poor prognosis

1094
Q

What is death in most patients with emphysema caused by?

A

1) coronary artery disease
2) respiratory failure
3) right-sided heart failure
4) massive collapse of the lungs secondary to pneumothorax

1095
Q

What are the treatment options for emphysema?

A
  • Smoking cessation
  • Oxygen therapy
  • Long-acting bronchodilaters with inhaled corticosteroids
  • Physical therapy
  • Bullectomy
  • Lung volume reduction surgery
  • Lung transplantation
1096
Q

Between predominantly bronchitis and predominantly emphysema, how do the following things differ:
* Age
* Dyspnea
* Cough
* Infections
* Respiratory insufficiency
* Cor pulmonale
* Airways resistance
* Elastic recoil
* CXR
* Appearance

A
1097
Q

What is compensatory hyperinflation?

A

Compensatory hyperinflation is sometimes used to designate dilation of alveoli in response to loss of lung substance elsewhere. It is best exemplified by the hyperexpansion of residual lung parenchyma following surgical removal of a diseased lung or lobe

1098
Q

What is obstructive overinflation? Why does it happen?

A

In this condition the lung expands because air is trapped within it. A common cause is subtotal obstruction of the airways by a tumour or foreign object.
It occurs either:
(1) because the obstructive agent acts as a ball valve, allowing air to enter on inspiration while preventing its exodus on expiration or
(2) because collaterals bring in air behind the obstruction. These collaterals consist of the pores of Kohn and other direct accessory bronchioalveolar connections

1099
Q

Is obstructive overinflation serious?

A

Yes! It can be a life-threatening emergency, because the affected portion distends sufficiently to compress the remaining lung

1100
Q

What is bullous emphysema?

A

This is a descriptive term for large subpleural blebs or bullae (spaces greater than 1cm in diameter in the distended state) that can occur in any form of emphysema
These localised accentuations of emphysema occur near the apex, sometimes near old TB scarring.
On occasion, rupture of the bullae may give rise to PTx

1101
Q

What is interstitial emphysema? What causes it?

A
  • Entrace of air into the connective tissue stroma of the lung, mediastinum, or subcutaneous tissue produces interstitial emphysema.
  • In most instances, alveolar tears into pulmonary emphysema provide the avenue of entrance of air into the stroma of the lung, but rarely, chest wounds that allow entry of air or fractured ribs that puncture the lung substance underlie this disorder.
  • Alveolar tears are usually caused by rapid increases in pressure within alveolar sacs, such as occurs when there is a combination of coughin and bronchiolar obstruction.
1102
Q

What is chronic bronchitis?

A

Chronic bronchitis is defined clinically as persistent cough with sputum production for at least 3 months in at least 2 consecutive years, in the absence of any other causes

1103
Q

What are the consequences of chronic bronchitis that lasts for years?

A

It may accelerate decline in lung function, lead to cor pulmonale and heart failure, or cause atypical metaplasia and dysplasia of the respiratory epithelium, providing a rich soil for cancerous transformation

1104
Q

What are the primary or initiating factors of chronic bronchitis?

A

The primary or initiating factor in the genesis of chronic bronchitis is exposure to noxious or irritating inhaled substances such as tobacco smoke (90% of patients are smokers) and dust from grain, cotton and silica.

1105
Q

What is the pathogenesis of chronic bronchitis?

A
  • Mucus hypersecretion - the earliest feature is hypersecretion of mucus in the large airways, associated with hypertrophy of the submucosal glands in the trachea and bronchi. With time, there is also a marked increase in goblet cells in small airways leading to excessive mucus production that contributes to airway obstruction.
  • Inflammation - inhalants that induce chronic bronchitis cause cellular damage, eliciting both acute and chronic inflammatory responses involving neutrophils, lymphocytes, and macrophages
  • Infection does not initiarte chronic bronchitis, but is probably significant in maintaining it and may be critical in producing acute exacerbations
1106
Q

What are the gross morphological features of chronic bronchitis?

A

Grossly, there is hyperemia, swelling and edema of the mucous membranes, frequently accompanied by excessive mucinous or mucopurulent secretions.
Sometimes, heavy casts of secretions and pus fill the bronchi and bronchioles.

1107
Q

What are the microscopic changes associated with chronic bronchitis?

A

The characteristic features are mild chronic inflammation of the airways (predominantly lymphocytes) and enlargement of the mucus-secreting glands of the trachea and bronchi.
Although the numbers of goblet cells increase slightly, the major change is in the size of the mucous glands (hyperplasia).
The bronchial epithelium may exhibit squamous metaplasia and dysplasia. There is marked narrowing of bronchioles caused by mucus plugging, inflammation, and fibrosis.

1108
Q

What is the Reid index and how does it change in chronic bronchitis?

A

The Reid index is the ratio of the thickness of the mucous gland layer to the thickness of the wall between the epithelium and the cartilage.

The Reid index (normally 0.4) is increased in chronic bronchitis, usually in proportion to the severity and duration of the disease.

1109
Q

What are the clinical features of chronic bronchitis?

A
  • The cardinal symptom of chronic bronchitis is a persistent cough productive of sparse sputum.
  • For many years no other respiratory functional impairment is present, but eventually dyspnea on exertion develops.
  • With the passage of time, and usually continued smoking, other elements of COPD may appear, including hypercapnia, hypoxaemia, and mild cyanosis
  • Long standing severe chronic bronchitis commonly leads to cor pulmonale and cardiac failure.
  • Death may also result from further impairment of respiratory function due to superimposed acute infections.
1110
Q

What is asthma?

A

Asthma is a chronic disorder of the conducting airways, usually caused by an immunological reaction, which is marked by episodic bronchoconstriction due to increased airway sensitivity to a variety of stimuli; inflammation of the bronchial walls; and increased mucus secretion

1111
Q

What are the main symptoms of asthma?

A

The disease is manifested by recurrent episodes of wheezing, breathlessness, chest tightness, and cough, particularly at night and/or in the early morning.

1112
Q

What are the two categories of asthma?

A

Asthma may be categorised as atopic - evidence of allergen sensitisation and immune activation, often in a patient with allergic rhinitis or eczema
OR
non-atopic - no evidence of allergen sensitsation

1113
Q

In either types of asthma, episodes of bronchospasm can have diverse triggers. Give some examples of what?

A

Respiratory infections (especially viral infections), exposure to irritants (eg, smoke, fumes), cold air, stress and exercise

1114
Q

Asthma may also be classified according to the agents or events that trigger bronchoconstriction. What are the classifications?

A

Seasonal, exercise-induced, drug-induced (eg, aspirin), and occupational asthma, and asthmatic bronchitis in smokers

1115
Q

What is the most common type of asthma? What type of reaction is this?

A

Atopic asthma is the most common type of asthma and is a classic example of IgE-mediated (type I) hypersensitivity reaction.

1116
Q

When does atopic asthma usually begin? Is it linked with family history?

A
  • The disease usually begins in childhood and is triggered by environmental allergens, such as dusts, pollens, cockroach or animal dander, and foods, which most frequently act in synergy with other proinflammatory environmental cofactors, most notably viral infections
  • A positive family history of asthma is common
1117
Q

How do you diagnose atopic asthma?

A
  • A skin test with the offending antigen in these patients results in an immediate wheal-and-flare reaction.
  • It may also be diagnosed based on high total serum IgE levels or evidence of allergen sensitisation by serum radioallergosorbent tests (called RAST), which can detect the presence of IgE antibodies that are speciufic for individual allergens.
1118
Q

What do the allergen sensitisation and skin tests show with non-atopic asthma? Do they often have a family history?

A

Individuals with non-atopic asthma do not have evidence of allergen sensitisation and skin test results are usually negative.
A positive family history of asthma is less common in these patients

1119
Q

What are the common triggers for non-atopic asthma?

A

Respiratory infections due to viruses (eg, rhinoviruses, parainfluenza virus and RSV) are common triggers in non-atopic asthma.
Inhaled air pollutants, such as smoking, sulfur dioxide, ozone, and nitrogen dioxide, may also contribute to the chronic airway inflammation and hyperreactivity in some cases.

1120
Q

What are the symptoms of aspirin-sensitive asthma?

A

It is an uncommon type, occurring in individuals with recurrent rhinitis and nasal polyps. These individuals are exquisitely sensitive to small doses of aspirin as well as other NSAIDs, and they experience not only asthmatic attacks but also urticaria.

1121
Q

How does aspirin trigger asthma?

A

Aspirin and NSAIDs trigger asthma in these patients by inhibiting the cyclooxygenase pathway of arachidonic acid metabolism, leading to a rapid decrease in prostaglandin E2.
Normally prostaglandin E2 inhibits enzymes that generate proinflammatory mediators such as leukotrienes B4, C4, D4 and E4, which are believed to have central toles in aspirin-induced asthma

1122
Q

What might trigger occupational asthma?

A

This form of asthma may be triggered by fumes (epoxy resins, plastics), organic and chemical dusts (wood, cotton, platinum), gases (toluene), or other chemicals (formaldehyde, penicillin products)
Only minute quantities of chemicals are required to induce the attack, which usually occurs after repeated exposure.

1123
Q

Broadly, what is the pathogenesis of atopic asthma?

A
  • Atopic asthma is cause by a TH2 and IgE response to environmental allergens in genetically pre-disposed individuals.
  • Airway inflammation is central to disease pathophysiology and causes airway dysfunction partly through the release of potent inflammatory mediators and partly through remodeling of the airway wall.
  • As the disease becomes more severe, there in increased local secretion of growth factors, which induce mucous gland hypertrophy, smooth muscle proliferation, angiogenesis, fibrosis and nerve proliferation.
1124
Q

A fundamental abnormality in asthma is an exaggerated TH2 reponse to normally harmless environmental antigens. What happens in this process?

A
  • TH2 cells secrete cytokines that promote inflammation and stimulate B cells to produce IgE and other antibodies.
  • These cytokines include IL-4, which stimulates the production of IgE; IL-5, which activates locally recruited eosinophils; and IL-13, which stimulates mucus secretion from bronchial submucosal glands and also promotes IgE production by B cells.
  • The T cells and epithelial cells secrete chemokines that recruit more T cells and eosinophils, thus exacerbating the reaction.
  • IgE binds to the Fc receptors on submucosal mast cells, and repeat exposure to the allergen triggers the mast cells to release granule contents and produce cytokines and other mediators, which collectively induce the early-phase (immediate hypersnesitivity) reaction and the **late-phase reaction.
1125
Q

What happens during the early reaction in asthma?

A
  • The early reaction is dominated by bronchoconstriction, increased mucus production, variable degrees of vasodilation, and increased vascular permeability
  • Bronchoconstriction is triggered by direct stimulation of subepithelial vagal (parasympathetic) receptors through both central and local reflexes trigerred by mediators produced by mast cells and other cells in the reaction.
1126
Q

What happens during the late-phase reaction in asthma?

A

The late-phase reaction is dominated by recruitment of leukocytes, notably eosinophils, neutrophils, and more T cells.

1127
Q

Many mediators produced by leukocytes and epithelial cells have been implicated in the asthmatic response. The long list of “suspects in acute asthma can be ranked based on the clinical efficacy of pharmacological intervention with antagonists of specific mediators. Tell me about these…

A
  • Mediators whose role in bronchospasm is clearly supported by efficacy of intervention are: 1) leukotrienes C4, D4, E4,, which cause prolonged bronchonconstriction as well as increased vascular permeability and mucus secretion and 2) acetylcholine, released from intrapulmonary parasympathetic nerves, which can cause airway smooth muscle constriction by directly stimulating muscarinic receptors
  • Agents present in asthma but whose role seems fairly minor: 1) histamine, a potent bronchoconstrictor; 2) prostaglandin D2, which elicits bronchoconstriction and vasodilation; and 3) platelet-activating factor, which causes aggregation of platelets and release of serotonin from their granules
1128
Q

Susceptibility to atopic asthma is multigenic and often associated with increased incidence of other allergic disorders, such as allergic rhinitis and eczema. What are some of the stronger assocations?

A
  • One susceptibility locus for asthma is located on chromosome 5q. Among the genes in this cluster, polymorphisms in the IL13 gene have the strongest and most consistent associations with asthma or allergic disease.
  • Particular class II HLA alleles are linked to production of IgE antibodies against some antigens, such as ragweed pollen.
  • Polymorphisms in the gene encoding ADAM33, a metalloproteinase, may be linked to increased proliferation of bronchial smooth muscle cells and fibroblasts, thus contributing to bronchial hyperreactivity and subepithelial fibrosis.
  • β2-adrenergic receptor gene variants are associated with differential in vivo airway hyper-responsiveness and in vitro response to β-agonist stimulation
1129
Q

Asthma is a disease of industrialised societies where the majority of people live in cities. This likely hasv two main explanations. What are they?

A
  • Firstly, industralised environments contain many airborne pollutants that can serve as allergens to initiate the TH2 response.
  • Secondly, city life tends to limit the exposure of very young children to certain antigens, particularly microbial antigens, and exposure to such antigens seems to protect children from asthma and atopy. This protective effect is even more apparent if the microbial exposure occurred throughout the mother’s pregnancy.
1130
Q

What is the hygiene hypothesis?

A

The idea that microbial exposure during early development reduces the later incidence of allergic (and some autoimmune) diseases has been popularised as the hygiene hypothesis.

1131
Q

What happens during “airway remodelling” in asthma?

A

Over time, repeated bouts of allergen exposure and immune reactions result in structural changes in the bronchial wall, referred to as “airway remodelling”. These changes include:
* Thickening of the airway wall
* Subbasement membrane fibrosis (due to deposition of type I and III collagen)
* Increased vascularity
* An increase in the size of submucosal glands and number of airway goblet cells
* Hypertrophy and/or hyperplasia of the bronchial wall muscles

1132
Q

What is the morphology of patients dying of severe asthma (status asthmaticus)?

A
  • The lungs are distended by overinflation and contain small areas of atelectasis.
  • The most striking gross finding is occlusion of bronchi and bronchioles by thick, tenscious mucous plugs, which often contain shed epithelium.
  • A characteristic finding in sputum and bronchoalveolar lavage specimens is Curschmann spirals, which may result from extrusion of mucus plugs from subepithelial mucous gland ducts or bronchioles
  • Also present are numerous eosinophils and Charcot-Leyden crystals; the latter are composed of an eosinophil protein called galectin-10.
1133
Q

What is the clinical course of asthma?

A
  • An acute attack can last up to several hours.
  • In some patients, however the cardinal symptoms of chest tightness, dyspnea, wheezing and coughing (with or without sputum production) are present at a low level constantly
  • In its most severe form, status asthmaticus, the paroxysm persists for days and even weeks, sometimes causing airflow obstruction that is so extreme that makred cyanosis or even death ensues.
1134
Q

What is the diagnosis of asthma based on?

A

The diagnosis is based on demonstration of an increase in airflow obstruction (from baseline levels), difficulty with exhalation (prolonged expiration, wheeze), peripheral blood eosinophilia, and the finding of eosinophils, Curschmann spirals, and Charcot-Leyden crystals in the sputum (particularly in patients with atopic asthma)

1135
Q

What is bronchiectasis?

A

Bronchiectasis is a disorder in which destruction of smooth muscle and elastic tissue by chronic necrotising infections leads to permanent dilation of bronchi and bronchioles

1136
Q

Is bronchiectasis common? What diseases does it usually occur with?

A

Because of better control of lung infections, bronchiectasis is now uncommon. It may still develop in association with a variety of conditions, including the following:
* Congenital or hereditary conditions such as CF, immunodeficiency states, primary ciliary dyskinesia
* Infections, including necrotising pneumonia caused by bacteria, viruses, or funghi
* Bronchial obstruction, due to tremor, foreign bodies, or mucus impaction
* Other conditions, including rheumatoid arthritis, SLE, IBD, COPD and post-transplantation

1137
Q

Obstruction and infection are the major conditions associated with bronchiectasis, and it is likely that both are necessary for the development of full-fledged lesions. How do they cause it?

A
  • After bronchial obstruction, normal clearing mechanisms are impaired, resulting in pooling of secretions distal to the obstruction and secondary infection and inflammation
  • Conversely, severe infections of the bronchi lead to inflammation, often with necrosis, fibrosis and eventually dilation of airways.
1138
Q

How does cystic fibrosis cause bronchiectasis?

A
  • In CF, the primary defect in ion transport leads to defective mucociliary action and airway obstruction by thick viscous secretions.
  • This sets the stage for chronic bacterial infections, which cause widespread damage to airway walls.
  • With destruction of supporting smooth muscle and elastic tissue, the bronchi become markedly dilated, while smaller bronchioles are progressively obliterated as a result of fibrosis (bronchiolitis obliterans)
1139
Q

How is primary ciliary dyskinesia inherited? How common is it?

A

It is an autosomal recessive syndrome with a frequency of 1 in 15,000 to 40,000 births

1140
Q

How does primary ciliary dyskinesia cause bronchiectasis?

A

Ciliary dysfunction due to defects in ciliary motor proteins (eg, mutations involving dynein) contributes to the retention of secretions and recurrent infections that in turn lead to bronchiectasis.

1141
Q

What is Kartagener syndrome? How often does it occur with primary ciliary dyskinesia?

A

It is marked by situs inversus or a partial lateralising abnormality associated with bronchiectasis and sinusitis. The lack of ciliary activity interferes with bacterial clearance, predisposes the sinuses and bronchi to infection, and affects cell motility during embyrogenesis, resulting in the situs inversus.
Males with this condition tend to be infertile, as a result of sperm dysmotility.

1142
Q

What is allergic bronchopulmonary aspergillosis? What patient population does it occur in?

A

It is a condition that results from a hypersensitivity reaction to the fungus Aspergillus fumigatus.
It occurs in patients with asthma and CF who develop periods of exacerbation and remission that may lead to proximal bronchiectasis and fibrotic lung disease.

1143
Q

What is the pathogenesis of allergic bronchopulmonary aspergillosis? How does this show in the bloods?

A

Sensitisation to Aspergillus in the allergic host leads to activation of TH2 cells, which play a key role in recruiting eosinphils and other leukocytes.
Characteristically, there are high serum IgE levels, serum antibodies to Aspergillus, intense airway inflammation with eosinophils, and the formation of mucus plugs, which play a primary role in its pathogenesis.

1144
Q

Where in the lung does bronchiectasis usually occur?

A

It usually affects the lower lobes bilaterally, particularly air passages that are vertical, and is most severe in more distal bronchi and bronchioles.
When tumours of aspiration of foreign bodies lead to bronchiectasis, the involvement may be localised to a single lung segment.

1145
Q

How does bronchiectasis effect the size of the airways?

A

The airways are dilated, sometimes up to four times the normal size.
Characteristically, the bronchi and bronchioles are so dilated that they can be followed almost to the pleural surfaces.
On the cut surface of the lung, the dilated bronchi appears cystic and are filled with mucopurulent secretions

1146
Q

What are the histological findings in bronchiectasis?

A
  • The histologic findings vary with the activity and chronicity of the disease
  • In the fullblown, active case there is an intense acute and chronic inflammatory exudation within the walls of the bronchi and bronchioles, associated with desquamation of the lining epithelium and extensive areas of ulceration.
  • There may be pseudostratification of the columnar cells or squamous metaplasia of the remaining epithelium.
  • In some instances necrosis destroys the bronchial or bronchial walls and forms a lung abscess.
  • Fibrosis of the bronchial and bronchiolar walls and peribronchiolar fibrosis develop in the more chronic cases, leading to varying degrees of subtotal or total obliteration of bronchiolar lumens
1147
Q

A large variety of bacteria can be found in the usual case of bronchiectasis. What are they?

A

They include staphylococci, streptococci, pneumococci, enteric organisms, anaerobis and micro-aerophilic bacteria, and (particularly in children) Haemophilus influenzae and Pseudomonas aerunginosa.

1148
Q

What is acute lung injury and acute respiratory distress syndrome?

A

Acute lung injury is characterised by the abrupt onset of significant hypoxaemia and bilateral pulmonary infiltrates in the absence of cardiac failure.
Acute respiratory distress syndrome is a manifestation of severe ALI.

1149
Q

What is the mechanisms behind behind ARDS and ALI?

A

Both ARDS and ALI are associated with inflammation-associated increases in pulmonary vascular permeability, oedema and epithelial cell death

1150
Q

Acute Lung Injury (ALI) is a well-recognised complication of diverse conditions. Name me some

A

Infection
Sepsis
Diffuse pulmonary infections
Gastric aspiration
Physical/Injury
Mechanical trauma, including head injuries
Pulmonary contusions
Near-drowning
Fractures with fat embolism
Burns
Ionising radiation
Inhaled irritants
oxygen toxicity
smoke
irritant gases and chemicals
Chemical injury
heroin or methadone overdose
acetylsalicylic acid
barbiturate overdose
paraquat
Other things
DIC
Pancreatitis
Uraemia
CABG

1151
Q

ALI/ARDS is initiated by injury of pneumocytes and pulmonary endothelium, setting in motion a vicious cycle of increasing inflammation and pulmonary damage. What are the steps involved in this?

A

Endothelial activation
Adhesion and extravasation of neutrophils
Accumulation of intraalveolar fluid and formation of hyaline membranes
Resolution of injury

1152
Q

What happens in endothelial activation during ARDS?

A
  • In some instances, endothelial activation is secondary to pneumocyte injury, which is sensed by resident alveolar macrophages.
  • In response, these immune sentinels secrete mediators such as TNF that act on the neighbouring endothelium.
  • Alternatively, circulating inflammatory mediators may activate pulmonary endothelium directly in the setting of severe tissue injury or sepsis.
  • Some of these mediators injure endothelial cells, while others (notably cytokines) activate endothelial cells to express increased levels of adhesion molecules, procoagulant proteins and chemokines
1153
Q

What happens during adhesion and extravasation of neutrophils in ARDS?

A
  • Neutrophils adhere to the activated endothelium and migrate into the interstitium and the alveoli, where they degranulate and release inflammatory mediators, including proteases, ROS and cytokines.
  • Macrophage migration inhibitory factor (MIF) released into the local milieu also helps to sustain the ongoing pro-inflammatory response.
  • The result is increased recruitment and adhesion of leukocytes, causing more endothelial injury, and local thrombosis.
  • This cycle of inflammation and endothelial damage lies at the heart of ALI/ARDS.
1154
Q

What happens regarding accumulation of alveolar fluid and formation of hyaline membranes during ARDS?

A
  • Endothelial activation and injury make pulmonary capillaries leaky, allowing interstitial and intraalveolar oedema fluid to form.
  • Damage and necrosis of type II alveolar pnuemocytes leads to surfactant abnormalities, further compromising alveolar gas exchange.
  • Ultimately, the inspissated protein-rich oedema fluid and debris from dead alveolar epithelial cells organise into hyaline membranes, a characteristic feature of ALI/ARDS.
1155
Q

What happens regarding resolution of injury during ARDS?

A
  • It is impeded in ALI/ARDS due to epithelial necrosis and inflammatory damage that impairs the ability of remaining cells to assist with oedema resorption
  • Eventually, however, if the inflammatory stimulus lessens, macrophages remove intraalveolar debris and release fibrogenic cytokines such as transforming growth factor β and platelet-derived growth factor (PDGF).
  • These factors stimulate fibroblast growth and collagen deposition, leading to fibrosis of alveolar walls.
  • Bronchiolar stem cells proliferate to replace pneumocytes. Endothelial restoration occurs through proliferation of uninjured capillary endothelium
1156
Q

What populations is ARDS worse in?

A

More common and associated with a worse prognosis in chronic alcoholics and in smokers

1157
Q

What is the morphology in the acute stage on ARDS?

A

In the acute stage, the lungs are heavy, firm, red and boggy.
They exhibit congestion, interstitial and intra-alveolar oedema, inflammation, fibrin deposition, and diffuse alveolar damage.
The alveolar walls become lined with waxy hyaline membranes that are morphologically similar to those seen in hyaline membrane disease of neonates.
Alveolar hyaline membranes consist of fibrin-rich oedema fluid mixed with the cytoplasmic and lipid remnants of necrotic epithelial cells.

1158
Q

What is the morphology of the organising stage of ARDS?

A

Type II pneumocytes proliferate, and granulation tissue forms in the alveolar walls and spaces.
In most cases the granulation tissue resolves, leaving minimal functional impairment.
Sometimes, however, fibrotic thickening (scarring) of the alveolar septa ensues.

1159
Q

What are the clinical and radiographic symptoms of ALI/ARDS?

A
  • Profound dyspnoea and tachypnoea herald ALI, followed by increasing cyanosis and hypoxaemia, respiratory failure
  • The appearance of diffuse bilateral infiltrates on radiographic examination
1160
Q

Does ALI/ARDS effect the whole lung equally?

A

The functional abnormalities in ALI are not evenly distributed throughout the lungs.
The lungs have areas that are infiltrated, consolidated, or collapsed (and thus poorly aerated and poorly compliant) and regions that have nearly normal levels of compliance and ventilation.
Poorly aerated regions continue to be perfused, producing ventilation-perfusion mismatch and hypoxaemia

1161
Q

What is pulmonary oedema? What causes it?

A
  • Pulmonary oedema is leakage of excessive interstitial fluid which accumulates in alveolar spaces
  • It can result from haemodynamic disturbances (haemodynamic or cardiogenic pulmonary oedema) or from direct increases in capillary permeability as a result of microvascular injury
1162
Q

What is haemodynamic pulmonary oedema? Where does the fluid accumulate?

A
  • Haemodynamic pulmonary oedema is due to increased hydrostatic pressure, as occurs most commonly in left sided CHF or decreased osmotic pressure, as occurs less commonly in nephrotic syndrome and liver disease.
  • Fluid accumulates initially in the basal regions of the lower lobes because hydrostatic pressure is greatest in these sites *dependent oedema).
1163
Q

How does haemodynamic pulmonary oedema appear histologically? Give me both acute and chronic

A
  • The alveolar capillaries are engorged, and an intra-alveolar transudate appears as finely granular pale pink material
  • Alveolar micro haemorrhages and hemosiderin-laden macrophages may be present.
  • Chronically, hemosiderin-laden macrophages are abundant, and fibrosis and thickening of the alveolar walls cause the soggy lungs to become firm and brown (brown induration).
1164
Q

How do the hemosiderin-laden macrophages in APO change the lungs?

A

These changes not only impair normal respiratory function but also predispose to infection

1165
Q

What are the causes of oedema due to alveolar wall injury?

A

Direct injury such as infections, inhaled gases, liquid aspiration, raditation
Indirect injury such as septicaemia, blood transfusion related, burns, drugs and chemicals

1166
Q

How does microvascular alveolar injury cause pulmonary oedema?

A
  • Non-cardiogenic pulmonary oedema is due to injury to the alveolar septa
  • Primary injury to the vascular endothelium or damage to alveolar epithelial cells (with secondary microvascular injury) produces an inflammatory exudate that leaks into the interstitial space and, in more severe cases, into the alveoli.
1167
Q

How does pneumonia cause pulmonary oedema?

A
  • In most forms of pneumonia the oedema remains localised and is overshadowed by the manifestations of infection.
  • When diffuse, however, alveolar oedema is an important contributor to a serious and often fatal condition.
1168
Q

Restrictive lung disorders occur in two general conditions. What are they?

A
  1. Chronic interstitial and infiltrative diseases, such as pneumoconioses and interstitial fibrosis of unknown ethology
  2. Chest wall disorders such as neuromuscular diseases such as poliomyelitis, severe obesity, pleural diseases and kyphoscoliosis
1169
Q

What are chronic interstitial pulmonary diseases?

A
  • They are a heterogeneous group of disorders characterised predominantly by inflammation and fibrosis of the pulmonary interstitium
  • Many of the entities are of unknown cause and pathogenesis, and some have an intra-alveolar as well as an interstitial component.
1170
Q

What are the symptoms of chronic interstitial pulmonary diseases? Including functional abilities

A
  • In general, the clinical and pulmonary functional changes are those of restrictive lung disease
  • Patients have dyspnoea, tachypnea, end-inspiratory cracked and eventual cyanosis, without wheezing or other evidence of airway obstruction.
  • The classic functional abnormalities are reductions in diffusion capacity, lung volume and lung compliance.
1171
Q

What are the classes CXR features of chronic diffuse interstitial lung diseases? What are the complicates of them?

A
  • Chest radiographs show bilateral lesions that take the form of small modules, irregular lines, or ground-glass shadows, all corresponding to areas of interstitial fibrosis
  • Eventually, secondary pulmonary HTN and right-sided heart failure associated with cor pulmonale may result
1172
Q

What are the major categories of chronic interstitial lung diseases and give some examples of each?

A

Fibrosing - idiopathy pulmonary fibrosis, radiation pneumonitis, non-specific interstitial pneumonia, connective tissue diseases-associated
Granulomatous - sarcoidosis, hypersensitivity pneumonitis
Eosinophilic
Smoking related - desquamative interstitial pneumonia
Other - Langerhands cell histiocytosis

1173
Q

What is idiopathic pulmonary fibrosis?

A

Idiopathic pulmonary fibrosis refers to a clinicopathologic syndrome marked by progressive interstitial pulmonary fibrosis and respiratory failure.
It has characteristic radiologic, pathologic and clinical features.

1174
Q

While the cause of idiopathic pulmonary fibrosis remains unknown, it spread that the fibrosis arises in genetically predisposed individuals who are prone to aberrant repair of recurrent alveolar epithelial cell injuries caused by environmental exposures. What are the implicated factors?

A
  • Environmental factors - cigarette smoking, exposure to metal fumes or wood dust. It is thought that exposure to these things causes recurrent alveolar epithelial cells damage.
  • Genetic factors - potentially germline loss-of-function mutates in the TERT and TERC gibers, which encode components of telomerase, making pneumocytes more sensitive to environmental insults, enhancing the consequences of cellular dysfunction and injury
  • Age - occurs in older people
1175
Q

What is the gross morphology of idiopathic pulmonary fibrosis?

A

The pleural surfaces of the lung are cobblestones as a result of the retraction of scars along the interlobular septa.
The cut surface shows firm, rubbery white areas of fibrosis, which occurs preferentially in the lower lobes, the subpleural regions, and making the interlobular septa.

1176
Q

What are the microscopic features of idiopathic pulmonary fibrosis?

A
  • The hallmark is patchy interstitial fibrosis, which varies in intensity and age.
  • The earliest lesions contain exuberant fibroblastic proliferation (fibroblastic foci). With time these areas become more collagenous and less cellular. Quite typical is the coexistence of both early and late lesions.
  • The dense fibrosis causes the destruction of alveolar architecture and formation of cystic spaces lined by hyperplasticity type II pneumocytes or bronchiolar epithelium (honeycomb fibrosis).
1177
Q

What are some histological changes with idiopathic pulmonary fibrosis?

A
  • There is mild to moderate inflammation within the fibrotic areas, consisting of mostly lymphocyte admixed with a few plasma cells, neutrophils, eosinophils and mast cells
  • Foci of squamous metaplasia and smooth muscle hyperplasia may be present, along with pulmonary arterial hypertensive changes (intimal fibrosis and medial thickening).
  • In acute exacerbation diffuse alveolar damage may be superimposed on these chronic changes.
1178
Q

What is the clinical course of idiopathic pulmonary fibrosis?

A
  • IPF begins insidiously with gradually increasing dyspnoea on exertion and dry cough
  • Most patients are 55-75 years old at presentation
  • Hyperaemia, cyanosis, and clubbing occur late in the course.
  • The progression in an individual patient is unpredictable Usually there is a gradual deterioration in pulmonary status despite medical treatment with immunosuppressive drugs such as steroids, cyclophosphamide and azathioprine.
  • Other IPF patients have acute exacerbations of the underlying disease and follow a rapid downhill clinical course
  • The median survival is about 3 years after diagnosis. Lung transplantation is the only definitive therapy.
1179
Q

What is non-specific interstitial pneumonia?

A

Diffuse interstitial lung disease with lung biopsies lack the diagnostic featured of any of the other well-characterised interstitial diseases.

1180
Q

How does the prognosis of non-specific interstitial pneumonia differ from the usual interstitial pneumonia?

A

Patients with non-specific interstitial pneumonia have a much better prognosis than do those with usual interstitial pneumonia.

1181
Q

On the basis of its histology, non-specific interstitial pneumonia is divided into cellular and fibrosing patterns. What are these?

A
  • The cellular pattern consists primarily of mild to moderate chronic interstitial inflammation, containing lymphocytes and a few plasma cells, in a uniform or patchy distribution.
  • The fibrosing pattern consists of diffuse or patchy interstitial fibrotic lesions of roughly the same stage of development, an important distinction from usual interstitial pneumonia. Fibroblastic foci, honeycombing, hyaline membranes and granolas are absent.
1182
Q

What is the clinical course of non-specific interstitial pneumonia? What population does it occur in? What do you see on CT?

A
  • Patients present with dyspnoea and cough of several months duration.
  • They are more likely to be female non-smokers in their sixth decade of life. Those having the cellular pattern are somewhat younger than those with the fibrosing pattern and have a better prognosis
  • Key features on high-resolution CT are bilateral, symmetric, predominantly lower lobe reticular opacities.
1183
Q

What is cryptogenic organising pneumonia? What do patients present with?

A
  • A clinicopathologic syndrome of unknown etiology.
  • Patients present with cough and dyspnea and have patchy subpleural or peri-bronchial areas of airspace consolidation radiographically.
1184
Q

What are the histological findings in cryptogenic organising pneumonia?

A
  • It is characterised by the presence of polypoid plugs of loose organising connective tissue (Masson bodies) within alveolar ducts, alveoli, and often bronchioles.
  • The connective tissue is all of the same age, and the underlying lung architecture is normal. There is no interstitial fibrosis or honeycomb lung.
1185
Q

What is the treatment of cryptogenic organising pneumonia?

A

Some patients recover spontaneously, but most need treatment with oral steroids for 6 months or longer for complete recovery.

1186
Q

What usually triggers a cryptogenic organising pneumonia?

A
  • Organising pneumonia with intra-alveolar fibrosis is more often seen as a response to infections or inflammatory injury of the lungs
  • These include viral and bacterial pneumonia, inhaled toxins, drugs, connective tissue disease, and graft-versus-host disease in bone marrow transplant recipients.
  • The prognosis for these patients is dependent on the underlying disorder
1187
Q

Many autoimmune diseases can involve the lung at some point in their course. Pulmonary involvement can take different histologic patterns; non-specific interstitial pneumonia, usual interstitial pneumonia, vascular sclerosis, organising pneumonia and bronchiolitis are the most common. Tell me about the different lung changes in common autoimmune diseases.

A
  • Rheumatoid arthritis: pulmonary involvement may occur in 30-40% of patients as 1) chronic pleuritic, with or without effusion; 2) diffuse interstitial pneumonitis and fibrosis; 3) intrapulmonary rheumatoid nodules; 4) follicular bronchiolitis; or 5) pulmonary hypertension
  • Systemic sclerosis: diffuse interstitial fibrosis and pleural involvement
  • Lupus erthematosus: patchy, transient parenchymal infiltrates, or occasionally severe lupus pneumonitis as well as pleurisy and pleural effusions.
1188
Q

What is sarcoidosis? What organs does it effect?

A
  • Sarcoidosis is a systemic granulomatous disease of unknown cause that may involve different tissues and organs
  • It presents in many clinical pattens, but bilateral hilar lymphadenopathy or lung involvement is most common, occurring in 90% of cases. Eye are skin lesions are next in frequency.
1189
Q

What populations does sarcoidosis usually occur in?

A
  • Sarcoidosis usually occurs in adults younger than 40 years of age, but can affect any group
  • The prevalence is higher in women but caries widely in different countries and populations.
  • In the US, the rates are higher in black people than white people and it rare among Chinese and Southeast Asians.
  • Patterns of organ involvement also vary with race
1190
Q

What is the pathogenesis of sarcoidosis?

A

Although the etiology of sarcoidosis remains unknown, several lines of evidence suggest that it is a disease of disordered immune regulation in genetically predisposed individuals. It is not clear whether exposure to any environmental or infectious agent has a role in its pathogenesis.

1191
Q

There are several immunological abnormalities in the local milieu of sarcoid granulomas that suggest the involvement of a cell-mediated immune response to an unidentified antigen. What are these abnormalities?

A
  • Intra-alevolar and interstitial accumulation of CD4+ T cells, resulting in CD4/CD8 T-cell ratios ranging from 5:1 to 15:1; suggesting pathogenic involvement of CD4+ helper T cells. It is likely antigen-driven proliferation
  • Increased levels of T cell-derived TH1 cytokines such as IL-2 and IFN-γ
  • Increased levels of several cytokines in the local environment (IL-8, TNF) that favour recruitment of additional T cells and monocytes.
  • Impaired dendritic cell function
1192
Q

What are the genetic factors of sarcoidosis?

A

Familial and racial clustering of cases and association with certain HLA genotypes.

1193
Q

What are the cell changes in sarcoidosis involving all tissues affected by it?

A
  • Regardless of the tissue, involved tissues contain well-formed non-necrotising granulomas composed of aggregates of tightly clustered epithelioid macrophages, often with giant cells. Central necrosis is unusual
  • With chronicity, the granulomas may become enclosed within fibrous rims or may eventually be replaced by hyaline fibrous scars.
  • Laminated concentrations composed of calcium and proteins known as Schaumann bodies and stellate inclusions known as asteroid bodies are found within giant cells in approx 60% of the granulomas
1194
Q

The lungs are a common organ effected by sarcoidosis, what are the macroscopic changes?

A
  • Macroscopically, there is usually no demonstrable alteration, although in advanced cases the coalescence of granulomas produces small bodies that are palpable or visual as 1-2cm non-caseating, non-cavitated consolidations.
  • The lesions are distributed primarily along the lymphatics around bronchi and blood vessels, although alveolar lesions and pleural involvement are also seen
  • The relatively high frequency of granulomas in the bronchial submucosa accounts for the high diagnostic yield of bronchoscopic biopsies.
  • There seems to be strong tendency for lesions to heal in the lungs, so varying stages of fibrosis and hyalinisation are often found.
1195
Q

Lymph nodes are involved in almost all cases of sarcoidosis. What are the changes to them?

A

Nodes are characteristically enlarged, discrete, and sometimes calcified.

1196
Q

How is the spleen and liver affected in sarcoidosis?

A
  • The spleen is affected in about 3/4 of cases, but it is enlarged in only one fifth. On occasion, granulomas may coalesce to form small modules that are visible macroscopically
  • The liver is affected slightly less often than the spleen. It may be moderately enlarged and typically contains scattered granulomas, more in portal triads than in the lobular parenchyma.
1197
Q

The bone marrow is involved in about 1/5th of cases of sarcoidosis. What will you see radiologically and which bones?

A

Radiologically visible bone lesions have a particular tendency to involve phalangeal bones of the hands and feet, creating small circumscribed areas of bone resorption within the marrow cavity and diffuse reticulated pattern throughout the cavity, with widening of the bony shaft or new bone formation on the outer surfaces.

1198
Q

Skin lesions are encountered in 1/4 of sarcoidosis cases, how do they appear?

A

They assume a variety of appearances, including:
* discrete subcutaneous nodules, * focal, slightly elevated, erythematous plaques
* flat lesions that are slightly reddened and scaling, resembling those of SLE
* they may also appear on the mucous membranes of the oral cavity, larynx and upper resp tract

1199
Q

Ocular involvement is seen in 1/4 sarcoidosis cases. How? What are the consequences?

A
  • It takes the form or iritis or iridocyclitis, either bilaterally or unilaterally.
  • Consequently, corneal opacities, glaucoma and total loss of vision may occur
1200
Q

Why is Mikulicz syndrome?

A

Bilateral sarcoidosis of the parotid, submaxillary and sublingual glands constituting the combined uveoparotid involvement.

1201
Q

How does sarcoidosis present?

A
  • Because of it’s varying severity and inconsistent tissue distribution, sarcoidosis may present with diverse features.
  • It may be discovered unexpectedly on routine chest films as bilateral pilar adenopathy or may present with peripheral lymphadenopathy, cutaneous lesions, eye involvement, splenomegaly, or hepatomegaly.
  • Mostly, individuals seek medical attention because of the insidious onset of respiratory abnormalities (SOB, cough, chest pain, haemoptysis) or of constitutional signs and symptoms (fever, fatigue, weight loss, anorexia, night sweats)
1202
Q

Sarcoidosis follows as unpredictable course. Tell me about some of them.

A
  • It may be inexorably progressive or marked by periods of activity interspersed with remission, sometimes permanent, that may spontaneous or induced steroid therapy
  • Overall, 65-70% of affected patients recover with minimal or no residual manifestations
  • 20% have permanent loss of some lung function or some permanent visual impairment.
  • Of the remaining 10-15%, some die of cardiac or CNS damage, but most succumb to progressive pulmonary fibrosis and cor pulmonale.
1203
Q

What is hypersensitivity pneumonitis?

A

It describes a spectrum of immunologically mediated, predominantly interstitial, lung disorders caused by intense, often prolonged exposure to inhaled organic antigens.

1204
Q

What is the pathogenesis of hypersensitivity pneumonitis?

A

Affected individuals have an abnormal sensitivity or heightened reactivity to the causative agent, which, in contrast to asthma, leads to pathological changes that primarily involve the alveolar walls.

1205
Q

Why is it important to recognise hypersensitivity pneumonitis early in its course?

A

Because progression to serious chronic fibrotic lung disease can be prevented by removal of the environmental agent.

1206
Q

What are the main causes of hypersensitivity pneumonitis?

A
  • Most commonly, hypersensitivity results from the inhalation of organic dust containing antigens made up of the spores of thermophilic bacteria, fungi, animal proteins, or bacterial products
  • Farmer’s lung results from exposure to dusts generated from humid, warm, newly harvested hay that permits rapid proliferation of the spores of thermophilic actinomycetes
  • Pigeon breeder’s lung is provoked by proteins from serum, excreta, or feathers of birds
  • Humidifier lung is caused by thermophilic bacteria in heated water reservoirs
1207
Q

Several lines of evidence suggest that hypersensitivity pneumonitis is an immunologically mediated disease. What are they?

A
  • Bronchoalveoalr lavage specimens from the acute phase show increased levels of pro inflammatory chemokines such as IL-8 and also consistently demonstrate high level of CD4 and CD8 T lymphocytes.
  • Complement and immunoglobulins are within vessel walls
  • The presence on non-caseating granulomas in 2/3rds of patients suggest that T-cell-mediated (type IV0 hypersensitivity reactions against the implicated antigens are also common and have a pathogenic role.
1208
Q

What are the morphological changes of hypersensitivity pneumonitis?

A

Histologic changes are characteristically centred on bronchioles. They include
1) interstitial pneumonitis, consisting primarily of lymphocytes, plasma cells, and macrophages (eosinophils are rare)
2) non-caseating granulomas in 2/3rds of patient
3) interstitial fibrosis with fibroblastic foci, honeycombing, and obliterative bronchiolitis (in late stages)

1209
Q

What are the clinical features of hypersensitivity pneumonia?

A
  • Acute attacks, which follow inhalation of antigenic dust in sensitised patients, consisting of recurring episodes of fever, dyspnoea, cough and leukocytosis.
  • Symptoms usually appear 4-6 hours after exposure and may last for 12 hours to several days. They recur with re-exposure.
  • If exposure is continuous and protracted, a chronic form of the disease supervenes, leading to progressive respiratory failure, dyspnoea, and cyanosis and a decrease in total lung capacity and compliance
1210
Q

What are the CXR and pulmonary functions tests with hypersensitivity pneumonitis?

A
  • Micronodular interstitial infiltrates may appear in the CXR
  • PFTs show an acute restrictive disorder
1211
Q

Although relatively rare, there are several clinical and pathological pulmonary entities that are characterised by an infiltration of eosinophils (pulmonary eosinophilia). What causes them? What are the different categories of pulmonary eosinophilia?

A

They are characterised by an infiltration of eosinophils, recruited in part by elevated alveolar levels of eosinophil attractant such as IL-5.
Pulmonary eosinophilia is divided into:
* Acute eosinophilic pneumonia with respiratory failure
* Secondary eosinophilia
* Idiopathic chronic eosinophilic pneumonia

1212
Q

What is acute eosinophilic pneumonia with respiratory failure? i.e. symptoms, imaging, hisotlogy and prognosis

A
  • It is an acute illness of unknown cause
  • It has a rapid onset with fever dyspnoea, and hypoxemic respiratory failure
  • The CXR shows diffuse infiltrates, and bronchoalveolar lavage fluid contains more than 25% eosinophils
  • Histology shows diffuse alveolar damage and many eosinophils
  • There is a prompt response to corticosteroids.
1213
Q

What causes secondary pulmonary eosinophilia?

A

It occurs in a number of parasitic, fungal and bacterial infections, in hypersensitivity pneumonitis; in drug allergies; and in association with asthma, allergic bronchopulmonary aspergillosis, or vasculitis (Churg-Strauss syndrome)

1214
Q

What are the cellular characteristics of idiopathic chronic eosinophilic pneumonia? What are the symptoms and treatments?

A
  • It is characterised by focal areas of cellular consolidation of the lung substance distributed chiefly in the periphery of the lung fields.
  • Prominent in these lesions are heavy aggregates of lymphocytes and eosinophils within both the septal walls and the alveolar spaces.
  • Interstitial fibrosis and organising pneumonia are often present
  • These patients have cough, fever, night sweats, dyspnoea, and weight loss, all of which response to corticosteroid therapy.
1215
Q

What are some smoking-related pulmonary interstitial diseases?

A
  • Smoking-related diseases can be grouped into obstructive diseases (emphysema and chronic bronchitis) and restrictive or interstitial diseases.
  • A majority of individuals with idiopathic pulmonary fibrosis are smokers
  • Desquamative interstitial pneumonia and respiratory bronchiolitis-associated lung diseases are also smoking-associated lung diseases
1216
Q

What is desequamative interstitial pneumonia? What are its clinical features? and in who does it usually present?

A
  • It is characterised by large collections of macrophages in the airspaces in a current or former smoker.
  • Presenting symptoms include an insidious onset of dyspnoea and dry cough over weeks or mon the, often associated with clubbing of digits.
  • It usually presents in the fourth or fifth decade of life, and is now equally common in men and women
1217
Q

What is the morphological of desquamative interstitial pneumonia?

A
  • The most striking finding is the accumulation of a large number of macrophages with abundant cytoplasm containing dusty brown pigment (smokers macrophages) in the airspaces
  • Finely granular iron may be seen in the macrophage cytoplasm.
  • Some of the macrophages contain lamellar bodies (composed of surfactant) within phagocytic vacuoles, presumably derived from necrotic type 2 pneumoncytes.
  • The alveolar septa are thickened by a sparse inflammatory infiltrate of lymphocytes, plasma cells, and occasional eosinophils.
  • The septa are lined by plump, cuboidal pnuemocytes.
  • Interstitial fibrosis, when present, is mild. Emphysema is often present
1218
Q

What is respiratory bronchiolitis-associated interstitial lung disease?

A
  • Respiratory bronchiolitis-associated interstitial lung disease is marked by chronic inflammation and peribronchiolar fibrosis.
  • It is a common histologic lesion in cigarette smokers.
  • It is characterised by the presence of pigmented intraluminal macrophages within first- and -second order respiratory bronchioles.
  • Symptoms are usually mild, consisting of gradual onset of dyspnea and cough in patients who are typically current smokers in the fourth or fifth decade of life with average exposures of over 30 pack-years of cigarette smoking
1219
Q

What are the morphological changes in respiratory bronchiolitis-associated interstitial lung disease?

A
  • The changes are patchy at low magnification and have a bronchiolocentric distribution.
  • Respiratory bronchioles, alveolar ducts, and peribronchiolar spaces contain aggregates of dusty brown macrophages (smokers macrophages) similar to those seen in desquamative interstitial pneumonia.
  • There is a patchy submucosal and peribonchiolar infiltrate of lymphocytes and histiocytes.
  • Mild peribronchiolar fibrosis is also seen, which expands contiguous alveolar septa.
  • Desquamative interstitial pneumonia is often found in different parts of the same lung
1220
Q

What is Pulmonary Langerhans cell histiocytosis? What are Langerhans cells?

A
  • Pulmonary Langerhans cell histiocytosis is a rare disease characterised by focal collections of Langerhans cells (often accompanied by eosinophils).
  • As these lesions progress scarring occurs, leading to airway destruction and alveolar damage the result in the appearance of irregular cystic spaces.
  • Langerhans cells are immature dendritic cells with grooved, indented nuclei and abundant cytoplasm. They are positive for S100, CD1s, and CD207 (langerin) and are negative for CD68.
1221
Q

What does the imaging show in pulmonary langerhans cell histiocytosis?

A

It shows characteristic cystic and nodular abnormalities.

1222
Q

What is pulmonary alveolar proteinosis? How is is characterised radiologically?

A
  • Pulmonary alveolar proteinosis is a rare disease caused by defects related to granulocyte-macrophage-colony-stimulating factor (GM-CSF) or pulmonary macrophage dysfunction that results in the accumulation of surfactant in the intra-alveolar and bronchiolar spaces
  • PAP is characterised radiologically by bilateral patchy asymmetric pulmonary opacifications.
1223
Q

There are three distinct classes of pulmonary alveolar proteinosis. What are they and how are they different?

A
  • Autoimmune PAP is caused by circulating neutralising antibodies specific for GM-CSF. It occurs primarily in adults, represented 90% of all cases of PAP, and lacks any familial predisposition. Loss if GM-CSF signalling blocks the terminal differentiation of alveolar macrophages impairing their ability to catabolise surfactant
  • Secondary PAP is uncommon and associated with haematopoietic disorders, malignancies, immunodeficiency disorders, and acute silicosis. It is speculated these diseases somehow impair macrophage maturation or function
  • Hereditary PAP is extremely rare occurs in neonates and is caused by mutations that disrupt genes involved in GM-CSF signalling
1224
Q

What is the morphology of pulmonary alveolar proteinosis?

A
  • The disease is characterised by a peculiar homogenous, granular precipitate containing surfactant proteins within the alveoli, causing focal-to-confluent consolidation of large areas of the lungs with minimal inflammatory reaction.
  • As a consequence there is a marked increase in the size and weight of the lungs
  • The alveolar precipitate is periodic acid-Schiff positive and contains cholesterol clefts and surfactant proteins
  • Ultrastructurally, the surfactant lamellae in type II pneumoncytes are normal
1225
Q

What are the clinical features of pulmonary alveolar proteinosis?

A
  • Adult patents, for the most part, present with cough and abundant sputum that often contains chunks of gelatinous material.
  • Some have symptoms lasting for years, often with febrile illness
  • Progressive dyspnoea, cyanosis, and respiratory insufficiency may occur, but other patients follow a benign course, with eventual resolutions of the lesions
1226
Q

What is the treatment of pulmonary alveolar proteinosis?

A
  • Whole-lung lavage is the standard of care and provides benefit regardless of the underling defect.
  • GM-CSF therapy is safe and effective in more than half of the patients with autoimmune PAP while therapy directed at the underlying disorder may be helpful in secondary PAP.
1227
Q

Surfactant dysfunction disorders are diseases caused by mutations in genes encoding proteins involved in surfactant trafficking or secretion. What are the mutated genes involved?

A
  • ATP-binding cassette protein member 3 (ATPCA3) is the most frequently mutated gene in surfactant dysfunction disorders. It is an autosomal recessive disorder and usually presents in the first few months of life with rapidly progressive respiratory failure followed by death.
  • Surfactant protein C is the second most commonly mutated gene in surfactant dysfunction disorders. It is autosomal dominant with variable penetrance and severity in 45% and sporadic in 55%. It has a highly variable course
  • Surfactant protein B is the least commonly mutated gene and is associated with an autosomal recessive form of surfactant dysfunction disorder. Typically, the infant is full term and rapidly develops progressive respiratory distress shortly after birth. Death ensues between 3 and 6 months of age
1228
Q

What is the morphology of surfactant dysfunction disorders?

A
  • There is variable amount of intra-alveolar pink granular material, type II pneumocyte hyperplasia, interstitial fibrosis and alveolar simplification.
  • Immunohistochemical stains show the lack of surfactant proteins C and B in their respective deficiencies.
  • Ultrastructurally, abnormalities in lamellar bodies in type II pneumocytes can be seen in all three; small lamellar bodies with electron dense cores are diagnostic for ABCA3 mutation
1229
Q

Pulmonary emboli have two deleterious pathophysiologic consequences. What are they?

A
  • Respiratory compromise due to the non-perfused, although ventilated, segment
  • Haemodynamic compromise due to increased resistance to pulmonary blood flow caused by the embolic obstruction.
1230
Q

Where are large and small pulmonary emboli

A
  • Large emboli lodge in the main pulmonary artery or its major branches or at the bifurcation as a saddle embolus.
  • Smaller emboli travel out into the more peripheral vessels, where they may cause haemorrhage or infarction.
1231
Q

How do patient withs adequate cardiovascular function cope with PE compared with those who do not have adequate cardiovascular function?

A
  • In patient with adequate cardiovascular function, the bronchial artery supply sustains the lung parenchyma; in this instance, haemorrhage may occur, but there is no infarction
  • In those in whom the cardiovascular function is already compromised, such as patient with heart of lung disease, infarction may occur
1232
Q

What proportion of PEs cause infarction? Where in the lung is this most commonly?

A
  • Overall, about 10% of emboli cause infarction.
  • About three fourths of infarcts affect the lower lobes, and in more than half, multiple lesions occur.
  • they vary in size from barely visible to massive lesions involving large parts of a lobe.
  • Typically, they extend to the periphery of the lung as a wedge with the apex pointing toward the hilus of the lung.
  • In many cases, an occluded vessel is identified near the apex of the infarct.
1233
Q

What is the morphological changes in the time following a PE?

A
  • The pulmonary infarct is classically haemorrhagic and appears as a raised, red-blue area in the early stages.
  • Often, the apposed pleural surface is covered by a fibrinous exudate.
  • The red cells begin to lyse with 48 hours, and the infarct becomes paler and eventually red-brown as hemosiderin is produced.
  • With the passage of time, fibrous replacement begins at the margins as a grey-white peripheral zone and eventually converts the infarct into a contracted scar
1234
Q

What are the histological changes of a PE? How is it different with an infected embolus?

A
  • Histologically, the haemorrhagic area shows ischaemic necrosis of the alveolar walls, bronchioles, and vessels,
  • If the infarct is caused by an infected embolus, the neutrophilic inflammatory reaction can be intense
  • Such lesions are referred to as septic infarcts, some which turn into abscesses
1235
Q

What is a common arrest rhythm with PE?

A

The patient is frequently said to have electromechanical dissociation, in which the ECG has a rhythm but no pulses are palpated because no blood is entering the pulmonary arterial circulation.

1236
Q

What are the clinical features of PE?

A
  • In the patient survives a sizeable PE, the clinical syndrome may mimic MI, with severe chest pain, dyspnoea and shock
  • Small emboli are silent of indue only transient chest pain and cough
  • Pulmonary infarcts manifest as dyspnoea, tachypnoea, fever, chest pain, cough and haemoptysis.
  • An overlying fibrinous pleuritic may produce a pleural friction rub
1237
Q

What are the radiological findings in PE?

A
  • Findings on CXR are variable, and can be normal or dispose a pulmonary infarct, usually 12-36 hours after it has occurred, as a wedge-shaped infiltrate.
  • You’ll see it on a CTPA
  • DVT can be diagnosed with duplex US
1238
Q

What are the consequences of a PE after the initial insult?

A
  • After the initial acute insult, emboli often resolves via contraction and fibrinolysis, particularly in the relatively young.
  • If unresolved, over the course of time multiple small emboli may lead to pulmonary HTN and chronic cor pulmonale.
  • Perhaps most important is that a small embolus may presage a larger one.
  • In the presence on an underlying predisposing condition, patients with a pulmonary embolus have a 30% chance of suffering a second embolus.
1239
Q

What is the prophylaxis and treatment for PE?

A
  • Prevention of pulmonary embolism is a major clinical challenge for which there is no easy solution. Prophylactic therapy includes early ambulation in post-op and postpartum patients, elastic stockings and graduated compression stockings for bedridden patients, and anticoagulation in high-risk individuals.
  • Treatment of PE includes anticoagulation and supportive measures; thrombolysis may have some benefit in those with severe complications (e.g, shock), but carries a high risk of bleeding. Those at risk of recurrent PE in whom anticoagulation is contraindicated may be fitted with an IVC filter that catches clots before the reach the lungs
1240
Q

What is pulmonary hypertension?

A

It is defined as a mean pulmonary artery pressure greater than or equal to 25 mm Hg at rest.

1241
Q

Based on underlying mechanisms, the WHO has classified pulmonary HTN into five groups. What are they?

A

1) Pulmonary arterial HTN, a diverse collection of disorders that all primarily impact small pulmonary muscular arteries
2) Pulmonary HTN secondary to left-heart failure
3) Pulmonary HTN stemming from lung parenchymal disease or hypoxaemia
4) Chronic thromboembolic pulmonary HTN
5) Pulmonary HTN of multifactorial basis

1242
Q

Pulmonary HTN has diverse causes. What are the most frequently associated conditions?

A

It is most frequently associated with structural cardiopulmonary conditions that increase pulmonary blood flow, pulmonary vascular resistance, or left heart resistance to blood flow.

1243
Q

What are the some of the common causes of pulmonary hypertension?

A
  • Chronic obstructive or interstitial lung diseases. These diseases obliterate alveolar capillaries, increasing pulmonary resistance to blood flow and, secondarily, pulmonary blood pressure
  • Antecedent congenital or acquired heart disease. Mitral stenosis, for example, causes an increase in left atrial pressure and pulmonary venous pressure that is eventually transmitted to the arterial side of the pulmonary vasculature, leading to HTN.
  • Recurrent thromboemboli. Recurrent pulmonary emboli may cause pulmonary HTN by reducing the functional cross-sectional area of the pulmonary vascular bed, which in turn leads to an increase in pulmonary vascular resistance
  • Autoimmune diseases. Several of these diseases (most notably systemic sclerosis) involve the pulmonary vasculature and/or the interstitium, leading to increased vascular resistance and pulmonary HTN
  • Obstructive sleep apnoea is a common disorder that is associated with obesity and hypoxaemia. It is not recognised to be a significant contributor to the development of pulmonary HTN and cor pulmonale.
1244
Q

Uncommonly, pulmonary HTN is encountered in patients in whom all known causes are excluded. What is this called? Is it really not known about the cause?

A
  • This is referred to as an idiopathic pulmonary arterial HTN
  • However, this is a bit of misnomer, as up to 80% of “idiopathic” pulmonary HTN has a genetic basis, sometimes being inherited in families as an autosomal dominant trait.
  • Within these families, there is incomplete penetrance, and only 10% to 20% of the family members actually develop overt disease
1245
Q

What are the genes associated with pulmonary HTN?

A
  • Inactivating germline mutations in the BMPR2 gene are found in 75% of the familial cases of pulmonary hypertension, and 25% of sporadic cases.
  • BMPR2 is down-regulated in lungs from some idiopathic pulmonary arterial HTN patients without mutation in its gene.
  • A two-hot model has been proposed whereby a genetically susceptible individual with BMPR2 mutation requires additional genetic or environmental insults to develop the disease
1246
Q

What is BMPR2? What is its role?

A
  • A cell surface protein belonging to the TGF-β receptor superfamily, which binds a variety of cytokines, including TGF-β, bone morphogenetic protein (BMP), activin, inhibin.
  • BMP-BMPR2 signalling is nor know to be important for embryogenesis, apoptosis, and cell proliferation and differentiation.
  • Haploinsufficiency for BMPR2 leads to dysfunction and proliferation of endothelial cells and vascular smooth muscle cells.
1247
Q

What is the gross morphology of pulmonary HTN?

A

Regardless of their etiology, all forms of pulmonary hypertension are associated with medial hypertrophy of the pulmonary muscular and elastic arteries, pulmonary arterial atherosclerosis, and right ventricular hypertrophy.

1248
Q

How can you tell the cause of the pulmonary hypertension by the morphology?

A

The presence of many organising and recanalised thrombi favours recurrent PE as the cause, and the co-existence of diffuse pulmonary fibrosis, or severe emphysema and chronic bronchitis, points to chronic hypoxia as the initiating event.

1249
Q

What part of the respiratory vascular system does pulmonary hypertension effect? What changes does it have in each part?

A
  • The vessels changes can involve the entire arterial tree, from the main pulmonary arteries down to the arterioles.
  • In most severe cases, atheromatous deposits form the pulmonary artery and its major branches, resembling systemic atherosclerosis
  • The arterioles and small arteries are most prominently affected by striking medial hypertrophy and intimal fibrosis, sometimes narrowing the lumens to pinpoint channels.
1250
Q

One extreme pathological change in pulmonary hypertension is the plexiform lesion. What is this and what are the most common causes of pulmonary HTN that cause it?

A
  • It is a tuft of capillary formations, producing a network, or web, that spans the lumens of dilated, thin-walled, small arteries and may extend outside the vessel.
  • Plexiform lesions are most prominent in idiopathic and familial pulmonary hypertension, unrepaired congenital heart disease with left-to-right shunts, and pulmonary hypertension associated with HIV
1251
Q

What is the clinical course of pulmonary hypertension?

A
  • Idiopathic pulmonary HTN is most common in women who are 20-40 years of age and is occasionally seen in young children
  • Clinical signs and symptoms in all forms are only evident in advanced disease
  • The presenting features are usually dyspnoea and fatigue, but some patient have chest pain of the anginal type.
  • Over time, severe respiratory distress, cyanosis, and right ventricular hypertrophy occur, and death from decompensated cor pulmonale, often with superimposed thromboembolism and pneumonia, usually ensues within 2-5 years in 80% of patients
1252
Q

How do you treat pulmonary hypertension?

A
  • Treatment choices depend on the underlying cause
  • For those with secondary disease, therapy is directed ay the trigger.
  • A variety of vasodilators have been used with varying success
  • Lung transplantation provides definitive treatment for selected patients
1253
Q

What are some diffuse pulmonary haemorrhage syndromes?

A
  • Pulmonary haemorrhage is a dramatic complication of some interstitial lung disorders.

Some examples include:
1) Goodpasture syndeom
2) Idiopathic pulmonary haemosiderosis
3) Vasculitis-associated haemorrhage, which is found in conditions such as hypersensitivity angiitis, Wegener granulomatosis, and SLE

1254
Q

What is Goodpasture syndrome?

A

Goodpasture syndrome is an uncommon autoimmune disease in which kidney and lung injury are caused by circulating autoantibodies against the non-collagenous domain of the α3 chain of collagen IV.

1255
Q

What is Goodpasture syndrome that only affected the kidney? what percentage of patients have lung disease as well?

A
  • When only renal disease is caused by the antibody, it is called anti-glomerular basement membrane disease.
  • The term Goodpasture syndrome designates the 40-60% of patients who develop pulmonary haemorrhage in addition to renal disease.
1256
Q

How do the antibodies against the non-collagenous domain of the α3 chain of collagen IV cause damage to the kidneys and lung in Goodpasture syndrome?

A

The antibodies initiate inflammatory destruction of the basement membrane in renal glomeruli and pulmonary alveoli, giving rise to rapidly progressive glomerulonephritis and a necrotising haemorrhage interstitial pneumonitis

1257
Q

What population does Goodpasture syndrome effect?

A

Although any age can be affected, most cases occur in the teens or 20s, and in contrast to many other autoimmune diseases, there is a male preponderance.
The majority of patients are active smokers

1258
Q

What is the pathogenesis of Goodpasture syndrome?

A
  • The trigger that initiates the production of anti-basement membrane antibodies is still unknown.
  • Because the epitopes that evoke anti collagen antibodies are normally hidden within the molecule, it is presumed that some environmental insult such as viral infection, exposure to hydrocarbon solvents, or making is required to unmask the cyrptic epitopes.
1259
Q

What is the morphology o the lungs in Goodpasture syndrome?

A
  • In the classic case, the lungs are heavy, with areas of red-brown consolidation
  • Histologically, there is focal necrosis of alveolar walls associated with intra-alveolar haemorrhages. Often the alveoli contain hemosiderin-laden macrophages.
  • In later stages, there may be fibrous thickening of the septa, hypertrophy of type II pneuomocytes and organisation of blood in alveolar spaces.
  • In many cases, immunofluorescence studies reveal linear deposits of immunoglobulins along the basement membranes of the septal walls
1260
Q

What is the morphology of the kidneys in Goodpasture syndrome?

A
  • The kidneys have the characteristic findings of focal proliferator glomerulonephritis in early cases or crescentic glomerulonephritis in patients with rapidly progressive glomerulonephritis.
  • Diagnostic linear deposits of immunoglobulins and complement are seen along the glomerular basement membranes even in the few patients without renal disease
1261
Q

What are the clinical features of Goodpasture syndrome?

A
  • Most cases begin clinically with respiratory symptoms, principally haemoptysis, and radiographic evidence of focal pulmonary consolidations.
  • Soon, manifestations of glomerulonephritis appear, leading to rapidly p-rogressive renal failure.
1262
Q

What is the most common cause of death in Goodpasture syndrome? What is the treatment for it?

A
  • The most common cause of death is uraemia.
  • The prognosis have been improved by plasmapheresis, which is though to be beneficial by removing circulating anti-basement membrane antibodies as well as chemical mediators of immunologic injury.
  • Simultaneous immunosuppressive therapy inhibits further antibody production, ameliorating both lung haemorrhage and glomerulonephritis
1263
Q

What is idiopathic pulmonary haemosiderosis?

A
  • Idiopathic pulmonary hemosiderosis is a rare disorder characterised by intermittent alveolar haemorrhage.
  • The cause and pathogenesis are unknown, and no anti-basement membrane antibodies are detectable in serum or tissues.
1264
Q

What population does idiopathy pulmonary hemosiderosis occur in? What are the symptoms?

A
  • Most cases occur in young children, although the disease has been reported in adults as well.
  • It usually presents with an insidious onset of productive cough, haemoptysis and anaemia associated with diffuse pulmonary infiltrations similar to Goodpasture syndrome.
1265
Q

What is the treatment for idiopathic hemosiderosis?

A
  • Favourable response to long-term immunosuppression with prednisolone and azathioprine.
  • In addition, long-term follow-up of patients shows that some of them develop other immune disorders
1266
Q

Pneumonia can result whenever the pulmonary local defence mechanisms are impaired. This can be compromised by many factors. What are some?

A
  • Loss or suppression of the cough reflex as a result of coma, anaesthetic , neuromuscular disorders, drugs, or chest pain
  • Injury to the mucociliary apparatus by either impairment of ciliary function or destruction of ciliated epithelium, due to cigarette smoke, inhalation of hot or corrosive gases, viral diseases, or genetic defects
  • Accumulation of secretions in conditions such as CF and bronchial obstruction
  • Interference with the phagocytic or bactericidal action of alveolar macrophages by alcohol, tobacco smoke, anoxia, or oxygen intoxication
  • **Pulmonary congestion and oedema
1267
Q

How do defects in innate immunity vs cell-mediated immune defects change the type of pneumonia ?

A
  • Defects in innate immunity (including neutrophil and complement defects) and humeral immunodeficiency typically lead to an increased incidence of infections with pyogenic bacteria
  • Germline mutations in MyD88 (an adaptor for several TLRs that is important for activation of the transcription factor NFκB) are also associated with destructive bacterial (pneumococcal) pneumonias.
  • Cell-mediated immune defects (congenital and acquired) lead to increased infections with intracellular microbes such as mycobacteria and HSV as well as with microorganisms of very low virulence, such as Pneumocystis jiroveci
1268
Q

What is community-acquired bacterial pneumonia?

A
  • Community-acquired acute pneumonia refers to lung infection in otherwise healthy individuals that is acquired from the normal environment.
1269
Q

Are CAPs viral or bacterial? How can you tell?

A
  • It may be bacterial or viral
  • Clinical and radiologic features are usually insensitive in differentiating between them
  • CRP and procalcitonin, both acute-phase reactants produced primarily in the liver, are significantly elevated in bacterial more than in viral infections.
1270
Q

How do bacterial CAPs cause damage?

A

Bacterial invasion of the lung parenchyma causes the alveoli to be filled with an inflammatory exudate, thus causing consolidation of the pulmonary tissue.

1271
Q

What are some predisposing conditions that increase the risk of CAPs in patients?

A
  • extremes of age
  • chronic disease (CHF, COPD, T1/2DM)
  • congenital or acquired immune deficiencies
  • decreased or absent splenic function (sickle cell or post-splenectomy)
1272
Q

What are some of the microbes that cause CAPs?

A
  • Strep. pneumoniae
  • Haemophilus influenzae
  • Moraxella catarrhalis
  • Staphylococcus aureus
  • Legionella pneumophilia
  • Klebsiella pneumoniae
  • Pseudomonas
  • Mycoplasma pneumonia
  • Chlamydia species
  • Viruses: RSV, parainfluenza virus, influenza A and B
1273
Q

What is the most common cause of CAP? How do you confirm the diagnosis?

A
  • Streptococcus pneumoniae is the most common cause of CAP
  • Examination of Gram-stained sputum is an important step in the diagnosis of acute pneumonia.
  • The presence of numerous neutrophils containing the typical gram-positive lancet-shaped diplococci supports the diagnosis of pneumococcal pneumonia
  • It must be remmerberd that S.pneumoniae is a part of the endogenous flora in 20% of adults and therefore false-positive results may be obtained.
  • Isolation of pneumococci from blood cultures is more specific but less sensitive (in the early phase, only 20-30% of patients have positive BCs)
1274
Q

What are some causes of health care-associated pneumonia?

A
  • Staph aureus, methicillin-sensitive
  • Staph aureus, methicillin-resistant
  • Pseudomonas aeruginosa
  • Streptococcus pneumoniae
1275
Q

What are some causes of hospital-acquired pneumonia?

A
  • Gram-negative rods, enterobacteriaecae (Klebsiella, E.coli, Pseudomonas)
  • Staph aureus (usually methicillin-resistant)
1276
Q

What are some microorganisms that cause aspiration pneumonia?

A

Anaerobic oral flora (Bacteroides, Prevotella, Furobcterium, Peptostreptococcus), admixed with aerobic bacteria (Streptococcus pneumoniae, Staph aureus, H.influenzae, Pseudomonas aeruginosa

1277
Q

What are some micro-organisms that cause chronic pneumonia?

A
  • Nocardia
  • Actinomyces
  • Granulomatous: Mycobacterium tuberculosis and atypical mycobacteria, Histoplasma capsulated, Coccidoides immitis, Blastocymes dermatitidis
1278
Q

What are some micro-organisms that cause necrotising pneumonia and lung abscesses?

A
  • Anaerobic bacteria, with or without mixed aerobic infection
  • Staph. aura, Klebsiella pneumoniae, Streptococcus progenies, and type 3 pneumococcus
1279
Q

What are some micro-organisms that cause pneumonia in the immunocompromised host?

A
  • CMV
  • Pneumocystitis jiroveci
  • Mycobacterium avian-intracellulare
  • Invasive aspergillosis
  • Invasive candidiasis
  • “Usual” bacterial, viral and fungal organisms
1280
Q

What vaccine can be used against strep pneumococcal?

A

Pneumococcal vaccines containing capsular piolysaccharides from the common serotypes are used in individuals at high risk for pneumococcal sepsis

1281
Q

What type of organism is Haemophilus influenzae? What are the subtypes? Which one is most virulent?

A
  • Haemophilus influenzae is a pleomorphic, gram-negative organism that occurs in encapsulated and non-encapsulated forms.
  • There are six serotypes of the encapsulated form (types a to f), of which type b is the most virulent.
1282
Q

How does the vaccine for haemophilus influenza work? Which subtype does it target?

A
  • Antibodies against the capsule protect the host from H.influenze infection; hence the capsular polysaccharide b is incorporated in the widely used vaccine against H.influenzae.
  • With the routine use of H.influenzae conjugate vaccines, the incidence of disease caused by the b serotype has declined significantly.
1283
Q

Infections with non-encapsulated forms, also called non-typeable forms of H.influenzae are increasing. How do these spread? Who do they affect?

A
  • They are less virulent, spread among the surface of the upper respiratory tract, and produce otitis media, sinusitis, and bronchopneumonia
  • Neonates and children with co-morbidities such as prematurity, malignancy and immunodeficiency are at high risk for development of invasive infection
1284
Q

Is H.influenze pneumonia critical in children? What areas of the lungs does it effect and how?

A
  • H.influenzae pneumonia, which may follow a viral respiratory infection, is a paediatric emergency and has a high mortality rate.
  • Descending laryngotracheobronchitis results in airway obstruction as the smaller bronchi are plugged by dense, fibrin-rich exudates containing neutrophils, similar to that seen in pneumococcal pneumonias.
  • Pulmonary consolidation is usually lobular and patchy but may be confluent and involve the entire lung lobe.
1285
Q

Apart from pneumonia, what other organs can H.influenzae effect in different populations?

A

H.influenzae also causes an acute, purulent conjunctivitis in children and, in predisposed older patients, may cause septicaemia, endocarditis, pyelonephritis, cholecystitis, and suppurative arthritis.

1286
Q

What is the most common bacterial cause of IECOPD?

A

H. influenzae

1287
Q

When is Moraxella catarrhalis pneumonia common?

A
  • Moraxella catarrhalis is being increasingly recognised as a cause of bacterial pneumonia, especially in the elderly.
  • It is the second most common bacterial cause of IECOPD.
  • Along with S.pneumoniae and H.influenzae, M.catarrhalis constitutes one of the three most common causes of otitis media in children.
1288
Q

In what situations is staphylococcus aureus more common as a cause of pneumonia? What are some of its common complications?

A
  • Staph. aureus is an important cause of secondary bacterial pneumonia in children and healthy adults following viral respiratory illnesses (e.g. measles in children and influenzae in both children and adults)
  • Staphylococcal pneumonia is associated with high incidence of complications, such as lung abscesses and empyema
  • IVDUs are at high risk for development of staph pneumonia in association with endocarditis.
1289
Q

In what populations is Klebsiella pneumoniae more common? How does it present?

A
  • Klebsiella is the most frequent cause of gram-negative bacteria pneumonia
  • It commonly afflicts debilitated and malnourished people, particularly chronic alcoholics.
  • Thick, mucoid, (often blood-tinged) sputum is characteristic, because the organism produces an abundant viscid capsular polysaccharide, which the patient may have difficulty expectorating
1290
Q

What population is more commonly affected by pseudomonas aeruginosa? Why is it’s spread important?

A
  • It is most common in CF and immunocompromised patients.
  • It is common in patients who are neutropenic and it has a propensity to invade blood vessels with consequent extra pulmonary spread.
  • Pseudomonas septicaemia is a very fulminant disease
1291
Q

In what population is Legionella pneumonia most common? Is it severe?

A
  • The mode of transmission is either inhalation of aerosolised organisms or aspiration of contaminated drinking water
  • Legionella pneumonia is common in individuals with predisposing conditions such as cardiac, renal, immunologic or haematologic disease.
  • Organ transplant recipients are particularly susceptible.
  • It can be quite severe, frequently requiring hospitalisation, and immunosuppressed patients may have fatality rates of up to 50%
1292
Q

How do you diagnose Legionella?

A
  • Rapid diagnosis is facilitated by demonstration of Legionella antigens in the urine or by a positive fluorescent antibody test on sputum samples
  • Culture remains the diagnostic gold standard
1293
Q

Bacterial pneumonia has two patterns of anatomic distribution. What are they? What’s the difference?

A
  • Patchy consolidation of the lung is the dominant characteristic of bronchopneumonia. The patchy involvement may become confluent, producing virtually total lobar consolidation; however, effective antibiotic therapy may limit involvement to a subtotal consolidation.
  • Consolidation of a large portion of a lobe or of an entire lobe defines lobar pneumonia.
1294
Q

In bacterial lobar pneumonia, there are four stages of the inflammatory response. What are they?

A
  • In the first stage, congestion, the lung is heavy, boggy, and red. It is characterised by vascular engorgement, intra-alveolar fluid with few neutrophils, and often the presence of numerous bacteria.
  • The stage of red hepatisation that follows is characterised by massive confluent exudation, as neutrophils, red cells, and fibrin fill the alveolar spaces. On gross examination, the lobe is red, firm and airless, with a liver-like consistency, hence the term hepatisation.
  • The stage of **gray hepatisation that follows is marked by progressive disintegration of red cells and the persistence of fibrinosuppurative exudate resulting in a colour change to grayish-brown.
  • In the final stage of resolution the exudate within the alveolar spaces is broken down by enzymatic digestion to produce granular, semi-fluid debris is resorbed, ingested by macrophages, expectorated, or organised by fibroblasts growing into it.
1295
Q

What is the pleural reaction to bacterial pneumonia?

A
  • Pleural fibrinous reaction to the underlying inflammation, often present in the early stages if the consolidation extends to the surface (pleuritis), may similarly resolve.
  • More often, it undergoes organisation, leaving fibrous thickening or permanent adhesions.
1296
Q

What is the morphology of bacterial bronchopneumonia?

A
  • Foci of bronchopneumonia are consolidated areas of acute suppurative inflammation.
  • The consolidation may be confined to one lobe but is more often multi lobar and frequently bilateral and basal because of the tendency of secretions to gravitate to the lower lobes.
  • Well-developed lesions are slightly elevated, dry, granular, grey-red to yellow, and poorly delimited at their margins.
  • Histologically, the reaction usually elicits a neutrophil-rich exudate that fills the bronchi, bronchioles, and adjacent alveolar spaces.
1297
Q

What are some of the complications of bacterial pneumonia?

A

1) Tissue destruction and necrosis, causing abscess formation (particularly common with type 3 pneumococci or Klebsiella infections)
2) Spread of infection to the pleural cavity, causing the intrapleural fibrinoscuppurative reactions known as empyema
3) Bacteremic dissemination to the heart valves, pericardium, brain, kidneys, spleen or joints, causing metastatic abscesses, endocarditis, meningitis, or suppurative arthritis.

1298
Q

What is the clinical course of bacterial CAP?

A
  • The major symptoms of CAP are abrupt onset of high fever, shaking chills, and cough producing mucopurulent sputum; occasional patients may have haemoptysis.
  • When pleuritis is present it is accompanied by pleuritic pain and pleural friction rub.
  • The whole lobe is radiopaque in lobar pneumonia, whereas there are focal opacities in bronchopneumonia.
1299
Q

What are some common community-acquired viral pneumonias?

A

Common viral infections include influenza virus types A and B, RSV, HMV, adenovirus, rhinoviruses, and varicella viruses.

1300
Q

Although the molecular details vary, all of the viruses that cause pneumonia produce disease through similar general mechanisms. What are these?

A
  • These viruses have tropisms that allow them to attach to and enter respiratory lining cells.
  • Viral replication and gene expression leads to cytopathic changes, inducing cell death and secondary inflammation.
  • The resulting damage and impairment of local pulmonary defences, such as mucociliary clearance, may predispose to bacterial superinfections, which are often more serious than the viral infection itself.
1301
Q

What species does influenza A infect?

A
  • Humans, pigs, horses, and birds
  • The are the major cause of pandemic and epidemic influenza infections.
1302
Q

The influenza genome encodes a number of proteins. What are they?

A

The most important from the vantage point of viral virulence are the haemagglutin and neuroaminidase proteins.

1303
Q

What does hemagglutinin do?

A
  • Hemagglutinin has three major subtypes (H1-H3).
  • It is a component of the influenza virus envelope, which consists of a lipid bilayer.
  • Hemagglutinin serves to attach the virus to its cellular target via sialic acid residues on surface polysaccharides
  • Following uptake of the virus into endosomal vesicles, acidification of the endoscope triggers a conformational change in hemagglutinin that allows the viral envelope to fuse with the host cell membrane, releasing the viral genomic RNAs into the cytoplasm of the cell.
1304
Q

What does neuraminidase do?

A
  • Neuraminidase in turn facilitates the release of newly formed virions that are budding from infected cells by cleaving sialic acid residues.
1305
Q

How do antibodies react to influenza viruses?

A

Neutralising host antibodies against viral hemagglutinin and neuraminidase prevent and ameliorate, respectively, infection with the influenza virus by interfering with these functions.

1306
Q

The viral genome is composed of eight single-stranded RNAs. Where are they? What do they determine?

A

The RNAs are packaged into helices by nucleoproteins that determine the influenza virus type (A, B, or C).

1307
Q

What causes epidemics of influenza?

A
  • Epidemics of influenza are caused by spontaneous mutations that alter antigenic epitopes on the viral hemagglutinin and neuraminidase proteins.
  • These antigenic changes (antigenic drift) result in new viral strains that are sufficiently different to elude, at least in part, anti-influenza antibodies produced in members of the population in response to prior exposures to other flu strains.
  • Usually, however, these new strains near sufficient resemblance to prior strains that some members of the population are at least partially resistant to infection.
1308
Q

What causes pandemics of influenza?

A
  • Pandemics, which are longer and more widespread than epidemics, occur when both the hemagglutinin and the neuraminidase genes are replaced through recombination with animal influenza viruses (antigenic shift).
  • In this instance, essentially all individuals are susceptible to the new influenza virus.
1309
Q

How does the unusual genome of influenza virus ensure that antigenic shifts leading to pandemics are inevitable?

A

Viral assembly involves packaging go each of the 8 viral RNAs into single burins, and it is easy to see how infection of an animal by two different flu types could lead to swapping of genetic material within co-infected cells, creating a completely new viral strain.

1310
Q

What happens on a cellular level after an influenza virus has infected someone if the host lacks protective antibodies?

A
  • The virus infects the pneumocytes and elicits several cytopathic changes.
  • Shortly after entry into pneumocytes, the viral infection inhibits sodium channels, producing electrolyte and water shifts that lead to fluid accumulation in the alveolar lumen.
  • This is followed by the death of the infected cells through several mechanisms, including inhibition of host cell mRNA translation and activation of caspases leading to apoptosis.
  • The death of epithelial cells exacerbates the fluid accumulation and releases “danger signals” that activate resident macrophages
  • In addition, prior to their death, infected epithelial cells release a variety of inflammatory mediators, including several chemokines and cytokines.
  • Mediators released from epithelial cells and macrophages activate the nearby pulmonary endothelium, allowing neutrophils to attach and extravasate into the interstitium within the first day or two of infection.
1311
Q

What are the clinical consequences of viral pneumonia?

A

In some cases, viral infection may cause sufficient lung injury to produce the acute respiratory distress syndrome, but more often severe and sometimes fatal pulmonary disease stems from a superimposed bacterial pneumonia. Of these, secondary pneumonias caused by S.aureus are particularly common and often life-threatening.

1312
Q

Control of a viral influenza pneumonia relies on several host mechanisms. What are they?

A
  • The presence of viral products induces innate immune responses in infected cells, such as the production of α- and β-interferon.
  • These mediators upregulate the expression of the MX1 gene, which encodes a GTPase that interferes with influenza gene transcription and viral replication.
  • As with other viral infections, NK cells and cytotoxic T cells can recognise and kill infected host cells, limiting viral replication and viral spread to adjacent pneumocytes.
  • The cellular immune response is eventually augmented by development of antibody responses to the viral hemagglutinin and neuraminidase proteins.
1313
Q

What is human metapneumovirus? Who does it affect?

A
  • It is a paramyxovirus discovered in 2001, is found worldwide and is associated with upper and lower respiratory tract infections.
  • It can infect any age groups but is most commonly seen in young children, elderly subjects, and immunocompromised patients.
1314
Q

What are the symptoms of human metapneumovirus? How is it diagnosed? How do you treat it?

A
  • Infections are clinical indistinguishable from those caused by RSV and are often mistaken for influenza.
  • Diagnostic methods include PCR test for viral RNA and direct immunofluorescence.
  • Ribavirin is the only antiviral treatment that is currently available for human MPV infections, it is used most commonly in immunocompromised patients with severe disease
1315
Q

All viral infections produce similar morphological changes. What are they?

A
  • Upper respiratory infections are marked by mucosal hyperaemia and swelling, lymphonocytic and plasmacytic infiltration of the submucosa, and overproduction of mucus secretions
  • The swollen mucose and viscous exudate may plug the nasal channels, sinuses or the Eustachian tubes, leading to suppurative secondary bacterial infection.
  • Virus-induced tonsilitis cuasing hyperplasia of the lymphoid tissue within the Waldeyer ring is frequent in children.
1316
Q

What are the morphological changes in viral laryngotracheobronchitis and bronchiolitis?

A
  • There is vocal cord swelling and abundant mucus production
  • Impariemtn of bronchocilliary function invites bacterial superinfection with more marked suppuration.
  • Plugging of small airways may give rise to focal lung atelectasis.
  • With more severe bronchiolar involvement, widespread plugging of secondary and terminal airways by cell debris, fibrin and inflammatory exudate may, if prolonged, lead to organisation and fibrosis, resulting in obliterative bronchiolitis and permanent lung damage.
1317
Q

The histological pattern of viral pneumonia depends on the severity of the disease. Tell me about these changes.

A
  • Predominant is an interstitial inflammatory reaction involving the walls of the alveoli.
  • The alveolar septa are widened and oedematous and usually have a mononuclear inflammatory infiltrate of lymphocytes, macrophages, and occasionally plasma cells.
  • In acute cases, neutrophils may also be present.
  • The alveoli may be free of exudate, but in many patients there is intra-alveolar proteinaceous material and a cellular exudate.
1318
Q

What is the morphology of the complications of viral pneumonia?

A
  • When complicated by ARDS, pink hyaline membranes line the alveolar walls.
  • Superimposed bacterial infection modifies this picture by causing ulcerative bronchitis, bronchiolitis, and bacterial pneumonia.
  • Some viruses, such as herpes simplex, varicella, and adenovirus, may be associated with necrosis of bronchial and alveolar epithelium and acute inflammation.
1319
Q

What is health care-associated pneumonia?

A

Health-care associated pneumonia is described as a distinct clinical entity associated with significant risk factors. These are:
* hospitalisation of at least 2 days within the recent past
* presentation from a nursing home of long-term care facility
* attending a hospital or haemodialysis clinic
* recent IV antibiotic therapy, chemotherapy or wound care.

1320
Q

Hospital-acquired pneumonias are defined as pulmonary infections acquired in the course of a hospital stay. What populations of patients are they common in?

A
  • Patients with severe underlying disease
  • Immunosuppression
  • Prolonged antibiotic therapy
  • Invasive access devices such as lines
  • Mechanical ventilation
1321
Q

What type of pneumonia is aspirin pneumonia commonly?

A

This type of pneumonia is often necrotising, pursues a fulminant clinical cause and has a common complication of lung abscess

1322
Q

What is microaspiration? What ar the consequences of it?

A
  • It occurs frequently in almost all people, especially those with GORD
  • It usually results in small, poorly formed non-necrotising granulomas with multinucleate foreign body giant cell reaction.
  • It is usually inconsequential, but may exacerbated other pre-existing lung diseases such as asthma, interstitial fibrosis, and lung rejection.
1323
Q

What is a lung abscess? What often triggers their development?

A
  • It is a local suppurative process that produces necrosis of lung tissue.
  • Oropharyngeal, surgical or dental procedures, sinobronchial infections, and bronchiectasis play important roles in their development.
1324
Q

Although under appropriate circumstances any pathogen can produce an abscess, what are the commonly isolated organisms?

A
  • The commonly isolated organisms include aerobic and anaerobic streptococci, S.aureus, and a host of gram-negative organisms.
  • Mixed infections often occur because of the causal role played by inhalation of foreign material.
  • Anaerobic organisms normally found in the oral cavity including members of the Bacteroides, Fusobacterium,and Peptococcus species, are the exclusive isolates in about 60% of causes.
1325
Q

What mechanisms introduce the causative organisms in lung abscesses?

A
  • Aspiration of infective material is common in acute alcoholism, coma, anaesthesia, sinusitis, gingivodental sepsis, and debilitation in which the cough reflexes are depressed. Aspiration first causes pneumonia, which progresses to tissue necrosis and formation of lung abscess
  • Antecedent primary lung infection - post-pneumonic abscess formations are usually associated with S.aureua, K. pneumoniae, and type 3 pneumococcus
  • Septic embolism from thombophlebitis in any portion of the systemic venous circulation or from the vegetations of infective bacterial endocarditis on the right side of the heart are trapped in the lung
  • Neoplasia - infection is particular common in the bronchopulmonary segment obstructed by a primary or secondary malignancy
  • Miscellaneous - direct traumatic penetrations of the lungs; spread of infections from a neighbouring organ, such as suppuration in the oesophagus, spine, subphrenic space, or pleural cavity
1326
Q

What is a primary cryptogenic lung abscess?

A

When all the known causes for a lung abscess are excluded, there are still cases in which no discernible basis for the abscess formation can be identified. These are referred to as primary cryptogenic lung abscesses.

1327
Q

Where do lung abscesses generally form?

A
  • Abscesses vary in diameter from a few mm to large 5-6cm cavities.
  • They may affect any part of the lung and may be single or multiple.
  • Pulmonary abscesses due to aspiration are more common on the right and are most often single
  • Abscesses that develop in the course of pneumonia or bronchiectasis are usually multiple, basal, and diffusely scattered.
  • Septic emboli and pyemic abscesses are multiple and may affect any region of the lungs
1328
Q

What are the histological changes in lung abscesses?

A
  • The cardinal histologic change in all abscesses is suppurative destruction of the lung parenchyma within the central area of cavitation
  • The abscess cavity may be filled with suppurative debris or, if there is communication with an air passage, may be partially drained to create an air-containing cavity.
  • Superimposed saphrophytic infections are prone to develop within the necrotic debris.
  • Continued infection leads to large, poorly demarcated, fetid, green-black, multilocular cavities designated gangrene of the lung.
  • In chronic cases considerable fibroblastic proliferation produces a fibrous wall
1329
Q

What are the clinical presentations of lung abscesses?

A
  • The manifestations of pulmonary abscesses are much like those of bronchiectasis and are characterised principally by cough, fever and copious amounts of foul-smelling purulent or sanguineous sputum.
  • Feverm, chest pain, and weight loss are common
  • Clubbing of the fingers and toes may appear within a few weeks after the onset of an abscess.
1330
Q

How do you diagnose lung abscesses? What do you need to rule out following a diagnosis?

A
  • The diagnosis can be only suspected from clinical findings and must be confirmed radiologically
  • Whenever an abscess is discovered in older individuals, it is important to rule out an underlying carcinoma, which is present in 10-15% of cases/
1331
Q

What is the clinical course of an abscess ? How do you treat it?

A
  • The course of abscesses is variable
  • With antimicrobial therapy, most resolve leaving behind a scar.
  • Complications include extension of the infection into the pleural cavity, haemorrhage, the development of brain abscesses or meningitis from septic emboli, and (rarely) secondary amyloidosis
1332
Q

What population does chronic pneumonia often occur in? What type of inflammation is it? What causes it?

A
  • Chronic pneumonia is most often a localised lesion in the immunocompetent patient, with or without regional lymph node involvement.
  • Typically, the inflammatory reaction is granulamatous, and is caused by bacteria (e.g., M. tuberculosis) or fungi (e.g, Histoplasma capsulated).
1333
Q

What are some fungal causes of chronic pneumonia?

A

Histoplasmosis
Blastomycosis
Coccidioidomycosis

1334
Q

What is histoplasma capsulatum? Where do you catch it from? Where is it common?

A

*Histoplasma capsulatum infection is acquired by inhalation of dust particles from soil contaminated with bird or bat droppings that contain small spores (microconidia), the infectious form of the fungus
* It is endemic along the Ohio and Mississippi rivers and the Caribbean. It is also found in Mexico, Central and South America, parts of eastern and southern Europe, Africa, Eastern Asia and Australia

1335
Q

What type of pathogen is Histoplasma capsulatum? What are the clinical presentations and morphological lesions associated with it?

A
  • It is an intracellular pathogen that is found mainly in phagocytes
  • The clinical presentations and morphological lesions of histoplasmosis also strikingly resemble those of TB including:
    1) a self-limited and often latent primary pulmonary involvement, which may result in coin lesions on CXR
    2) chronic, progressive, secondary lung disease, which is localised to the lung apices and causes cough, fever and night sweats
    3) spread to extra-pulmonary sites including mediastinum, adrenals, liver or meninges
    4) widely disseminated disease in immunocompromised patients
1336
Q

What is the pathogenesis of histoplasmosis?

A
  • The pathogenesis of histoplasmosis is incompletely understood
  • It is known that macrophages are the major target of infection
  • H.capsulatum may be internalised into macrophages after opsonisation with antibody
  • Histoplasma yeasts can multiply within the phagosome, and lyse the host cells.
  • Histoplasma infections are controlled by helper T cells that recognise fungal cell wall antigens and heat-shock proteins and subsequently secrete IFN-γ, which activated macrophages to kill intracellular yeasts.
  • In addition, Histoplasma induces macrophages to secrete TNF, which recruits and stimulates other macrophages to kill Histoplasma
1337
Q

What is the morphology of histoplasmosis in the lungs of otherwise healthy adults?

A
  • Histoplasma infections produce granulomas, which usually undergo caseation necrosis and coalesce to produce large areas of consolidation, but may also liquefy to form cavities (particularly in patients with COPD)
  • With spontaneous resolution or effective treatment, these lesions undergo fibrosis and concentric calcification (tree-bark appearance).
1338
Q

How do you histologically differentiate between histoplasmosis and TB, sarcoidosis and coccidiomycosis?

A

It requires identification of the 3- and 5-μm thin-walled yeast forms, which may persist in tissues for years

1339
Q

What is the morphology of fulminant disseminated histoplasmosis, which occurs in immunosuppressed individuals?

A

Granulomas do not form; instead, there are focal accumulations of mononuclear phagocytes with fungal yeasts throughout the body

1340
Q

How do you diagnose histoplasmosis?

A

The diagnosis of histoplasmosis is established by culture or identification of the fungus in tissue lesions.
In addition, serologic tests for antibodies and antigen are also available.
* Antigen detection in body fluids is most useful in the early stages, because antibodies are formed 2-6 weeks after infection.

1341
Q

What is blastomycosis? Where is it common?

A
  • Blastomyces dermatitidis is a soil-inhabiting dimorphic fungus.
  • It causes disease in the central and southeastern US; infection also occurs in Canada, Mexico, the Middle East, Africa, and India
1342
Q

There are three clinical forms of blastomycosis. What are they and describe them to me? How do they present clinically and radiographically?

A
  • Pulmonary blastomycosis, disseminated blastomycosis, and a rare primary cutaneous form that results from direct inoculation of organisms into the skin
  • Pulmonary blastomycosis most often presents as an abrupt illness with productive cough, headache, chest pain, weight loss, fever, abdominal pain, night sweats, chills, and anorexia
    *CXR reveal lobar consolidation, multi-lobar infiltrates, peri-hilar infiltrates, multiple nodules, or military infiltrates.
  • The upper lobes are most frequently involved.
  • The pneumonia most often resolved spontaneously, but it may persist, or progress to a chronic lesion
1343
Q

What is the morphology of blastomycosis?

A
  • In the normal host, the lung lesions of blastomycosis are suppurative granulomas.
  • Macrophages have a limited ability to ingest and kill B. dermatitidis, and the persistence of the yeast cells leads to continued recruitment of neutrophils.
  • In tissue, B.dermatitidis is a round, 5-15μm yeast cell that divides by broad-based budding.
  • It has a thick, double-contoured cell wall, and visible nuclei.
  • Involvement of the skin and larynx is associated with marked epithelial hyperplasia, which may be mistaken for squamous cell carcinoma
1344
Q

What is coccidioidomycosis? Who is infected by it?

A
  • Almost everyone who inhales the spores of Coccidioides immitis becomes infected and develops a delayed-type hypersensitivity reaction to the fungus.
  • Ended, more than 80% of people in endemic areas of southwestern and western US and in Mexico have a positive skin test reaction.
1345
Q

What is the pathogenesis of coccidioidomycosis?

A
  • One reason for eh infectivity of C.immitis is that infective arthroconidia, when ingested by alveolar macrophages, block fusion of the pgaosome and lysosome and so resist intracellular killing.
  • As is the case with Histoplasma, most primary infections with C.immitis are asymptomatic, but 10% of infected people develop lung lesions, fever, cough, and pleuritic pains, accompanied by erythema nodosum, or erythema multiforme.
1346
Q

What is the morphology of lung lesions of C.immitis?

A
  • The primary and secondary lung lesions of C.immitis are similar to the granulomatous lesions of Histoplasma
  • Within macrophages or giant cells, C. immitis is present as thick-walled, non-budding spherules 20-60μm in diameter, often filled with small endospores. a
1347
Q

What is the morphology of the complications of coccidioidomycosis?

A
  • A progeny reaction is superimposed when the spherules rupture to release the endospores.
  • Rare progressive C. immitis disease involves the lungs, meninges, skin, bones, adrenals, lymph nodes, spleen, or liver.
  • At all these sites, the inflammatory response may be purely granulomatous pyogenic, or mixed.
  • Purulent lesions dominate in patients with diminished resistance and with widespread dissemination.
1348
Q

What are some of the common organisms that cause diffuse infiltrates in immunocompromised hosts?

A
  • CMV
  • Pneumocystis jiroveci
  • Drug reaction
1349
Q

What are some of the common organisms that cause focal infiltrates in immunocompromised hosts?

A
  • Gram-negative bacterial infections
  • Staph. aureus
  • Aspergillus
  • Candida
  • Malignancy
1350
Q

What are some of the uncommon organisms that cause diffuse infiltrates in immunocompromised hosts?

A
  • Bacterial pneumonia
  • Aspergillus
  • Cryptococcus
  • Malignancy
1351
Q

What are some of the uncommon organisms that cause focal infiltrates in immunocompromised patients?

A
  • Cryptococcus
  • Mucor
  • Pneumocystis jiroveci
  • Legionella pnuemophila
1352
Q

Pulmonary disease accounts for 30-40% of hospitalisations in HIV-infected individuals. What are the most common organisms that cause pneumonia in HIV-positive patients?

A
  • Despite the emphasis on opportunistic infections, it must be remembered that bacterial LRTIs caused by the “usual” pathogens are among the most serious pulmonary disorders in HIV infection.
  • The implicated organisms include S.pneumoniae, S.aureus, H.influenzae, and gram-negative rods.
  • Bacterial pneumonias in HIV-infected persons are more common, more severe, and more often associated with bacteraemia than in those without HIV infection.
1353
Q

Not all pulmonary infiltrates in HIV-infected individuals are infectious in etiology. What are some other diseases that cause them?

A

A host of non-infectious diseases, including Capos sarcoma, non-hodgkin lymphoma, and lung cancer, occur with increased frequency and must be excluded

1354
Q

A CD4+ T cell count determines the risk of infection with specific organisms. Which organisms are more common at which CD4 levels?

A
  • Bacterial and tubercular infections are more likely at higher CD4+ counts (>200 cells/mm3)
  • Pneumocystis pneumonia usually strikes at CD4+ counts less than 200 cells/mm3
  • CMV, fungal and Mycobacterium avium complex infections are uncommon until the very late stages of immunosuppression (CD4+ counts less than 50 cells/mm3)
1355
Q

What are the most common indications are lung transplantation?

A

The most common indications are end-stage emphysema, idiopathic pulmonary fibrosis, CF, and idiopathic/familial pulmonary arterial hypertension.

1356
Q

Are bilateral lung transplants possible?

A
  • While bialteral lung and heart-lung transplants are possible, in many cases a single lung transplant is performed, offering sufficient improvement in pulmonary function for two recipient from a single donor
  • When bilateral chronic infection is present (e.g, CF, bronchiectasis), both lungs of the recipient must be replaced to remove the reservoir of infection
1357
Q

With improving surgical and organ preservation techniques, post-operative complications (e.g, anastomotic dehiscence, vascular thrombosis, primary graft dysfunction) are fortunately becoming rare. The transplanted lung is subject to two major complications. What are they and how do they happen?

A
  • Pulmonary infections are essentially those of any immunocompromised host
  • Acute rejection of the lung occurs to some degree in all patients despite routine immunosuppression.
1358
Q

What pulmonary infections happen at what point following a lung transplant?

A
  • In the early post-transplant period (the first few weeks), bacterial infections are most common
  • With ganciclovir prophylaxis and matching of donor recipient CMV status, CMV pneumonia occurs less frequently, and is less severe, although some resistant strains are emerging.
  • Most infections occur in the 3rd to 12th month after transplantation
  • Pnuemocystis jeiroveci pneumonia is rare, since almost all patients receive adequate prophylaxis, usually with Bactrim
  • Fungal infections are mostly due to Aspergillus and Candida species, and they involve the bronchial anastomotic site and/or the lung
1359
Q

When does acute rejection of the lung following a transplant occur? What are the symptoms?

A
  • It most often appears several weeks to months after surgery but also may present years later or whenever immunosuppression is decreased.
  • Patients present with fever, dyspnoea, cough, and radiologic infiltrates.
  • Since there are similar to the picture of infections, diagnosis often relies on transbronchial biopsy
1360
Q

When does chronic rejection take place after a lung transplant?

A
  • It is a significant problem in at least half of all patients by 3-5 years post-transplant.
  • It is manifested by cough, dyspnoea, and an irreversible decrease in lung function tests due to pulmonary fibrosis
1361
Q

What are the morphological features of acute rejection following lung transplant?

A

They are primarily those of inflammatory infiltrates (lymphocytes, plasma cells,a dn few neutrophils and eosinophils), either around small vessels, in the submucosa of airways or both.

1362
Q

What are the morphological changes of chronic rejection following a lung transplant?

A
  • The major morphologic correlate of chronic rejection is bronchiolitis obliterans, the partial or complete occlusion of small airways by fibrosis, with or without active inflammation.
  • Bronchiolitis obliterates is patchy and therefore difficult to diagnose via transbronchial biopsy
1363
Q

How do you treat acute rejcetion of a lung transplant?

A
  • Acute cellular airways rejection is generally response to therapy, but the treatment of established bronchiolitis obliterans has been disappointing.
  • Its progress may be slowed or even halted for some times, but it cannot be reversed.
1364
Q

What is the most frequently diagnosed cancer in the world and the most common cause of cancer mortality? Why?

A

Lung cancer, largely due to the carcinogenic effects of cigarette smoke.

1365
Q

In what age does lung cancer appear most often?

A
  • Cancer of the lung occurs most often between ages 40 and 70 years, with a peak incidence in the 50s or 60s
1366
Q

Lung cancers can be broadly classified into what groups?

A

Small cell and non-small cell types (made up of squamous and adenocarcinomas)

1367
Q

Does smoking increase the risk of lung cancer in men or women more?

A

For reasons not entirely clear, women have a higher susceptibility to carcinogens in tobacco than men.

1368
Q

Although the duration and intensity of smoking are well correlated with cancer risk, not all smokers develop cancer. Some of this may be a matter of chance, but it is also likely that the mutagenic effect of carcinogens in smoke is modified by genetic variants. What are some of these?

A
  • Many chemicals (procarcinogens) are converted into carcinogens via activation by the highly polymorphic P450 monooxygenase enzyme system
  • Specific P-450 polymorphisms have an increased capacity to activate procarcinogens in cigarette smoke, and smokers with these genetic variants appear to incur a greater risk of lung cancer.
  • Individuals whose peripheral blood lymphocytes show more numerous chromosomal breakages after exposure to tobacco-related carcinogens have a greater than 10-fold higher risk of developing lung cancer as compared with controls, presumably because of genetic variation in genes involved in DNA repair.
1369
Q

There is a linear correlation between the intensity of exposure to cigarette smoke and the appearance of ever more worrisome epithelial changes. What are these changes?

A

These begin with rather innocuous-appearing basal cell hyperplasia and squamous metaplasia and progress to squamous dysplasia and carcinoma in situ, the last stage before progression to invasive cancer.

1370
Q

Certain industrial exposure increase the risk of developing lung cancer. What are some of these?

A

Asbestos, arsenic, chromium, uranium, mickel, vinyl chloride and mustard has

1371
Q

Asbestos exposure increases the risk for lung cancer development. How soon after does it happen and how much does it increase your risk?

A
  • The latent period before the development of lung cancer is 10 to 30 years
  • Asbestos workers who do not smoke have a five-fold greater risk of developing lung cancer than do non-smoking control subjects, and those who smoke have a 55-fold greater risk
1372
Q

It is uncertain whether air pollution, by itself, increases the risk of lung cancer, but it likely adds to the risk in those who smoke or are exposed to second-hand smoke. It may do so through several different mechanisms. What is the most likely one of these?

A
  • Chronic exposure to air particulates in smog may cause lung irritation, inflammation and repair, which increases the risk of a variety of cancers
1373
Q

What is a specific form of air pollution that may contribute to an increased risk of lung cancer? How?

A

Radon gas is a ubiquitous radioactive as that has been linked epidemiologically to increased lung cancer in uranium miners, particularly those who smoke.
This has generated concern that low-level exposure may also increase the incidence of lung cancers

1374
Q

How do molecular genetics effect smoking-related lung cancers?

A
  • As with other cancers, smoking-related carcinomas of the lung arise by a stepwise accumulation of oncogenic “driver” mutations that result in the neoplastic transformation of pulmonary epithelial cells.
  • Some of the genetic changes associated with cancers can be fund in the “benign” bronchial epithelium of smokers without lung cancers, suggesting that large areas of the respiratory mucosa are mutagenised by exposure to carcinogens in tobacco smoke (“field effect”).
  • On this fertile soil, those few cells that accumulate a sufficient panoply of complementary driver mutations to acquire all of the hallmarks of cancer to develop into invasive carcinomas.
1375
Q

Lung carcinomas fall into several major histologic sub-groups, each with distinctive molecular features. What are some of these?

A
  • Squamous cell carcinoma is highly associated with exposure to tobacco smoke and harbours divers genetic aberrations, many of which are chromosome deletions involving tumour suppressor loci.
  • Small cell carcinoma shows the strong associated with smoking and despite its divergent histologic features shares many molecular features with SCC
  • Adenocarcinoma is marked by oncogenic gain-of-function mutations involving components of growth factor receptor signalling pathways.
1376
Q

Squamous cell carcinoma is highly associated with exposure to tobacco smoke and harbours diverse genetic aberrations, many of which are chromosome deletions involving tumour suppressor loci. What are some molecular changes in lung SCC and when do they happen?

A
  • These chromosome deletions, especially those involving 3p, 9p (site of the CDKN2A gene) and 17p (site of the TP53 gene) are early events in tumour evolution, being detected at an appreciable frequency in the histologically normal respiratory mucosal cells of smokers.
  • Squamous cell carcinomas show the highest frequency of TP53 mutations of all histological types of lung carcinoma
  • p53 protein over expression, a marker of TP53 mutations, is also an early event.
  • Loss of expression of the retinoblastoma tumour suppressor is identified in 15% of SCCs
  • The cyclin-dependent kinase inhibitor gene CDKN2A is inactivated and its protein product p16, is lost in 65% of tumours
  • Many SCCs have amplification of FGFR1, a gene encoding the fibroblast growth factor receptor tyrosine kinase.
1377
Q

What molecular changes occur in small cell carcinoma?

A

Despite it’s divergent histologic features, it shares many molecular features with squamous cell carcinoma
* This includes frequent loss-of-function aberrations involving *TP53
(75-90% of tumour), RB (close to 100% of tumours) and chromosome 3p deletions. Also common is amplification of genes of the MYC family.

1378
Q

Lung adenocarcinoma is marked by oncogenic gain-of-function mutations involving components of growth factor receptor signalling pathways. All are found in minority of tumours, but together they make up a substantial fraction of tumours as a whole. What are some of these gain-of-function mutations?

A
  • These include gain-of-function mutations in multiple genes encoding receptor tyrosine kinases, including EGFR, ALK, ROS, MET and RET, which are all also mutated in other forms of cancer.
  • Tumours without tyrosine kinase gene mutations often have mutations in the KRAS gene, which lies downstream of receptor tyrosine kinases in growth factor signalling pathways.
1379
Q

The WHO estimates that 25% of lung cancer worldwide occurs in never smokers. What populations do these occur in? What mutations do they have?

A
  • These cancers occur more commonly in women and most are adenocarcinomas
  • Cancers in no-smokers are more likely to have EGFR mutations, and almost never have KRAS mutations, TP53 mutations are not uncommon, but occur less frequently than in smoking-related cancers
1380
Q

What is the histologic classification of malignant epithelial lung tumours?

A

Squamous cell carcinoma - papillary, clear cell, small cell, basaxoid
Small-cell carcinoma - combined small-cell carcinoma
Adenocarcinoma - minimally invasive adenocarcinoma (non-mucinous, mucinous); lepidi, acinar - papillary, solid; mucinous adenocarcinoma
Large-cell carcinoma - large-cell neuroendocrine carcinoma
Adenosquamour carcinoma
**Carcinomas with pleomorphic sarcomatous or sarcomatous elements
Carcinoid tumour - typical, atypical
Carcinomas of salivary gland type

1381
Q

Four types of morphologic precursor epithelial lesions are recognised in lung cancer. What are they?

A

1) squamous dysplasia and carcinoma in situ
2) atypical adenomatous hyperplasia
3) adenocarcinoma in situ
4) diffuse idiopathic pulmonary neuroendocrine cell hyperplasia

1382
Q

What are the relative proportions of the major categories of lung cancer?

A
  • Adenocarcinoma (38%)
  • Squamous cell carcinoma (20%)
  • Small cell carcinoma (14%)
  • Large cell carcinoma (3%)
  • Other (25%)

There may be mixtures of histologic patterns, even in the same cancer. Thus, combinations of squamous cell carcinoma and adenocarcinoma or small-cell and squamous cell carcinoma occur in about 10% of patients.

1383
Q

The incidence of adenocarcinoma has increased significantly in the last 2 decades and is now the most common form of lung cancer in women and, in many studies, men as well. Why?

A
  • The basis for this change is unclear
  • A possible factor is the increase in women smokers, but this only highlights out lack of knowledge about why women tend to develop more adenocarcinomas
  • One possibility is that changes in cigarette type (filter tips, lower tar, and nicotine) have caused smoked to inhale more deeply and thereby expose more peripheral airways and cells (with a predilection to adenocarcinoma) to carcinogens.
1384
Q

Where in the lung is associated with what type of cancer?

A

Lung carcinomas may arise in the peripheral lung (more often associated with adenocarcinomas) or in the central/hilar region (more often squamous cell carcinomas)< sometimes in association with recognisable precursor lesions

1385
Q

What is the morphology of atypical adenomatous hyperplasia in the lung?

A
  • A small lesion (<5mm) characterised by dysplastic pneumocytes lining alveolar walls that are mildly fibrotic.
  • It can be single or multiple and can be in the lung adjacent to invasive tumour or away from it
1386
Q

What is the morphology of lung adenocarcinoma in situ?

A
  • A lesion that is less than 3cm and is composed entirely of dysplastic cells growing along pre-existing alveolar septae
  • The cells have more dysplasia than atypical adenomatous hyperplasia and may or may not have intracellular mucin (mucinous and non-mucinous, resectively)
1387
Q

What is the morphology of lung adenocarcinoma?

A
  • It is an invasive malignant epithelial tumour with glandular differentiation or mucin production by the tumour cells
  • Adenocarcinomas grow in various patterns, including acinar, lepidic, papillary, micropapillar, and solid with mucin formation.
  • Compared with squamous cell cancers, the lesions are more peripherally located and tend to be smaller.
  • They vary histologically from well-differentiated tumours with obvious glandular elements (A) to papillary lesions resembling other papillary carcinomas to solid masses with only occasional mucin-producing glands and cells
  • At the periphery of the tumour there is often a lepidic pattern of spread, in which the tumour cells “crawl” along normal-appearing alveolar septae
1388
Q

What transcription factor do adenocarcinomas often express? What is is needed for?

A

The majority express thyroid transcription factor-1; first identified in the thyroid, this factor is required for normal lung development

1389
Q

What are microinvasive lung adenocarcinomas vs mucinous lung adenocarcinomas?

A
  • Tumours (<3cm) with a small invasive component (<5mm) associated with scarring and a peripheral lepidic growth pattern are called microinvasive adenocarcinomas. They have a far better outcome than invasive carcinomas of the same size
  • Mucinous adenocarcinomas tend to spread aerogenously, forming satellite tumours. These may present as a solitary nodule or as multiple nodules, or an entire lobe may be consolidated by tumour, resembling lobar pneumonia and thus are less likely to be cured by surgery.
1390
Q

What is the morphology of early lung squamous cell carcinoma development?

A
  • Precursor lesions that give rise to invasive squamous cell carcinoma are well characterised.
  • Squamous cell carcinomas are often antedated by squamous metaplasia or dysplasia in the bronchial epithelium, which then transforms to carcinoma in situ, a phase that may last for several years.
  • By this time, atypical cells may be identified by cytologic smears of sputum or in bronchial leverage fluids or brushings, although the lesion in asymptomatic and undetectable on CXR.
  • Eventually, an invasive squamous cell carcinoma appears
1391
Q

Once an invasive squamous cell lung carcinoma appears, the tumour may then follow a variety of paths. What are some of these patterns?

A
  • It may grow exophytically into the bronchial lumen, producing an intraluminal mass. With further enlargement, the bronchus become obstructed, leading to distal atelectasis and infection
  • The tumour may also penetrate the wall of the bronchus and infiltrate along the peri-bronchial tissue in the adjacent carina or mediastinum
  • In other instances, the tumour grows along a broad front to produce a cauliflower-like intraparenchymal mass that pushes lung substance ahead of it.
1392
Q

What is the gross morphology of distinguished invasive squamous cell lung carcinoma?

A
  • As in almost all types of lung cancer, the neoplastic tissue is gray-white and firm to hard.
  • Especially when the tumours are bulky, focal areas of haemorrhage or necrosis may appear to produce red or yellow-white mottling and softening
  • Sometimes, these necrotic foci cavitate
1393
Q

What are the histological patterns of squamous cell lung carcinoma? How is it different in well-differentiated and undifferentiated tumours?

A
  • Histologically, squamous cell carcinoma is characterised by the presence of keratinisation and/or intercellular bridges.
  • Keratinisation may take the form of squamous pearls or individual cells with marked eosinophilic dense cytoplasm.
  • These features are prominent in well-differentiated tumour, are easily seen but not extensive in moderately differentiated tumours, and are focally seen in poorly differentiated tumours.
  • Mitotic activity is higher in poorly differentiated tumours.
1394
Q

Where in the lung is squamous cell carcinoma most common? What may be seen next for the tumour mass?

A
  • In the past, most squamous cell carcinomas were seen to arise centrally from the segmental or sub-segmental bronchi
  • However, the incidence of squamous cell carcinoma of the peripheral lung is increasing
  • Squamous metaplasia, epithelial dysplasia, and foci of frank carcinoma in situ may be seen in bronchial epithelium adjacent to the tumour mass.
1395
Q

Where is small cell carcinoma most often found in the lung? Is it aggressive?

A
  • Small cell carcinoma is a highly malignant tumour with a strong relationship to cigarette smoking; only about 1% occurs in non-smokers
  • They may arise in major bronchi or in the periphery of the lung
  • There is no known pre-invasive phase.
  • They are the most aggressive of lung tumours, metastasising widely and virtually always proving to be fatal.
1396
Q

What is the cell size and shape of small cell carcinoma?

A
  • Small cell carcinoma is comprised of relatively small cells with scant cytoplasm, ill-defined cell borders, finely granular nuclear chromatin (salt and pepper pattern), and absent or inconspicuous nucleoli.
  • The cells are round, oval or spindle-shaped, and nuclear molding is prominent.
  • There is no absolute size for the tumour cells, but in general they are smaller than three times the diameter of a small resting lymphocytes (a size of about 25 microns).
1397
Q

What are the cell characteristics of small cell carcinoma?

A
  • The mitotic count is high
  • The cells grow in clusters that exhibit neither glandular nor squamous organisation
  • Basophilic staining of vascular walls due to encrustation by CNA from necrotic tumour cells (Azzopardi effect) is frequently present.
  • All small cell carcinomas are high grade
1398
Q

Electron microscopy shows dense-core neurosecretory granules, about 100nm in diameter, in 2/3rds of cases of small cell lung cancer. What are these and what do they do?

A
  • The occurrence of neurosecretory granules, expression of neuroendocrine markers such as chromogranin, synaptophysin, and CD57, and the ability of some of these tumours to secrete hormones (e.g, parathormone-related protein, a cause of paraneoplastic hypercalcaemia) suggests that this tumour originates from neuroendocrine progenitor cels, which are present in the bronchial epithelium
  • Among the various types of lung cancer, small cell is the one that is most commonly associated with ectopic hormone production
  • Immunohistochemistry demonstrates high levels of the anti-apoptotic protein BCL2 in 90% of tumours.
1399
Q

What is the morphology of large cell lung carcinoma?

A
  • It is an undifferentiated malignant epithelial tumour that lacks the cytologic features of other forms of lung cancer.
  • The cells typically have large nuclei, prominent nucleoli, and a moderate amount of cytoplasm
  • On histologic varient is large cell neuroendocrine carcinoma, which has molecular features similar to those that characterise small cell carcinoma, but is comprised of tumour cells of larger size
1400
Q

What type of diagnosis is large cell carcinoma?

A

It is a diagnosis of exclusion since it expresses none of the markers associated with adenocarcinoma (TTF-1, napsin A) and squamous cell carcinomas (p63, p40).

1401
Q

Where does lung cancer spread locally?

A
  • Any type of lung carcinoma may extend on to the pleural surface and then within the pleural cavity or into the pericardium
  • Metastases to the bronchial, tracheal, and mediastinal nodes can be found in most cases.
  • The frequency of nodal involvement varies slightly with histologic pattern but averages greater than 50%
1402
Q

How do lung carcinomas spread distally? Where do they go?

A
  • Distant spread on lung carcinoma occur through both lymphatic and haematogenous pathways
  • These tumours often spread early throughout the body except for squamous cell carcinoma, which metastasises outside the thorax late.
  • No organ or tissue is spared in the spread of these lesions, but the adrenals, for obscure reasons, are involved in more than half the cases. The liver (30-50%), brain (20%) and bone (29%) are additional favoured sites of metastases
1403
Q

Lung carcinomas have local effects that may cause several pathologic changes in the lung distal to the point of bronchial involvement. What are some of these changes?

A
  • Partial obstruction may cause focal emphysema
  • Total obstruction may lead to atelectasis
  • The impaired damage of the airways is a common cause for severe suppurative or ulcerative bronchitis or bronchiectasis
  • Pulmonary abscesses sometimes call attention to na otherwise silent carcinoma
  • Compression of invasion of the SVC can cause venous congestion and oedema of the head and arm, and, ultimately, circulatory compromise - the superior vena cava syndrome
  • Extension to the pericardial or pleural sacs may cause pericarditis or pleuritis with significant effusions.
1404
Q

What are the T stages of TNM staging of the lung?

A

Tis - carcinoma in situ
T1 - tumour <3cm without pleural or mainstay bronchus involvement (T1a, <2cm; T1b, 2-3cm)
T2 - tumour 3-7cm or involvement of mainstream bronchus 2cm from carina, visceral pleural involvement or lobar atelectasis (T2a, 3-5cm; T2b, 5-7cm)
T3 - tumour >7cm or one with involvement of parietal pleura, chest wall, diaphragm, phrenic nerve, mediastinal pleura, parietal pericardium, mainstream bronchus <2cm from carina but without involvement of carina, or entire lung atelectasis or separate tumour nodules in the same lobe
T4 - Amy tumour with invasion of mediastinum, heart, great vessels, trachea, recurrent laryngeal nerve, oesophagus, vertebral body, or carina, or separate tumour nodules in a different ipsilateral lobe

1405
Q

What are the N stages in TNM staging of the lung?

A

N0 - no metastasis to regional lymph nodes
N1 - ipsilateral hilar or peribronchial nodal involvement
N2 - metastasis to ipsilateral mediastinal or subcarinal lymph nodes
N3 - metastasis to contralateral mediastinal or hilar lymph nodes, ipsilateral or contralateral scalene, or supraclavicular lymph nodes

1406
Q

What is the stage grouping for lung cancers?

A

Stage IA - T1N0M0
Stage IB - T2aN0M0
Stage IIA - T2bN0M0 or T1/2aN1M0
Stage IIB - T2bN0M0 or T3N0M0
Stage IIIA - T1/2N2M0 or T3N1/2M0 or T4N0/1M0
Stage IIIB - AnyTN3M0, T4N2/3M0
Stage IV - any T, any N, M1

1407
Q

When and how do lung cancers present?

A
  • In the usual case it is discovered in patients in their 50s or older whose symptoms are of several months’ duration
  • The major presenting complaints are cough (75%), weight loss (40%), chest pain (40%) and dyspnoea (20%)
  • Not infrequently the tumour is discovered by its secondary spread during the course of investigation of an apparent primary or metastatic neoplasm elsewhere
1408
Q

What are some clinic features of lung tumour spread and their causes?

A

Cough - involvement on central airways
Haemoptysis - haemorrhage from tumour in airway
Chest pain - extension of tumour into mediastinum, pleura or chest wall
Pneumonia, abscess, lobar collapse - airway obstruction by tumour
Lipoid pneumonia - tumour obstruction; accumulation of cellular lipid in foamy macrophages
Pleural effusion - tumour spread into pleura
Hoarseness - recurrent laryngeal nerve invasion
Dysphagia eosophageal invasion
Diaphragm paralysis - phrenic nerve invasion
Rib destruction - chest wall invasion
SVC syndrome - SVC compression by tumour
Horner syndrome - sympathetic ganglian invasion
Pericarditis, tamponade - pericardial involvement

1409
Q

What is the prognosis of lung cancer?

A
  • Even with many incremental improvements in thoracic surgery, radiation therapy, and chemotherapy, the overall 5-year survival rate is only 16%.
  • The 5-year survival rate is 52% for cases detected when the disease is still localised, 22% when there is regional metastasis and only 4% with distant metastases
1410
Q

What lung cancer mutations point toward a better prognosis and which point toward a worse one?

A
  • Targeted treatment of patients with adenocarcinoma and activating mutations in EGFR or in other tyrosine kinases with specific inhibitors of the mutates kinases prolongs survival
  • Many tumours that recur carry new mutations that generate resistance to these inhibitors, proving that these drugs are “hitting” their target
  • In contrast, activating KRAS mutations appear to be associated with a worse prognosis, regardless of treatment, in an already grim disease
1411
Q

Lung carcinoma can be associated with several paraneoplastic syndromes, some of which may antedate the development of a detectable pulmonary lesions. What are the hormones or hormone-like factors elaborated?

A
  • Antidiuretic hormone, inducing hyponatraemia due to inappropriate ADH secretion
  • Adrenocorticotropic hormone, producing Cushing syndrome
  • Parathormone, parathyroid hormone-related peptide, prostaglandin E, and some cytokines all implicated in the hypercalcaemia often seen with lung cancer
  • Calcitonin causing hypocalcaemia
    Gonadotropins causing gynaecomastia
  • Serotonin and bradykinin, associated with the carcinoid syndrome
1412
Q

Any histologic type of lung tumour may occasionally produce any of the hormones but which tumours often produce which hormones?

A
  • Tumours that produce ACTH and ADH are predominantly small cell carcinomas
  • Those that produce hypercalcaemia are mostly squamous cell carcinomas
1413
Q

What are some systemic manifestations of lung carcinoma?

A
  • Lambert-Eaton myasthenic syndrome, in which muscle weakness is caused by auto-antibodies (possibly elicited by tumour ionic channels) directed to the neuronal calcium channel
  • Peripheral neuropathy, usually purely sensory
  • Dermatologic abnormalities, including acanthosis nigricans
  • Haematologic abnormalities, such as **Leukemoid reactions
  • Hypercoagulable states, such as Trousseau syndrome (DVT and thromboembolism)
  • A peculiar abnormality of connective tissue called hypertrophic pulmonary osteoarthropathy, associated with clubbing of the fingers
1414
Q

What are some of the complications of apical lung cancers?

A

Apical lung cancers in the superior pulmonary sulcus tend to invade the neural structures around the trachea, including the cervical sympathetic plexus, and produce a group of clinical findings that includes severe pain in the distribution of the ulnar nerve and Horner syndrome (enophtalmos, ptosis, mitosis, and anhidrosis) on the same side as the lesion.

1415
Q

The normal lung contains neuroendocrine cells within the epithelium as single cells or as clusters, the neuroepithelial bodies. What is neuroendocrine cell hyperplasia? What causes it?

A

While virtually all pulmonary neuroendocrine cell hyperplasias are secondary to airway fibrosis and/or inflammation, a rare disorder called diffuse idiopathic pulmonary neuroendocrine cell hyperplasia seems to be a precursor to the development of multiple tumorlets and typical or atypical carcinoids.

1416
Q

What are some neoplasms of neuroendocrine cells in the lung?

A

They include benign tumourlets, small, inconsequential, hyperplasticity nests of neuroendocrine cells seen in areas of scarring or chronic inflammation; carcinoids and the highly aggressive small cell carcinoma nd large cell neuroendocrine carcinoma of the lung.

1417
Q

Carcinoid tumours are classified separately, since they differ significantly from carcinomas with evidence of neuroendocrine differentiation in terms of incidence and clinical, epidemiological, histologic, and molecular characteristics. What are the characteristics of carcinoid tumours?

A
  • They represent 1-5% of all lung tumours.
  • Most patients with thee tumours are younger than 40 years of age, and the incidence is equal for both sexes.
  • Approx 20-40% of patients are non-smokers
1418
Q

Where in the lungs do carcinoid tumours grow? What is their gross morphology?

A
  • Carcinoids may arise centrally or may be peripheral.
  • On gross examination, the central tumours grow as fingerlike or spherical polypoid masses that commonly project into the lumen of the bronchus and are usually covered by an intact mucosa
  • They rarely exceed 3-4cm in diameter
  • Most are confined to the mainstream bronchi. Others, however, penetrate the bronchial wall to fan out in the peribronchial tissue, producing the so-called collar-button lesion
1419
Q

What is the histologic changes of carcinoid tumours?

A
  • Histologically, the tumour is composed of organdie, trabecular, palisading, ribbon, or rosette-like arrangements of cells separated by a delicate fibrovascular stroma.
  • In common with the lesions of the GI tract, the individual cells are quite regular and have uniform round nuclei and a moderate amount of eosinophilic cytoplasm.
1420
Q

How do typical and atypical carcinoids differ histologically?

A
  • Typical carcinoids have fewer than two mitoses per 10 high-power fields and lack necrosis, while atypical carcinoids have between two and 10 mitoses per 10 high-power fields and/or foci of necrosis.
  • Atypical carcinoids also show increased pleomorphism, have more prominent nucleoli, and are more likely to grow in a disorganised fashion and evade lymphatics.
  • On electron microscopy the cells exhibit the dense-core granules characteristic of other neuroendocrine tumours and, by immunohistochemistry, are found to contain serotonin, Neuro-specific enolase, bombesin, calcitonin, or other peptides
1421
Q

What are the clinical symptoms of carcinoid symptoms?

A
  • The clinical manifestations of bronchial carcinoids emanate from their intraluminal growth, their capacity to metastasise, and the ability of some of the lesions to elaborate vasoactive amines.
  • Persistent cough, haemoptysis, impairment of drainage of respiratory passaged with secondary infections, bronchiectasis, emphysema, and atelectasis are all by-products of the intraluminal growth of these lesions.
1422
Q

What is carcinoid syndrome? How many carcinoid tumours cause it?

A
  • It is characterised by intermittent attacks of diarrhoea, flushing, and cyanosis.
  • Approximately, 10% of bronchial carcinoids give rise to this syndrome.
  • Overall, most bronchial carcinoids do not have secretory activity and do not metastasise to distant sites but follow a relatively benign course for long periods and are therefore amenable to resection.
1423
Q

What is the prognosis of carcinoid tumours?

A

The reported 5-year survival rates are 95% for typical carcinoids, 70% for atypical carcinoids, 30% for large cell neuroendocrine carcinoma, and 5% for small cell carcinoma

1424
Q
A
1425
Q
A
1426
Q

What is the histology of a lung hamartoma?

A

The epithelial clefts are lined by ciliated columnar epithelium and non-ciliated epithelium and probably represent entrapment of respiratory epithelium.

1427
Q

What is lymphangioleiomyomatosis?

A
  • A pulmonary disorder that primarily affects young women of childbearing age.
  • It is characterised by a proliferation of perivascular epithelioid cells that express markers of both melanocytes and smooth muscle cells.
  • The proliferation distorts the involved lung, leading to cystic, emphysema-like dilation of terminal air-spaces, thickening of the interstitium, and obstruction of lymphatic vessels
1428
Q

What are the mutations and other hormonal factors that are associated with lymphangioleiomyomatosis?

A
  • The lesion epithelioid cells appear to frequently harbour loss of function mutations in the tumour suppressor TSC2, one of the loci linked to tuberous sclerosis.
  • The protein encoded by TSC2, tuberin, is a negative regulator of mTOR, a key regulator of cellular metabolism. while TSC2 mutations points to increased mTOR activity as a contributing factor, the disorder remains poorly understood.
  • The strong tendency to affect young women suggests the oestrogen contributes to the proliferation of perivascular epithelioid cells, which often express oestrogen receptors.
1429
Q

What is the clinical course of lymphangioleiomyomatosis?

A
  • Patients mostly commonly present with dyspnoea or spontaneous pneumothorax, the latter related to the emphysematous changes
  • The disease tends to be slowly progressive over a period of several decades.
1430
Q

What are some common mediastinal masses and tumours? Where in the mediastinum are they commonly?

A
  • Anterior mediastinum - thymoma, teratoma, lymphoma, thyroid lesions, parathyroid tumours, metastatic carcinoma
  • Posterior mediastinum - neurogenic tumours (schwannoma, neurofibroma), lymphoma, metastatic tumur (mostly from the lung), bronchogenic cyst, gastroenteritis hernia
  • Middle mediastinum - bronchogenic cyst, pericardial cyst, lymphoma
1431
Q

Which organ is the most common site of metastatic neoplasms? How do tumours spread there?

A
  • The lung is the most common site of metastatic neoplasms
  • Both carcinomas and sarcomas arising anywhere in the body may spread to the lungs via the blood or lymphatics or by direct continuity
  • Growth of contiguous tumours into the lungs occurs most often with oesophageal carcinomas and mediastinal lymphomas
1432
Q

The pattern of metastatic growth morphologically within the lungs is quite variable. What are some of the options?

A
  • In the usual case, multiple discrete nodules (cannonball lesions) are scattered throughout all lobes, more being at the periphery
  • Other patterns include solitary nodule, endobronchial, pleural, pneumonic consolidations nd combinations of these.
  • Foci of lepidic growth similar to adenocarcinoma in situ are seen occasionally with metastatic carcinomas and may be associated with any of the listed patterns
1433
Q

Pathological involvement of the pleura is, most often, a secondary complication of some underlying disease. What are some important primary disorders of the pleura?

A

1) Primary intrapleural bacterial infections that imply seeding of this space as an isolated focus in the course of a transient bacteraemia
2) A primary neoplasm of the pleural : mesothelioma

1434
Q

Pleural effusion is a common manifestation of both primary and secondary pleural diseases. What setting cause accumulation of pleural fluid?

A
  • Increased hydrostatic pressure, as in congestive heart failure
  • Increased vascular permeability, as in pneumonia
  • Decreased osmotic pressure, as in nephrotic syndrome
  • Increased intrapleural negative pressure, as in atelectasis
  • Decreased lymphatic drainage, as in mediastinal carcinomatosis
1435
Q

Serous, serofibrinous and fibrinous pleuritis all have an inflammatory basis, differing only in the intensity and duration of the process. What are some primary causes of all of the sub-categories above?

A
  • The most common causes of pleuritis are disorders associated with inflammation of the underlying lung, such as TB, pneumonia, lung infarcts, lung abscess and bronchiectasis.
  • RA, SLE, uraemia, diffuse systemic infections, other systemic disorders, and metastatic involvement of the pleura can also cause serous or serofibrinous pleuritis.
  • Radiation used in therapy for tumours in the lung or mediastinum often causes a serofibrinous pleuritis
1436
Q

What is the outcome of many inflammatory pleuritis cases?

A
  • In most of these disorders, the serofibrinous reaction is only minimal, and the fluid exudate is resorbed with either resolution or organisation of the fibrinous component.
  • However, large amounts of fluid sometimes accumulate and compress the lung, causing respiratory distress
1437
Q

What usually causes a purulent pleural exudate (empyema)?

A
  • It usually results from bacterial or mycotic seeding of the pleural space
  • Most commonly, this seeding occurs by contiguous spread of organisms from intrapulmonary infection, but occasionally it occurs through lymphatic or haematogenous dissemination from a more distant source
  • Rarely, infections below the diaphragm, such as a liver abscess, may extend by continuity through the diaphragm into the pleural spaces, more often on the right side
1438
Q

What is the morphology of empyema? What volume are they normally?

A
  • Empyema is characterised by located, yellow-green, creamy pus composed of masses of neutrophils admixed with other leukocytes.
  • Although empyema may accumulate in large volumes (up to 500ml-1L), usually the volume is small, and the pus becomes localised.
1439
Q

What are the outcomes of empyema?

A

Empyema may resolve, but more often the exudate organises into dense, tough fibrous adhesions that frequently obliterate the pleural space or envelop the lungs; either can seriously restrict pulmonary expansion.

1440
Q

What is haemorrhagic pleuritis? What should you differentiate it from and what should you look for?

A
  • Haemorrhagic pleuritis manifested by sanguineous inflammatory exudates is infrequent and is found in haemorrhagic diatheses, rickettsial diseases, and neoplastic involvement of the pleural cavity.
  • The sanguineous exudate must be differentiated from haemothorax.
  • When haemorrhagic pleuritis is encountered, careful search should be made for the presence of exfoliated tumour cells.
1441
Q

What is a hydrothorax? Where is it? What is the most common cause?

A
  • Non-inflammatory collections of serous fluid within the pleural cavities are called hydrothorax.
  • The fluid is clear and straw coloured.
  • Hydrothorax may be unilateral or bilateral, depending on the underlying cause
  • The most common cause of hydrothorax is cardiac failure, and for this reason it is usually accompanied by pulmonary congestion and oedema.
1442
Q

What is a chylothorax? What causes it?

A
  • An accumulation of milky fluid, usually of lymphatic origin, in the pleural cavity.
  • Chyle is milky white because it contains finely emulsified fats
  • Chylothorax is most often caused by thoracic duct trauma or obstruction that secondarily causes rupture of major lymphatic ducts. This disorder is typically caused by malignancies that obstruct the major lymphatic ducts
1443
Q

Where are the malignancies that most commonly cause a chylothorax?

A

Usually such cancers arise within the thoracic cavity and invade the lymphatics locally, but occasionally more distant cancers metastasise via the lymphatics and grow within the right lymphatic or thoracic duct, producing obstruction

1444
Q

Pneumothorax refers to air or gas in the pleural cavities. What is it commonly associated with? What are the different types?

A
  • It is most commonly associated with emphysema, asthma and TB
  • It may be spontaneous, traumatic or therapeutic
1445
Q

What may cause a spontaneous pneumothorax?

A
  • It may complicate any form of pulmonary disease that causes rupture of an alveolus
  • An abscess cavity that communicates either directly with the pleural space or with the lung interstitial tissue may also lead to the space of air
  • In the latter circumstance, the air may dissect through the lung substance or back through the mediastinum (interstitial emphysema), eventually entering the pleural cavity.
1446
Q

What type of trauma often causes a traumatic pneumothorax?

A
  • Traumatic pneumothorax is usually caused by some perforating injury to the chest wall, but sometimes the trauma pierces the lung and thus provides two avenues for the accumulation of air within the pleural spaces.
1447
Q

What causes a spontaneous idiopathic pneumothorax? How does it resolve? Is it likely to come back?

A
  • This entity is encountered in relatively young people, seems to be due to rupture of small, peripheral, usually apical sub pleural blebs, and usually subsides spontaneously as the air is resorbed
  • Recurrent attacks are common and can be quite disabling
1448
Q

What is a tension pneumothorax?

A

When the defect acts as a flap valve and permits the entrance of air during inspiration but fails to permit its escape during expiration, it effectively acts as a pump that created the progressively increasing pressures of tension pnuemothorax, which may be sufficient to compress vital medicinal structures and contralateral lung

1449
Q

The pleura may be involved by primary or secondary tumours. Which is most common?

A

Secondary metastatic involvement is far more common than are primary tumours

1450
Q

Where are the most frequent metastatic malignancies of the pleura from?

A
  • The most frequent metastatic malignancies arise from primary neoplasms of the lung and breast
  • In addition to these cancers, malignancy from any organ of the body may spread to the pleural spaces
  • Ovarian carcinomas tend to cause widespread implants in both the abdominal and thoracic cavities
1451
Q

What pleural complication usually follows a metastasis?

A

In most metastatic involvements, a serous or sero-sanguineous effusion follows that often contains neoplastic cells.
For this reason, careful cytologic examination of the sediment is of considerable diagnostic value.

1452
Q

What is a solitary fibrous tumour? How big is it?

A
  • Solitary fibrous tumour is a soft-tissue tumour with a propensity to occur in the pleura and, less commonly, in the lung, as well as other sites
  • The tumour is often attached to the pleural surface by a pedicle.
  • It may be small (1-2cm) or may reach an enormous size, but it tends to remain confined to the surface of the lung
1453
Q

What is the morphology of solitary fibrous tumours?

A
  • Grossly, solitary fibrous tumour consists of dense fibrous tissue with occasional cysts with viscid fluid
  • Microscopically, the tumour shows whorls of reticulin and collagen fibers among which are interspersed spindle cells resembling fibroblasts
1454
Q

Are solitary fibrous tumours malignant? If so, what indicates this?

A
  • Rarely, this tumour may be malignant, with pleomorphism, mitotic activity, necrosis and large size (>10cm)
  • The tumour cells are CD34+ and keratin-negative by immunostaining
1455
Q

Malignant mesotheliomas, although rare, have assumed great importance in the past few decades because of their increased incidence among people with heavy exposure to asbestos. Which part of the pleura are they in? Where in the world do they happen? What are they associated with?

A
  • Thoracic mesothelioma arises from either the visceral or the parietal pleura
  • In coastal areas with shipping industreis in the US and UK, and Canadian, Australian, and South African mining areas, as many as 90% of mesotheliomas are asbestos related.
1456
Q

What is the increase in chance of mesothelioma in asbestos workers and smokers? How is this different to lung cancer?

A
  • The lifetime risk of developing mesothelioma in heavily exposed individuals is as high as 7-10%.
  • There is a long latent period of 25-45 years for the development of asbestos-related mesothelioma
  • There seems to be no increased risk of mesothelioma in asbestos workers who smoke.
  • This is in contrast to the risk of asbestos-related lung carcinoma, already high, which is markedly magnified by smoking
  • Thus, for asbestos workers (particularly smokers), the risk of dying of lung carcinoma far exceeds that of developing mesothelioma
1457
Q

What do you find morphologically in patients with asbestos exposure?

A
  • Asbestos bodies are found in increased numbers in the lungs of patients with mesothelioma
  • Another marker of asbestos exposure is the asbestos plaque
1458
Q

Although several cytogenic abnormalities have been associated with mesothelioma, what is the most common?

A
  • A homozygous deletion of the tumour suppressor gene CDKN2A/INK4a, which occurs in about 80% of mesotheliomas
1459
Q

What is the gross morphology of mesothelioma?

A
  • Malignant mesothelioma is a diffuse lesion arising either from the visceral or parietal pleura, that spreads widely in the pleural space and is usually associated with extensive pleural effusion and direct invasion of thoracic structures.
  • The affected lung becomes ensheathed by a thick layer of soft, gelatinous, greyish, pink tumour tissue
1460
Q

What is the microscopic morphology in mesothelioma?

A
  • Microscopically, malignant mesotheliomas may be epithelioid (60%), sarcomatous (20%), ox mixed (20%). This is in keeping with the fact that mesothelial cells have the potential to develop as epithelium-like cells or mesenchymal striatal cells
1461
Q

What is the morphology of the epithelioid type of mesothelioma?

A
  • The epithelioid type of mesothelioma consists of cuboidal, columnar, or flattened cells forming tubular or papillary structures resembling adenocarcinoma.
  • Immunohistochemical stains are very helpful in differentiating it from pulmonary adenocarcinoma
1462
Q

What proteins do mesotheliomas show strong positivity for?

A
  • Most mesotheliomas show strong positivity for keratin proteins, calretinin, Wilms tumour 1, cytokeratin 5/6 and D2-40.
  • This panel of antibodies is diagnostic in a majority of causes when interpreted in the context of morphology and clinical presentation.
1463
Q

What is the morphology of the sarcomatoid type of mesothelioma?

A
  • The mesenchymal type of mesothelioma appears as a spindle cell sarcoma, resembling fibrosarcoma
  • Sarcomatoid mesotheliomas tend to have lower expression of many of the markers described previously and some may be positive only for keratin
1464
Q

What are the common clinical presentations of mesothelioma?

A
  • The presenting complaints are chest pain, dyspnea, and, as noted, recurrent pleural effusions.
  • Concurrent pulmonary asbestosis (fibrosis) is present in only 20% of individuals with pleural mesothelioma.
1465
Q

Where does mesothelioma metastasise to? What is its prognosis? How do we treat it?

A
  • The lung is invaded directly, and there is often metastatic spread to the hilar lymph nodes and, eventually to the liver and other distant organs.
  • Fifty percent of patients die within 12 months of diagnosis, and few survive longer than 2 years
  • Aggressive therapy (extra pleural pneumonectomy, chemotherapy, radiation therapy) seems to improve this poor prognosis in some patients.
1466
Q

Apart from the pleura, where else are mesotheliomas?

A
  • Mesotheliomas also arise in the peritoneum, pericardium, tunica vaginalis, and genital tract (benign adenomatoid tumour).
  • Peritoneal mesotheliomas are related to heavy asbestos exposure in 60% of male patients (the number is much lower in females).
  • Although in about half cases the disease remains confined to the abdominal cavity, intestinal involvement frequently leads to death from intestinal obstruction or inanition.
1467
Q

Glomeruli may be injured in a variety of factors and in the course of several systemic diseases. What are some of them?

A
  • Systemic immunological diseases such as SLE, vascular disorders such as HTN, metabolic diseases such as diabetes mellitus, and some hereditary conditions such as Fabry disease often affect the glomerulus. These are termed secondary glomerular diseases
  • Disorders in which the kidney is the only or predominant organ involved constitute the various types of primary glomerulonephritis or, because some do not have a cellular inflammatory component, glomerulopathy
1468
Q

What are some primary glomerulopathies?

A

Acute proliferative glomerulonephritis
Rapidly progressive (crescentic) glomerulonephritis
Membranous nephropathy
Minimal-change disease
Focal segmental glomerulosclerosis
Membranoproliferative glomerulonephritis
Dense deposit disease
IgA nephropathy
Chronic glomerulonephritis

1469
Q

What are some systemic diseases with glomerular involvement?

A

SLE
Diabetes mellitus
Amyloidosis
Goodpasture syndrome
Microscopic polyarteritis/polyangiitis
Wegener granulomatosis
Henoch-Schonlein purpura
Bacterial endocarditis

1470
Q

What are some hereditary glomerular diseases?

A

Alport syndrome
Thin basement membrane disease
Fabry disease

1471
Q

What are the manifestations of the glomerular syndromes?

A

Nephritic syndrome - haematuria, azotemia, variable proteinuria, oliguria, oedema, and HTN
Rapidly progressive glomerulonephritis - acute nephritis, proteinuria and AKI
Nephrotic syndrome - >3.5gm.day proteinuria, hypoalbuminaemia, hyperlipidaemia, lipiduria
*Chronic renal failure** - azotemia -> uraemia progressing for months to years
Isolated urinary abnormalities - glomerular haematuria and/or subnephrotic proteinuria

1472
Q

What does the glomerulus consist of?

A
  • An anastomosing network of capillaries lined by fenestrated endothelium invested by two layers of endothelial cells
  • The visceral epithelial cells (podocytes) are incorporated into and become an intrinsic part of the capillary wall, separated from endothelial cells but a basement membrane
  • The parietal epithelium, situated on the Bowman’s capsule, lines the urinary space, the cavity in which the plasma filtrate first collects
1473
Q

The glomerular capillary wall is the filtering membrane in the glomerulus. What are the structures that it consists of?

A
  • There is a thin layer of fenestrated **endothelial cells*, which each fenestra being about 70-100nm in diameter
  • A glomerular basement membrane with a thick electron-dense central layer, the lamina densa and thinner electron-lucent peripheral layers, the lamina rara interna and lamina rara externa
  • The visceral epithelial cells (podocytes)
  • The entire glomerular tuft is supported by mesangial cells lying between the capillaries. Basement membrane-like mesangial matrix forms a meshwork in which the mesangial cells are embedded
1474
Q

What is the glomerular basement membrane made of?

A
  • The GBM consists of collagen (mostly type IV), laminin, polyanionic proteoglycans (mostly heparan sulfate) fibronectin, entactin, and several other glycoproteins
1475
Q

Type IV collagen forms a network suprastructure in the glomerular basement membrane. What is the structure of this?

A
  • The building block (monomer) of this network is a triple-helical molecule composed of one or more of six types of α chains
  • Each molecule consists of a 7S domain at the N terminus, a triple-helical domain in the middle, and a globular non-collagenous domain (NC1) at the C terminus
  • The NC1 domain is important for helix formation and for assembly of collagen monomers into the basement membrane suprastructure
  • Glycoproteins (laminin, entactin) and proteoglycans (heparan sulfate, perlecan) attach to the collagenous suprastructure
1476
Q

Why are the biochemical properties of the structural components of the glomerular important to know? What are they?

A
  • They are critical to understanding glomerular diseases
  • Angtigens in the NC1 domain are the targets of antibodies in anti-GBM nephritis
  • Genetic defects in the α-chains underlie some forms of hereditary nephritis
  • The proteoglycan content of the GBM may contribute to its permeability characteristics
1477
Q

What is the structure of the visceral epithelial cells of the glomerular capillary wall?

A
  • The visceral epithelial cells (podocytes) possess interdigitating processes embedded in and adherent to the lamina rara externa of the basement membrane
  • Adjacent **foot processes* are separated by 20- to 30-nm-wide filtration slits, which are bridged by a thin diaphragm
1478
Q

What are the characteristics of mesangial cells?

A
  • These cells, of mesenchymal origin, are contractile, phagocytic, and capable of proliferation, of laying down both matrix and collagen, and of secreting several biologically active mediators
  • Biologically, they are most akin to vascular smooth muscle cells and pericytes. They are important in many forms of glomerulonephritis
1479
Q

What is the normal glomerulus permeable to and not permeable to?

A

The normal glomerulus is highly permeable to water and small solutes, because of the fenestrated nature of the endothelium, and impermeable to proteins of the size of albumin or larger

1480
Q

What molecular characteristics make a molecule more and less likely to be permeable through the glomerulus? Why is this important?

A
  • The permeability characteritsics of the glomerular filtration barrier allow discrimination among various protein molecules, depending on their size (the larger, the less permeable) and charge (the more cationic, the more permeable)
  • This size- and charge-dependent barrier function is accounted for by the structure of the capillary wall.
  • The charge-dependent restriction is important in the virtually complete exclusion of albumin from the filtrate, because albumin is an anionic molecule
1481
Q

Why is the visceral epithelial cell importatn for the maintenance of glomerular barrier function?

A

Its slit diaphragm presents a size-selective distal diffusion barrier to the filtration of proteins, and it is the cell type that is largely responsible for the synthesis of GBM components

1482
Q

What are the proteins located in the slit diaphragm or present in assemblies of molecules within visceral epithelial cells in the glomerulus that are attached to the slit diaphragm?

A
  • Nephrin is a transmembrane protein with a large extracellular portion made up of Ig-like domains.
  • Nephrin molecules extend toward each other from neighbouring foot processes and dimerise across the slit diaphragm
  • Within the cytoplasm of the foot processes, nephron forms molecular connections with podocin, CD2- associated protein, and ultimately the actin cytoskeleton of the visceral epithelial cells
1483
Q

Various types of glomerulonephropathies are characterised by one or more of four basic tissue reactions. What are these?

A
  • Hypercellularity - some inflammatory diseases of the glomerulus are characterised by an increase in the number of cells in the glomerular tufts
  • Basement membrane thickening - by light microscopy, this change appears at thickening of the capillary walls, best seen in sections stained with periodic acid-Schiff (PAS)
  • Hyalinosis denotes the accumulation of material that is homogenous and eosinophilic by light microscopy.
  • Sclerosis is characterised by deposition of extracellular collagenous matrix
1484
Q

Hypercellularity - some inflammatory diseases of the glomerulus are characterised by an increase in the number of cells in the glomerular tufts. Why does this hypercellularity occur?

A
  • Proliferation of mesangial and endothelial cells
  • Infiltration of leukocytes, including neutrophils, monocytes, and in some diseases, lymphocytes. The combination of infiltration of leukocytes and swelling and proliferation of mesangial and/or endothelial cells is often referred to as endocapillary proliferation
  • Formation of crescents - these are accumulations of cells composed of proliferating glomerular epithelial cells (predominantly parietal but including some visceral cells) and infiltrating leukocytes.
1485
Q

How do crescents form as part of the hypercellularity in glomerular injury?

A
  • The epithelial cell proliferation that characterises crescent formation occurs following an immune/inflammatory injury involving the capillary walls
  • Plasma proteins leak into the urinary space, where it is believed that exposure to procoagulants such as tissue factor leads to fibrin deposition.
  • Activation of coagulation factors such as thrombin is suspected of being a trigger for crescent formation, but the actual mechanism is still unknown
  • Molecules that have been implicated in recruitment of leukocytes into crescents include multiple pro-inflammatory cytokines.
1486
Q

By electron microscopy, basement membrane thickening in glomerular disease takes one of three forms. What are these?

A
  • Deposition of amorphous electron-dense material, most often immune complexes, on the endothelial or epithelial side of the basement membrane or within the GBM itself. Fibrin, cryoglobulins and abnormal fibrillary proteins may also deposit in the GBM
  • Increased synthesis of the protein components of the basement membrane, as occurs in diabetic glomerulosclerosis
  • Formation of additional layers of basement membrane matrices, which most often occupy sub endothelial locations and may range from poorly organised matrix to fully duplicated lamina densa, as occurs in membrane-proliferative glomerulonephritis
1487
Q

**Hyalinosis, as applied to the glomerular in response to injury, denotes the accumulation of material hat is homogenous and eosinophilic by light microscopy. How does this take place? What is it usually in response to?

A
  • Hyalin is an extracellular, amorphous material composed of plasma proteins that have insulated from the circulation into glomerular structures.
  • When extensive, these deposits may obliterate the capillary lumens of the glomerular tuft
  • Hyalinosis is usually a consequence of endothelial or capillary wall injury and typically the end result of various forms of glomerular damage
1488
Q

Sclerosis in response to glomerular injury is characterised by deposition of extracellular collagenous matrix. Where and why does this happen?

A
  • It may be confined to mesangial areas, as is often the case in diabetic glomerulosclerosis, involve the capillary loops or both
  • The sclerosis process may also result in obliteration of some or all of the capillary lumens in affected glomeruli
1489
Q

Many primary glomerulopathies are classed by their histology. The histologic changes can be further subdivided by their distribution into what categories?

A
  • Diffuse - involving all of the glomeruli in the kidney
  • Global - involving the entirety of individual glomeruli
  • Focal - involving only a fraction of the glomeruli in the kidney
  • Segmental - affecting a part of each glomerulus
  • Capillary loop or mesangial - affecting predominantly capillary or mesangial regions.
1490
Q

Although much remains unknown about etiologic agents and triggering events, it is clear that immune mechanisms underlie most forms of primary glomerulopathy and many of the secondary glomerular diseases. What are some immune mechanisms of glomerular injury

A

Antibody-mediated injury
In situ immune complex deposition
Fixed intrinsic tissue antigens - NC1 domain fo type IV collagen antigen (anti-GBM nephritis); PLA2R antigen (membranous glomerulopathy); mesangial antigens
Planted antigens - exogenous (infectious agents, drugs); endogenous (DNA, nuclear proteins, immunoglobulins, immune complexes, IgA)
Circulating immune complex deposition
Endogenous antigens (e.g. DNA, tumour antigens)
Exogenous antigens (e.g. infectious products)
Cell-mediated injury
**Activation of alternative complement pathway

1491
Q

Two forms of antibody associated glomerular injury have been established. What are these? Which one is more common?

A

1) injury by antibodies reacting in situ within the glomerulus, either binding to insoluble fixed (intrinsic) glomerular antigens or extrinsic molecules planted within the glomerulus
2) injury results from deposition of circulating antigen-antibody complexes in the glomerulus.

It is clear that the major cause of glomerunlonephrtiis resulting from formation of antigen-antibody complexes is the consequence of in situ immune complex formation, and not deposition of circulating complexes as was once thought

1492
Q

How is glomerular disease caused by in situ formation of immune complexes? What disease is a classic example of this?

A
  • In this form of injury, immune complexes are formed locally by antibodies that react with intrinsic tissue antigen or with extrinsic antigens “planted” in the glomerulus from the circulation
  • Membranous nephropathy is the classic example of glomerular injury resulting from local formation of immune complexes
1493
Q

Glomerular disease caused by in situ formation of immune complexes has a well-studied experimental counterpart in the Hermann nephritis rat model, from which much of the underlying pathophysiology of glomerular immune complex-mediated diseases has been deduced. What is the Heymann model of glomerulonephritis?

A
  • It is induced by immunising rats with an antigen, megalin, that is present in epithelial cell foot processes.
  • The rats develop antibodies to this antigen, and disease develops from the reaction of antibody with the megalin-containing protein complex located on the basal surface of visceral epithelial cells, leading to localised immune complex formation
  • Antibody binds to PLA2R (phospholipase A2 receptor) present in the glomerular epithelial cell membrane is followed by complement activation and then shedding of the immune aggregates from the cell surface to form characteristic deposits of immune complexes along the sub-epithelial aspect of the basement membrane
  • These sub-epithelial complexes, with resultant host responses, can result in a thickened basement membrane appearance; hence the term membranous nephropathy
1494
Q

How does Heymann nephritis appear on immunofluorescence and electron microscopy?

A
  • On electron microscopy, the glomerulopathy is characterised by the presence of numerous discrete sub epithelial electron-dense deposits (made up largely by immune reactants
  • The pattern of immune deposition by immunofluorescence microscopy is granular rather than linear, reflective of the very localised antigen-anti-body interaction
1495
Q

In humans, what causes primary and secondary membranous nephropathy?

A
  • Primary membranous nephropathy is an autoimmune disease, caused by antibodies to endogenous tissue components. What triggers these autoantibodies is unclear
  • Secondary forms of membranous nephropathy can be experimentally induced by drugs (eg, mercuric chloride), and graft-versus-host disease. In some of these situations there may be uncontrolled B-cell activation, leading to the production of autoantibodies that react with renal antigens.
1496
Q

Antibodies can react in suit with antigens that are not normally present in the glomerulus but are “planted” there. How does this happen?

A
  • Such antigens may localise in the kidney by interacting with various intrinsic components of the glomerulus
  • Planted antigens include cationic molecules that bind to anionic components of the glomerulus; DNA, nucleosomes, and other nuclear proteins, which have an affinity for GBM components; bacterial products, large aggregated proteins (eg, aggregated immunoglobulins), which deposit in the mesangium because of their size; and immune complexes themselves, since they continue to have reactive sites for further interactions with free antibody, free antigen or complement.
1497
Q

An example of antibodies against planted antigens that cause membranous nephropathy is how it develops in a small number of infants fed cow’s milk. How does this happen?

A
  • These children have been found to have antibodies to bovine albumin and their lesions contain bovine milk antigens, which presumably become lodged in the glomerular basement membrane following intestinal absorption, where they serve as the substrate for immune complex formation in situ.
  • Antibodies that bind to these planted antigens induce a discrete pattern of Ig deposition as granular staining by immunofluorescence microscopy that is indistinguishable from the pattern of staining observed with immune complexes formed from intrinsic antigens
1498
Q

How is disease caused by antibodies directed against normal components of the glomerular basement membrane? How does this compare with other nephropathies?

A
  • In anti-GBM antibody induced glomerulonephritis, antibodies bind to intrinsic antigens homogenously distributed along the entire length of the GBM, resulting in a diffuse linear pattern of staining for the antibodies by immunofluorescence techniques.
  • This contrasts with the granular pattern of immunofluorescence staining corresponding to the discrete immune complexes seen in membranous nephropathy, or other glomerular disease in which large complexes of antigens and antibodies form in situ.
  • These intrinsic fixed antigens cannot be mobilised to form large, discrete complexes
1499
Q

Do the anti-GBM antibodies in anti-GBM antibody induced glomerulonephritis only react with the GBM?

A
  • Often the anti-GBM antibodies cross-react with other basement membranes, especially this in the lung alveoli, resulting in simultaneous lung and kidney lesions (Goodpasture syndrome).
  • The GBM antigen that is responsible for classic anti-GBM antibody-induced glomerulonephritis and Goodpasture syndrome is a component of the non-collagenous domain (NC1) of the α3 chain of type IV collagen that is critical for maintenance of GBM suprastructure.
  • Although anti-GBM antibody-induced glomerulonephritis accounts for fewer than 5% of cases of human glomerulonephritis, it causes severe necrotising and crescentic glomerular damage and the clinical syndrome of rapidly progressive glomerulonephritis
1500
Q

How does glomerulonephritis resulting from deposition of circulating immune complexes happen?

A
  • In this type of nephritis, glomerular injury is caused by the trapping of circulating antigen-antibody complexes within glomeruli
  • The antibodies have no immunologic specificity for glomerular constituents, and the complexes localise within the glomeruli because of their physicochemical properties and the haemodynamic factors peculiar to the glomerulus.
1501
Q

The antigens that trigger the formation of circulating immune complexes in glomerulonephritis may be of endogenous or exogenous origin. What conditions are associated with these?

A
  • The antigens that trigger the formation of circulation immune complexes may be of endogenous origin, as in the glomerulonephritis associated with SLE and in IgA nephropathy
  • Or they may be exogenous, as may occur in the glomerulonephritis that follows certain infections. Microbial antigens that are implicated include bacterial products (streptococcal proteins), the surface antigen of hepatitis B virus, hepatitis C virus antigens, and antigens of Treponema pallidum, Plasmodium falciparum, and several viruses
  • Some tumour antigens are also though to cause immune complex-mediated nephritis
1502
Q

What are the mechanisms of glomerular injury following immune complex formation?

A
  • Whatever the antigen may be, antigen-antibody complexes formed or deposited in the glomeruli may elicit a local inflammatory reaction that produces injury
  • It has long been thought that the inflammation and injury are mediated and amplified by the binding of complement, but recent studies in knockout mice also point to the important of engagement of Fc receptors on leukocytes and perhaps glomerular mesangial or other calls as mediators of the injury process
  • The glomerular lesions may exhibit leukocytic infiltration and proliferation of mesangial and endothelial cells
  • Once deposited in the kidney, immune complexes may eventually be degraded, mostly by infiltrating neutrophils and monocytes/macrophages, mesangial cells, and endogenous proteases, and the inflammatory reaction may then subside
1503
Q

What does electron microscopy and immunofluorescence show in glomerular injury following immune complex formation?

A
  • Electron microscopy reveals electron-dense deposits, presumably containing immune complexes, that may lie in the mesangium, between the endothelial cells and the GBM (subendothelial deposits), or between the outer surface of the GBM and the podocytes (subepithelial deposits). Deposits may be located at more than one site in a given case.
  • By immunofluorescence microscopy, the immune complexes are seen as granular deposits along the basement membrane, in the mesangium, or in both locations.
1504
Q

Several molecular factors affect glomerular localisation of antigen, antibody or immune complexes in glomerular disease. What are these?

A
  • The molecular charge and size of these reactants are clearly important
  • Highly cationic antigens tend to cross the GBM, and the resultant complexes eventually reside in subepithelial location
  • Highly anionic macromolecules are excluded from the GBM and are trapped subendothelially or are not nephritogenic at all
  • Molecules of neutral charge and immune complexes containing these molecules tend to accumulate in the mesangium
  • Large circulating complexes are not usually nephritogenic, because they are cleared by the mononuclear phagocyte system and do not enter the GBM in significant quantities
1505
Q

The pattern of glomerular localisation of antigen, antibody or immune complexes is also affected by organ factors. What are these?

A
  • The pattern of localisation is affected by changes in glomerular haemodynamics, mesangial function, and integrity of the charge-selective barrier in the glomerulus.
  • These influences may underlie the variable pattern of immune reactant deposition in various forms of glomerulonephritis
1506
Q

The distinct patterns of glomerular localisation of immune complexes is a key determinant of the injury response and the histologic features that subsequently develop. What locations lead to what features?

A
  • Immune complexes located in subendothelial portions of capillaries and in mesangial regions accessible to the circulation are more likely to be involved in inflammatory processes that require interaction and activation of circulating leukocytes
  • Diseases in which immune complexes are confined to the subepithelial locations and for which the capillary basement membranes may be a barrier to interaction with circulating leukocytes, as in the case of membranous nephropathy, typically have a non-inflammatory pathology.
1507
Q

What is the cause of most cases of immune complex mediated glomerulonephritis?

A

*It is a consequence of deposition of discrete immune complexes, which give rise to granular immunofluorescence staining along the basement membranes or in the mesangium
* However, it may be difficult to determine whether the deposition has occurred in situ, by circulating complexes, or by both mechanisms because trapping of circulating immune complexes can initiate further in situ complex formation.

1508
Q

What factors contribute to the diverse morphologic and functional alterations in glomerulonephritis?

A

It is best to consider that antigen-antibody deposition in the glomerulus is a major pathway of glomerular injury and that in situ immune reactions, trapping or circulation complexes, interactions between these two events, and local haemodynamic and structural determinants in the glomerulus all contribute to the diverse morphologic and functional alterations in glomerulonephritis

1509
Q

Although antibody-mediated mechanisms may initiate many forms of glomerulonephritis, there is evidence that sensitised T cells cause glomerular injury and are involved in the progression of some glomerulonephritides. What is the evidence for cell-mediated immunity in glomerulonephritis?

A
  • Clues to the role of cellular immunity include the presence of activated macrophages and T cells and their products in the glomerulus in some forms of human and experimental glomerulonephritis; abrogation of glomerular injury by lymphocyte depletion; and experiments in which glomerular injury may be induced by transfer of T cells from nephritic animals to normal recipients.
1510
Q

Once immune reactants or sensitised T cells have localised in the glomerulus, what are the cellular mediators that are involved in acute and chronic inflammation? What do they do?

A
  • Neutrophils and monocytes infiltrate the glomerulus in certain types of glomerulonephritis, largely as a result of activation of complement, resulting in generation of chemotactic agents (mainly C5a), but also by Fc-mediated adherence and activation. Neutrophils release proteases, which cause GBM degradation; oxygen-derived free radicals, which cause cell damage; and arachidonic acid metabolites, which contribute to the reductions in GFR
  • Macrophages and T lymphocytes, which infiltrate the glomerulus in antibody- and cell-mediated reactions, when activated, release a vast number of biologically active molecules
  • Platelets may aggregate in the glomerulus during immune-mediated injury. Their release of eicosanoids, growth factors and other mediators may contribute to vascular injury and proliferation of glomerular cells
  • Resident glomerular cells, particular mesangial cells, can be stimulated to produce several inflammatory mediators, including ROS, cytokines, chemokines, growth factors, eicosanoids, NO and endothelin
1511
Q

Virtually all the known inflammatory chemical mediators have been implicated in glomerular injury. What are some soluble mediators and what do they do?

A
  • Complement activation leads to the generation of chemotactic products that induce leukocyte influx (complement-neutrophil-dependent injury) and the formation of C5b-C9, the membrane attack complex. C5b-C9 causes cell lysis but, in addition, stimulates mesangial cells to produce oxidants, proteases, and other mediators
  • Eicosanoids, NO, angiotensin and endothelin are involved in the haemodynamic changes
  • Cytokines, particularly IL-1 and TNF, which may be produced by infiltrating leukocytes and resident glomerular cells, induce leukocyte adhesion and a variety of other effects
  • Chemokines such as monocyte chemoattractant protein 1 promote monocyte and lymphocyte influx. Growth factors such as platelet-derived growth factor (PDGF) are involved in mesangial cell proliferation. TGF-β, connetcive tissue growth factors and FGF seem to be crtiical in the ECM deposition and hyalinisation leading to glomerulosclerosis in chronic injury. VEGF seems to maintain endothelial integrity and may help regulate capillary permeability
  • The coagulation system is also a mediator of glomerular damage. Fibrin is frequently present in the glomeruli and Bowman space in glomerulonephritis, indicative of coagulation cascade activation, and activated coagulation factors, particularly thrombin, may be a stimulus for crescent formation.
1512
Q

Podocyte injury is common to many forms of both primary and secondary glomerular diseases, of both immune and non-immune etiologies. The term podocytopathy has been applied to diseases with desperate etiologies whose principal manifestation is injury to podocytes. What are the causes of this? How is it reflected?

A
  • This can be induced by antibodies to podocyte antigens; by toxins, conceivably by certain cytokines; by certain viral infections such as HIV or still by inadequately characterised circulating factors, as in some cases of focal segmental glomerulosclerosis
  • Such injury is reflected by stereotypic morphologic changes in the podocytes, which include effacement of foot processes, vacuolisation, and retraction and detachment of cells from the GBM, and functionally by proteinuria
1513
Q

Can podocytes repair after injury? What diseases can the injury?

A

Loss of podocytes, which have only a very limited capacity for replication and repair, may be a feature of multiple types of glomerular injury including focal and segmental glomerulosclerosis and diabetic nephropathy.

1514
Q

In most forms of glomerular injury, loss of normal slit diaphragm is a key event in the development of proteinuria. How does it happen?

A
  • Functional abnormalities of the slit diaphragms may also result from mutations in its components, such as nephron and podocin, without actual inflammatory damage o the glomerulus.
  • Such mutations are the cause of rare hereditary forms of the nephrotic syndrome
1515
Q

What does the outcome of glomerular injury depend on?

A

The severity of the renal damage, the nature and persistence of the antigens, and the immune status, age, and genetic predisposition of the host

1516
Q

In renal disease, below what GFR level does progression to end-stage-renal failure occur at?

A

Once any renal disease, glomerular or otherwise, destroys functioning nephrons and reduces the GFR to about 30-50% of normal, progression to end-stage renal failure proceeds at a steady rate, independent of the original stimulus or activity of the underlying disease.

1517
Q

What are the two major histological characteristics of progressive renal damage?

A

Focal segmental glomerulosclerosis and tubulointerstitial fibrosis

1518
Q

Where does focal segmental glomerulosclerosis occur in the kidney? What does it lead to?

A

Progressive fibrosis involving portions of some glomeruli develops after many types of renal injury and leads to proteinuria and increasing functional impairment

1519
Q

Focal segmental glomerulosclerosis may be seen even in causes in which the primary disease was non-glomerular. How does this happen?

A
  • The glomerulosclerosis seems to be initiated by the adaptive change that occurs in the relatively unaffected glomeruli of disease kidneys. Such a mechanism is suggested by experiments in rats subjected to subtotal nephrectomy
  • Compensatory hypertrophy of the remaining glomeruli initially maintains the renal function in these animals, but proteinuria and segmental glomerulosclerosis soon develop, leading eventually to total glomerulosclerosis and uraemia.
  • The glomerular hypertrophy is associated with haemodynamic changes, including increases in glomerular blood flow, filtration, and transcapillary pressure (glomerular HTN), and often with systemic hypertension
1520
Q

What is the sequence of events that it thought to lead to focal segmental glomerulosclerosis?

A
  • It entails endothelial and visceral epithelial cell injury, visceral epithelial cell loss leading to segments of GBM denuded of overlying foot processes and consequently increased glomerular permeability to proteins, and accumulation of proteins in the mesangial matrix
  • This is followed by proliferation of mesangial cells, infiltration by macrophages, increased accumulation of extracellular matrix (ECM), and segmental and eventually global sclerosis of glomeruli
  • With increasing reductions in nephron mass and ongoing compensatory changes, a vicious cycle of continuing glomerulosclerosis sets in.
  • Most of the mediators of chronic inflammation and fibrosis, particularly TGF-β, play a role in the induction of sclerosis
1521
Q

What are the most successful interventions to interrupt the mechanisms of progressive glomerulosclerosis?

A

They involve treatment with inhibitors of the renin-angiotensin system, which not only reduce intraglomerular hypertension, but also have direct effect on each of the mechanisms identified in the flow chart.
Importantly, these agents have been shown to ameliorate progression of sclerosis in both animal and human studies

1522
Q

Tubulointerstitial injury, manifested by tubular damage and interstitial inflammation, is a component of many acute and chronic glomerulonephritides. What diseases is it involved it? How does it correlate with renal function compared to glomerular injury?

A
  • Tubulointerstitial fibrosis contributes to progression in both immune and non-immune glomerular diseases, for example, diabetic nephropathy
  • There is often a much better correlation of decline in renal function with the extent of tubulointerstitial damage than with the severity or glomerular injury
1523
Q

Many factors may lead to tubulointerstitial injury. What are some?

A
  • They include ischaemia of tubule segments downstream from sclerotic glomeruli, acute and chronic inflammation in the adjacent interstitium, and damage of loss of the peritubular capillary blood supply
  • It appears that proteinuria can cause direct injury to and activation of tubular cells
  • Activated tubular cells in turn express adhesion molecules and elaborate pro-inflammatory cytokines, chemokines, and growth factors that contribute to interstitial fibrosis.
  • Filtered proteins that may produce these tubular effects include cytokines, complement products, the iron in haemoglobin, immunoglobulins, lipid moieties, and oxidatively modified plasma proteins
1524
Q

What are the clinical and pathological characteristics of nephritic syndrome?

A
  • Glomerular diseases presenting with a nephritic syndrome are often characterised by inflammation in the glomeruli
  • The nephritic patient usually presents with haematuria, red cell casts in the urine, azotemia, oliguria, and mild to moderate hypertension
  • Proteinuria and oedema are common, but not as severe as those encountered in the nephrotic syndrome
1525
Q

What conditions are associated with nephritic syndrome?

A
  • The acute nephritic syndrome may occur in such multi-system diseases as SLE and microscopic polyangiitis
  • Typically however, it is characteristic of acute proliferative and exudative glomerulonephritis and is an important component of crescentic glomerulonephritis
1526
Q

What are the characteristics of acute proliferative (post-streptococcal, post-infectious) glomerulonephritis?

A
  • This cluster of diseases is characterised histologically by diffuse proliferation of glomerular cells associated with influx (exudation) of leukocytes
  • These lesions are typically caused by immune complexes.
1527
Q

What are the causes of acute proliferative glomerulonephritis?

A
  • The inciting antigen may be exogenous or endogenous
  • The prototypic exogenous antigen-induced disease pattern in post-infectious glomerulonephritis
  • An example of an endogenous antigen-induced disease is the nephritis of SLE
  • The most common underlying infections are streptococcal, but the disorder may also be assocated with other infections
1528
Q

What does post-strep glomerulonephritis present? How? in who?

A
  • It usually appears 1-4 weeks after a streptococcal infection of the pharynx or skin (impetigo)
  • Skin infections are commonly associated with overcrowding and poor hygiene
  • Post-streptococcal glomerulonephritis occurs most frequently in children 6-10 years of age, but children and adults of any any can also be affected
1529
Q

What is the pathogenesis of post-streptococcal glomerulonephritis?

A

Post-streptococcal GN is caused by immune complexes containing streptococcal antigens and specific antibodies, which are formed in situ

1530
Q

What strains of strep cause post-strep GN?

A
  • Only certain strains of group A β-haemolytic streptococci are nephritogenic, more than 90% of cases being traced to types 12, 4 and 1, which can be identified by typing of the M protein of the bacterial cell walls
1531
Q

Many lines of evidence support an immunologic basis for post-streptococcal glomerulonephritis. What are these?

A
  • The latent period between infection and onset of nephritis is compatible with the time required for the production of antibodies and the formation of immune complexes.
  • Elevated titres of antibodies against one or more streptococcal antigens are present in a great majority of patients.
  • Serum complement levels are low, compatible with activation of the complement system and consumption of complement components
  • There are granular immune deposits in the glomeruli, indicative of an immune complex-mediated mechanisms
1532
Q

The streptococcal antigenic responsible for the immune reaction had long eluded identification, but the what is the most likely one? What does it do to cause post-strep GN?

A
  • Streptococcal pyogenic exotoxin B (SpeB) has been identified as the principal antigenic determinant in most but not all cases of post-streptococcal glomerulonephritis.
  • This protein can directly active complement, is commonly secreted by nephritogenic strains of streptococci, and has been localised to the “hump-like” deposits characteristic of this disease.
  • At the outset, the inciting antigens are exogenously planted from the circulation in subendothelial locations in glomerular capillary walls, leading to in situ formation of immune complexes, where they elicit an inflammatory response
  • Subsequently, the antigen-antibody complexes dissociate, migrate across the GBM, and re-form on the subepithelial side of the GBM
1533
Q

What is the classic histologic picture of post-strep glomerulonephritis?

A
  • The classic histologic picture is one of enlarged, hypercellular glomeruli.
  • The hypercellularity is caused by 1) infiltration by leukocytes, both neutrophils and monocytes; 2) proliferation of endothelial and mesangial cells; and 3) in severe cases by crescent formation
  • The proliferation and leukocyte infiltration are typically global and diffuse, that is involving all lobules of all glomeruli
  • There is also swelling of endothelial cells, and the combination of proliferation, swelling, and leukocyte infiltration obliterates the capillary lumens.
  • There may be interstitial oedema and inflammation, and the tubules often contain red cell casts.
1534
Q

What do you see on immunofluorescence microscopy and electron microscopic findings of post-strep GN?

A
  • By immunofluorescence microscopy, there are granular deposits of IgG, and C3, and sometimes IgM in the mesangium and along the GBM. Although immune complex deposits are almost universally present, they are often focal and sparse
  • The characteristic electron microscopic findings are discrete, amorphous, electron-dense deposits on the epithelial side of the membrane, often having the appearance of “humps”, presumably representing the antigen-antibody complexes at the subepithelial cell surface. Subendothelial complexes are commonly seen, typically early in the disease course, and mesangial and intramembranous deposits may be presents.
1535
Q

What is the classical presentation clinically and in FWT of post-strep GN? In children and in adults please

A
  • In the typical case, a young child abruptly develops malaise, fever, nausea, oliguria, and haemturia (smoky or cola-coloured urine) 1-2 weeks after recovery from a sore throat.
  • The patients have dysmorphic red cells or red cell casts in the urine, mild proteinuria (usually less than 1gm/day), periorbital oedema, and mild to moderate hypertension.
  • Inducts, the onset is more likely to be atypical, such as the sudden appearance of HTN or oedema, frequently with elevation of BUN
1536
Q

What are some important lab findings for post-strep GN?

A

Important laboratory findings include elevations of anti-streptococcal antibody titres and a decline in the serum concentration of C3 and other components of the complement cascade.

1537
Q

What is the outcome of post-strep GN in children?

A
  • More than 95% of affected children eventually recover renal function with conservative therapy aimed at maintaining sodium and water balance.
  • A small minority of children (perhaps fewer than 1%) do not improve, become severely oliguric, and develop a rapidly progressive form of glomerulonephritis
  • Some of the remaining patents undergo slow progression to chronic glomerulonephritis with or without recurrence of an active nephritic picture.
    *Prolonged and persistent heavy proteinuria and abnormal GFR mark patents with an unfavourable prognosis
1538
Q

What is the outcome of post-strep GN in adults?

A
  • In adults the disease is less benign that in children
  • Although the overall prognosis in epidemics is good, in only about 60% of sporadic cases do the patents recover promptly
  • In the remainder the glomerular lesions fail to resolve quickly, as manifested by persistent proteinuria, haematuria, and HTN.
  • In some of these patients, the lesions eventually clear, but others develop chronic glomerulonephritis or even rapidly progressive glomerulonephritis
1539
Q

What is non-streptococcal acute glomerulonephritis (post-infectious glomerulonephritis)? What causes it? What do you see on immunofluorescence?

A
  • A similar form of glomerulonephritis to post-strep GN that occurs sporadically in association with other infections, including those of bacterial (eg, staph endocarditis, pneumococcal pneumonia, meningococcaemia), viral (hep B, hep C, mumps, HIV, varicella), and parasitic (malaria, toxoplasmosis) origin.
  • In these settings, granular immunofluorescent deposits and subepithelial humps characteristic of immune complex nephritis are present
  • Post-infectious glomerulonephritis due to staph infections differs by sometimes producing immune deposits containing IgA rather than IgG
1540
Q

What is rapidly progressive (crescentic) glomerulonephritis?

A
  • Rapidly progressive glomerulonephritis (RPGN), is a syndrome associated with severe glomerular injury, but does not denote a specific etiologic form of glomerulonephritis
  • It is characterised by rapid and progressive loss of renal function associated with severe oliguria and signs of nephritic syndrome if untreated, death from renal failure occurs within weeks to months
1541
Q

What is the most common histologic picture of rapidly progressive (crescentic) glomerulonephritis?

A
  • The most common histologic picture is the presence of crescents in most of the glomeruli (crescentic glomerulonephritis)
  • There are produce predominantly by the proliferation of parietal epithelial cells lining Bowman capsule and by the infiltration of monocytes and macrophages
1542
Q

Rapidly progressive glomerulonephritis may be caused by a number of different diseases, some restricted to the kidney and others systemic. Although no single mechanism can explain all causes there is little doubt that in most cases the glomerular injury is immunologically mediated. A practical classification divides RPGN into three groups on the basis of immunologic findings. What are these groups? What diseases are in each one?

A

Type 1 (Anti-GBM antibody)
Renal limited
Goodpasture syndrome
Type 2 (Immune complex)
Idiopathic
Post-infectious glomerulonephritis
Lupus nephritis
Henoch-Schonlein purpurn
IgA nephropathy
Type 3 (Pauci-immune)
ANCA-associated
Idiopathic
Granulomatosis with polyangiitis
Microscopic polyangiitis

1543
Q

What are the characteristics of anti-GBM antibody-mediated rapidly progressive glomerulonephritis? How is it treated?

A
  • Anti-GBM antibody-mediated disease, characterised by linear deposits of IgG and, in many cases, C3 in the GBM
  • In some of these patients, the anti-GBM antibodies cross-react with pulmonary alveolar basement to produce the clinical picture of pulmonary haemorrhage associated with renal failure (Goodpasture syndrome)
  • Plasmapheresis to remove the pathogenic circulating antibodies is usually part of the treatment, which also includes therapy to suppress the underlying immune response
1544
Q

What is the antigen that causes anti-GBM antibody-mediated disease? What triggers the formation of the antibodies? Is it genetically linked?

A
  • The antigen common to the alveoli and GBM is a peptide within the non-collagenous portion of the α3 chain of collagen type IV.
  • What triggers the formation of these antibodies is unclear in most patients
  • Exposure to viruses or hydrocarbon solvents (found in paints and dyes) has been implicated in some patients, as have various drugs and cancers
  • There is a high prevalence of certain HLA subtypes and haplotypes (eg, HLA-DRB1) in affected patients, a finding consistent with the genetic predisposition to autoimmunity
1545
Q

What is the immunofluorescence staining of rapidly progressive glomerulonephritis caused by immune complex deposition? What diseases is it associated with? How do you treat it?

A
  • RPGN can be a complication of any of the immune complex nephritides, including post-infectious GN, lupus nephritis, IgA nephropathy, and HSP.
  • In all these cases, immunofluorescence studies reveal all the granular pattern of staining characteristic immune complex deposition.
  • This type of of RPGN frequently demonstrates cellular proliferation and influx of leukocytes within the glomerular tuft, in addition to crescent formation
  • These patients usually cannot be helped by plasmapheresis, and they require treatment of the underlying disease
1546
Q

What is pauci-immune rapidly progressive glomerulonephritis? What antibodies are associated with it?

A
  • It is defined by the lack of detectable anti-GBM antibodies or immune complexes by immunofluorescence and electron microscopy.
  • Most patient with this type of RPGN have circulating antineutrophil cytoplasmic antibodies (ANCAs) that produce cytoplasmic (c) or perinuclear (p) staining pattern and are known to play a role in some vasculatides
1547
Q

What diseases are associated with pauci-immune rapidly progressive glomerulonephritis?

A
  • This type of RPGN may be a component of a systemic vasculitis such as granulomatosis with polyangiitis or microscopic polyangiitis.
  • In many cases, however, pauci-immune crescentic glomerulonephritis is limited to the kidneys and hence idiopathic
1548
Q

What is the gross morphology of rapidly progressive (crescentic) glomerulonephritis?

A
  • The kidneys are enlarged and pale, often with petechial haemorrhages on the cortical surfaces
  • Depending on the underlying cause, the glomeruli often show focal and segmental necrosis, and variably show diffuse or focal endothelial proliferation, and mesangial proliferation
  • Segmental glomerular necrosis adjacent to glomerular segments uninvolved by inflammatory or proliferative changes is the feature most typical of pauci-immune RPGN
1549
Q

What is the histologic picture of rapidly progressive (crescentic) glomerulonephritis?

A
  • The histologic picture is dominated by distinctive crescents.
  • Crescents are formed by proliferation of parietal cells and by migration of monocytes and macrophages into the urinary space.
  • Neutrophils and lymphocytes may be present
  • The crescents may obliterate the urinary space and compress the glomerular tuft
  • Fibrin strands are frequently prominent between the cellular layers in the crescents.
1550
Q

What is the immunofluorescence picture of rapidly progressive (crescentic) glomerulonephritis?

A
  • Immune complex-mediated cases show granular immune deposits
  • Goodpasture syndrome cases show linear GBM fluorescence for Ig and complement
  • Pauci-immune cases have little or no deposition of immune reactants
1551
Q

What is the electron microscopy picture of rapidly progressive (crescentic) glomerulonephritis?

A
  • Electron microscopy discloses deposits in those cases due to immune complex deposition (type II)
  • Regardless of type, electron microscopy may show rupture in the GBM, a severe injury that allows leukocytes, plasma proteins such as coagulation factors and complement, and inflammatory mediators to reach the urinary space, where they trigger crescent formation
1552
Q

Over time, what happens to most crescents in rapidly progressive (crescentic) glomerulonephritis?

A

In time, most crescents undergo organisation and foci of segmental necrosis resolve as segmental scars (a type of segmental sclerosis), but restoration of normal glomerular architecture may be achieved with early aggressive therapy

1553
Q

What are the renal clinical manifestations of rapidly progressive glomerulonephritis?

A

The renal manifestations of all forms of crescentic glomerulonephritis include haematuria with red blood cell casts in the urine, moderate proteinuria occasionally reaching the nephrotic range, and variable hypertension and oedema

1554
Q

What are some useful blood tests for crescentic glomerulonephritis?

A

Serum analyses for anti-GBM antibodies, antinuclear antibodies, and ANCAs are helpful in the diagnosis of specific subtypes

1555
Q

What is the treatment and progression of rapidly progressive glomerulonephritis?

A
  • Although milder forms of glomerular injury may subside, the renal involvement is usually progressive over a matter of weeks and culminates in severe oliguria
  • Recovery of renal function may follow early intensive plasmapheresis combined with steroids and cytotoxic agents in Goodpasture syndrome
  • Other forms of RPGN also response well to steroids and cytotoxic agents
  • However, despite therapy, many patients eventually require chronic dialysis or transplantation, particularly in the disease is discovered at a late stage
1556
Q

What causes nephrotic syndrome? What are the manifestations of this?

A

Nephrotic syndrome is caused by a derangement in glomerular capillary walls resulting in increased permeability to plasma proteins. The manifestations of the syndrome include:
* Massive proteinuria, with the daily loss of 3.5gm or more of proteins
* Hypoalbuminaemia, with plasma albumin levels less that 3gm/dL
* Generalised oedema
* Hyperlipidaemia and lipiduria

1557
Q

What causes the hypoalbuminaemia in nephrotic syndrome?

A
  • Heavy proteinuria depletes serum albumin levels at a rate beyond the compensatory synthetic capacity of the liver, resulting in hypoalbuminaemia
  • Increased renal catabolism of filtered albumin also contributes to the hypoalbuminaemia
1558
Q

What causes the generalised oedema in nephrotic syndrome?

A
  • The generalised oedema is a direct consequence of decreased intravascular colloid osmotic pressure
  • There is also sodium and water retention, which aggravates the oedema. This seems to be due to several factors, including compensatory secretion of aldosterone, mediated by the hypovolaemia-enhanced renin secretion; stimulation of the sympathetic system; and a reduction in the secretion of natriuretic factors such as atrial peptides
1559
Q

What proteins are lost in nephrotic syndrome?

A
  • The largest proportion of protein lost in the urine is albumin, but globulins are also excreted in some diseases
  • The ratio of low- to high-molecular-weight proteins in the urine in various cases of nephrotic syndrome in a manifestation of the selectivity of proteinuria
  • A highly selective proteinuria consists mostly of low-molecule-weight proteins
  • A poorly selective proteinuria consists of higher molecular-weight globulins in addition to albumin
1560
Q

What shows that is hyperlipidaemia linked to nephrotic syndrome?

A

Most patients with nephrotic syndrome have increased blood levels of cholesterol, triglyceride, very-low-density lipoprotein, low-density lipoprotein, Lp(a) lipoprotein, and apoprotein, and there is a decrease in high-density lipoprotein concentration in some patients

1561
Q

Why is hyperlipidaemia linked with nephrotic syndrome?

A
  • Hyperlipidaemia (particularly LDL) defects seem to be due to a combination of increased synthesis of lipoproteins in the liver, abnormal transport of circulating lipid particles, and decreased lipid catabolism.
  • Lipiduria follows the hyperlipidaemia, because lipoproteins also leak across the glomerular capillary wall
  • The lipid appears in the urine either as free fat or oval fat bodies, representing lipoprotein resorbed by tubular epithelial cells and then shed along with injured tubular cells that have detached from the basement membrane
1562
Q

What are the complications of nephrotic patients?

A
  • Nephrotic paients are particularly vulnerable to infection, especially staph or pneumococcal infections, probably due to loss of immunoglobulins in the urine
  • Thrombotic and thromboembolic complications are also common in nephrotic syndrome, due in part to loss of endogenous anticoagulants (eg, antithrombin III) in the urine
  • Renal vein thrombosis is most often a consequence of this hypercoagulable state, particularly in patients with membranous nephropathy
1563
Q

What are the most frequent systemic causes and most important primary glomerular lesions causes of nephrotic syndrome?

A
  • The most frequent systemic causes are diabetes, amyloidosis and SLE
  • The most important primary glomerular lesions are minimal-change disease, membranous glomerulopathy, and focal segmental glomerulosclerosis
1564
Q

What is membranous nephropathy? Is it often primary or secondary?

A
  • Membranous nephropathy is characterised by diffuse thickening of the glomerular capillary wall due to the accumulation of deposits containing Ig along the subepithelial side of the basement membrane.
  • Approximately 75% of cases of membranous nephropathy are primary
  • The remaining cases occur in association with other systemic diseases and have identifiably etiologic agents, and are referred to as secondary membranous nephropathy
1565
Q

What are some of the secondary causes of glomerular nephropathy?

A
  • Drugs (penicillin, NSAIDs)
  • Underlying malignant tumours, particularly carcinomas of the lung and colon, and melanoma.
  • SLE - about 10-15% of glomerulonephritis in SLE is of the membranous type
  • Infections - chronic hep B, hep C, syphilis, schistosomiasis, malaria
  • Other autoimmune disorders such as thyroiditis can be associated with secondary membranous nephropathy
1566
Q

Membranous nephropathy is a form of chronic immune complex-mediated disease. In secondary, the inciting antigens can sometimes by identified in the immune complexes. Where do these antigens come from in particular diseases?

A
  • The antigens may be endogenous or exogenous
  • The endogenous antigens may be renal or non-renal. For example, membranous nephropathy in SLE is associated with deposition of complexes of self nuclear proteins and autoantibodies. Another example is neutral endopeptidase, a membrane protein recognised by place tally transferred maternal antibodies in cases of neonatal membranous nephropathy
  • Exogenous antigens include those derived from hepatitis B and Treponema pallidum in patients infected with these microbes
1567
Q

Primary membranous nephropathy was long thought to be of unknown cause. But what is now known about the pathogenesis?

A
  • It is now considered to be an autoimmune disease linked to certain HLA alleles such as HLA-DQA1.
  • In many adult cases the auto antigen is the phospholipase A2 receptor.
  • The lesions bear a striking resemblance to those of experimental Heymann nephritis
1568
Q

How does the glomerular capillary wall become leaky in membranous nephropathy? What is the main immunoglobulin deposited?

A
  • There is a paucity of neutrophils, monocytes, or platelets in glomeruli
  • It is postulated that C5b-C9 complex activates glomerular epithelial and mesangial cells, inducing them to liberate proteases and oxidants, which cause capillary wall injury and increased protein linkage.
  • A subclass of IgG, IgG4, which differs from other IgG subclasses in being a poor activator of the classical complement pathway, is the principal immunoglobulin deposited in cases of primary membranous nephropathy.
1569
Q

What is the light and electron microscopy morphologic findings in membranous nephropathy?

A
  • By light microscopy, the glomeruli either appear normal in the early stages of the disease or exhibit uniform, diffuse thickening of the glomerular capillary wall
  • By electron microscopy (B), the thickening is seen to be caused by irregular electron dense also deposits containing immune complexes between the basement membrane and the overlying epithelial cells, with effacement of podocyte foot processes
  • Basement membrane material is laid down between these despotic, appearing as irregular spikes protruding from the GBM. These spikes are best seen by silver stains (A), which colour the basement membrane, but not the deposits, black.
  • In time, these spikes thicken to produce dome-like protrusions and eventually close over the immune deposits, burying them within a markedly thickened, irregular membrane.
1570
Q

What do you see on immunofluorescence microscopy of membranous nephropathy?

A
  • Immunofluorescence microscopy (C) demonstrates that the granular deposits contain both immunoglobulins and complement
  • As the disease advances segmental sclerosis may occur; in the course of time glomeruli may become completely sclerosed
  • The epithelial cells of the proximal tubules contain protein reabsorption droplets, and there may be considerable interstitial mononuclear cell inflammation
1571
Q

How does membranous nephropathy usually present?

A
  • This disorder usually present with the insidious onset of the nephrotic syndrome or, in 15% of patient, with non-nephrotic proteinuria.
  • Haematuria and mild HTN are present in 15-35% of cases
1572
Q

What is the clinical course of membranous nephropathy?

A
  • The course of the disease is variable but generally indolent
  • In contrast to minimal-change disease, the proteinuria is non-selective and usually does not response well to corticosteroid therapy
  • Complete or partial remissions may occur in up to 40% of patients, even in some patients without therapy
  • Progression is associated with increasing sclerosis of glomeruli, rising serum creatinine reflecting renal insufficiency, and development of HTN
1573
Q

What is the prognosis of membranous nephropathy?

A
  • Although proteinuria persists in more than 60% of patents, only about 10% die or progress to renal failure within 10 years, and no more than 40% eventually develop severe kidney disease or end-stage renal disease
  • The disease recurs in up to 40% of patients who undergo transplantation for end-stage renal disease
  • Spontaneous remissions are a relatively benign outcome occur more commonly in women and in those with proteinuria in the non-nephrotic range
1574
Q

What antibodies are a useful biomarker in membranous nephropathy?

A

The circulating antibodies to PLA2 receptor may be a useful biomarker of disease activity and thereby aid in the diagnosis and management of primary membranous nephropathy in the future

1575
Q

What is minimal-change disease? Who does it happen in?

A
  • This relatively benign disorder is characterised by diffuse effacement of foot processes of visceral epithelial cells (podocytes), detectable only by electron microscopy, in glomeruli that appear virtually normal by light microscopy
  • It is the most frequent cause of nephrotic syndrome in children, but it is less common in adults
  • The peak incidence in between 2 and 6 years old
  • The disease sometimes follows a respiratory infection or routine prophylactic immunisation
1576
Q

Although the absence of immune deposits in the glomerulus excludes classic immune complex mechanisms, several features of the disease point to an immunologic basis. What are these?

A

1) The clinical association with respiratory infections and prophylactic immunisation
2) The response to corticosteroids and/or other immunosuppressive therapy
3) The association with other atopic disorders (e.g, eczema, rhinitis)
4) The increase prevalence of certain HLA haplotypes in patients with minimal-change disease associated with atopy (suggesting a genetic predisposition)
5) The increased incidence of minimal-change disease in patients with Hodgkin lymphoma, in whom defects in T cell-mediated immunity are well recognised

1577
Q

The current leading hypothesis is that minimal-change disease involves some immune dysfunction that results in the elaboration of factors that damage visceral epithelial cells and cause proteinuria. What are these factors and changes?

A
  • The ultrastructural changes point to a primary visceral epithelial cell injury (podocytopathy), and studies in animal models suggest the loss of glomerular polyanions.
  • Thus, defects in the charge barrier may contribute to the proteinuria
  • The actual route by which protein traverses the epithelial cell portion of the capillary wall remains an enigma. Possibilities include transcellular passage through the epithelial cells, passage through residual spaces between remaining but damaged foot processes or through abnormal spaces developing underneath the portion of the foot process that directly abuts the basement membrane, or leakage through foci in which the epithelial cells have become detached from the basement.
1578
Q

In minimal-change disease, the glomeruli are normal by light microscopy. What are the changes in electron microscopy?

A
  • The GBM appears normal, and no electron-dense material is deposited
  • The principal lesion is in the visceral epithelial cells, which show a uniform and diffuse effacement of foot processes, these being reduced to a rim of cytoplasm with loss of recognisable intervening slit diaphragms.
  • This change represents simplification of the epithelial cell architecture with flattening, retraction, and swelling of foot processes.
  • Foot process effacement is also present in other proteinuric states; it is onlt when effacement is associated with normal glomeruli by Leith microscope that the diagnosis of minimal-change disease can be made.
  • The cells of the proximal tubules are often laden with lipid and prtein, reflecting tubular reabsorption of lipoproteins passing through diseased glomeruli
1579
Q

What are the clinical features of minimal-change disease?

A
  • Despite massive proteinuria, renal function remains good, and there is commonly no hypertension or haematuria
  • The proteinuria usually is highly selective, most of the protein being albumin
1580
Q

Is minimal-change disease responsive to steroids? What is the prognosis?

A
  • A characteristic feature it its usually dramatic response to corticosteroid therapy
  • Most children (>90%) with minimal-change disease respond rapidly to this treatment
  • However, proteinuria may recur, and some patients may become steroid-dependent or resistant.
  • Nevertheless, the long-term prognosis for patients is excellent, and even steroid dependent disease usually resolves when children reach puberty.
  • Although adults are slower to respond, their long-term prognosis is also excellent
1581
Q

What diseases is minimal-change disease associated with in adults?

A

Hodgkin lymphoma and, less frequently, other lymphomas and leukaemias

1582
Q

What is focal segmental glomerulosclerosis? Is it common?

A
  • Primary focal segmental glomerulosclerosis is the most common cause of nephrotic syndrome in adults in the US
  • The lesion is characterised by sclerosis of some, but not all, glomeruli (thus, it is focal); and in the affected glomeruli, only a portion of the capillary tuft is involved (thus, it is segmental).
1583
Q

How does focal segmental glomerulosclerosis frequently manifest?

A
  • It frequently manifests clinically by the acute or sub-acute onset of nephrotic syndrome or non-nephrotic proteinuria.
  • HTN, microscopic haematuria, and some degree of azotemia are commonly present when the disease is first clinically recognised
1584
Q

What are the settings in which focal segmental glomerulosclerosis?

A
  • As a primary disease (idiopathic focal segmental glomerulosclerosis)
  • In association with other known conditions, such as HIV, heroin addiction, sickle-cell disease, and massive obesity
  • As a secondary event, reflecting scarring of previously active necrotising lesions, in cases with focal glomerulonephritis (IgA nephropathy)
  • As a component of the adaptive response to loss of renal tissue (renal ablation), whether from congenital abnormalities (unilateral renal agenesis or renal dysplasia), or acquired cause (eg, reflux nephropathy), or in advanced disorders
  • In uncommon inherited forms of nephrotic syndrome where the disease may be caused by mutations in genes that encode proteins localised to the slit diaphragms e.g, podocin, α-actinin-4 and TRPC6 (transient receptor potential calcium channel-6)
1585
Q

How does idiopathic focal segmental glomerulosclerosis differ from minimal-change disease?

A

1) there is a higher incidence of haematuria, reduced GFR and HTN
2) proteinuria is more often non-selective
3) there is poor response to corticosteroid therapy
4) there is progression to CKD, with at least 50% developing ESRD within 10 years

1586
Q

What is the pathogenesis of focal segmental glomerulosclerosis?

A
  • The characteristic degeneration and focal disruption of visceral epithelial cells with effacement of foot processes resemble the diffuse epithelial cell change typical of minimal-change disease and other podocytopathies. It is the epithelial damage that is hallmark of FSGS
  • Multiple difference mechanisms can cause such epithelial damage, including circulating factors and genetically determined defects affecting components of the slit diaphragm complex.
  • The hyalinosis and sclerosis stem from entrapment of plasma proteins in extremely hyperpermeable foci and increased ECM deposition.
  • The recurrence of proteinuria after transplantation, sometimes within 24 hours, with subsequent progression to overt lesions of FSGS, suggests that an unknown circulating factor is the cause of the epithelial damage in some patients
1587
Q

The discovery of a genetic basis for some cases of FSGS and other causes of the nephrotic syndrome has improved the understanding of the pathogenesis of proteinuria in the nephrotic syndrome and have provided new methods for diagnosis and prognosis of affected patients. What are the useful genes identified? Why are they useful?

A
  • NPHS1, maps to chromosome 19q13 and encodes the protein nephrin. Nephrin is a key component of the slit diaphragm, the structure that controls glomerular permeability. Several mutations of the NPHS gene have been identified that give rise to congenital nephrotic syndrome
  • A distinctive pattern of autosomal recessive FSGS results from mutations in the NPHS2 gene, which maps to chromosome 1q25-q31 and encodes the protein product podocin. Podocin has also been localised to the slit diaphragm
  • A third set of mutations in the gene encoding the podocyte actin-binding protein α-actinin 4 underlies some cases of autosomal dominant FSGS, which can be insidious in onset but has a high rate of progression to renal insufficiency
  • A fourth type of mutation was found in the gene encoding TRPC6, which is widely expressed, including in podocytes, and the pathogenic mutations may perturb podocyte function by increasing calcium flux in these cells
1588
Q

What is renal ablation focal segmental glomerulosclerosis?

A
  • A secondary form of FSGS, which occurs as a complication of glomerular and non-glomerular diseases causing reduction in functioning renal tissue
  • Particularly striking examples where this occurs are reflux nephropathy and unilateral agenesis.
  • These may lead to progressive glomerulosclerosis and renal failure.
1589
Q

What is the morphology by light microscopy in focal segmental glomerulosclerosis?

A
  • By light microscopy, the focal and segmental lesions may involve only a minority of the glomeruli and may be missed if the biopsy specimen contains an insufficient number of glomeruli
  • In the sclerotic segments there is collapse of capillary loops, increase in matrix and segmental deposition of plasma proteins along the capillary wall (hyalinosis), which may become so pronounced as to occlude capillary lumens
  • Lipid droplets and foam cells are often present
  • Glomeruli that do not show segmental lesions usually appear normal on light microscopy but may show increased mesangial matrix
1590
Q

How does the morphology of focal segmental glomerulosclerosis change over time?

A
  • With the progression of the disease, increased numbers of glomeruli become involved and sclerosis spreads within each glomerulus
  • In time, this leads to total *i.e. global) sclerosis of glomeruli, with pronounced tubular atrophy and interstitial fibrosis
1591
Q

What is the morphology by electron microscopy and immunofluorescence in focal segmental glomerulosclerosis?

A
  • On electron microscopy both sclerotic and non-sclerotic areas show diffuse effacement of foot processes, and there may also be focal detachment of the epithelial cells and denudation of the underlying GBM
  • By immunofluorescence microscopy IgM and C3 may be present in the sclerotic areas and/or in the mesangium
  • In addition to the focal sclerosis, there may be pronounced hyalinosis and thickening of afferent arterioles
1592
Q

There is a morphologic variant of focal segmental glomerulosclerosis called collapsing glomerulopathy. What are the characteristics of this? What disease is it associated with?

A
  • It is characterised by retraction and/or collapse of the entire glomerular tuft, with or without additional normal FSGS lesions.
  • A characteristic feature is proliferation and hypertrophy of glomerular visceral epithelial cells. This lesion may be idiopathic, but it also has been associated with some drug toxicities (eg, pamidronate), and it is the most characteristic lesion of HIV-associated neuropathy
  • Collapsing glomerulopathy is typically associated with prominent tubular injury with formation of microcysts. It has a particularly poor prognosis
1593
Q

What is the clinical course of focal segmental glomerulosclerosis? Is it better in adults or children?

A
  • There is little tendency for spontaneous remission in idiopathic FSGS, and responses to corticosteroid therapy are variable
  • In general, children have a better prognosis than adults do
  • Progression to renal failure occurs at variable rates.
  • About 20% of patients follow an unusually rapid course, with intractable massive proteinuria ending in renal failure within 2 years
  • Recurrences are seen in 25-50% of patients receiving allografts
1594
Q

What are some factors associated with rapid progression into renal failure from focal segmental glomerulosclerosis?

A

Factors associated with rapid progression include:
* The degree of proteinuria,
* The degree of renal insufficiency at diagnosis,
* Histologic subtype (the collapsing variant has an unfavourable course; the tip variant has a relatively good prognosis)

1595
Q

HIV infection can directly and indirectly cause several renal complications. What are some of these?

A
  • Acute renal failure or acute interstitial nephritis induced by drugs or the infection
  • Thrombotic microangiopathies
  • Post-infectious glomerulonephritis
  • Most commonly, a severe form of the collapsing variant of FSGS, termed HIV-associated nephropathy
1596
Q

What are the morphological features of HIV-associated nephropathy?

A
  • A high frequency of the collapsing variant of FSGS
  • A striking focal cystic dilation of tubule segments, which are filled with proteinaceous material, and inflammation and fibrosis
  • The presence of large numbers of tubuloreticular inclusions within endothelial cells, detected by electron microscopy. Such inclusions, also present in SLE, have been shown to be modifications of endoplasmic reticulum induced by circulating interferon-α. They are not usually present in idiopathic FSGS and therefore may have diagnostic value in a biopsy specimen
1597
Q

Membranoproliferative glomerulonephritis is best considered a pattern of immune-mediated injury rather than a specific disease. An emerging consensus on classification separates one group of disorders into two groups. What are they and how are they different?

A
  • Type 1 is characterised by deposition of immune complexes containing IgG and complement
  • Type 2 (often called dense deposit disease) in which activation of complement appears to be the most important factor. This belongs to a group of disorders called C3 glomerulopathies
1598
Q

What is membranoproliferative glomerulonephritis histological characterisation?

A

It is characterised histologically by alterations of the glomerular basement membrane, proliferation of glomerular cells, leukocyte infiltration, and the presence of deposits in mesangial regions and glomerular capillary walls

1599
Q

How are type 1 and type 2 membranoproliferative glomerulonephritis different?

A
  • The deposits are made up of immune complexes in type 1 MPGN and some unknown material in type 2 MPGN.
  • In type II, C3 is present on the GBM but not in the dense deposits
  • In both types, because the proliferation is predominantly in the mesangium but also may involve the capillary loops, a frequently used synonym is mesangiocapillary glomerulonephritis
1600
Q

What is the pathogenesis of type 1 membranoproliferative glomerulonephritis?

A
  • There is evidence of immune complexes in the glomerulus and activation of both classical and alternative complement pathways
  • The antigens involved in MPGN are unknown
  • In many cases they are believed to be proteins derived from infectious agents such as hepatitis C and B viruses, which presumably behave either as “planted” antigens after first binding to or becoming trapped within glomerular structures or are contained in preformed immune complexes deposited from the circulation
1601
Q

What is the morphology of membranoproliferative glomerulonephritis?

A
  • The glomeruli are large and hypercellular. The hypercellularity is produced both by proliferation of cells in the mesangium and so-called endocapillary proliferation involving capillary endothelium and infiltrating leukocytes
  • The glomeruli have an accentuated “lobular” appearance due to the proliferating mesangial cells and increased mesangial matrix.
  • The GBM is thickened, and often shows a “double-contour” or “tram-track” appearance, especially evident in silver or PAS stains. The is caused by “duplication” of the basement membrane, usually as a result of new basement membrane synthesis in response to subendothelial deposits of immune complexes.
  • Between the duplicated basement membranes there is inclusion or interposition of cellular elements, which can be of mesangial, endothelial, or leukocytic origin. Such interposition also gives rise to the appearance of “split” basement membranes. Crescents are present in many cases
1602
Q

What morphology is characteristic of type 1 membranoproliferative glomerulonephritis?

A
  • It is characterised by the presence of discrete subendothelial electron-dense deposits
  • Mesangial and occasional subepithelial deposits may also be present
  • By immunofluorescence, IgG and C3 are deposited in a granular pattern, and early complement components (C1q and C4) are often also present indicative of an immune complex pathogenesis
1603
Q

What are the clinical features of membranoproliferative glomerulonephritis?

A
  • Most patients with primary membranoproliferative glomerulonephritis present in adolescence or as young adults with nephrotic syndrome and a nephritic component manifested by haematuria or, more insidiously, as mild proteinuria.
  • Few remissions occur spontaneously in either type, and the disease follows a slowly progressive but unremitting course
  • Some patients develop numerous crescents and a clinical picture of RPGN.
  • About 50% develop chronic renal failure within 10 years
  • Treatment with steroids, immunosuppressive agents and anti platelet drugs have not proven to be of any benefit
1604
Q

Secondary membranoproliferative glomerulonephritis invariably happens in type 1 MPGN and is more common in adults. What are the settings in which it occurs?

A
  • Chronic immune complex disorders, such as SLE; hep B; hep C, usually with cryoglobulinaemia; endocarditis; infected ventriculoatrial shunts; chronic visceral abscesses; HIV; schistosomiasis
  • α1-Antitrypsin deficiency
  • Malignant diseases, particularly lymphoid tumours such as CLL, which are commonly complicated by development of autoantibodies
1605
Q

What are the abnormalities that cause dense deposit disease?

A
  • Most patients with dense-deposit disease (formerly called type 2 membranoproliferative glomerulonephritis) have abnormalities resulting in excessive activation of the alternative complement pathway.
  • These patients have a consistently decreased serum C3 but normal C1 and C4, the early components of complement
  • They also have diminished serum levels of Factor B and properdin, components of the alternative complement pathway.
1606
Q

How does the diminished complement in dense deposit disease contribute to the pathogenesis?

A
  • In the alternative complement pathway, C3 is directly cleaved to C3b.
  • The reaction depends on the initial activation of C3 by such substances as bacterial polysaccharides, endotoxin, and aggregates of IgA via a pathway involving Factors B and D.
  • This leads to the generation of C3bBb, the alternative pathway C3 convertase.
  • Normally, this C3 converts is labile, but more than 70% of patients with dense-deposit disease have a circulating autoantibody termed C3 nephritic factor (C3NeF) that binds the alternative pathway C3 convertase and protects it from inactivation
  • This favours persistent C3 activation and hypocomplementaemia
  • There is also decreased C3 synthesis by the liver, further contributing to the profound hypocomplementaemia.
1607
Q

While some cases of dense deposit disease share histologic features with MPGN, there is a wider spectrum of histologic alterations in dense deposit disease. What are some of these alterations?

A
  • Many cases have a predominantly mesangial proliferative pattern of injury, while others have an inflammatory and focally crescentic appearance
  • In some cases, dense deposits of a cellular material can be seen permeating the glomerular basement membranes in histologic sections
1608
Q

What is the defining feature of dense deposit disease by electron microscopy?

A

Permeation of the lamina densa of the GBM by a ribbon-like homogenous, extremely electron-dense material of unknown composition

1609
Q

What are the morphological features of dense deposit disease by immunofluorescence? How is this different from other C3 glomerulopathies?

A
  • C3 is present in irregular granular or linear foci in the basement membranes on either side but not within the dense deposits
  • C3 is also present in the mesangium in characteristic circular aggregates (mesangial rings)
  • IgG is usually absent, as are the components of the classical pathway of complement activation (such as C1q and C4)
  • C3 glomerulopathies other than dense deposit disease can have a similar distribution, with mesangial and capillary wall involvement, but lack the extremely electron dense deposits that define dense deposit disease
1610
Q

What are the clinical features of dense deposit disease? Who does it affect? What is the prognosis?

A
  • Dense deposit disease primarily affects children and young adults
  • The clinical presentation of nephritic syndrome with haematuria and/or nephrotic syndrome with proteinuria overlaps with that of MPGN
  • The prognosis is poor, with about half of these patients progressing to end-stage renal disease
  • There is a high incidence of recurrence in transplant recipients; dense deposits may recur in 90% of such patients, although renal failure in the allograft is much less common
1611
Q

What is IgA nephropathy? How do you diagnose it?

A
  • IgA nephropathy, characterised by the presence of prominent IgA deposits in the mesangial regions and recurrent haematuria, is the most common type of glomerulonephritis worldwide
  • The disease can by suspected by light microscopic examination, but the diagnosis is made only by the detection of glomerular IgA deposition.
  • Mild proteinuria is usually present, and the nephrotic syndrome may occasionally develop. Rarely, patients may present with crescentic RPGN.
1612
Q

How is IgA nephropathy linked with Henoch-Schonlein purpura?

A

Where IgA nephropathy is typically an isolated renal disease, similar IgA deposits are present in HSP, which have many overlapping features with IgA nephropathy.

1613
Q

What is IgA and what form of it is increased in IgA nephropathy?

A
  • IgA, the main Ig in mucosal secretions, is present in plasma at low concentrations, mostly in monomeric form, the polymeric forms being catabolised in the liver
  • In patient with IgA nephropathy, levels of plasma polymeric IgA are increased, but increased production is not sufficient to cause this disease.
  • The glomerular deposits consist predominantly of polymeric IgA molecules with aberrant glycosylation.
1614
Q

It is believed that a key facet of IgA nephropathy is a hereditary or acquired defect in the normal formation of what? Why is this important?

A
  • A defect in the normal formation or attachment of galactose-containing sugar chains called O-linked glycans to the hinge region of the IgA molecule (particularly to those of the IgA1 subclass) prior to their secretion by B cells.
  • This aberrantly glycosylated IgA1 is either deposited by itself in glomeruli or it elicits an autoimmune response and forms immune complexes in the circulation with IgG autoantibodies directed against abnormal IgA molecules.
  • The immune complexes are deposited in the mesangium with subsequent formation of immune complexes in situ.
1615
Q

What do the immune complex deposits in IgA nephropathy cause?

A
  • The mesangial immune deposits activate mesangial cells to proliferate, produce increased amounts of extracellular matrix, and secrete numerous cytokines and growth factors
  • These secreted mediators may not only participate in further mesangial cell activation but may also recruit inflammatory cells into the glomeruli
  • The recruited leukocytes contribute to the glomerular injury and also to a reparative response, which can include opsonisation and removal of the immune complexes.
  • The deposited IgA and IgA-containing immune complexes active the complement system via the alternate pathway, and hence the presence of C3 and the absence of C1q and C4 in glomeruli are typical of this disorder.
1616
Q

Is there a genetic influence to IgA nephropathy?

A
  • A genetic influence is suggested by the occurrence of this condition in families and in HLA-identical siblings
  • The increased frequency of certain HLA and complement genotypes in some populations, and the findings of genome wide association studies linking specific MHC Class II loci to disease susceptibility
1617
Q

What are the triggers of IgA nephropathy?

A
  • Epidemiological features of this disorder indicate that the increased synthesis of abnormal IgA may occur in response to respiratory or GI exposure to environmental agents (eg, viruses, bacteria, food proteins).
  • The specific initiating antigens are unknown, and several infectious agents and food products have been implicated
  • IgA nephropathy occurs with increased frequency in individuals with gluten enteropathy (coeliac disease), in whom intestinal mucosal defects are well defined, and in liver disease, in which there is defective hepatobiliary clearance of IgA complexes (secondary IgA nephropathy)
1618
Q

What are the histologic features of IgA nephropathy?

A
  • On histologic examination the lesions vary considerably.
  • The glomeruli may be normal or show mesangial widening and endocapillary proliferation (mesangioproliferative glomerulonephritis), segmental proliferation confined to some glomeruli (focal proliferative glomerulonephritis), or rarely, overt crescentic glomerulonephritis
  • The presence of leukocytes within glomerular capillaries is a variable feature
  • The mesangial widening may be the result of cell proliferation, accumulation of matrix, immune deposits of some combination of these abnormalities
  • Healing of the focal proliferative lesion may lead to secondary focal segmental sclerosis
1619
Q

What are the immunofluorescent and electron microscopy findings of IgA nephropathy?

A
  • The characteristic immunofluorescent picture is often of mesangial deposition of IgA, often with Cr and properdin and lesser amounts of IgG and IgM. Early complement components are usually absent
  • Electron microscopy confirms the presence of electron-dense deposits predominantly in the mesangium; capillary wall deposits, if present, are usually sparse
1620
Q

What are the clinical symptoms of IgA nephropathy? Who does it usually affect?

A
  • The disease affects people of any age, most commonly older children and young adults
  • Many patients present with gross haematuria after an infection of the respiratory or, less commonly, GI or GU tract; 30-40% have only microscopic haematuria, with or without proteinuria; and 5-10% develop acute nephritic syndrome, including some with rapidly progressive glomerulonephritis.
1621
Q

What is the clinical course of IgA nephropathy? What is linked with increased risk of progression?

A
  • The haematuria typically lasts for several days and then subsides, only to return every few months.
  • The subsequent course is highly variable. Many patients maintain normal renal function for decades
  • Slow progression to chronic renal failure occurs in 15-40% of cases over a period of 20 years
  • Onset in old age, heavy proteinuria, HTN, and the extent of glomerulosclerosis on biopsy are clues to an increase risk of progression
  • Recurrence of IgA deposits in transplanted kidneys is frequent, and in approximately 15% of those with recurrent IgA deposits, the disease runs the same slowly progressive course as that of primary IgA nephropathy
1622
Q

What is hereditary nephritis? What are two important examples?

A
  • Hereditary nephritis refers to a group of heterogenous familial renal diseases associated with mutations in collagen genes that manifest primarily with glomerular injury
  • Alport syndrome and thin basement membrane lesion are two examples
1623
Q

What is Alport syndrome?

A

Alport syndrome, when fully developed, is manifest by haematuria with progression to chronic renal failure, accompanied by nerve deafness and various eye disorders, including lens dislocation, posterior cataracts and corneal dystrophy.

1624
Q

How is Alport syndrome inherited?

A
  • The disease is inherited as an X-linked trait in approximately 85% of cases
  • In the X-linked form, males express the full syndrome, while female heterozygous typically present with haematuria
  • Autosomal recessive and autosomal dominant pedigrees also exist, in which males and females are equally susceptible to the full syndrome
1625
Q

What is the pathogenesis of Alport syndrome?

A
  • The disease manifestations are due to mutations in one of several genes coding for subunits of the collagen IV molecule
  • More than 500 mutations resulting in disease have been identified, resulting in defective assembly of type IV collagen, which is crucial for function of the GBM, the lens of the eye, and the cochlea.
  • Because the GBM consists of networks of trimeric collagen IV molecules composed of α3, α4 and α5 chains, mutations affecting any one chain result in defective assembly of the collagen network
  • Since type IV collagen chains are encoded on autosomes (chromosomes 2 and 13) and the X-chromosome, the inheritance can be autosomal or X-linked.
1626
Q

What are the electron microscopic features of Alport syndrome?

A
  • The GBM shows irregular foci of thickening alternating with attenuation (thinning), and pronounced splitting and lamination of the lamina dense, often producing a distinctive basket-weave appearance.
  • Similar alterations can be found in the tubular basement membrane
1627
Q

What are the immunohistochemistry findings in Alport syndrome?

A
  • Immunohistochemistry can be helpful in cases with absent or borderline basement membrane lesions, because antibodies to α3, α4 and α5 collagen fail to stain both glomerular and tubular basement membranes in the classic X-linked form.
  • There is also absence of α5 staining in skin biopsy specimens from these patients.
  • As the disease progresses there is development of focal segmental and global glomerulosclerosis and other changes of progressive renal injury, including vascular sclerosis, tubular atrophy and interstitial fibrosis
1628
Q

What are the clinical features and prognosis of Alport syndrome?

A
  • The most common presenting sign is gross or microscopic haematuria, frequently accompanied by red cell casts.
  • Proteinuria may develop later, and rarely, the nephrotic syndrome develops
  • Symptoms appear at ages 5 to 20 years, and the onset of overt renal failure is between ages 20 and 50 years in men
  • The auditory defects may be subtle, requiring sensitive testing
  • Approximately 90% of affected males progress to ESRD before 40 years of age
1629
Q

What is thin basement membrane lesion (benign familial haematuria)?

A
  • This is a fair common hereditary entity manifested clinically by familial asymptomatic haematuria and morphologically by diffuse thinning of the GBM to widths between 150 and 225nm (compared to 300-400nm in healthy adults).
  • Although mild or moderate proteinuria may also be present, renal function is normal and prognosis is excellent.
1630
Q

What is the pathogenesis and genetic influence in thin membrane basement lesion?

A
  • The anomaly in thin basement membrane lesion has been traced to mutations in genes encoding α3 or α4 chains of type IV collagen.
  • The disease most often has an autosomal inheritance and most patients are heterozygous for the defective gene
1631
Q

What is chronic glomerulonephritis?

A

Chronic glomerulonephritis refers to end-stage glomerular disease that may result from specific types of glomerulonephritis or may develop without antecedent history of any of the well-recognised forms of acute glomerulonephritis

1632
Q

What types of acute glomerulonephritis are associated with chronic glomerulonephritis?

A
  • Post-streptococcal glomerulonephritis is a rare antecedent of chronic glomerulonephritis, except in adults
  • Patiens with crescentic glomerulonephritis, if they survive the acute episode, usually progress sot chronic glomerulonephritis
  • Membranous nephropthy, membranoproliferative glomerulonephritis, IgA nephropathy and focal segmental glomerulosclerosis all may progress to chronic renal failure
1633
Q

What are the morphological features of chronic glomerulonephritis?

A
  • The kidneys are symmetrically contracted and have diffusely granular cortical surfaces
  • On section, the cortex is thinned, and there is an increase in peripelvic fat.
  • The glomerular histology depends on the stage of the disease. In early cases, the glomeruli may still show evidence of the primary disease.
  • However, there eventually ensues obliteration of the glomeruli, transforming them into cellular eosinophilic masses, representing a combination of trapped plasma proteins, increased mesangial matrix, basement membrane-like material, and collagen
  • Because HTN is an accompaniment of chronic glomerulonephritis, arterial and arteriolar sclerosis may be conspicuous.
  • Marked atrophy of associated tubules, irregular interstitial fibrosis and mononuclear leukocytic infiltration of the interstitium also occurs
1634
Q

How does chronic glomerulonephritis often present?

A
  • Patients present with non-specific complaints such as loss of appetite, anaemia, vomiting or weakness.
  • In some, renal disease is suspected with the discovery of proteinuria, HTN, or azotemia
  • In others, the underlying renal disorder is discovered in the course of investigation of oedema
  • Most patients are hypertensive, and sometimes the dominant clinical manifestation relate to cerebral or cardiovascular disease.
1635
Q

How does chronic glomerulonephritis progress?

A
  • In most individuals, chronic glomerulonephritis develops insidiously and slowly progresses to renal insufficiency or death from uraemia during a span or years or possibly decades
  • In all, the disease is relentlessly progressive, though at widely varying rates
  • If patients with chronic glomerulonephritis do not receive dialysis or if they do not receive a renal transplant, they invariably succumb to their disease.
1636
Q

Many immunologically mediated, metabolic or hereditary systemic disorders are associated with glomerular injury; in some (eg, SLE and diabetes), the glomerular involvement is a major clinical manifestation. What are some of these disorders?

A
  • Lupus nephritis
  • Henoch-schonlein purpura
  • Bacterial endocarditis
  • Diabetic nephropathy
  • Fibrillary glomerulonephritis
  • Goodpasture syndrome
  • Microscopic polyangiitis
  • Granulomatosis with polyangiitis
1637
Q

SLE gives rise to a wide variety of renal lesions and clinical presentations. What are some of them?

A
  • Recurrent microscopic or gross haematuria
  • Nephritic syndrome
  • Rapidly progressive glomerulonephritis
  • Nephrotic syndrome
  • Acute and chronic renal failure
  • HTN
1638
Q

What are the clinical features of Henoch-Schonlein purpura?

A
  • This childhood syndrome consist of purpuric skin lesions, abdominal pain and intestinal bleeding, and arthralgias along with renal abnormalities
  • Skin lesions characteristically involve the extensor surfaces of arms and legs as well as buttocks
  • Abdominal manifestations include pain, vomiting and intestinal bleeding
  • The renal manifestations occur in 1/3rd of patients and include gross or microscopic haematuria, nephritic syndrome, nephrotic syndrome, or some combination of these
1639
Q

Can patients with HSP develop rapidly progressive glomerulonephritis? Who is this most common in?

A

A small number of patients, mostly adults, develop a rapidly progressive form of glomerulonephritis with many crescents

1640
Q

Do you need all components of HSP to have a diagnosis?

A

No, not all components of the syndrome need to be present for the diagnosis and individual patients may have purpura, abdominal pain or urinary abnormalities as the dominant feature

1641
Q

Who is HSP most common in? Both age and characteristics that increase likelihood?

A
  • The disease is most common in children 3-8 years old, but it also occurs in adults, in whom, the renal manifestations are usually more severe
  • There is a strong background of atopy in about 1/3rd of patients, and onset often follows an URTI
1642
Q

What is the pathogenesis of HSP?

A
  • IgA is deposited in the glomerular mesangium, in a distribution similar to that of IgA nephropathy
  • This has led to the concept that IgA nephropathy and HSP are manifestations of the sam disease
  • The finding of Ig and C3 deposits in the glomeruli suggests that immune complexes are involved in the disease
1643
Q

What is the morphology of HSP?

A
  • On histologic examination, the renal lesions vary from mild focal mesangial proliferation to diffuse mesangial proliferation and/or endocapillary proliferation to crescentic glomerulonephritis
  • Whatever the histologic lesions, the pathognomic feature by fluorescence microscopy is the deposition of IgA, sometimes with IgG and C3, in the mesangial region, sometimes with deposits extending to the capillary loops
  • The skin lesions consist of subepidermal haemorrhages and a necrotising vasculitis involvement the small vessels of the dermis.
  • Deposits of IgA along with IgG and C3, are also present in such vessels. Vasculitis also occurs in other organs, such as the GI tract, but is rare in the kidney
1644
Q

What is the pathogenesis of glomerulonephritis associated with bacterial endocarditis and other systemic infections? How do they present?

A
  • The lesions represent a type of immune complex nephritis initiated by complexes of bacterial antigen and antibody.
  • Haemturia and proteinuria of various degrees characterise this entity clinically, but an acute nephritic presentation is not uncommon, and even rapidly progressive glomerulonephritis may occur in rare instances.
1645
Q

What are the morphological features of glomerulonephritis associated with bacterial endocarditis and other systemic infections?

A
  • The histologic lesions may vary from a focal and segmental necrotising glomerulonephritis to a diffuse and more global exudative and proliferative glomerulonephritis, which may have a MPGN pattern.
  • More severe forms show a diffuse proliferative glomerulonephritis.
  • the lesions may be acute (influx of neutrophils) or chronic (fully developed MPGN pattern with basement membrane changes); the rapidly progressive forms show large numbers of crescents.
  • Immunofluorescence and electron microscopy show the presence of glomerular immune deposits
1646
Q

What is fibrillary glomerulonephritis?

A
  • It is a morphologic variant of glomerulonephritis, in which patients develop nephrotic syndrome, haematuria and progressive renal sufficiency.
  • The pathogenesis of this entity is unknown
  • The disease recurs in kidney transplants
1647
Q

What is the morphologic variant that is characteristic of fibrillary glomerulonephritis?

A
  • It is associated with characteristic fibrillar deposits in the mesangium and glomerular capillary walls that resemble amyloid fibrils superficially but differ ultrastructurally and do not stain with Congo red.
  • The glomerular lesions usually show membranoproliferative or mesangioproliferative patterns by light microscopy
  • By immunofluorescence microscopy, there is selective deposition of polyclonal IgG, often of the OgG4 subclass, complement C3, Igκ and Igλ light chains.
1648
Q

Goodpasture syndrome, microscopic polyangiitis, and granulomatosis with polyangiitis are commonly associated with glomerular lesions that can be histologically similar. What are these histological features?

A
  • They are principally characterised by foci of glomerular necrosis and crescent formation.
  • In the early or mild forms of renal involvement, there is focal and segmental, sometimes necrotising glomerulonephritis, and most of these patients will have haematuria with mild decline in GFR
  • In more severe causes, with may be associated with RPGN, there is more extensive necrosis, fibrin deposition, and extensive formation of epithelial (cellular) crescents, which can become organised to form fibrocellular and fibrous crescents if the glomerular injury evolves into segmental or global scarring (sclerosis)
1649
Q

How does essential mixed cryoglobulinaemia cause glomerulonephritis?

A
  • Deposits of cryoglobulins composed principally of IgG-IgM complexes induce cutaneous vasculitis, synovitis and a proliferative glomerulonephritis, typically membranoproliferative glomerulonephritis.
  • Most causes of essential mixed cryoglobulinaemia have been associated with infection with hep C, and this condition in particular is associated with glomerulonephritis, usually MPGN type 1
1650
Q

What is acute tubular injury?

A

A clinicopathologic entity characterised clinically by acute renal failure and often, but no invariably, morphologic evidence of tubular injury, int he form of necrosis of tubular epithelial cells

1651
Q

What can cause acute tubular injury?

A
  • Ischaemia, due to decreased or interrupted blood flow, examples of which include diffuse involvement of the intrarenal blood vessels such as in microscopic polyangiitis, malignant hypertension, microangiopathies and systemic conditions associated with thrombosis (DIC, TTP), or decreased effective circulating blood volume
  • **direct toxic injury to the tubules by endogenous (eg, myoglobin, Hb, monoclonal light chains, bile/bilirubin) or exogenous agents (eg, drugs, radiocontrast dyes, heavy metals)
1652
Q

What are the two patterns of acute tubular injury?

A
  • Ischaemic ATI caused by inadequate blood flow to the peripheral organs, usually accompanied by marked hypotension and shock.
  • Nephrotoxic ATI is caused by a multitude of durgs, such as gentamicin; radiographic contract agents; poisons, including heavy metals (eg, mercury) and organic solvents
  • Combinations of ischaemic and nephrotoxic ATI can also occur, exemplified by mismatched blood transfusions and other haemolytic crises caused haemoglobinuria and skeletal muscle injuries causing myoglobulinuria
1653
Q

What are the critical events in the pathogenesis of both ischaemic and nephrotoxic acute tubular injury?

A

1) tubular injury
2) persistent and severe disturbance in blood flow

1654
Q

Tubular epithelial cells are particularly sensitive to ischaemia and vulnerable to toxins. Several factors predispose the tubules to toxic injury. What are they?

A
  • Increased surface area for tubular reabsorption
  • Active transport systems for ions and organic acids
  • A high rate of metabolism and oxygen consumption that is required to perform these transport and reabsorption functions, and the capability for resorption and concentration of toxins
1655
Q

Ischaemia causes numerous structural and functional alterations in epithelial cells. What is an early reversible result of ischaemic in the tubules?

A
  • One early reversible result of ischaemia is loss of cell polarity due to redistribution of membrane proteins (eg, the enzyme Na/K/ATPase) from the basolateral to the luminal surface of the tubular cells, resulting in abnormal ion transport across the cells and increased sodium delivery to distal tubules
  • The latter incites vasoconstriction via tubuloglomerular feedback
  • In addition, ischaemic tubular cells express cytokines and adhesion molecules, thus recruiting leukocytes that appear to participate in the subsequent injury
1656
Q

Over time , what happens to the tubular cells following ischaemia?

A
  • In time, injured cells detach from the basements membranes and cause luminal obstruction, increased intratubular pressure, and decreased GFR.
  • In addition, glomerular filtrate in the lumen of the damaged tubules can leak back into the interstitium, resulting in interstitial oedema, increased interstitial pressure, and further damage to the tubule.
1657
Q

How do disturbances in blood flow cause acute tubular injury?

A
  • Ischaemic renal injury is characterised by haemodynamic alterations that cause reduced GFR.
  • The major one is intrarenal vasoconstriction, which results in both reduced glomerular blood flow and reduced oxygen delivery to the functionally important tubules in the outer medulla (thick ascending limb and straight segment of the proximal tubule)
1658
Q

Several vasoconstrictor pathways have been implicated in the intrarenal vasoconstriction that takes place in acute tubular injury. What are they?

A
  • The renin-angiotensin system, stimulated by increased distal sodium delivery (via tubuloglomerular feedback), * Sublethal endothelial injury, leading to increased release of the vasoconstrictor endothelin and decreased production of the vasodilators nitric oxide and prostacyclin.
1659
Q

Can recovery or renal function happen following acute tubular injury? Why?

A
  • The patchiness of tubular necrosis and maintenance of the integrity of the of the basement membrane along many segments allow repair of the injured foci and recovery of function if the precipitating cause is removed
  • This repair is dependent on the capacity of reversibly injured epithelial cells to proliferate and differentiate.
  • Re-epithelialisation is mediated by a variety of growth factors and cytokines produced locally but he tubular cells themselves or by inflammatory cells in the vicinity of necrotic foci.
1660
Q

What are the morphological characteristics of acute tubular injury?

A
  • ATI is characterised by focal tubular epithelial necrosis at multiple points along the nephron, with large skip areas in between, often accompanied by rupture of basement membranes (tubulorrhexis) and occlusion of tubular lumens by casts
1661
Q

Which part of the nephron is particularly susceptible to morphological changes of acute tubular injury? Do these correlate with clinical manifestations?

A
  • The straight portion of the proximal tubule and the thick ascending limb in the renal medulla are especially vulnerable, but focal lesions may also occur in the distal tubule, often in conjunction with casts.
  • It should be noted that the severity of the morphological findings often do not correlate well with the severity of the clinical manifestations.
1662
Q

What are the histological changes in ischaemic acute tubular injury?

A
  • Eosinophilic hyaline cases, as well as pigmented granular casts, are common, particularly in distal tubules and collections tubules
  • These casts consist principally of Tamm-Horsfall protein (a urinary glycoprotein normally secreted by the cells of ascending thick limb and distal tubules) in conjunction with other plasma proteins
  • Other findings in ischaemic ATI are interstitial oedema and accumulations of leukocytes within dilated vasa recta.
  • There is also evidence of epithelial regeneration in the form of flattened epithelial cells with hyper chromatic nuclei and mitotic figures. In the course of time this regeneration repopulates the tubules so that no residual evidence of damage is seen
1663
Q

What is the histological changes in toxic acute tubular injury?

A
  • Toxic ATI is manifested by acute tubular injury, most obvious in the proximal convoluted tubules
  • On histologic examination the tubular necrosis may be non-specific, but it is somewhat distinctive in poisoning with certain agents.
  • With mercuric chloride, severely injured cells may contain large acidophilic inclusions. Later, these cells become necrotic, are desquamated into the lumen, and may undergo calcification
  • Carbon tetrachloride poisoning, in contrast, is characterised by the accumulation of neutral lipids in injured cells; again, such fatty change is followed by necrosis
1664
Q

The clinical course of acute tubular necrosis is highly variable, but the classic case may be divided into three stages. What are they and what happens during them?

A
  • Initiation phase, lasting about 36 hours, is dominated by the inciting medical, surgical, or obstetric event. The only indication of renal involvement is a slight decline in urine output with a rise in BUN.
  • Maintenance phase is characterised by sustained decreases in ruin output to between 40 and 400ml/day, salt and water overload, rising BUN concentrations, hyperkalaemia, metabolic acidosis and other manifestations of uraemia.
  • Recovery phase is ushered in by a steady increase in urine volume that may reach up to 3L/day. The tubules are still damaged to large amounts of water, sodium and potassium are lost in the flood on urine. Eventually, renal tubular function is restored and concentrating ability improves.
1665
Q

How does the potassium change throughout the phases of acute tubular injury?

A
  • As the GFR drops, the potassium is elevated, as can happen in renal failure
  • In the recovery phase, the tubules are still damaged so large amount of water, sodium and potassium are lost on the flood of urine so hypokalaemia becomes a clinical problem
1666
Q

How long is the recovery of acute tubular injury?

A

Subtle functional impairment may persist for months, but most patients who reach the recovery phase eventually recover completely

1667
Q

What is the prognosis of acute tubular injury?

A
  • The prognosis of ATI depends on the magnitude and duration of injury
  • Recovery is expected with nephrotoxic ATI when the toxin has not caused serious damage to other organs, such as the liver or heart
  • With current supportive care, 95% of those who do not succumb to the precipitating cause recover
  • Conversely, in shock related to sepsis, extensive burns, or other causes of multi-organ failure, the mortality rate can be more than 50%
1668
Q

What is tubulointerstitial nephritis?

A

This group of renal diseases involves inflammatory injuries of the tubules and interstitium that are often insidious in onset and are principally manifested by azotemia

1669
Q

What are some of the causes of tubulointerstitial nephritis?

A

Infections - acute bacterial pyelonephritis, chronic pyelonephritis
Toxins - drugs, acute-hypersensitivity interstitial nephritis, analgesics, heavy metals
Metabolic disease - rate nephropathy, nephrocalcinosis, acute phosphate nephropathy, hypokalaemic nephropathy
Physical factors - chronic urinary tract obstruction
Neoplasma - multiple myeloma
immunologic reactions - transplant rejection, Sjogren syndrome, sarcoidosis
Vascular diseases
Miscellaneous - Balkan nephropathy, “idiopathic” interstitial nephritis

1670
Q

Tubulointerstitial nephritis can be acute or chronic. How do these present differently clinically and on a cellular level?

A
  • Acute tubulointerstitial nephritis has a rapid clinical onset and is characterised histologically by interstitial oedema, often accompanied by leukocytic infiltration of the interstitium and tubules, and tubular injury
  • In chronic interstitial nephritis there is infiltration wit h predominancy mononuclear leukocytes, prominent interstitial fibrosis, and wide-spread tubular atrophy
1671
Q

What are some morphological features that are helpful in separating acute from chronic tubulointerstitial nephritis?

A

They include oedema and, when present, eosinophils and neutrophils in the acute form, while fibrosis and tubular atrophy characterise the chronic form

1672
Q

How would you clinically differentiate between glomerulonephritis and tubulointerstitial nephritis?

A
  • Absence of nephritic or nephrotic syndrome
  • The presence of defects in tubular function. The latter may be subtle and include impaired ability to concentrate urine, evidenced clinically by polyuria or nocturia; salt wasting; diminished ability to excrete acids (metabolic acidosis); and isolate defects in tubular reabsorption or secretion
1673
Q

What is pyelonephritis?

A

Pyelonephritis is one of the most common diseases of the kidney and is defined as inflammation affecting the tubules, interstitium, and renal pelvis

1674
Q

Pyelonephritis can be acute or chronic. What are the causes of both of these?

A
  • Acute - generally caused by bacterial infection and is associated with UTI
  • Chronic - a more complex disorder; bacterial infection plays a dominant role, but other factors (vesico-ureteral reflux, obstruction) predispose to repeat episodes of acute pyelonephritis
1675
Q

What is the difference between pyelonephritis and UTI?

A
  • Pyelonephritis is a serious complication of UTIs that affect the bladder (cystitis), the kidneys and their collecting systems (pyelonephritis), or both
  • Bacterial infections of the lower urinary tact may be asymptomatic and often remain localised to the bladder without development of renal infection
1676
Q

What bacteria can cause UTIs and pyelo? What about in immunocompromised people?

A
  • More than 85% of cases of UTI are caused by the gram-negative bacilli that are normal inhabitants of the intestinal tract
  • For most UTIs, the infecting organisms are derived from the patient’s own faecal flora
  • By far, the most common is E coli, followed by Proteus, Klebsiella, and Enterobacter.
  • Streptococcus faecalis, also of enteric origin, staphylococci, and virtually every other bacterial and fungal agent can also cause lower urinary tract and renal infections
  • Mycobacterial and fungal organisms induce caveating and non-caveating granulomatous inflammation, respectively.
  • In immunocompromised people, particularly those with transplanted organs, viruses such as polymavirus, CMV and adenovirus can also be a cause of renal infection
1677
Q

There are two routes by which bacteria can reach the kidneys. What are they?

A

1) through the bloodstream (haematogenous infection)
2) from the lower urinary tract (ascending infection)

1678
Q

Pyelonephritis caused by haematogenous spread is less common that from a UTI. How does it happen? In what diseases and what patients?

A
  • It results from seeding of the kidneys by bacteria from distant foci in the course of septicaemia or localised infections such as infective endocarditis
  • Haematogenous infections is more likely to occur in the presence of ureteral obstruction, and in debilitated patients
  • Typically, in patients receiving immunosuppressive therapy, non-enteric organisms, such as staphylococci and certain fungi and viruses, are involved
1679
Q

Ascending infection is the most common cause of clinical pyelonephritis. Normal human ladder and bladder urine are sterile. What steps must occur for the bacteria to spread into the urethra?

A
  • The first step in ascending infection is the colonisation of the distal urethra and introitus by coliform bacteria.
  • This colonisation is influenced by the degree of bacterial adherence to urethral mucosal epithelial, which involves adhesive molecules (adhesins) on the P-fimbriae (pili) of bacteria that interact with receptors on the surface of urothelial cells.
  • Specific adhesins (those encoded by the pyelonephritis-associated pili [pap] gene) are associated with infection.
  • In addition, certain type of fimbriae promote renal tropism, persistence of infection, or an enhanced inflammatory response
1680
Q

What are the steps involved in the bacteria moving from the urethra to the bladder in a UTI? Why is this more common in women?

A
  • Organisms gain entrance during urethral catheterisation, or other instrumentation
  • Long-term catheterisation, in particular, carries a risk of infection
  • In the absence of instrumentation, urinary infections are much more common in females, this has been ascribed to the shorter urethra in females, as well as the absence of antibacterial properties found in prostatic fluid, hormones changes affecting adherence of bacteria to the mucosa, and urethral trauma during sexual intercourse, or a combination of these factors
1681
Q

What are the mechanisms by which microbes more from the bladder to the kidneys in pyelonephritis?

A
  • Urinary tract obstruction and stasis of urine
  • Vesicoureteral reflux
  • Intrarenal reflux
1682
Q

How does urinary tract obstruction and stasis or urine aid bacterial spread from the bladder to the kidneys?

A
  • Ordinarily, organisms introduced into the bladder are cleared by continual voiding and by antibacterial mechanisms
  • However, outflow obstruction or bladder dysfunction results in incomplete emptying and residual urine
  • In the presence of stasis, bacteria introduced into the bladder can multiply unhindered
  • Accordingly, urinary tract infection is frequent among patients with lower urinary tract obstruction, such as may occur with benign prostatic hypertrophy, tumours, or calculi, or with neurogenic bladder dysfunction caused by diabetes or spinal cord injury
1683
Q

How does vesicoureteric reflux aid in the bacterial movement from the bladder to the kidneys in pyelonephritis?

A
  • Although obstruction is an important predisposing factor in ascending infection, it is incompetence of the vesicoureteral valve that allows bacteria to ascend the ureter into the renal pelvis
  • The normal ureteral insertion into the bladder in a one-way valve that prevents retrograde flow or urine when the intravesical pressure rises, as in micturition.
  • An incompetent vesicoureteral orifice allows the reflux of bladder urine into the ureters (vesicoureteral reflux).
  • The effect of vesicoureteral reflux is similar to that of an obstruction in that there is residual urine in the urinary tract after voiding, which favours bacterial growth
1684
Q

What are some of the causes if vesicoureteric reflux?

A
  • Reflux is most often due to congenital absence or shortening of the intravesical portion of the ureter, such that the ureter is not compressed during micturition
  • In addition, it may be acquired by bladder infection itself. It is postulated that the bacteria themselves or the associated inflammation can promote reflux by affecting ureteral contractility, particularly in children
  • Vesicoureteral reflux is estimated to affect 1-2% of otherwise normal children
  • Acquired vesicoureteral reflux in adults can result from persistent bladder atony cause by spinal cord injury
1685
Q

How does intrarenal reflux aid the movement of bacteria from the bladder to the kidneys in pyelonephritis?

A
  • Vesicoureteral reflux also affords a ready mechanism by which the infected bladder urine can be propelled up to the renal pelvis and deep into the renal parenchyma through open ducts at the tips of the papillae (intrarenal reflux).
  • Intrarenal reflux is most common in the upper and lower poles of the kidney, where papillae tend to have flattened or concave tips rather than the convex pointed type present in the mid zones of the kidney
1686
Q

How can you radiologically demonstrate intrarenal and vesicoureteral reflux?

A
  • By a voiding cystourethrogram, in which the bladder is filled with a radiopaque dye and films are taken during micturition.
  • Vesicoureteral reflux can be demonstrated by this method in about 30% of infants and children with UTIs
1687
Q

What are the morphologic hallmarks of acute pyelonephritis?

A
  • Patchy interstitial suppurative inflammation, intratubular aggregates of neutrophils, neutrophilic tubulitis and tubular necrosis
  • The suppuration may occur as discrete focal abscesses or large wedge-like areas and can involve one or both kidneys
1688
Q

What are the morphologic changes that happen during early glomerulonephritis?

A
  • In the early stages, the neutrophilic infiltration is limited to the tubules.
  • The tubular lumens are a conduit for the extension of the infection, and soon, the infection extends to the interstitium and produces abscesses that destroy the involved tubules
  • Characteristically, glomeruli are relatively resistant o the infection.
  • Extensive disease, however, eventually also destroys the glomeruli, and fungal pyelonephritis (eg, Candida) often affects glomeruli and results in granulomatous interstitial inflammation
1689
Q

Three complications of acute pyelonephritis can be encountered. What are they are how do they present morphologically?

A
  • Papillary necrosis is seen mainly in diabetics, sickle cell disease, and those with urinary tract obstruction. Papillary necrosis is usually bilateral but may be unilateral. One of all of the pyramids of the affected kidney may be involved. On cut section, the tips or distal two thirds of the pyramids have areas of gray-white examination the necrotic tissue shows characteristic ischaemic coagulative necrosis, with preservation of outlines of tubules. The leukocytic response is limited to the junctions between preserved and destroyed tissue
  • **Pyonephrosis is seen when there is total or almost complete obstruction, particularly when it is high in the urinary tract. The suppurative exudate is unable to drain and thus fills the renal pelvis, calyces, and ureter with pus
  • Perinephric abscess is an extension of suppurative inflammation through the renal capsule into the perinephric tissue
1690
Q

After the acute phase of pyelonephritis, healing occurs. How is this reflected morphologically?

A
  • The neutrophilic infiltrate is replaced by one that is predominantly composed of macrophages, plasma cells, and lymphocytes.
  • The inflammatory foci are eventually replaced by irregular scars that can be seen on the cortical surface as fibrous depressions
  • Such scars are characterised microscopically by tubular atrophy, interstitial fibrosis, and a lymphocytic infiltrate in a characteristic patchy, jigsaw pattern with intervening preserved parenchyma.
  • The pyelonephritis scar is almost always associated with inflammation, fibrosis, and deformation of the underlying calyx and pelvis, reflecting the role of ascending infection and vesicoureteral reflux in the pathogenesis of the disease.
1691
Q

What are some diseases that acute pyelonephritis is often associated with?

A
  • Urinary tract obstruction, either congenital or acquired
  • Instrumentation of the urinary tract, most commonly catheterisation
  • Vesicoureteral reflux
  • Pre-existing renal lesions, causing intrarenal scarring and obstruction
  • Diabetes mellitus, in which increased susceptibility to infection, neurogenic bladder dysfunction, and more frequent instrumentation are pre-disposing factors
  • Immunosuppression and immunodeficiency
1692
Q

What are some populations in which acute pyelonephritis is more common in?

A
  • Pregnancy - between 4-6% of pregnant women develop bacteriuria sometime during pregnancy, and 20-40% of these eventually develop symptomatic UTIs if not treated
  • Gender and age - after the first year of life (when congenital anomalies in males commonly become evident) and up to around age 40 years, infections are much more frequent in females
  • With increasing age the incidence in males rises as a result of prostatic hypertrophy and instrumentation
1693
Q

How does acute pyelonephritis usually present clinically and on FWT?

A
  • It usually presents with a sudden onset of pain at the costovertebral angle and systemic evidence of infection, such as fever and malaise
  • There are often indications of bladder and urethral irritations such as dysuria, frequency, and urgency
  • The urine contains many leukocytes (pyuria) derived from the inflammatory infiltrate, but pyuria does not differentiate from upper and lower tract infections.
  • The finding of leukocyte casts, typically rich in neutrophils (pus casts), indicates renal involvement, because casts are formed only in tubules
1694
Q

How does acute pyelonephritis resolve? What happens if it doesn’t?

A
  • Uncomplicated acute pyelonephritis follows a benign course, and symptoms disappear within a few days after the institution of appropriate antibiotic therapy.
  • Bacteria, however, may persist in the urine, or there may be recurrence of infection with new serologic types of E.coli or other organisms. Such bacteriuria then either disappears or may persist, sometimes for years
  • In the presence of unrelieved urinary obstruction, diabetes mellitus, or immunodeficiency, acute pyelonephritis may be more serious, leading to repeated septicaemia episodes.
  • The superimposition of papillary necrosis may lead to acute renal failure
1695
Q

An emerging viral pathogen causing pyelonephritis in kidney allografts is polyomavirus. Latent infection with polyomavirus is widespread in the general population, and immunosuppression of the allograft recipient can lead to reactivation of latent infection and the development of nephropathy resulting in allograft failure in up to 5% of kidney transplant recipients. What is this called? What is it’s morphology? How do you treat it?

A
  • This form of pyelonephritis, now referred to as polyomavirus nephropathy, is characterised by infection of tubular epithelial cell nuclei, leading to nuclear enlargement and intranuclear inclusions visible by light microscopy (viral cytopathic effect).
  • The inclusions are composed of virions by electron microscopy.
  • An interstitial inflammatory response is invariably present.
  • Treatment consists of a reduction in immunosuppression
1696
Q

What is chronic pyelonephritis? How do you differentiate it from other chronic tubulointerstitial diseases?

A
  • Chronic pyelonephritis is a disorder in which chronic tubulointerstitial inflammation and scarring involve the calyces and pelvis.
  • Although several diseases produce chronic tubulointerstitial alterations, only chronic pyelonephritis and analgesic nephropathy affect the calyces, making pelvocalyceal damage an important diagnostic clue
1697
Q

Chronic pyelonephritis can be divided into two forms. What are they? Which is most common?

A

Reflux nephropathy and chronic obstructive pyelonephritis

Reflux nephropathy is far more common form of chronic pyelonephritic scarring

1698
Q

How does reflux nephropathy cause chronic pyelonephritis?

A
  • Reflux nephropathy occurs early in childhood as a result of superimposition of a urinary infection on congenital vesicoureteral reflux and intrarenal reflux
  • Reflux may be unilateral or bilateral; thus, the continuous renal damage may cause scarring and atrophy of one kidney or involve both, leading to renal insufficiency.
  • Vessicoureteral reflux occasionally causes renal damage in the absence of infection (sterile reflux), but only when obstruction is severe
1699
Q

How does chronic obstructive pyelonephritis cause chronic pyelonephritis?

A
  • Recurrent infections caused by obstruction superimposed on diffuse or localised obstructive lesions lead to repeated bouts of renal inflammation and scarring, resulting in chronic pyelonephritis
  • In this condition, the effects of obstruction contribute to the parenchymal atrophy; indeed, it is sometimes difficult to differentiate the effects of bacterial infection from this of obstruction alone.
  • The disease can be bilateral, as with posterior urethral valves, resulting in renal insufficiency unless the anomaly is corrected, or unilateral, as occurs with calculi and unilateral obstructive anomalies of the ureter
1700
Q

What are the gross morphologic changes in chronic pyelonephritis? How does this compare to chronic glomerulonephritis?

A
  • The kidneys are irregularly scarred; if bilateral, the involvement is asymmetric.
  • In contrast, both kidneys in chronic glomerulonephritis are diffusely and symmetrically scarred.
1701
Q

What are the morphological hallmarks of chronic pyelonephritis?

A
  • Course, discrete, corticomedullary scars overlying dilated, blunted or deformed calyces, and flattening of the papillae.
  • The scars vary from one to several and most are in the upper and lower poles, consistent with the frequency of reflux in these sites
1702
Q

The microscopic changes in chronic pyelonephritis involve predominantly tubules and interstitium. What are these changes?

A
  • The tubules show atrophy in some areas and hypertrophy in diction in others
  • Dilated tubules with flattened epithelium may be filled with casts resembling thyroid colloid (thyroidisation).
  • There are varying degrees of chronic interstitial inflammation and fibrosis in the cortex and medulla.
1703
Q

Apart from in the tubules and interstitium, what are the other microscopic changes in chronic pyelonephritis?

A
  • Arcuate and interlobular vessels demonstrate obliterative intimal sclerosis in the scarred areas; and in the presence of hypertension, hyaline arteriosclerosis is seen in the entire kidney
  • There is often fibrosis around the calyces epithelium as well as a marked chronic inflammatory infiltrate
  • Glomeruli may appears normal except for a variety of ischaemic changes, including peri-glomerular fibrosis, fibrous obliteration and secondary changes related to hypertension
  • Individuals with chronic pyelonephritis and reflux nephropathy who develop proteinuria in advanced stages show secondary focal segmental glomerulosclerosis
1704
Q

What is Xanthogranulomatous pyelonephritis? How does it appear morphologically? What bacteria is it associated with?

A
  • It is a relatively rare form of chronic pyelonephritis characterised by accumulation of foamy macrophages intermingled with plasma cells, lymphocytes, polymorphonuclear leukocytes, and occasional giant cells
  • Often associated with Proteus infections and obstruction, the lesions sometimes produce large, yellowish orange nodules that may be grossly confused with renal cell carcinoma
1705
Q

What are the clinical features of chronic obstructive pyelonephritis?

A
  • It may have a silent onset or present with manifestations of acute recurrent pyelonephritis, such as back pain, fever, pyuria, and bacteriuria
  • These patients receive medical attention relatively late in their disease course because of the gradual onset of renal insufficiency and hypertension
  • Reflux nephropathy is often discovered in children when the cause of hypertension is investigated
  • Loss of tubular function - in particular of concentrating ability - gives rise to polyuria and nocturia
1706
Q

What are the radiographic findings of chronic pyelonephritis?

A

Radiographic studies show asymmetrically contracted kidneys with characteristic coarse scars and blunting and deformity of the calyceal system

1707
Q

How is chronic pyelonephritis linked to focal segmental glomerulosclerosis? What does this mean for the prognosis?

A
  • Although proteinuria is usually mild, some individuals with pyelonephritis scars develop secondary focal segmental glomerulosclerosis with significant proteinuria, even in the nephrotic range, usually several years after the scarring has occurred and often in the absence of continued infection or persistent vesicoureteral reflux
  • The onset of proteinuria and focal segmental glomerulosclerosis is a poor prognostic sign, which may progress to ESRD.
1708
Q

Drug and toxin-induced tubulointerstitial nephritis is the second most common cause of acute kidney injury. Toxins and drugs are injure kidneys in at least three ways. What are they?

A

1) Trigger an interstitial immunologic reactions, exemplified by the acute hypersensitivity nephritis induced by drugs such as methicillin
2) Cause acute tubular injury
3) cause subclinical but cumulative injury to tubules that takes years to result in chronic renal insufficiency. This type of damage is especially worrisome, because it may be unrecognised until irreversible renal damage has occurred

1709
Q

What is drug-induced acute interstitial nephritis?

A
  • Drug-induced acute interstitial nephritis begins about 15 days (range:2-40) after drug exposure * It characterised by fever, eosinophilia (which may be transient), a rash in about 25% of patients, and renal abnormalities
  • The latter takes the form of haematuria, mild proteinuria, and leukocyturia (often including eosinophils). A rising serum creatinine or acute kidney injury with oliguria develops in about 50% of cases, particularly in older patients
1710
Q

What is the pathogenesis of acute drug-induced interstitial nephritis?

A
  • The most likely sequence of events is that the drugs function as haptens and covalently bind to some plasma membrane or extracellular component of tubular cells.
  • These modified self antigens then become immunogenic
  • The resultant injury is due to IgE or cell-mediated immune reactions to tubular cells or their basement membrane
1711
Q

Many features of acute drug-induced interstitial nephritis suggest an idiosyncratic immune mechanism that is not dose-related. What features suggest this?

A
  • Clinical evidence of hypersensitivity includes the latent period, the eosinophilia and rash, the fact that the onset of nephropathy is not dose-related, and the recurrence to the same or a chemically related drug
  • In some patients, serum IgE levels are increased, and IgE-containing plasma cells and basophils are present in the lesions, suggesting the the late-phase reaction of an IgE-mediated (type I) hypersensitivity maybe involved in the pathogenesis
  • In other cases, a mononuclear or granulomatous reaction, together with positive result of skin tests to drug haptens, suggest a T-cell mediated (type IV) delayed hypersensitivity reaction
1712
Q

What are the histological features of acute drug-induced interstitial nephritis?

A
  • On histologic examination the interstitium shows variable but frequently pronounced oedema and infiltration by mononuclear cells, principally lymphocytes and macrophages.
  • Eosinophils and neutrophils may be present, often in clusters and large numbers, and smaller numbers of plasma cells and mast cells are sometimes are present.
  • Inflammation may be more prominent in the medulla where the inciting agent is often concentrated
  • With some drugs (eg, methicillin, thiazides), interstitial non-necrotising granulomas may be seen.
  • Tubulitis, the infiltration of tubules by lymphocytes, is common. Variable degrees of tubular necrosis and regeneration are present.
  • The glomeruli are normal except in some cases caused by NSAIDs, when minimal-change disease and the nephrotic syndrome develop concurrently
1713
Q

What drugs can cause acute drug-induced interstitial nephritis?

A
  • First reported after the use of sulfonamides, acute tubulointerstitial nephritis most frequently occurs with synthetic penicillins (methicillin, ampicillin), other synthetic antibiotics (rifampicin), diuretics (thiazides), NSAIDs, and miscellaneous drugs (allopurinol, cimetidine)
  • The chronic tubulonephritis caused by phenacetin-containing analgesics, termed analgesic nephropathy, is mostly of historical importance as its incidence has substantially diminished due to the withdrawal or restriction of phenacetin in most countries
1714
Q

Why is it important to identify acute drug-induced interstitial nephritis?

A
  • It is important to recognised drug-induced acute interstitial nephritis because withdrawal of the offending drug is followed by recovery, although it may take several months, and irreversible damage can occur
  • It is also important to remember that while drugs ae the leading identifiable cause of acute interstitial nephritis, in many affected patients (approximately 30-40% of an offending drug or mechanisms cannot be identified)
1715
Q

How may acute drug-induced interstitial nephritis present?

A
  • On occasion, necrotic papillae are excreted, and may cause grow haematuria or renal colic due to ureteric obstruction
  • Papillary necrosis is nor specific for analgesic nephropathy, and is also seen in diabetes mellitus, as well as in urinary tract obstruction, sickle cell disease or treat, and focally in renal TB
  • In all cases, papillary necrosis is caused by ischaemia resulting from compression or obstruction of small blood vessels in the medulla. Such compression may be caused by interstitial oedema (as in inflammatory reactions and urinary tract obstruction) or microvascular disease (as in diabetes)
  • A small percentage of patients with analgesia nephropathy develop urothelial carcinoma of the renal pelvis
1716
Q

NSAIDs, produce several forms of renal injury. How do they do this?

A
  • Many NSAIDs are non-selective COX inhibitors, and their adverse renal effects are related to their ability to inhibit COX-dependent prostaglandin synthesis.
  • The selective COX-2 inhibitors, while sparing the GI tract, do affect the kidneys because COXI-2 is expressed in human kidneys.
1717
Q

What are some NSAID-associated renal syndromes?

A
  • Acute kidney injury, due to the decreased synthesis of vasodilatory prostaglandins and resultant ischaemia. This is particularly likely to occur in the setting of other renal diseases or conditions causing volume depletion.
  • Acute hypersensitivity interstitial nephritis, resulting in renal failure
  • Acute interstitial nephritis and minimal change disease. This curious association of two diverse renal conditions, one leading to renal failure and the other to nephrotic syndrome, suggests a hypersensitivity reaction affecting the interstitium and possibly the glomeruli, but also is consistent with injury to podocytes mediated by cytokines released as part of the inflammatory process
  • Membranous nephropathy, with the nephrotic syndrome
1718
Q

What are the causes of papillary necrosis and their specific characteristics?

A
1719
Q

Three types of nephropathy can occur in persons with hyperuricaemic disorders. What are they?

A
  • Acute uric acid nephropathy
  • Chronic urate nephropathy
  • Nephrolithiasis
1720
Q

How does hyperuricaemia cause acute uric acid nephropathy?

A
  • It is caused by the precipitation of uric acid crystals in the renal tubules, principally in collecting ducts, leading to obstruction of nephrons and the development of acute renal failure
  • This particularly likely to occur in individuals with leukaemia or lymphomas who are undergoing chemotherapy (tumour lysis syndrome)
  • The drugs kill tumour cells, and uric acid is produced as released nucleic acids are broken down
  • Precipitation of uric acid is favoured by the acidic pH in collecting tubules
1721
Q

How does hyperuricaemia cause chronic urate nephropathy? What populations does it occur in?

A
  • It occurs in a subset of patients with protracted forms of hyperuricaemia
  • The monosodium urate crystals deposit in the acidic milieu of the distal tubules and collecting ducts as well as in the interstitium, and form distinct birefringent needle-like crystals either in the tubular lumens or in the interstitium
  • The urate deposits evoke a mononuclear response that contains foreign-body giant cells. This lesion is called a tophus
  • Tubular obstruction by the grates causes cortical atrophy and scarring.
  • Clinically, urate nephropathy is a subtle disease associated with tubular defects that may progress slowly. Some individuals with gout who develop a chronic neuropathy have evidence of increased exposure to lead
1722
Q

What proportion of people with gout and secondary hyperuricaemia develop nephrolithiasis?

A

Uric acid stones are present in 22% of individuals with gout and 42% of those with secondary hyperuricaemia

1723
Q

What diseases are associated with nephrocalcinosis? What is it?

A
  • Disorders with hypercalcaemia, such as hyperparathyroidism, multiple myeloma, vitamin D intoxication, metastatic cancer, or excess calcium intake (milk-alkali syndrome), may induce the formation of calcium stones and deposition of calcium in the kidney (nephrocalcinosis).
  • Extensive degrees of calcinosis, under certain conditions, may lead to chronic tubulointerstitial disease and renal insufficiency
1724
Q

What happens over time with nephrocalcinosis?

A
  • The earliest functional defect is an inability to concentrate the urine
  • Other tubular defects, such as tubular acidosis and salt-losing nephritis, may occur
  • With further damage, a slowly progressive renal insufficiency develops.
  • This is usually due to nephrocalcinosis, but many of these patients also have calcium stones and secondary pyelonephritis
1725
Q

What causes acute phosphate nephropathy?

A
  • Extensive accumulations of calcium phosphate crystals in tubules can occur in patients consuming high doses of select oral phosphate solutions in preparation for colonoscopy
  • These patients are not hypercalcaemic, but the excess phosphate load, perhaps complicated by dehydration, causes marked precipitation of calcium phosphate, typically presenting as renal insufficiency several weeks after the exposure.
  • Patients with acute and reversible injury typically recover partial renal function.
1726
Q

Non-renal malignant tumours, particularly those of haematopoietic origin, affect the kidneys in several ways. What are these?

A

** Direct or metastatic tumour invasion of renal parenchyma** - ureters (obstruction), artery (renovascular hypertension)
Hypercalcaemia
Hyperuricaemia
Amyloidosis
Excretion of abnormal proteins - multiple myeloma
Glomerulonephropathies - secondary membranous nephropathy (carcinomes), minimal change disease (Hodgkin disease), membranoproliferative glomerulonephritis (leukaemia and lymphomas), monoclonal immunoglobulin/light-chain desposition disease (multiple myeloma)
Effects of radiation therapy, chemotherapy, haematopoietic cell transplantation, secondary infection

1727
Q

Overt renal insufficiency occurs in half of those with multiple myeloma and related lymphoplasmacytic disorders. Several factors contribute to renal damage. What are they?

A
  • Amyloidosis of AL type, formed from free light chains (usually of λ type), which occurs in 6-24% of individuals with myeloma
  • Light-chain deposition disease - in some patients, light chains (usually of κ type) deposit in GBMs and mesangium in non-fibrillar forms, causing a glomerulopathy, and in tubular membranes, which may cause tubulointerstitial nephritis
  • Hypercalcaemia and hyperuricaemia are often present in these patients.
  • Bence-jones proteinuria and cast nephropathy - we’ll go on to this
1728
Q

The main cause of renal dysfunction in multiple myeloma is Bence-Jones proteinuria and cast nephropathy. Two mechanisms seem to account for the renal toxicity of Bence-Jones proteins. What are they?

A
  • First, some Ig light chains are directly toxic to epithelial cells, apparently becuase of their intrinsic physicochemical properties
  • Second, Bence-Jones proteins combine with the urinary glycoprotein (Tamm-Horsall protein) under acidic conditions to form large, histologically distinct tubular casts that obstruct the tubular lumens and induce a characteristic inflammatory reaction (light-chain cast nephropathy)
1729
Q

What is the morphology of light-chain cast nephropathy (“myeloma kidney”)

A
  • The tubulointerstitial changes in light-chain cast nephropathy are characteristic.
  • The Bence-Jones tubular casts appear as pink to blue amorphous masses, sometimes concentrically laminate and often fractures, which fit and distend the tubular lumens. Some casts are surrounded by multinucleate giant cells thats are derived from activated macrophages
  • The adjacent interstitial tissue usually shows an inflammatory response and fibrosis.
  • On occasion, the casts rupture the tubules, evoking a granulomatous inflammatory reaction.
1730
Q

Clinically, the renal manifestation of light chain cast nephropathy are of several types. What are these? What are the precipitating factors? What is in the urine?

A
  • In the most common form, chronic kidney disease develops insidiously and progresses slowly during a period of several months to years
  • Another form occurs suddenly and is manifested by acute kidney injury with oliguria
  • Precipitating factors include dehydration, hypercalcaemia, acute infection, and treatment with nephrotoxic antibodies.
  • **Bence-Jones proteinuria occurs in 70% of individuals with multiple myeloma; the presence of significant non-light-chain proteinuria (eg, albuminuria) suggests AL amyloidosis or light-chain deposition disease.
1731
Q

Hepatorenal syndrome refers to impairment of renal function in patients with acute or chronic liver disease with advance liver failure. Why does this happen?

A
  • In this setting, serum bilirubin levels can be markedly elevated, particularly in jaundiced patients, with bile cast formation (also known as cholemic nephrosis) in distal nephron segments.
  • The casts can extend to proximal tubules, resulting in both direct toxic effects on tubular epithelial cells and obstruction of the involved nephron.
  • This mechanism of injury is analogous to the with myeloma protein and myoglobin casts. The tubular bile casts can range from yellowish-green to pink and contain variable degrees of sloughed cells or cellular debris.
  • The reversibility of the renal injury depends upon the severity and duration of the liver dysfunction
1732
Q

What is nephrosclerosis? What is it associated with?

A
  • It is the term used for the renal pathology associated with sclerosis of renal arterioles and small arteries
  • It is strongly associated with HTN, which can be both a cause a consequence of nephrosclerosis. Diabetes mellitus also increases the incidence and severity of the lesions
1733
Q

What are the characteristic of the affects vessels in nephrosclerosis?

A
  • The affected vessels have thickened walls and consequently narrowed lumens, changes that result in focal parenchymal ischeamia
  • Ischaemia leads to glomerulosclerosis and chronic tubulointerstitial injury, and produces a reduction in functional renal mass
1734
Q

Two processes participate in the arterial lesions of nephrosclerosis. What are they?

A
  • Medial and intimal thickening, as a response to haemodynamic changes, aging, genetic defects, or some combination of these
  • Hyalinisation of arteriolar walls, caused by extravasation of plasma proteins through injured endothelium and by increased deposition of basement membrane matrix
1735
Q

How does the size of the kidneys changes in nephrosclerosis? Why?

A
  • The kidneys are either normal or moderately reduced in size, with average weights between 110 and 130gm
  • The cortical surfaces have a fine, even granularity that resembles grain leather.
  • The loss of mass is due mainly to cortical scarring and shrinking
1736
Q

What are the histologic examination findings of nephrosclerosis?

A
  • On histologic examination, there is narrowing of the lumens of arterioles and small arteries, caused by thickening and hyalinisation of the walls (hyaline arteriolosclerosis)
  • Corresponding to the finely granular surface are microscopic subcapsular scars with sclerotic glomeruli and tubular dropout, alternating with better preserved parenchyma
  • In addition, the interlobular and arcuate arteries show medial hypertrophy, replication of the internal elastic lamina, and increased myofibroblastic tissue in the intake, all of which narrow the lumen.
  • This change, called fibroelastic hyperplasia, often accompanies hyaline arteriolosclerosis and increases in severity with age and in the presence of hypertension
1737
Q

What are the morphological consequences of the vascular narrowing in nephrosclerosis?

A

There is patchy ischaemic atrophy, which consists of 1) foci of tubular atrophy and interstitial fibrosis and 2) a variety of glomerular alterations

  • The latter include collapse of the GBM, deposition of collagen within Bowman space, periglomerular fibrosis, and total sclerosis of glomeruli
  • When the ischaemic changes are pronounced and affect large areas of parenchyma, they can produce wedge shaped infarcts or regional scars with histologic alterations that may resemble those in renal ablation injury
1738
Q

It is unusual for uncomplicated nephrosclerosis to cause renal insufficiency or uraemia. However, three groups of hypertensive patients with nephrosclerosis are at increased risk of developing renal failure. Who are they?

A
  • People of African descent
  • People with severe blood pressure elevations
  • People with a second underlying disease, especially diabetes

In these groups renal insufficiency may supervene after prolonged hypertension, but rapid renal failure results from the development of the malignant or accelerated phase of hypertension

1739
Q

What is malignant nephrosclerosis? What and in who does it occur?

A
  • Malignant nephrosclerosis is a renal vascular disorder associated with malignant or accelerated hypertension
  • It occasionally develops suddenly in previously normotensive individuals but more often is superimposed on pre-existing essential hypertension, secondary forms of hypertension, or an underlying chronic renal disease, particularly glomerulonephritis or reflux nephropathy
  • It is a frequent cause of renal failure in individuals with systemic sclerosis
  • In its pure form it usually affects younger individuals, and occurs more often in men and in blacks
1740
Q

The fundamental lesion in malignant nephrosclerosis is vascular injury. How does this happen?

A
  • The initial insult seems to be some form of vascular damage to the kidneys that might result from a variety of disorders, including long-standing HTN, arteritis, or a coagulopathy, alone or in combination
  • The initiating event incurs endothelium and results in increased permeability of the small vessels to fibrinogen and other plasma proteins, focal death of cells of the vascular wall, and platelet deposition
  • This can lead to fibrinoid necrosis of arterioles and small arteries, with activation of platelets and coagulation factors causing intravascular thrombosis.
  • Mitogenic factors from platelets (PDGF), plasma, and other cells cause hyperplasia of intimal smooth muscle of vessels, resulting in the hyperplasticity arteriolosclerosis that is typical of malignant hypertension and responsible for further narrowing of the lumens.
  • The kidneys become markedly ischaemia
1741
Q

How is the renin level affected in malignant nephrosclerosis? How does this contribute to the pathology?

A
  • With severe involvement of the renal afferent arterioles, the renin-angiotensin system receives a powerful stimulus
  • Patients with malignant hypertension ahem markedly elevated levels of plasma renin.
  • This sets up a self-perpetuating cycle in which angiotensin II causes intrarenal vasoconstriction, and the attendant renal ischaemia perpetuates renin secretion.
1742
Q

What are the gross morphological changes in malignant nephrosclerosis?

A
  • The kidney size varies depending on the duration and severity of the hypertensive disease
  • Small, pinpoint petechial haemorrhages may appear not the cortical surface from rupture of arterioles or glomerular capillaries, giving the kidney a peculiar “flea-bitten” appearance
1743
Q

Two histologic alterations characterise renal blood vessels in malignant hypertension. What are these?

A
  • Fibrinoid necrosis of arterioles. In this form of necrosis, cytologic detail is lost and the vessel wall takes on a smudgy eosinophilic appearance due to fibrin deposition. Inflammation is usually not seen or is minimal. Sometimes the glomeruli become necrotic and infiltrated with neutrophils, and the glomerular capillaries may thrombose
  • In the interlobular arteries and arteries, there is intimal thickening caused by a proliferation of elongated, concentrically arranged smooth muscle cells, together with fine concentric layering of collagen and accumulation of pale-staining material that probably represents deposition of proteoglycans and plasma proteins. This alteration has been referred to as onion-skinning because of its concentric appearance. Th lesion, also called hyperplastic arteriolitis, correlates with renal failure.
1744
Q

What are the clinical features of malignant hypertension?

A
  • It is characterised by systolic pressures greater than 200mmHg and diastolic pressures greater than 120mmHg, papilloedema, retinal haemorrhages, encephalopathy, cardiovascular abnormalities and renal failure
  • Most often, the early symptoms are related to increased ICP and include headaches, nausea, vomiting and visual impairments, particularly scotomas or spots before the eyes.
  • “Hypertensive crises” are sometimes encountered, characterised by loss of consciousness or even convulsions.
  • At the onset, there may only be marked proteinuria and microscopic or macroscopic haematuria, but renal failure soon ensues.
1745
Q

What is the prognosis of malignant hypertension?

A
  • The syndrome is a medical emergency requiring aggressive and prompt antihypertensive therapy to prevent irreversible renal injury
  • Before the development of current antihypertensive drugs, malignant hypertension was associated with a 50% mortality rate within 3 months of onset, progressing to 90% within a year
  • At present, however, about 75% of patients survive 5 years, and 50% survive with restoration of pre-crisis renal function
1746
Q

What is the pathogenesis of renal artery stenosis?

A
  • Hypertension secondary to renal artery stenosis is caused by increased production of renin from the ischaemic kidney
  • Elevation in BP, at least initially, is due to stimulation of renin secretion by the juxtaglomerular apparatus and the subsequent production of the vasoconstrictor angiotensin II.
  • Other factors, however, may contribute to the maintenance of renovascular hypertension after the renin-angiotensin system has initiated it, including sodium retention
1747
Q

What is the most common cause of renal artery stenosis and what population does it occur in?

A
  • The most common cause of renal artery stenosis (70% of cases) is narrowing at the origin of the renal artery by an atheromatous plaque.
  • The plaque is usually concentrically placed, and superimposed thrombosis often occurs
  • This occurs more frequent in men, and the incidence increases with advancing age and diabetes mellitus
1748
Q

What is the second most frequent cause of renal artery stenosis? Who does it happen to?

A
  • The second most frequent cause of stenosis is fibromuscular dysplasia of the renal artery.
  • This heterogenous entity is characterised by fibrous or fibromuscular thickening that may involve the intima, the media, or the adventitia of the artery
  • The stenoses, as a whole, are more common in omen and tend to occur in younger age groups (30-40s)
1749
Q

What are the morphologic changes in renal artery stenosis?

A
  • The ischaemic kidney is reduced in size and shows signs of diffuse ischaemic atrophy, with crowded glomeruli, atrophic tubules, interstitial fibrosis, and focal inflammatory infiltrates
  • The arterioles in the ischaemic kidney are usually protected from the effects of high pressure, thus showing only mild arteriolosclerosis
  • In contrast, the contralateral non-ischaemic kidney may show more severe arteriosclerosis, depending on the severity of the hypertension
1750
Q

What is the clinical course of renal artery stenosis?

A
  • Few distinctive features suggest the presence of renal artery stenosis, and in general, these patients resemble those with essential hypertension
  • On occasion, a bruit can be head on auscultation of the affected kidneys
  • Elevated plasma or renal vein renin, response to angiotensin-converting enzyme inhibitor, renal scans or IV pyelography may aid with diagnosis, but arteriography is required to localise the stenotic lesion
  • The cure rate after surface is 70-80% in well-selected cases
1751
Q

What are thrombotic microangiopathies? What causes them

A
  • The term thrombotic microangiopathy encompasses a spectrum of clinical syndrome that includes thrombotic thrombocytopaenic purpura (TTP) and haemolytic-uraemic syndrome (HUS)
  • They are caused by diverse insults that lead to excessive activation of platelets, which deposit thrombi in capillaries and arterioles in various tissue beds, including those of the kidney
  • Wide-spread “consumption” of platelets leads to thrombocytopaenia, and the resulting thrombi create flow abnormalities that shear red cells, producing a microangiopathic haemolytic anaemia.
  • Of even greater importance, the thrombi produce microvascular occlusions that cause tissue ischaemia and organ dysfunction
1752
Q

Thrombotic microangiopathies is now classified according to the current understanding of their causes or associations. What are they? What are their associations?

A
  • Typical HUS (epidemic, classic, diarrhoea-positive), most frequently associated with consumption of food contaminated by bacteria producing Shiga-like toxins
  • **Atypical HUS (non-epidemic, diarrhoea-negative), associated with: - inherited mutations of complement-regulatory proteins - diverse acquired causes of endothelial injury including antiphospholipid antibodies, complications of pregnancy and oral contraceptives, vascular renal diseases such as scleroderma and hypertension, chemotherapeutic and immunosuppressive drugs
  • TTP, which is often associated with inherited or acquired deficiencies if ADAMTS13, a plasma metalloprotease that regulates the function of vWF.
1753
Q

Within thrombotic microangiopathies, two pathogenetic triggers dominate. What are they?

A

1) Endothelial injury - primary cause of HUS
2) Excessive platelet activation and aggregation - primary cause of TTP

1754
Q

How does endothelial injury trigger haemolytic-uraemic syndrome?

A
  • In typical (epidemic, classic, diarrhoea-positive) HUS, the trigger for endothelial injury and activation is usually Shiga-like toxin, while for inherited forms of atypical HUS the cause of the endothelial injury appears to be excessive, inappropriate activation of complement.
  • Many other exposures and condition occasionally precipitate a HUS-like picture, presumably also by injury the endothelium
  • The endothelial injury in HUS appears to cause platelet activation and thrombosis within microvascular beds.
  • There is evidence that reduced endothelial production of prostaglandin I2 and NO (both inhibitors of platelet aggregation) contributes to thrombosis.
  • The reduction in these two factors and increased production of endothelium-derived endothelin may also promote vasoconstriction, exacerbating the hypoperfusion of tissues
1755
Q

How does platelet aggregation cause thrombotic thrombocytopaenic purpura? Why does it happen?

A
  • In contrast the HUS, in TTP the initiating event appears to be platelet aggregation induced by very large multimers of vWF, which accumulate due to deficiency of ADAMTS13, a plasma protease that leaves vWF into smaller sizes.
  • The deficiency of ADAMTS13 is most often caused by autoantibodies that inhibit ADAMTS13 function.
  • Less commonly, a chronic relapsing and remitting form of TTP is associated with inherited deficiencies of ADAMTS13.
  • Very large vWF multimers can bind platelet surface glycoproteins and activate platelets spontaneously, providing a pathophysiologic explanation for the micro thrombi that are observed in vascular beds.
1756
Q

What are the clinical features of typical HUS? What populations is it most common?

A
  • Typical HUS can occur at any age, but children and older adults are at highest risk
  • Following a prodrome of influenza-like or diarrhoea symptoms, there is a sudden onset of bleeding manifestations (especially haematemesis and malaria), severe oliguria, and haemturia, associated with microangiopathic haemolytic anaemia, thrombocytopenia, and (in some patients) prominent neurologic changes
  • HTN is present in about half the patients.
1757
Q

How does Shiga-like toxin cause haemolytic uraemic syndrome?

A
  • According to one model the toxin “activates” endothelial cells, which respond by increasing their expression of leukocytes adhesion molecules and endothelin and decreasing nitric oxide production.
  • In the presence of cytokines such as TNF, Shiga-like toxin may cause endothelial apoptosis. These alterations lead to platelet activation and induce vasoconstriction, resulting in the characteristic microangiopathy.
  • There is some evidence that Shiga-like toxins may bind and activate platelets directly
  • They may bind the regulatory complement protein Factor H and inhibit its activity, causing hyperactivation of complement
1758
Q

What is the prognosis of typical haemolytic uraemic syndrome?

A
  • In typical HUS, if the renal failure is managed properly with dialysis, most patients recover normal renal function in a matter of weeks
  • However, due to underlying renal damage the long-term (15-25 years) outlook is more guarded, may go on to develop CKD
1759
Q

At what age do people develop atypical haemolytic-uraemic syndrome and what is the clinical course of the disease?

A
  • Patients with genetic mutations in complement-regulatory proteins (Factor H, Factor I and CD46) may develop HUS at any age
  • Roughly half of affected individuals have a course marked by multiple releases and progression to end-stage renal disease.
1760
Q

How does thrombotic thrombocytopaenic purpura typically present? In what populations?

A
  • TTP is classically manifested by the pentad of fever, neurologic symptoms, microangiopathic haemolytic anaemia, thrombocytopaenia, and renal failure
  • The most common cause of deficient ADAMTS13 activity is inhibitory autoantibodies, and the majority with such antibodies are women
  • Regardless of cause, most patients present as adults at younger than 40
  • In those with hereditary ADAMTS13 deficiency, the onset is often delayed until adolescence and the symptoms are episodic
1761
Q

What are the dominant clinical features of all the thrombotic thrombocytopaenia purpura symptoms? Why?

A
  • CNS involvement is the dominant feature, whereas renal involvement is seen in about 50% of patients
  • The clinical findings are dictated by the distribution of the micro-thrombi, which are found in arterioles throughout the body
1762
Q

How can you treat thrombotic thrombocytopaenic purpura?

A

With plasma exchange, which removes autoantibodies and provides functional ADAMTS13, TTP can be treated successfully in more than 80% of patients.

1763
Q

The morphologic findings in the various forms of HUS/TTP show considerable overlap, and vary mainly according to chronicity rather than cause. What are they in acute disease?

A
  • In acute, active disease the kidney may show patchy or diffuse cortical necrosis and subcapsular petechiae
  • On microscopic examination, the glomerular capillaries are occluded by thrombi composed of aggregated platelets and to a lesser extent fibrin
  • The capillary walls are thickened due to endothelial cell welling and subendothelial deposits of cell debris and fibrin.
  • Disruption of the mesangial matrix and damage to the mesangial cells often results in mesangiolysis.
  • Interlobular arteries and arterioles often show fibrinoid necrosis of the wall and occlusive thrombi
1764
Q

What are the morphologic findings in chronic atypical HUS and TTP?

A
  • It has features that stem from continued injury and attempts at healing. The renal cortex reveals various degrees of scarring
  • By light microscopy the glomeruli are mildly hypercellular and have marked thickening of the capillary walls associated with splitting or reduplication of the basement membrane (so called double contours or tram tracks).
  • The walls of arteries and arterioles often exhibit increased layers of cells and connective tissue (“onion-skinning”) that narrow the vessel lumens.
  • These changes lead to persistent hypoperfusion and ischaemic atrophy of the parenchyma, which manifests clinically as renal failure and hypertension
1765
Q

Embolisation of fragments of atheromatous plaques from the aorta or renal artery into intrarenal vessels occurs in older adults with severe atherosclerosis, especially after surgery on the abdominal aorta, aortography, or intra-aortic cannulisation. Where can these emboli be seen? What are the clinical consequences of them? and in whom are the consequences more severe?

A
  • These emboli can be recognised in the lumens of arcuate and interlobular arteries by their content of cholesterol crystals, which appear as rhomboid clefts.
  • The clinical consequences of atheroemboli vary according to the number of emboli and the pre-existing state of renal function.
  • Frequently they are of no significant. However, acute renal injury or failure may develop in older adults in whom renal function in already compromised.
1766
Q

Sickle cells disease (homozygous) or trait (heterozygous) may lead to a variety of alterations in renal morphology and function, some of which produce clinically significant abnormalities. The various manifestations are grouped under sickle-cell nephropathy. What are some of the most common abnormalities? Why do they happen?

A
  • The most common abnormalities are haematuria and a diminished concentrating ability (hyposthenuria). These are thought to be due to accelerated sickling in the hypertonic hypoxic milieu of the renal medulla; the hyperosmolarity dehydrates the red cells and increases intracellular HbS concentrations, which likely explains why even those with sickle trait are affected
  • Patchy papillary necrosis may occur in both homozygotes and heterozygotes; this is sometimes associated with cortical scarring
  • Proteinuria is also common in sickle-cell disease, occurring in about 30% of patients. It is usually mild to moderate, but on occasion the overt nephrotic syndrome arises, associated with sclerosing glomerular lesions
1767
Q

What is diffuse cortical necrosis? Who does it happen to? What is the prognosis?

A
  • This uncommon condition occurs most frequently after an obstetric emergency, such as abruption placentae (premature separation of the placenta), septic shock, or extensive surgery
  • The cortical destruction has the features of ischaemic necrosis. Glomerular and arteriolar microthrombi are often found and contribute to the necrosis and renal damage.
  • Massive acute cortical necrosis is of grave significance, since its gives rise to sudden anuria, terminating rapidly in uraemic death. Instances of unilateral or patchy involvement are compatible with survival
1768
Q

What are the morphological features of diffuse cortical necrosis?

A
  • The morphologic features considerably overlap with thrombotic microangiopathy and disseminated intravascular coagulation
  • The gross alterations of massive ischaemic necrosis are sharply limited to the cortex
  • The lesions may be patchy, with areas of coagulative necrosis and apparently better preserved cortex
  • Intravascular and intraglomerular thromboses may be prominent but are usually focal, and acute necroses of small arterioles and capillaries may occasionally be present
  • Haemorrhages occur into the glomeruli, together with the formation of fibrin plugs in the glomerular capillaries
1769
Q

The kidneys are common sites for the development of infarcts. Why?

A

Contributing to this predisposition is the extensive blood flow to the kidneys (1/4 of the cardiac output), but probably more important is the limited collateral circulation from extra renal sites (small blood vessels penetrating from the renal capsule supply only the very outer rim of the cortex)

1770
Q

Although thrombosis in advance atherosclerosis and the acute vasculitis of polyarteritis nodosa may occlude arteries, most renal infarcts are due to embolism. Where are they most likely to be from?

A
  • A major source of such emboli is mural thrombosis in the left atrium and ventricle as a result of myocardial infarction
  • Vegetative endocarditis, aortic aneurysms, and aortic atherosclerosis are less frequent sources of emboli
1771
Q

What are the clinical features of a renal infarct?

A
  • Many renal are clinically silent
  • Sometimes, pain with tenderness localised to the costovertebral angle occurs, associated with showers of red cells in the urine
  • Large infarcts of one kidney are probably associated with narrowing of the renal artery or one of its major branches, which in turn may cause HTN
1772
Q

What are the acute morphological changes of a renal infarct?

A
  • Because of the lack of a collateral blood supply, most renal infarcts are of the “white” anaemic variety
  • Within 24 hours infarcts become sharply demarcated, pale, yellow-white areas that may contain small irregular foci of haemorrhagic discolouration.
  • They are usually ringed by a zone of intense hyperaemia
1773
Q

What are the morphologic changes over time from a renal infarct?

A
  • The infarcts are usually wedge-shaped, with the base against the cortical surface and the apex pointing toward the medulla
  • In time these acute areas of ischaemic necrosis undergo progressive fibrous scarring, giving rise to depressed, pale, gray-white scars that assume a V-shape on section
  • The histologic changes in renal infants are those of ischaemic coagulative necrosis
1774
Q

About 10% of people are born with significant malformations of the urinary system. They can be hereditary but most often result from an acquired developmental defect during gestation. What are some examples of these?

A
  • Agenesis of the kidney
  • Hypoplasia
  • Ectopic kidneys
  • Horseshoe kidneys
1775
Q

You can get bilateral and unilateral agenesis of the kidneys. What is the difference in prognosis?

A
  • Bilateral agenesis is incompatible with life. It is often associated with other congenital disorders (limb defects, hypoplastic lungs)
  • Unilateral agenesis is uncommon and compatible with normal life if no other abnormalities exist
1776
Q

What is unilateral agenesis of the kidney? How does it affect the kidney? What is its clinical course?

A
  • It is where you only have one kidney
  • The solitary kidney enlarges as a result of compensatory hypertrophy.
  • Some patients eventually develop progressive glomerular sclerosis in the remaining kidney as a result of the adaptive changes in the hypertrophied nephrons, and in time, CKD ensues
1777
Q

What is kidney hypoplasia? In which population can you get true hypoplastic kidney and what will their kidney look like?

A
  • It refers to failure of the kidneys to develop to a normal size
  • This anomaly may occur bilaterally, resulting in renal failure in early childhood, but it is more commonly encountered as a unilateral defect
  • True renal hypoplasia is observed in low birth weight infants and may contribute to their increased lifetime risk for CKD. It shows no scars and has a reduced number of renal lobes and pyramids, usually six or fewer
1778
Q

What are ectopic kidneys? What are the complications?

A
  • The development of the metanephros into the kidneys may occur in ectopic foci.
  • These kidneys lie either just above the pelvic brim or sometimes within the pelvis
  • They are usually normal or slightly small in size but otherwise are not remarkable
  • Because of their abnormal position, kinking or tortuosity of the ureters may cause obstruction to urinary flow, which predisposes to bacterial infections.
1779
Q

What is a horseshoe kidney? How common is it?

A
  • Fusion of the upper (10%) or lower (90%) poles of the kidneys produces a horseshoe-shaped structure that is continuous across the midline anterior to the great vessels.
  • This anomaly is found in 1 in 500 to 1000 anomalies.
1780
Q

Cystic diseases of the kidney are heterogenous, comprising hereditary, developmental, and acquired disorders. They are important for several reasons. What are they?

A

1) They are reasonably common and often represent diagnostic problems for clinicians, radiologists, and pathologists
2) some forms, such as adult polycystic kidney disease, are major causes of CKD
3) they can occasionally be confused with malignant tumours

1781
Q

What are the different classifications of renal cysts?

A
  • Polycystic kidney disease (autosomal dominant (adult) and autosomal recessive (childhood)
  • Medullary cystic disease - medullary sponge kidney and nephronophthisis
  • Multicystic renal dysplasia
  • Acquired (dialysis-associated) cystic disease
  • Localised (simple) renal cysts
  • Renal cysts in hereditary malformation syndromes (eg, tubular sclerosis)
  • Glomerulocystic disease
  • Extraparenchymal renal cysts (pyelocalyceal cysts, hilar lymphangitic cysts)
1782
Q

What are the pathologic features of:
* Adult polycystic kidney disease
* Childhood polycystic kidney disease
* Medullary sponge kidney
* Familial juvenile nephronophthisis
* Adult-onset medullary cystic disease
* Multicystic renal dysplasia
* Acquired renal cystic disease
* Simple cysts

A
  • Adult polycystic kidney disease - large multicystic kidneys, liver cysts, berry aneurysms
  • Childhood polycystic kidney disease - enlarged, cystic kidneys at birth
  • Medullary sponge kidney - medullary cysts on excretory urography
  • Familial juvenile nephronophthisis - corticomedullary cysts, shrunken kidneys
  • Adult-onset medullary cystic disease - corticomedullary cysts, shrunken kidneys
  • Multicystic renal dysplasia - irregular kidneys with cysts of variable size
  • Acquired renal cystic disease - cystic degeneration in end-stage kidney disease
  • Simple cysts - single or multiple cysts in normal-sized kidney
1783
Q

What are the clinical features of complications of:
* Adult polycystic kidney disease
* Childhood polycystic kidney disease
* Medullary sponge kidney
* Familial juvenile nephronophthisis
* Adult-onset medullary cystic disease
* Multicystic renal dysplasia
* Acquired renal cystic disease
* Simple cysts

A
  • Adult polycystic kidney disease - haematuria, flank pain, urinary tract infection, renal stones, HTN
  • Childhood polycystic kidney disease - hepatic fibrosis
  • Medullary sponge kidney - haematuria, URI, recurrent renal stones
  • Familial juvenile nephronophthisis - salt wasting, polyuria, growth retardation, anaemia
  • Adult-onset medullary cystic disease - salt wasting, polyuria
  • Multicystic renal dysplasia - association with other renal anomalies
  • Acquired renal cystic disease - haemorrhage, erythrocytosis, neoplasia
  • Simple cysts - microscopic haematuria
1784
Q

What are the typical outcomes of:
* Adult polycystic kidney disease
* Childhood polycystic kidney disease
* Medullary sponge kidney
* Familial juvenile nephronophthisis
* Adult-onset medullary cystic disease
* Multicystic renal dysplasia
* Acquired renal cystic disease
* Simple cysts

A
  • Adult polycystic kidney disease - chronic renal failure beginning at ago 40-60 years
  • Childhood polycystic kidney disease - variable, death in infancy or childhood
  • Medullary sponge kidney - benign
  • Familial juvenile nephronophthisis - progressive renal failure beginning in childhood
  • Adult-onset medullary cystic disease - chronic renal failure beginning in adulthood
  • Multicystic renal dysplasia - renal failure if bilateral, surgically curable if unilateral
  • Acquired renal cystic disease - dependence on dialysis
  • Simple cysts - benign
1785
Q

What is the inheritance of the following:
* Adult polycystic kidney disease
* Childhood polycystic kidney disease
* Medullary sponge kidney
* Familial juvenile nephronophthisis
* Adult-onset medullary cystic disease
* Multicystic renal dysplasia
* Acquired renal cystic disease
* Simple cysts

A
  • Adult polycystic kidney disease - autosomal dominant
  • Childhood polycystic kidney disease - autosomal recessive
  • Medullary sponge kidney - none
  • Familial juvenile nephronophthisis - autosomal recessive
  • Adult-onset medullary cystic disease - autosomal dominant
  • Multicystic renal dysplasia - none
  • Acquired renal cystic disease - none
  • Simple cysts - none
1786
Q

What is autosomal dominant (adult) polycystic kidney disease? How common is it?

A
  • Autosomal dominant (adult) polycystic kidney disease is a hereditary disorder characterised by multiple expanding cysts of both kidneys that ultimately destroy the renal parenchyma and cause renal failure
  • It is a common condition affecting roughly 1 in every 400-1000 live births and accounting for about 5-10% of cases of end-stage renal disease requiring transplantation or dialysis
1787
Q

A wide range of different mutations in PKD1 and PKD2 has been described, and this allelic heterogeneity has complicated genetic diagnosis of autosomal dominant PCKD. What do the PKD1 and PKD2 genes do? Where are they?

A
  • The PKD1 gene is located on chromosome 16p13.3. It encodes a large integral membrane protein named polycystin-1, which is expressed in tubular epithelial cells, particularly those of the distal nephron. It contains domains that are usually involved in cell-cell and cell-matrix interactions.
  • The PKD2 gene, located on chromosome 4q21, produces polycystin-2, which is an integral membrane protein that is expressed in all segments of the renal tubules and in many extrarenal tissues. It functions as a Ca2+ permeable cation channel
1788
Q

Are mutations in PKD1 or 2 most common in autosomal dominant PCKD? Which one is more severe?

A
  • Mutations in PKD1 account for about 85% of cases. The majority of the rest are caused by PKD2 mutations.
  • In individuals with PKD1 mutations, the likelihood of developing of renal failure is less than 5% by 40 years of age, rising to more than 35% by 50 years, more than 70% at 60 years of age, and more than 95% by 70 years of age
  • Overall, the disease in PKD2 mutations is less severe than that associated with PKD1 mutations. Renal failure occurs in less than 5% of patients with PKD2 mutations at 50 years of age, but this rises to 15% at 60 years of age, and 45% at 70 years of age
1789
Q

The pathogenesis of autosomal dominant polycystic disease is not established, but the currently favoured hypothesis places the cilia-centrosome complex of tubular epithelial cells the centre of the disorder. What is the cilia-centrosome complex?

A
  • The tubular epithelial cells of the kidney contain a single non-motile primary cilium, a 2- to 3-μm long hairlike organelle that projects into the tubular lumen from the apical surface of the cells
  • The cilium is made of microtubules, and arises from and is attached to a basal body derived from the centriole. The cilia are part of a system of organelles and cellular structures that sense mechanical signals.
  • The apical cilia function in the kidney tubule as a mechanosensor to moniter changes in fluid flow and shear stress, while intercellular junctional complexes monitor forces between cells, and focal adhesions sense attachment to extracellular matrices.
  • In response to external signals, these sensors regulate ion flux (cilia can induce Ca2+ flux in cultured kidney epithelial cells) and cellular behaviour, including cell polarity and proliferation.
1790
Q

The idea that defects in mechanosensing, Ca2+ flux, and signal transaction underlie cyst formation in autosomal dominant PCKD is supported by several observations. What are they?

A
  • Both polycystin-2 and polycystin-2 are localised to the primary cilium
  • Other genes that are mutated in cystic diseases encode proteins that are localised to cilia and/or basal bodies
1791
Q

What do polycystin-1 and polycystin-2 do in the cilio-centrosome complex in the tubular epithelium? How is this changed in autosomal dominant PCKD?

A
  • Polycystin-1 and polycystin-2 appear to form a protein complex that regulates intracellular Ca2+ in response to fluid flow, perhaps because fluid moving through the kidney tubules cause ciliary bending than opens Ca2+ channels.
  • The consequent disruption of normal polycystin activity results in alterations on intracellular Ca2+, which regulating many downstream signalling events, including pathways thats directly or indirectly impact cellular proliferation, apoptosis, and secretory functions.
  • The increase in calcium is thought to stimulate proliferation and secretion from epithelial cells lining the cysts, which together result in progressive cyst formation and enlargement.
  • In addition, cyst fluids have been shown to harbour mediators, derived from epithelial cells that enhance fluid secretion and induce inflammation.
  • Finally, the calcium-induced signals also alter the interaction of epithelial cells with extracellular matrix, and this too is thought to contribute to the cyst formation and interstitial fibrosis that are characteristic of progressive polycystic kidney disease
1792
Q

What is the gross morphology of autosomal dominant PCKD?

A
  • In gross appearance, the kidneys are bilaterally enlarged and may achieve enormous sizes; weights as much as 4kg for each kidney have been reports
  • The external surface appears to be composed solely of a mass of cysts, up to 3-4cm in diameter, with no intervening parenchyma
1793
Q

What are the microscopic findings in autosomal dominant PCKD?

A
  • It reveals functioning nephrons dispersed between the cysts
  • The cysts may be filled with clear, serous fluid or with turbid, red to brown, sometimes haemorrhagic fluid
  • As these cysts enlarge, they may encroach on the calyces and pelvis to produce pressure defects
  • The cysts arise from the tubules throughout the nephron and therefore have variable lining epithelia.
  • On occasion, papillary epithelial formations and polyps project into the lumen. Bowman capsules are occasionally involved in cyst formation, ad glomerular tufts may be seen within the cystic space
1794
Q
A
1795
Q

What factors affect clinical course of autosomal dominant PCKD?

A
  • Patients with PKD2 mutations tend to have an older age at onset and later development of renal failure
  • Both genetic and environmental factors influence disease severity
  • Progression is accelerated in the black population (particularly in those with sickle-cell trait), in males, and in the presence of hypertension
1796
Q

Individuals with autosomal polycystic kidney disease also tend to have extrarenal congenital anomalies. What are these?

A
  • About 40% have one to several cysts in the liver (polycystic liver disease) that are usually asymptomatic. The cysts are derived from biliary epithelium
  • Cysts occur much less frequently in the spleen, pancreas, and lungs
  • Intracranial berry aneurysms, presumably from altered expression of polycystin in vascular smooth muscle, arise in the circle of Willis, and SAH from these account for death in 4-10% of individuals.
  • Mitral valve prolapse and other cardiac valvular anomalies occur in 20-25% of patients, but most are asymptomatic. The diagnosis is made by radiological imaging techniques
1797
Q

What do people with autosomal dominant PCKD die of?

A
  • This form of CKD is remarkable in that patients may survive for many years with azotemia slowly progressing to uraemia
  • Ultimately, about 40% of adult patients die or coronary or hypertensive heart disease, 25% of infection, 15% of ruptured berry aneurysm or hypertensive intracerebral haemorrhages and the rest of other causes.
1798
Q

Autosomal recessive (childhood) polycystic kidney disease is genetically distinct from adult polycystic kidney disease. What are the subcategories of childhood PCKD? How common are they and what are the outcomes?

A
  • Perinatal, neonatal, infantile, and juvenile subcategories have been defined, depending on the time of presentation and presence of associated hepatic lesions.
  • The first two are most common; serious manifestations are usually present at birth, and the young infant might succumb rapidly to renal failure
1799
Q

What are the genetic associations with childhood PCKD? Where is it? What does it do?

A
  • In most cases, the disease is caused by mutations of the PKHD1 gene, which maps to chromosome region 6p21-p23
  • The gene is highly expressed in adult and fetal kidney and also in liver and pancreas
  • The PKHD1 gene encodes fibrocystin. Like polycystin 1 and 2, fibrocystic also has been localised to the primary cilim of tubular cells
  • The function of fibrocystic is unknown, but its putative conformational structure indicates it may be a cell surface receptor with a role in collecting duct and biliary differentiation.
1800
Q

What is the gross morphology of childhood PCKD?

A
  • The kidneys are enlarged and have a smooth external appearance
  • On cut section, numerous small cysts in the cortex and medulla give the kidney a spongelike appearance
  • Dilated elongated channels are present at right angles to the cortical surface, completely replacing the medulla and cortex
1801
Q

What are the microscopic findings in childhood PCKD?

A
  • On microscopic examination, there is cylindrical or, less commonly, saccular dilation of all collecting tubules
  • The cysts have a uniform lining of cuboidal cells, reflecting their origin from the collection ducts
  • In almost all cases, the liver has cysts associated with portal fibrosis and proliferation of portal bile ducts
1802
Q

What are the liver changes that occur in childhood PCKD?

A
  • Patients who survive infancy (infantile and juvenile forms) may develop a peculiar hepatic injury characterised by bland periportal fibrosis and prolfieration of well-differentiated biliary ductules, now termed congenital hepatic fibrosis
  • In older children, hepatic disease is the predominant clinical concern. Such patients may develop portal hypertension with splenomegaly
  • Curiously, congenital fibrosis sometimes occurs in the absence of polycystic kidney disease or has been reported in the presence of adult polycystic kidney disease
1803
Q

There are three major types of medullary cystic disease. What are they and how common are they?

A
  • Medullary sponge kidney - relatively common and usually innocuous structural change
  • Nephronophthisis and adult-onset medullary cystic disease, which are almost always associated with renal dysfunction
1804
Q

What id medullary sponge kidney? What are the morphological changes?

A
  • The term medullary sponge kidney is restricted to multiple cystic dilations of the collecting ducts in the medulla
  • The condition occurs in adults and is usually discovered radiographically. Renal function is usually normal
  • On gross inspection the papillary ducts in the medulla are dilated and small cysts may be present
  • The cysts are lined by cuboidal epithelium or occasionally by transitional epithelium
  • Unless there is superimposed pyelonephritis, cortical scarring is absent
1805
Q

What is the pathogenesis of nephronophthisis and adult-onset medullary cystic disease?

A
  • These groups of progressive renal disorders are characterised by variable number of cysts in the medulla, usually concentrated at the corticomedullary junction.
  • Initial injury probably involves the distal tubules with tubular basement membrane disruption, followed by chronic and progressive tubular atrophy involving both medulla and cortex and interstitial fibrosis
  • Although the medulla cysts are important, the cortical tubulointerstitial damage is the cause of the eventual renal insufficiency
1806
Q

Three variants of the nephronophthisis disease complex are recognised. What are they? How are they inherited?

A

1) Sporadic, non-familial
2) Familial, juvenile nephronophthisis (most common)
3) Renal-retinal dysplasia (15%) in which the kidney disease is accompanied by ocular lesions.

The familial forms are inherited as autosomal recessive traits and usually become manifest in childhood or adolescence.

1807
Q

What are the common presentations in children with nephronophthisis? What is the prognosis?

A
  • They present first with polyuria and polydipsia, which reflect a marked defect in concentrating ability of renal tubules
  • Sodium wasting and tubular acidosis are also prominent
  • Some syndromic variants of nephronophthisis (Joubert syndrome, Meckel Gruber syndrome) can have extrarenal associations, including ocular motor abnormalities, retinal dystrophy, liver fibrosis, and cerebellar abnormalities.
  • The expected course is progression to ESRD in 5-10 years
1808
Q

What genes have been associated with nephronophthisis? Where are they?

A
  • Sixteen responsible gene loci , NPHP1 and NPHP11 (that encode proteins called nephrocystins) are mutated in the juvenile forms of nephronophthisis and the list continually expands as additional loci that contribute to this ciliopathy are identified
  • These proteins are present in the primary cilia, basal bodies attached to these cilia, or the centrosome organelle form which the basal bodies originate
1809
Q

What are the genes associated with adult-onset medially cystic disease? How is it inherited?

A
  • Adult-onset medullary cystic disease has an autosomal dominant pattern of transmission
  • At one time it was considered to be part of the nephronophthisis spectrum, but based on its distinctive genetics it is now considered a separate entity
  • Mutations in two genes (MCKD1 and LCKD2) have been identified as causing medullary cystic disease, which is characterised by progression to end-stage kidney disease in adult life
1810
Q

What is the morphology of nephronophthisis?

A
  • The kidneys are small, have contracted granular surfaces, and show cysts in the medulla, most prominently at the corticomedullary junction. Small cysts are also seen in the cortex
  • The cysts are lined by flattened or cuboidal epithelium and are usually surrounded by either inflammatory cells or fibrous tissue
  • In the cortex, there is widespread atrophy and thickening of the tubular basement membranes, together with interstitial fibrosis.
  • In general, glomerular structure is preserved.
1811
Q

What is multicystic renal dysplasia? What is the gross morphology?

A
  • Dysplasia is a sporadic disorder that can be unilateral or bilateral and is almost always cystic.
  • The kidney is usually enlarged, extremely irregular, and multicystic. The cysts vary in size from several millimetres to centimetres in diameter
1812
Q

What are the histologic findings in multicystic renal dysplasia?

A
  • The cysts are lined by flattened epithelium
  • Although normal nephrons are present, many have immature collecting ducts
  • The characteristic histologic feature is the presence of islands of undifferentiated mesenchyme, often with cartilage, and immature collecting ducts.
1813
Q

What are the trigger, clinical features and prognosis of multicystic renal dysplasia?

A
  • Most cases are associated with ureteropelvic obstruction, ureteral agenesis or atresia, and other anomalies of the lower urinary tract
  • When unilateral, the dysplasia may mimic a neoplasm and lead to surgical exploration and nephrectomy
  • The opposite kidney functions normally, and such patients have an excellent prognosis after surgical removal of the affected kidney.
  • In bilateral multicystic renal dysplasia, renal failure may ultimately result
1814
Q

What is acquired (dialysis-associated) cystic disease? Why does It happen? How does it present? What are the complications?

A
  • Patients with end-stage renal disease who have undergone prolonged dialysis sometimes show numerous cortical and medullary renal cysts.
  • The cysts measure 0.1-4cm in diameter, contain clear fluid, are lined by either hyperplastic or flattened tubular epithelium, and often contain calcium oxalate crystals
  • The probably form as a result of obstruction of tubules by interstitial fibrosis or by oxalate crystals
  • Most are asymptomatic, but sometimes the cysts bleed, causing haematuria
    *There is a 12-18 fold increased risk of RCC, which develops in 7% of dialysed patients observed for 10 years
1815
Q

What are simple kidney cysts? How big are they? What are they filled with and what are the microscopic findings?

A
  • Simple cysts may be single or multiple and usually involve the cortex. They are commonly 1-5cm, but may reach 10cm or more in size
  • They are translucent, lined by a grey, glistening, smooth membrane, and filled with clear fluid
  • On microscopic examination, these membranes are composed of a single layer of cuboidal or flattened cuboidal epithelium, which is many instances may be completely atrophic
1816
Q

What are the clinical and radiographic findings with simple cysts?

A
  • Simple cysts are common post-mortem findings without clinical significance.
  • On occasion, haemorrhage into them may cause sudden distension and pain, and calcification of the haemorrhage may give rise to bizarre radiographic shadows.
  • The main importance of cysts lie in their differentiation from kidney tumours
  • Radiologic studies show that in contrast to renal tumours, renal cysts have smooth contours, are almost always avascular, and give fluid rather than solid signals on US
1817
Q

What are the complications of obstructive lesions of the urinary tract?

A

Increased susceptibility to infection and to stone formation, and unrelieved obstruction almost always leads to permanent atrophy, termed hydronephrosis or obstructive uropathy. Fortunately, many causes of obstruction are surgically correctable or medically treatable

1818
Q

What are the different types of urinary tract obstructions? What are the different categories of causes?

A
  • They can be sudden or insidious, partial or complete, unilateral or bilateral; it may occur at any level of urinary tract from the urethra to the renal pelvis.
  • It can be caused by intrinsic lesions or the urinary tract or extrinsic lesions that compress the ureter
1819
Q

What are the common causes of urinary tract obstruction?

A
  • Congenital abnormalities: posterior urethral valves and urethral strictures, metal stenosis, bladder neck obstruction, ureteropelvic junction narrowing o obstruction, severe viscoureteral reflux
  • Urinary calculi
  • **Benign prostatic hypertrophy
  • Tumours: carcinoma of the prostate, bladder tumours, contiguous malignant disease (retroperitoneal lymphoma), carcinoma of the cervix or uterus
  • Inflammation: prostatitis urethritis, urethritis, retroperitoneal fibrosis
  • **Sloughed papillae or blood clots
  • **Pregnancy
  • **Uterine prolapse and cystocele
  • Functional disorders: neurogenic (spinal cord damage or diabetic nephropathy) and other functional abnormalities of the ureter or bladder (often termed dysfunctional obstruction)
1820
Q

What is hydronephrosis?

A

Hydronephrosis is the term used to describe dilation of the renal pelvis and calyces associated with progressive atrophy of the kidney due to obstruction to the outflow of the urine.

1821
Q

Why does hydronephrosis happen?

A
  • Even with complete obstruction, glomerular filtration persists for some some time because the filtrate subsequently diffuses back into the renal interstitium and perirenal spaces, from where it ultimately return to the lymphatic and venous systems
  • Because of this continued filtration, the affected calyces and pelvis become dilated, often markedly so.
  • The high pressure in the pelvis is transmitted back through the collecting ducts into the cortex, causing renal atrophy, but it also compresses the renal vasculature of the medially, causing a diminution in inner medially blood flow.
1822
Q

In hydronephrosis, at what stages do different mechanisms cause injury?

A
  • The medially vascular defects are initially reversible, but lead to medullary function disturbances
  • Accordingly, the initial functional alterations caused by obstruction are largely tubular, manifested primarily by impaired concentrating ability.
  • Only later does the GFR begin to fall
  • Obstruction also triggers an interstitial fibrosis, by similar mechanisms similar to those you already know
1823
Q

How does the morphology change when a urinary tract obstruction is sudden and complete opposed to subtotal or intermittent?

A
  • When the obstruction is sudden and complete, it leads to mild dilation of the pelvis and calyces and sometimes to atrophy of the renal parenchyma.
  • When the obstruction is subtotal or intermittent, progressive dilation ensues, giving rise to hydronephrosis.
  • Depending on the level of urinary block, the dilation may affect the bladder first, or the ureter and then the kidney
1824
Q

In urinary tract obstruction, the kidneys may be slightly to massively enlarged, depending on the degree and the duration of the obstruction. How does the gross morphology change over time with hydronephrosis?

A
  • The earlier features are those of simple dilation of the pelvis and calyces, but in addition there is often significant interstitial inflammation, even in the absence of infection
  • In chronic cases the picture is one of cortical tubular atrophy with marked diffuse interstitial fibrosis. Progressive blunting to the apices of the pyramids occurs, and these eventually become cupped.
  • In far-advanced cases, the kidney may become transformed into thin-walled cystic structure have a diameter of up to 15-20cm with striking parenchymal atrophy, total obliteration of the pyramids, and thinning of the cortex
1825
Q

What are the clinical features of acute urinary obstruction?

A
  • It may provoke pain attributed to distension of the collecting system or renal capsule.
  • Most of the early symptoms are produced by the underlying cause of the hydronephrosis
  • Thus, calculi lodged in the ureters may give rise to renal colic, and prostatic enlargements ma give rise to bladder symptoms
1826
Q

What are the clinical features of unilateral complete urinary tract obstruction or partial hydronephrosis?

A
  • It may remain silent for long periods, since the unaffected kidney can maintain adequate renal function.
  • Sometimes its existence first becomes apparent in the course of imaging studies
  • In its early stages, perhaps the first few weeks, relief of obstruction leads to reversion to normal function
1827
Q

What are the clinical features of bilateral partial urinary tract obstruction?

A
  • The earliest manifestation is inability to concentrate urine, reflected by polyuria and nocturia
  • Some patients develop distal tubular acidosis, renal salt wasting, secondary renal calculi, and chronic tubulointerstitial nephritis with scarring and atrophy of the papilla and medulla.
  • HTN is common
1828
Q

What are the clinical features of complete bilateral urinary tract obstruction?

A
  • Rapid onset results in oliguria or anuria and is incompatible with survival unless the obstruction is relieved.
  • Curiously, after relief of complete urinary tract obstruction, post-obstructive diuresis occurs
  • This can often be massive, with the kidney excreting large amounts of urine that is rich in sodium chloride
1829
Q

How common is urolithiasis? Where do the stones form? Who does it affect?

A
  • Urolithiasis affects 5-10% of Americans in their lifetime and the stones may form anywhere in the urinary tract, but most arise in the kidney.
  • Men are affected more often than women, and the peak ago of onset is between 20-30 years.
  • Familial and hereditary predisposition to stone formation has long been known
  • Many inborn errors of metabolism, such as cystinuria and primary hyperoxaluria, provide examples of hereditary disease characterised by excessive production and excretion of stone-forming substances
1830
Q

There are four main types of renal calculi. What are they?

A

1) Calcium stones (about 70%), composed largely of calcium oxalate or calcium oxalate mixed with calcium phosphate
2) another 15% are so-called triple stones or struvite stones composed of magnesium ammonium phosphate
3) 5-10% are uric acid stones
4) 1-2% are made up of cystine

An organic mucoprotein matrix, making up 1-5% of the stone but weight, is present in all calculi

1831
Q

Although there are many causes for the initiation and propagation of stones. What is the most important determinant?

A

An increased urinary concentration to the stones’ constituents, such that it exceeds their solubility (supersaturation).

1832
Q

What diseases are associated with calcium oxalate stones?

A
  • They are associated in about 5% of patients with hypercalcaemia and hypercalciuria, such as occurs in hyperparathyroidism, diffuse bone disease, sarcoidosis, and other hypercalcaemic states.
  • About 55% have hypercalciuria without hypercalcaemia. This is caused by several factors, including hyper absorption of calcium from the interstine (absorptive hypercalciuria), an intrinsic impairment in renal tubular reabsorption of calcium (renal hypercalciuria), or idiopathic fasting hypercalciuria with normal parathyroid function
  • As many as 20% of calcium oxalate stones are associated with increased uric acid secretion (*hyperuricosuric calcium nephrolithiasis), with or without hypercalciuria. The mechanism of stone formation in this setting involved “nucleation” of calcium oxalate by urea acid crystals in the collecting ducts
  • 5% are associated with hyperoxaluria, either hereditary (primary oxaluwria) or, more commonly, acquired by intestinal overabsorption in patients with enteric diseases
1833
Q

When and how are magnesium ammonium phosphate renal stones formed?

A
  • They are formed largely after infections by urea-splitting bacteria (eg, Proteus and some staphylococci) that convert urea to ammonia
  • The resultant alkaline urine causes the precipitation of magnesium ammonium phosphate salts.
  • These form some of the largest stones, so the amount of urea excreted normally is very large
  • Indeed, so-called staghorn calculi occupying large portions of the renal pelvis are frequently a consequence of infection
1834
Q

When and how do uric acid stones form?

A
  • They are common individuals with hyperuricaemia, such as patients with gout, and diseases involving rapid cell turnover, such as the leukaemias
  • However, more than half of all patient with uric acid calculi have neither hyperuricaemia nor increased urinary excretion of uric acid
  • In this group, it is thought that a tendency to excrete urine of pH below 5.5 may predispose to uric acid stones, because uric acid is insoluble in acidic urine.
  • In contrast to the radiopaque calcium stones, uric acid stones aer radiolucent
1835
Q

What causes cystine renal stones?

A
  • They are caused by genetic defects in the renal reabsorption of amino acids, including cystine, leading to cystinuria.
  • These stones also form at low urinary pH
1836
Q

What are some factors that influence the formation of calculi?

A
  • Increased concentrations of stone constituents
  • Changes in urinary pH
  • Decreased urine volume
  • The presence of bacteria

It has been postured that stone formation is enhanced by a deficiency in inhibitors of crystal formation in urine. This list of such inhibitors is long, including pyrophosphate, diphosphonate, nephrocalcin,

1837
Q

Where are stones often found? What is their gross morphologic appearance?

A
  • Stones are unilateral in about 80% of patients.
  • The favoured sites for their formation are within the renal calyces and pelves and in the bladder.
  • If formed in the renal pelvis they tend to remain small, having an average diameter of 2-3mm. These may have smooth contours or may take the form of an irregular jagged mass of spicules.
  • Often many stones are found within one kidney
  • On occasion, progressive accretion of salts leads to the development of branching structures known as staghorn calculi, which create a cast of the pelvic and calyceal system.
1838
Q

What are the clinical features of urolithiasis?

A
  • They may be asymptomatic, produce severe renal colic and abdominal pain, or may cause significant renal damage
  • Larger stones often manifest themselves by haematuria
  • Stones also predispose to superimposed infection, both by their obstructive nature and by the trauma they provide
1839
Q

What are some benign renal neoplasms?

A
  • Renal papillary adenom
  • Angiomyolipoma
  • Oncocytoma
1840
Q

What is a renal papillary adenoma?

A

It is a small discrete adenomas arising from the renal tubular epithelium are found commonly (7-22%) at autopsy. They are most frequently papillary and are therefore called papillary adenomas

1841
Q

What are the morphologic characteristics of renal papillary adenomas?

A
  • There are small tumours, usually less than 0.5cm in diameter
  • They are present invariably within the cortex and appear grossly as pale yellow-grey, discrete, well-circumscribed nodules
  • On microscopic examination, they are composed of complex, branching, papillomatous structures with numerous complex fronds.
  • Cells may also grow as tubules, glands, cords, and sheets of cells. The cells are cuboidal to polygonal in shape and have regular, small central nuclei, scanty cytoplasm and no atypia
1842
Q

How do renal papillary adenomas differ from low-grad papillary renal cell carcinoma?

A
  • By histologic criteria, these tumours do not differ from low-grade papillary renal cell carcinoma and indeed share some immunohistochemical and cryogenic features (trisomies 7 and 17) with papillary cancers
  • The size of the tumour is used as a prognostic feature, with a cutoff of 3cm separating those that metastasise from those that rarely do.
  • However, because of occasional reports of small tumours that have metastasised the current view is to regard all adenomas, regardless of size, as potentially malignant
1843
Q

What is an angiomyolipoma? What is it associated with? What is the clinical importance of them?

A
  • It is is a benign neoplasm consisting of vessels, smooth muscle, and fat originating from perivascular epithelia cells.
  • They are present in 25-50% of patients with tuberous sclerosis, a disease caused by loss-of-function mutations in the TSC1 or TSC2 tumour suppressor genes
  • Tuberous sclerosis is characterised by lesions of the cerebral cortex that produce epilepsy and learning difficulties, a variety of skin abnormalities, and unusual benign tumours at other sites, such as the heart
  • The clinical important of angiomyolipoma is due largely to they susceptibility to spontaneously haemorrhage
1844
Q

What is an oncocytoma? What is its gross and microscopic morphology?

A
  • This is an epithelial neoplasm composed of large eosinophilic cells having small, round, benign-appearing nuclei that have large nucleoli.
  • It is thought to arise from the intercalated cells of collecting ducts, and accounts for approximately 5-15% of renal neoplasms
  • Ultrastructurally the eosinophilic cells ahem numerous mitochondria
  • In gross appearance, the tumours are tan or mahogany brown, relatively homogenous, and usually well encapsulated with a central scar in 1/3rd of cases.
  • However, they may achieve a large size (up to 12cm in diameter)
1845
Q

How common is renal cell carcinoma? Who does it often happen in?

A
  • It represents about 3% of all newly diagnosed cancers in the US and accounts for 85% of renal cancers in adults
  • The tumours occur most often in older individuals, usually in sixth and seventh decades of life, and show a 2:1 male preponderance
1846
Q

What are the factors that increase your risk of renal cell carcinoma?

A
  • Tobacco is the most significant risk factor. Cigarette smokers have double the incidence of RCC, and pipe and cigar smokers are also more susceptible
  • An international study has identified additional risk factors, including obesity (particularly in women); hypertension; unopposed oestrogen therapy; and exposure to asbestos, petroleum products, and heavy metals
  • There is also an increased risk in patients with ESRD, CKD, acquired cystic disease and tuberous sclerosis.
1847
Q

Most renal cancer is sporadic, but unusual forms of autosomal dominant familial cancers occur, usually in younger individuals. Although they account for only 4% of renal cancers, familial variants have been instructive in understanding renal carcinogenesis. What are some conditions associated with RCC?

A
  • Von Hippel-Lindau (VHL) syndrome - half to 2/3rd of individuals with VHL develop renal cysts and bilateral, often multiple RCCs
  • Hereditary leiomyomatosis and renal cell cancer syndrome - this autosomal dominant disease is caused by mutations of the FH gene, which expresses fumarate hydratase, and is characterised by cutaneous and uterine leiomyomata and an aggressive type of papillary carcinoma with increased propensity for metastatic spread.
  • Hereditary papillary carcinoma - this autosomal dominant form is manifested by multiple bilateral tumours with papillary histology. These tumours show a series of cytogenic abnormalities and mutations in the MET port-oncogene
  • Birt-Hogg-Dube syndrome - the autosomal dominant inheritance pattern of this disease is due to mutations involving the BHD gene, which expresses follicular.
1848
Q

The classification of renal cell carcinoma is based on correlative cytogenic, genetic, and histologic studies of both familial and sporadic tumours. What are the major types of tumour?

A
  • Clear cell carcinoma (70-80%)
  • Papillary carcinoma
  • Chromophone carcinoema
  • Xp11 translocation carcinoma
  • Collecting duct (Bellini duct) carcinoma
1849
Q

What cells are clear cell carcinomas made of? Are they inherited? What gene is associated with them?

A
  • The tumours are made up of cells with clear or granular cytoplasm and are non-papillary
  • They can be familial but in most cases (95%) are sporadic.
  • In 98% of these tumours, there is loss of sequences on the short arm of chromosome 3. The deleted region harbours the VHL gene, which acts as a tumour suppressor gene in both sporadic and familial cancers.
1850
Q

What does the VHL gene that is often deleted in clear cell carcinomas do? What happens when it is inactive?

A
  • The VHL gene encodes a protein that is part of a ubiquitin ligase complex involved in targeting other proteins for degradation.
  • Important among the targets of the VHL protein is the transcription factor hypoxia-inducible factor-1 (HIF1).
  • When VHL is inactive, HIF-1 level remains high, even under normoxic conditions, causing inappropriate expression of a number of genenes that are turned on by HIF. These include genes that promote angiogenesis, such as VEGF, and genes that stimulate cell growth, scubas insulin-like growth factor (IGF-1).
  • In addition, HIF collaborates in complex ways with the oncogenic factor MYC to “reprogram” cellular metabolism in a way that favour growth.
1851
Q

How common is papillary renal carcinoma? What genes is it associated with? What does this gene do?

A
  • Papillary carcinoma accounts for 10-15% of renal cancers.
  • It is characterised by a papillary growth pattern and also occurs in both familial and sporadic forms. * These tumours are NOT associated with 3p deletions. The most common cytogenic abnormalities are trisomies 7 and 18, and loss of Y in male patients in the sporadic form, and trisomy 7 in the familial form.
  • The gene on chromosome 7 for the familial form has been mapped to MET, a proton-oncogene that encodes the tyrosine kinase receptor for hepatocyte growth factor. MET is also mutated in a small proportion of sporadic papillary carcinomas.
  • Hepatocyte growth factor (also called scatter factor) mediates growth, cell, mobility, invasion and morphogenetic differentiation.
1852
Q

How does the distribution of papillary carcinomas differ to clear cell carcinomas?

A

Unlike clear cell carcinomas, papillary carcinomas are frequently multifocal in origin

1853
Q

How common are chromophore carcinomas? What is their morphology, pathogenesis and prognosis?

A
  • Chromophobe carcinoma represented 5% of renal cell cancers and is composed of cells with prominent cell membranes and pale eosinophilic cytoplasm, usually with a halo around the nucleus
  • On cytogenic examination thee tumours show multiple chromosome losses and extreme hypodiploidy.
  • Like the benign oncocytoma, they are thought to grow from intercalated cells of collecting ducts and have an excellent prognosis compared with that of the clear cell and papillary cancers.
1854
Q

What is Xp11 translocation renal carcinoma? Who does it occur in? What is the morphology?

A
  • It is a genetically distinct subtype of RCC. It often occurs in young patients and is defined by translocations of the TFE3 gene located at Xp11.2 with a number of partner genes, all of which result in overexpression of the TFE3 transcription factor. The neoplastic cells consist of clear cytoplasm with a papillary architecture
1855
Q

How common is collecting duct (Bellini duct) carcinoma? What is their histology? What is it similar to?

A
  • It represents approximately 1% of less of renal epithelial neoplasms.
  • They arise from collecting duct cells in the medulla.
  • Histologically these tumours characterised by malignant cells forming glands enmeshed within a prominent fibrotic stroma, typically in a medullary location.
  • Medullary carcinoma is a morphologically similar neoplasm that is seen in patients with sickle cell trait
1856
Q

Where in the kidney does RCC usually affect?

A

RCCs may arise in any portion of the kidney, but more commonly affects the poles

1857
Q

What is the gross morphology of clear cell carcinomas? Where do they arise from?

A
  • They most likely arise from proximal tubular epithelium, and usually occur as solitary unilateral lesions
  • They are bright yellow-gray-white spherical masses of variable size distort the renal outline.
  • The yellow colour is a consequence of the prominent lipid accumulations in tumour cells.
  • There are commonly large areas of grey-white necrosis and foci of haemorrhagic discolouration.
  • The margins are usually sharply defined and confined within the renal capsule
  • The growth pattern varies from solid to trabecular (cord-like) or tubular (resembling tubules)
1858
Q

What are the microscopic findings in clear cell carcinoma?

A
  • The tumour cells have a rounded or polygonal shape and abundant clear or granule cytoplasm, which contains glycogen and lipids
  • The tumours have delicate branching vasculature and may show cystic as well as solid areas.
  • Most tumour are well differentiated, but some show nuclear atypia with bizarre nuclei and giant cells.
1859
Q

As RCC tumours enlarge, what do they bulge and invade into ?

A
  • As tumours enlarge that may bulge into the calyces and pelvis and eventually fungate through the walls of the collecting system to extend into the ureter.
  • One of the striking characteristics of renal cell carcinoma is its tendency to invade the renal vein, in which is may grow as a solid column of cells that extends up the inferior vena cava, sometimes as far as the right side of the heart
1860
Q

What are the morphological changes in papillary carcinomas? Both gross and microscopic! What do they arise from?

A
  • They are thought to arise from distal convoluted tubules, can be multifocal or bilateral.
  • They are typically haemorrhagic and cystic, especially the large
  • The tumour is composed of cuboidal or low columnar cells arranged papillary formations.
  • Interstitial foam cells are common in the papillary cords.
  • Psammoma bodies may be present. The stroma is usually scanty but highly vascularised
1861
Q

What is the morphology of chromophobe renal carcinoma?

A

It is made up of pale eosinophilic cells, often with a perinuclear halo, arranged in solid sheets with a concentration of the largest cells around the blood vessels

1862
Q

What are the classic clinical features of renal cell carcinoma?

A
  • Costovertebral pain, palpable mass, and haematuria, but all three are seen in only 10% of cases
  • The most reliable clue is haematuria, but it is usually intermittent and may be microscopic; thus, the tumour may remain silent until it attains a large size, often greater than 10cm
  • At this time it is often associated with generalised constitutional symptoms, such as fever, malaise, weakness, and weight loss.
  • This pattern of asymptomatic growth occurs in many patients, so the tumour may have reached a diameter of more than 10cm when it is discovered.
1863
Q

Renal cell carcinoma is considered one of the great mimics in medicine, because it tends to produce a diversity of systemic symptoms not related to the kidney. What aer these?

A

In addition to fever and constitutional symptoms, RCC produce a number of syndromes ascribed to abnormal hormone production, including polycythaemia, hypercalcaemia, HTN, hepatic dysfunction, feminisation or masculinisation, Cushing syndrome, eosinophilia, leukemoid reactions, and amyloidosis

1864
Q

A particularly troublesome feature of renal cell carcinoma is its tendency to metastasise widely before giving rise to any local symptoms or signs. Where does it metastasise to?

A
  • In 25% of new patients with renal cell carcinoma, there is radiologic evidence of metastases at the time of presentation.
  • The most common locations of metastasis are the lungs (more than 50%) and bones (33%), followed in frequency by the regional lymph nodes, liver, adrenal and brain
1865
Q

What is the prognosis of RCC? What is the treatment?

A
  • The average 5-year survival rate of persons with RCC is about 70% and as high as 95% in the absence of distant metastases. With renal vein invasion or extension into the perinephric fat, the figure is reduced to approximately 60%.
  • Radical nephrectomy has been the treatment of choice, but partial nephrectomy to preserve renal function is recommended for T1a tumours (<4cm) as well as larger tumours when technically feasible.
  • Drugs that inhibit VEGF and various tyrosine kinases are used as an adjunct to therapy in patients with metastatic disease
1866
Q

Approximately 5-10% of primary tumours originate from the urothelium of the renal pelvis. What are some of these?

A

There tumours range from apparently benign papillomas to invasive urothelial (transitional cell) carcinomas

1867
Q

How do renal pelvic tumours present? What are they similar to histologically?

A

They usually become clinically apparent within a short time, because they lie within the pelvis and, by fragmentation, produce noticeable haemturia.
* They are almost invariably small when discovered
* They may block the urinary outflow and lead to palpable hydronephrosis and flank pain.
* On histologic examination, pelvic tumours are the exact counterpart of those found in the urinary bladder

1868
Q

What is the prognosis of urothelial tumours? Why?

A
  • Infiltration of the wall of the pelvis and calyces is common
  • For this reason, despite their apparently small, deceptively benign appearance, the prognosis for these tumours is not good
  • Reported 5-year survival rates vary from 50-100% for low-grade noninvasive lesions to 10% with high-grade infiltrating tumours
1869
Q

What diseases are associated with urothelial tumours?

A

Lynch syndrome and analgesic nephropathy

1870
Q

The liver has dual blood supply. Where from and what proportion? How do they enter the liver?

A
  • The portal vein provides 60-70% of hepatic blood flow
  • The hepatic artery supplying the remaining 30-40%
  • The portal vein and the hepatic artery enter the inferior aspect of the liver through the hilum, or porta hepatis
1871
Q

The most common terminology of the hepatic microarchitecture is based on the lobular model. What is this model?

A
  • The liver is divided into 1- to 2-mm in diameter lobules that are orientated around the terminal tributaries of the hepatic vein (terminal hepatic veins), with portal tracts at the lobule’s periphery.
  • The hepatocytes in the vicinity of the terminal hepatic vein are called “centrilobular”; those near the portal tract are “peri-portal”
1872
Q

How does the layout of the liver lobules enable well-mixed portal venous and hepatic arterial blood?

A
  • Within the lobule, hepatocytes are organised into anastomosing sheets or “plates” extending from portal tracts to the terminal hepatic veins. Between the trabecular plates of hepatocytes are vascular sinusoids.
  • Blood traverses the sinusoids and exits into the terminal hepatic veins through numerous orifices in the vein wall
  • Hepatocytes are thus bathed on two sides by well-mixed, portal venous and hepatic arterial blood.
1873
Q

The liver sinusoids are lined by fenestrated endothelial cells. What space is beneath this and what cells are in it?

A
  • Beneath the endothelial cells lies the space of Disse, into which protrude abundant hepatocyte microvilli.
  • Scattered Kuppfer cells of the mononuclear phagocyte system are attached to the luminal face of endothelial cells, and fat-containing myofibroblastic hepatic stellate cells are found in the space of Disse
1874
Q

Where does the bile travel and drain to in the liver lobules?

A
  • Between abutting hepatocytes are bile canaliculi, which are channels 1 to 2µm in diameter, formed by grooves in the plasma membranes of facing hepatocytes and separated from the vascular space by tight junctions.
  • These channels drain into the canals of Hering that, in turn, connect to bile ductules in the periportal region.
  • The ductules empty into the terminal bile ducts within the portal tracts.
1875
Q

What happens in hepatic necrosis? What diseases is it most common in?

A
  • The cell swells due to defective osmotic regulation at the cell membrane: fluid flows into the cell, which swells and ruptures.
  • Even before rupture, membrane blebs form, carrying off cytoplasmic contents (without organelles) into the extracellular compartment.
  • Macrophages cluster at such sites of injury and mark the sites of hepatocyte necrosis since the dying cells essentially burst and disappear.
  • This form of injury is the predominant mode of death in ischaemic/hypoxic injury and a significant part of the response to oxidative stress
1876
Q

Apoptotic hepatocytes were first described in yellow fever. While apoptosis occurs in many forms of liver disease, by convention apoptotic hepatocytes are called something else in other diseases. What is this? What other diseases does it occur in?

A

In more frequent settings in which apoptotic hepatocytes are seen (eg, acute and chronic hepatitis), the term acidophil bodies is used, due to their deeply eosinophilic staining characteristics

1877
Q

What is hepatic confluent necrosis? When does it happen? What cells are involved?

A
  • When there is widespread parenchymal loss there is often evidence of confluent necrosis, a severe, zonal loss of hepatocytes
  • This may be seen in acute toxic or ischaemic injuries or in severe viral or autoimmune hepatitis
  • Confluent necrosis may begin as a zone of hepatocyte dropout around the central vein
  • The resulting space is filled by cellular debris, macrophages, and remnants of the reticulin meshwork.
1878
Q

What happens in hepatic bridging necrosis?

A
  • In bridging necrosis this zone may link central veins to portal tracts (often with inapparent central vein within the zone of injury).
  • Even in diseases such as viral hepatitis in which hepatocytes are the principal targets of attack, vascular insults - via inflammation or thrombosis - lead to parenchymal extinction due to large areas of contiguous hepatocyte death.
1879
Q

How does regeneration of the hepatocytes occur?

A
  • Regeneration of lost hepatocytes occurs primarily by mitotic replication of hepatocytes adjacent to those that have died, even when there is significant confluent necrosis.
  • Hepatocytes are almost stem cell-like in their ability to continue to to replicate even in the setting of years of chronic injury and thus stem cell replenishment is usually not a significant part of parenchymal repair
1880
Q

What happens on a cellular level during the most severe forms of acute liver failure?

A
  • There is activation of the primary intrahepatic stem cell niche, namely the canal of Hering, but the contribution of stem cells to the replenishment of hepatocytes in such a setting remains unclear.
  • Eventually, however, in many individuals with chronic disease the hepatocytes do reach replicative senescence and then there is clear evidence of stem cell activation seen in the form of ductular reactions. These duct like structures, sometimes without any lumens develop from stem cells and contribute significantly to parenchymal restoration.
1881
Q

What is the principal cell type in hepatic scar deposition? What does it normally do? What happens when it’s activated?

A
  • The principal cell type involved in scar deposition is the hepatic stellate cell.
  • In its quiescent form, it is a lipid (vitamin A) storing cell
  • However, in several forms of acute and chronic injury, the stellate cells can become activated and are converted into highly fibrogenic myofibroblasts.
1882
Q

Proliferation of hepatic stellate cells and their activation into myofibroblasts is initiated by a series of changes. What are these?

A
  • An increase in the expression of platelet-derived growth factor receptor β (PDGFR-β) in the stellate cells
  • At the same time, Kupffer cells and lymphocytes release cytokines and chemokines that modulate the expression of genes in stellate cells that are involved in fibrogenesis. The include, transforming growth factor β (TGF-β) and its receptors, metalloproteinase 2 (MMP-2), and tissue inhibitors of metalloproteinases 1 and 2 (TIMP-1 and -2)
1883
Q

Once stellate cells are converted into myofibroblasts. What do they release and what do they do?

A
  • As they are converted to myofibroblasts, the cell release chemotactic and vasoactive factors, cytokines, and growth factors.
  • Myofibroblasts are contractile cells; their contraction is stimulated by endothelin-1 (ET-1)
1884
Q

The stimuli for stellate cell activation may originate from several sources. What are these?

A

1) Chronic inflammation, with production of inflammatory cytokines such as tumour necrosis factor (TNF), lymphotoxin and IL-1β, and lipid peroxidation products
2) Cytokine and chemokine production by Kupffer cells, endothelial cells, hepatocytes, and bile duct epithelial cells
3) In response to disruption of the ECM
4) Direct stimulation of stellate cells by toxins

1885
Q

Where in the liver does scar deposition take place? What diseases does it occur in?

A
  • If injury persists, scar deposition begins, often in the space of Disse
  • This is particularly important in alcoholic and non-alcoholic fatty liver diseases, but it also a generalised mechanism of scar formation in other forms of chronic liver injury
1886
Q

How are fibrous septa created in hepatic scar formation? What happens following this?

A
  • Zones of parenchymal loss transform into dense fibrous septa through a combination of the collapse of the underlying reticulin where large swaths of hepatocytes have irrevocably disappeared and hepatic stellate cells have been activated.
  • Eventually, these fibrous septa encircle surviving, regenerating hepatocytes in the late stages of chronic liver diseases that give rise to diffuse scarring described as cirrhosis
1887
Q

What happens if the chronic injury leading to hepatic scar formation is interrupted (eg, clearance of hepatitis virus infection, cessation of alcohol use)?

A
  • Stellate cell activation ceases, scars condense, becoming more dense and thin, and then, due to metalloproteinases produced by hepatocytes, begin to break apart.
  • In this way, scar formation can be reversed.
  • It should be kept in mind that in any chronic liver disease there are probably areas of both fibrotic progression and regression, but the balance in active disease favours the former and with remission of disease the latter is favoured.
1888
Q

What is acute liver failure?

A

It is defined as an acute liver illness associated with encephalopathy and coagulopathy that occurs within 26 weeks of the initial liver injury in the absence of pre-existing liver disease

1889
Q

What causes acute liver failure? How do the time frames differ with different causes?

A
  • It is caused by massive hepatic necrosis, most often induced by drugs or toxins.
  • Accidental or deliberate ingestion of acetaminophen accounts for almost 50% of cases in the US, while autoimmune hepatitis, other drugs/toxins and acute hepatitis A and B infections account for the rest
  • With acetaminophen toxicity, the liver failure occurs within a week of the onset of symptoms, whereas failure due to hepatitis viruses takes longer to develop.
1890
Q

What is the morphology of acute liver failure?

A
  • Acute liver failure usually displays massive hepatic necrosis, with broad regions of parenchymal loss surrounding islands of regenerating hepatocytes.
  • These livers are small and shrunken. The prominence of scar and of ductular reactions in these livers depends on the nature and duration of the insult
1891
Q

How does the morphology change in acute liver failure caused by acetaminophen overdose and hepatitis?

A
  • Toxic injuries, such as acetaminophen overdose, usually take place within hours to days, too brief a period to allow time for scar formation or regeneration
  • Acute viral infections may cause failure over weeks to a few months, so that while hepatocyte injury continues to outpace repair, regeneration is often demonstrable. Also this time scale allows for early scarring in areas of parenchymal loss
1892
Q

Apart from acetaminophen overdose and hepatitis what are the rare causes of acute liver failure? What is the morphology of these causes?

A
  • Rarely, there may be diffuse poisoning of liver cells without obvious cell death and parenchymal collapse, such as in diffuse micro vesicular steatosis related to fatty liver of pregnancy or idiosyncratic reactions to toxins (e.g., valproate, tetracycline). In these settings, usually related to primary mitochondrial dysfunction, hepatocytes are unable to perform their usual metabolic functions.
  • In states of immunodeficiency such as untreated HIV or post-transplant, non-hepatotropic viruses, particularly CMV, HSV, and adenovirus, can cause fulminant liver failure with histologic features specific to each of those viruses.
1893
Q

How does acute liver failure first manifest clinically?

A
  • It manifests first with nausea, vomiting, jaundice, followed by life-threatening encephalopathy, and coagulation defects
1894
Q

How do serum liver transaminases change throughout the course of acute liver failure? Why?

A
  • Typically, serum liver transaminases are markedly elevated. The liver is initially enlarged due to hepatocyte swelling, inflammatory infiltrates and oedema; as parenchyma is destroyed, however, the liver shrinks dramatically.
  • Decline of serum transaminases as the liver shrinks if often not, therefore, a sign of improvement, but is rather an indication that there are few viable hepatocytes left; this suspicion is confirmed if there is worsening jaundice, coagulopathy, and encephalopathy.
1895
Q

What are some manifestations of acute liver failure?

A
  • Jaundice and cholestasis
  • Hepatic encephalopathy
  • Coagulopathy
  • Portal hypertension
  • Hepatorenal syndrome
1896
Q

Why do you get jaundice and cholestasis with acute liver failure?

A
  • Alterations of bile formation and flow become clinically evident as yellow discolouration of the skin and sclera (jaundice and icterus, respectively) due to retention of bilirubin, and as cholestasis due to systemic retention of not only bilirubin but also other solutes eliminated in bile.
  • In the setting of acute liver failure, there is classic yellowing of skin, sclerae, and mucous membranes; cholestasis increases the risk of life-threatening bacterial infection
1897
Q

What is hepatic encephalopathy? When and why does it happen? What blood test is associated with it?

A
  • Hepatic encephalopathy is a spectrum of disturbances in consciousness, ranging from subtle behavioural abnormalities, to marked confusion and stupor, to deep coma and death
  • It may progress over days, weeks, or months following acute injury
  • Associated fluctuating, neurologic signs including rigidity and hyperreflexia.
  • Astrexis, a particularly characteristic sign, is manifested as non-rhythmic, rapid extension-flexion movements of the head and extremities, best seen when the arm are held in extention with dorsiflexion wrists
  • Hepatic encephalopathy is regarded as a disorder of neurotransmission in the CNS and neuromuscular system.
  • Elevated ammonia levels in blood and the CNS correlate with impaired neuronal function and cerebral oedema.
1898
Q

Why do you get coagulopathy with acute liver failure?

A
  • The liver is responsible for production of vitamin K-dependent and independent clotting factors. Thus, with massively impaired hepatic synthetic function, coagulopathy develops.
  • Easy bruisability is an early sign of this process, which can lead to life-threatening or fatal intracranial bleeding.
  • The liver is also responsible for helping to remove activated coagulation factors from the circulation, and loss of this function in some instances can lead to DIC, further exacerbating the bleeding tendency
1899
Q

Why do you get portal hypertension in acute liver failure? Does it always happen?

A
  • It arises with there is diminished flow through the portal venous system, which may occur because of obstruction at the pre-hepatic, intrahepatic and post-hepatic.
  • While it can occur in acute liver failure, it is more commonly seen with chronic liver failure
  • In acute liver failure, if portal hypertension develops within days to weeks, obstruction is predominantly intrahepatic and the major clinical consequences are ascites and hepatic encephalopathy.
1900
Q

What is hepatorenal syndrome?

A
  • It is a form of renal failure occurring in individuals in liver failure in whom there are no intrinsic morphologic or functional causes for kidney dysfunction
  • Sodium retention, impaired free-water excretion, and decreased renal perfusion and glomerular filtration rate are the main renal functional abnormalities
  • There is decreased renal perfusion pressure due to systemic vasodilation, activation of the renal sympathetic nervous system with vasoconstriction of the afferent renal arterioles, and increased activation of the renin-angiotensin axis, causing vasoconstriction that further decreases glomerular filtration.
  • The syndrome’s onset begins with a drop in urine output and rising blood urea nitrogen and creatinine levels
1901
Q

What are the leading causes of chronic liver failure?

A

Worldwide, they include chronic hepatitis B, chronic hepatitis C, NAFLD, and alcoholic liver disease

1902
Q

Liver failure in chronic liver disease is most often associated with cirrhosis. What is cirrhosis?

A

It is a condition marked by the diffuse transformation of the entire liver into regenerative parenchymal nodules surrounded by fibrous bands and variable degrees of vascular (often portosystemic) shunting

1903
Q

Not all cirrhosis leads inexorably to chronic liver failure and not all end-stage chronic liver disease is cirrhotic. Give some examples of both

A
  • For example, chronic diseases such as primary biliary cirrhoses, primary sclerosing cholangitis, nodular regenerative hyperplasia, chronic schistosomiasis and fibropolycystic liver disease are often not accompanied by fully established cirrhosis, even at end stage
  • On the other hand, patients with well-treated autoimmune hepatitis or those with suppressed hepatitis B or cured hepatitis C often do not progress to end stage, even though they are cirrhotic.
1904
Q

What is the Child-Pugh classification of cirrhosis?

A

Class A (well compensated)
Class B (partially decompensated)
Class C (decompensated)

1905
Q

What does all cirrhosis have morphologically in common and how is it different?

A
  • Cirrhosis occurs diffusely throughout the liver, which is comprised of regenerating parenchymal nodules surrounded by dense bands of scar and variable degrees of vascular shunting
  • The size of the nodules, the pattern of scarring (linking portal tracts to each other vs linking portal tracts to central veins), the degree of parenchymal collapse in which no viable liver tissue present, the range of macroscopic vascular thrombosis (particularly of the portal vein) all vary between diseases and even, in some cases, between individuals with the same disease
1906
Q

What biopsy results show good and bad prognosis?

A
  • Biopsy specimens demonstrating narrow, densely compacted fibrous septa separated by large islands of intact hepatic parenchyma are likely to have less portal hypertension
  • Those with broad bands of dense scar, often with dilated lymphatic spaces, with less intervening parenchyma are likely to be progressing toward portal hypertension and therefore, to end-stage disease
1907
Q

In chronic liver disease ductular reactions increase with advancing stage of disease and are usually most prominent in cirrhosis. There are two correlates of ductular reactions. What are they?

A
  • The role of liver stem cells in parenchymal regeneration increases as the preexisting hepatocytes undergo replicative senescence after years to decades of high turnover
  • Ductular reactions may incite some of the scarring in chronic liver disease and thus may have a negative effect on progressive liver disease
1908
Q

Although uncommon regression of fibrosis, albeit rarely, in fully established cirrhosis, does occur. What are the morphologic changes you would see?

A
  • Scars can become thinner, more densely compacted, and eventually fragment
  • As fibrous senate break apart, adjacent nodules of regenerating parenchyma coalesce into larger islands
  • All cirrhotic livers show elements of both progression and regression, the balance determined by the severity and persistence of the underlying disease
1909
Q

About 40% of individuals with cirrhosis are asymptomatic until the most advances stages of the disease. What are the symptoms when they are there? What are the ultimate causes of death?

A
  • When symptomatic, they present with non-specific manifestations: anorexia, weight loss, weakness, and, in advanced disease, symptoms and signs of liver failure
  • The ultimate causes of death in chronic liver failure, whether cirrhotic or not, include those seen in acute liver failure and additional outcomes such as development of HCC
  • Hepatic encephalopathy, bleeding for oesophageal varices and bacterial infections (resulting from damage to mucosal barrier in the gut and Kupffer cell dysfunction) are often the terminal events.
1910
Q

How is jaundice different in chronic liver failure to acute liver failure?

A
  • Jaundice, when chronic, can lead to pruritus, that is, itching, the intensity of which can be profound
  • Some patients may even scratch their skin raw and risk repeated bouts of potentially life-threatening infection
  • Pruritus can be so severe that it can be relieved only by liver transplantation
1911
Q

What are the outcomes of impaired oestrogen metabolism and consequent hyperoestrogenaemia from chronic liver disease?

A
  • It can give rise to palmar erythema (a reflection of local vasodilation) and spider angiomas of the skin
  • Each angioma is a central, pulsating, dilated arteriole from which small vessels radiate.
  • In men, it also leads to hypogonadism and gynaecomastia.
  • Hypogonadism can also occur in women from disruption of hypothalamic-pituitary axis function, either through nutritional deficiencies associaetd with the chronic liver disease or primary hormonal alterations.
1912
Q

Increased resistance to portal blood flow may develop in a variety of circumstances, which can be divided into pre-hepatic, intrahepatic and post-hepatic. What are examples of all of these?

A
  • The major pre-hepatic conditions are obstructive thrombosis, narrowing of the portal vein before it ramifies within the liver or massive splenomegaly with increased splenic vein blood flow
  • The main post-hepatic causes are severe right-sided heart failure, constrictive pericarditis, and hepatic vein outflow obstruction.
  • The dominant intrahepatic cause is cirrhosis, accounting for most causes of portal hypertension. Far less frequent intrahepatic causes are schistosomiasis, massive fatty change, diffuse fibrosing granulomatous disease such as sarcoidosis and diseases affecting the portal microcirculation such as nodular regenerative hyperplasia
1913
Q

The pathophysiology of portal hypertension is complex and involves resistance to portal flow at the level of the sinusoids and an increase in portal flow caused by hyperdynamic circulation. What causes the increased resistance to portal flow?

A
  • The increased resistance to portal flow at the level of the sinusoids is caused by contraction of vascular smooth muscle cells and myofibroblasts, and disruption of blood flow by scarring and the formation of parenchymal nodules.
  • Alterations in sinusoidal endothelial cells that contribute to the intrahepatic vasoconstriction associated with portal hypertension include a decrease in nitric oxide production, and increased release of endothelin-1 (ET-1), angiotensinogen, and eicosanoids.
  • Sinusoidal remodelling and anastomosis between the arterial and portal system in the fibrous septa contribute to portal hypertension by imposing arterial pressures on the low pressure portal venous system.
  • Sinusoidal remodelling and intrahepatic shunts also interfere with the metabolic exchange between sinusoidal blood and hepatocytes.
1914
Q

Alongside increased resistance to portal flow, another major factor in the development of portal hypertension is an increase in portal venous blood flow resulting from a hyperdynamic circulation. What causes this?

A
  • This is caused by arterial vasodilation, primarily in the splanchnic circulation
  • The increased splanchnic arterial blood flow in turn leads to increase venous efflux into the portal venous system.
  • While various mediators such as prostacyclin and TNF have been implicated in the causation of the splanchnic arterial vasodilation, nitric oxide has emerged as the most significant ones.
1915
Q

There are four major clinical consequences of portal hypertension. What are they?

A

1) Ascites
2) The formation of portosystemic venous shunts
3) Congestive splenomegaly
4) Hepatic encephalopathy

1916
Q

What are ascites? How much fluid usually has to accumulate before it becomes clinically detectable? What is the most common cause?

A
  • The accumulation of excess of fluid in the peritoneal cavity is called ascites
  • In 85% of cases, ascites is caused by cirrhosis
  • Ascites usually becomes clinically detectable when at least 500ml have accumulated
1917
Q

What type of fluid does ascites consist of? What cells are normally in it? What cells point to what diagnosis?

A
  • The is generally serous, having less than 3gm/dL of protein (largely albumin), and a serum to ascites albumin gradient of >1.1gm/dL.
  • The fluid may contain a scant number of mesothelial cells and mononuclear leukocytes.
  • Influx of neutrophils suggests infection, whereas the presence of blood cells points to possible disseminated intra-abdominal cancer
1918
Q

What is a complication of long-standing ascites?

A

With long-standing ascites, seepage of peritoneal fluid through trans-diaphragmatic lymphatics may produce hydro-thorax, more often on the right side.

1919
Q

The pathogenesis of ascites is complex. What are the mechanisms that cause it?

A
  • Sinusoidal hypertension, altering Starling’s forces and driving fluid into the space of Disse, from where it is removed by hepatic lymphatics; this movement of fluid is also promoted by hypoalbuminaemia
  • Percolation of hepatic lymph into the peritoneal cavity - normal thoracic duct lymph flow approximates 800-1000mL/day. With cirrhosis, hepatic lymphatic flow may approach 20L/day, exceeding thoracic duct capacity. Hepatic lymph is rich in proteins and low in triglycerides, which explains the presence of protein in the ascitic fluid.
    Splanchnic vasodilation and hyperdynamic circulation - arterial vasodilation in the splanchnic circulation tends to reduce arterial blood pressure. With worsening of the vasodilation, the heart rate and cardiac output are unable to maintain the blood pressure. This triggers the activation of vasoconstrictors, including the renin-angiotensin system, and also increases the secretion of ADH. The combination of portal hypertension, vasodilation, and sodium and water retention increases the perfusion pressure of interstitial capillaries, causing extravasation of fluid into the abdominal cavity.
1920
Q

How does portal hypertension cause portosystemic shunts?

A
  • With the rise in portal system pressure, the flow is reversed from portal to systemic circulation by dilation of collateral vessels and development of new vessels
  • Venous bypasses develop wherever the systemic and portal circulation share a common capillary beds.
  • Principal sites are veins around and within the rectum (manifest as haemorrhoids), the oesophagogastric junction (producing varices), the retroperitonium, and the falciform ligament of the liver (involving periumbilical and abdominal wall collaterals).
1921
Q

What is the complication of portosystemic shunts?

A
  • Although hemorrhoidal bleeding may occur, it is rarely massive or life-threatening.
  • Much more important are the oesophagogastric varices that appear in about 40% of individuals with advanced cirrhosis and cause massive haematemesis and death in about half of them. Each episode of bleeding is associated with a 30% mortality.
  • Abdominal wall collaterals appear as dilated subcutaneous veins extending from the umbilicus toward the rib margins (caput medusae) and constitute an important clinical hallmark of portal hypertension.
1922
Q

Long-standing moral congestion may cause congestive splenomegaly. What may this induce?

A

The massive splenomegaly may secondarily induce haematologic abnormalities attributable to hypersplenism, such as thrombocytopaenia or even pancytopenia.

1923
Q

What is hepatopulmonary syndrome? How common is it? What is unusual about the hypoxia?

A
  • It is seen in up to 30% patients with cirrhosis of the liver and portal hypertension.
  • These patients develop intrapulmonary vascular dilations involving capillary and pre-capillary vessels up to 100μM in size.
  • The blood flows rapidly through such dilated vessels, giving inadequate time for oxygen diffusion and leading to ventilation-perfusion mismatch and right-to-left shunting, manifesting as hypoxia.
  • Hypoxia and resultant dyspnoea occur preferentially in an upright position rather than in the recumbent position, as gravity exacerbates the ventilation-perfusion mismatch.
1924
Q

What is portopulmonary hypertension? What causes it? How does it manifest clinically?

A

*It refers to pulmonary arterial hypertension arising in liver disease and portal hypertension.
* Poorly understood, it seems to depend on concomitant portal hypertension and excessive pulmonary vasoconstriction and vascular remodelling.
* The most common clinical manifestations are dyspnoea on exertion and clubbing of the fingers

1925
Q

What happens in acute-on chronic liver failure?

A
  • There is often established cirrhosis with extensive vascular shunting
  • Thus, large volumes of functioning liver parenchyma have a borderline vascular supply, leaving them vulnerable to superimposed, potentially lethal insults.
1926
Q

What is the virus that causes acute hepatitis?

A

Hepatotropic viruses

1927
Q

What is the clinical course of Hepatitis A virus? What is the incubation period and prognosis?

A
  • Hepatitis A virus *HAV) is a usually benign, self-limited disease with an incubation period or 2-6 weeks.
  • HAV does not cause chronic hepatitis or a carrier state and only uncommonly causes acute hepatic failure, so the fatality rate associated with HAV is only about 0.1-0.3%
1928
Q

How common is HAV in developing and developed countries? What are the symptoms at the different ages?

A
  • HAV occurs through the world and is endemic in countries with poor hygiene and sanitation. Many individuals in these countries have detectable anti-HAV antibodies by the time they are 10 years old. Clinical disease tends to be mild or asymptomatic and is rare after childhood
  • In developed countries, the prevalence of seropositivity (indicative of previous exposure) increases gradually with age, reaching 50% by age 50 in the US. Affected individuals have non-specific symptoms such as fatigue and loss of appetite, and often develop jaundice.
1929
Q

What is the structure of hepatitis virus? Is there a virus?

A
  • HAV is a small, non enveloped positive-strand RNA picornavirus that occupies its own genus, Hepatovirus.
  • The receptor for HAV is HAVcr-1, a class I integral-membrane mucin-like glycoprotein.
  • HAV vaccine, available since 1992, is effective in preventing infection
1930
Q

How is hepatitis A virus spread? Who is therefore at risk?

A
  • HAV is spread by ingestion of contaminated water and foods and is shed in the stool for 2-3 weeks before and 1 week after the onset of jaundice
  • Thus, close personal contact with an infected individual or faecal-oral contamination during this period accounts for most cases and explains the outbreaks in institutional settings such as schools and nurseries, and in the water-borne epidemics in places where people live in overcrowded, unsanitary conditions.
1931
Q

What cellular factors affect any hepatocellular injury during HAV infection?

A

HAV itself does not seem to be cytopathic.
Cellular immunity, particularly CD8+ T cells, plays a key role in hepatocellular injury during HAV infection

1932
Q

How does the IgM antibody against hepatitis A change over time and compare to faecal shedding and IgG anti-HAV?

A
  • Specific IgM antibody against HAV appears with the onset of symptoms, constituting a reliable marker of acute infection.
  • Faecal shedding of the virus ends as the IgM titer rises.
  • The IgM response usually begins to decline in a few motnhs and is followed by the appearance of IgG anti-HAV
  • IgG persists for years, perhaps conferring lifelong immunity against reinfection by all strains of HAV.
  • Since there are no routinely available tests for IgG anti-HAV, the presence of IgG anti-HAV is inferred from the difference between total and IgM anti-HAV.
1933
Q

What are the different outcomes of hepatitis B virus?

A

1) Acute hepatitis followed by recovery and clearance of the virus
2) Non-progressive chronic hepatitis
3) Progressive chronic disease ending in cirrhosis
4) Acute hepatic failure with massive liver necrosis
5) An asymptomatic, “healthy carrier state

HBV-induced chronic liver disease is also an important precursor for the development of HCC even in the absence of cirrhosis

1934
Q

The mode of transmission of hepatitis B virus varies with geographic areas. How?

A
  • In high prevalence areas, transmission during childbirth accounts for 90% of cases.
  • In areas with intermediate prevalence, horizontal transmission, especially in early childhood, is the dominant mode of transmission. Such spread occurs through skin or mucous membranes among children with close bodily contact
  • In low prevalence areas, unprotected sex and IV drug use (sharing of needles) are the chief modes of spread.
1935
Q

What is the incubation period for hepatitis B virus? Does it remain in the blood?

A
  • HBV has a prolonged incubation period (2-26 weeks).
  • Unlike HAV, HBV remains in the blood until and during active episodes of acute and chronic hepatitis.
1936
Q

What proportion of people with HBV have symptoms? What is the outcome of most cases?

A
  • Approximately 65% of adult newly acquiring HBV have mild or no symptoms and do not develop jaundice.
  • The remaining 25% have non-specific constitutional symptoms such and anorexia, fever, jaundice and RUQ pain
  • In almost all cases the infection is self-limited and resolves without treatment. Chronic disease occurs in 5-10% of infected individuals. Fulminant hepatitis is rare, occurring in approximately 0.1-0.5% of acutely infected individuals
1937
Q

What is the structure of the hepatitis B virus?

A
  • The virus is a member of the Hepadnaviridae, a family of DNA viruses that cause hepatitis in multiple animal species.
  • There are 8 HBV genotypes that are distributed around the globe
  • The mature HBV vision is a spherical double-layered “Dane particle” that has an outer surface envelope of protein, lipid and carbohydrate enclosing an electron-dense, slightly hexagonal core.
1938
Q

The genome of HBV is a partially double-stranded circular DNA molecule having 3200 nucleotides with four open reading frames. What do those four open reading frames code for?

A
  • A nucleocapsid “core” protein (HBcAg) and a longer polypeptide transcript with a precore and core region, designated HBeAg. The precore region directs the secretion of the HBeAg polypeptide, whereas HbcAg remains in hepatocytes, where is participates in the assembly of complete virions
  • Envelope glycoproteins (HBsAg), which consists of three related proteins: large, middle and small HBsAg. Infected hepatocytes are capable of synthesising and secreting massive quantities of non-infective surface protein.
  • A polymerase (Pol) that exhibits both DNA polymerase activity and reverse transcriptase activity. Replication of the viral genome occurs via an intermediate RNA template, through a unique replication cycle: DNA -> RNA -> DNA
  • HBx protein, which is necessary for virus replication and may act as a transcriptional transactivator of both viral genes and a subset of host genes. It has been implicated in the pathogenesis of HCC in HBV infection
1939
Q

The natural course of hepatitis B virus can be followed by serum markers. How? When would you expect the different ones?

A
  • HBsAg appears before the onset of symptoms, peaks during overt disease, and then often declines to undetectable levels in 12 weeks, although it may persist in some individuals for as long as 24 weeks.
  • Anti-HBs antibodies does not rise until the acute disease is over, concomitant with the disappearance of HBsAg. In some cases, however, Anti_HBs antibody is not deteactable for a few weeks to several months fter the disappearance of HBsAG. During this window, serologic diagnosis can be made by detection of IgM anti-HBc antibodies. Anti-HBs may persist for life, conferring protection
  • HBeAg, HBV-DNA and DNA polymerase appear in serum soon after HBsAg, and all signify active viral replication. Persistence of HBeAg is an important indicator of continued viral replication, infectivity and probable progression to chronic hepatitis. The appearance of anti-hue antibodies implies that an acute infection has peaked and is on the wane.
  • IgM anti-HBc antibody becomes detectable in serum shortly before he onset of symptoms, concurrent with the onset of elevated serum aminotransferase levels (indicative of hepatocyte destruction). Over a period of months the IgM anti-HBc antibody is replaced by IgG anti-HBc. As in the case of anti-HAV, there is direct assay for IgG anti-HBc; its presence is inferred from decline of IgM anti-HBc in the face of rising total anti-HBc
1940
Q

The host immune response to the hepatitis B virus is the main determinant of the outcome of the infection. What is the host response?

A
  • Innate immune mechanisms protect the host during the initial phases of the infection, and a strong response by virus specific CD4+ and CD8+ interferon (IFN)-γ-producing cells is associated with the resolution of acute infection.
  • HBV generally does not cause direct hepatocyte injury. Instead, injury is caused by CD8+ cytotoxic T cells attacking infected cells.
1941
Q

Age at the time of hepatitis B infection is the best predictor chronicity. How?

A

The younger the age at the time of HBV infection, the higher the age of chronicity.

1942
Q

What aspects of the hepatitis B virus make infection difficult to treat?

A
  • Despite progress in the treatment of chronic HBV infection, complete cure is extremely difficult to achieve even when treated with highly effective antiviral agents.
  • The difficulty in achieving cure has been attributed to the ability of the virus to insert itself in the host DNA, thus limiting the development of an effective immune response (HBsAb development).
  • This allows the virus to persist in the face of drugs that impair its replication
  • Hence, the goal of the treatment of chronic hepatitis B is to slow disease progression, reduce liver damage, and prevent liver cirrhosis or liver cancer
1943
Q

How is hepatitis B prevented?

A
  • It can be prevented by vaccination and by the screening of donor blood, organs, and tissues
  • Vaccination induces a protective anti-HBs antibody response in 95% of infants, children, and adolescents
1944
Q

What are the most common risk factors for hepatitis C infection?

A
  • IVDU (54%)
  • Multiple sex partners (36%)
  • Having had surgery within the last 6 motnhs (16%)
  • Needle stick injury (10%)
  • Multiple contacts with an HCV-infected person (10%)
  • Employment in medical or dental fields (1.5%)
  • Unknown (32%)
1945
Q

How does the risk for acquiring hepatitis C infection by needle stick compare with the risk for acquiring HIV?

A

The risk for acquiring HCV by needle stick is about six times higher than the same risk for acquiring HIV (1.8% vs 0.3%)

1946
Q

What is the structure of the hepatitis C virus? How does it vary within a person

A
  • HCV is a member of the Flaviviridae family.
  • It is a small, enveloped, single-stranded RNA virus with a genome that codes for a single polyprotein with one open reading frame, which is subsequently processed into functional proteins.
  • Because of the low fidelity of the HCV RNA polymerase, the virus is inherently unstable, giving rise to multiple genotypes and subtypes. Indeed, within any given individual, HCV exists as closely related genetic variants known as quasispecies
1947
Q

Over time, dozens of quasispecies of hepatitis C virus can be detected within one individual all derived from the original HCV strain that infected the patient. What is the most variable region? Why is this relevant?

A
  • The E2 protein of the envelope is the target of many anti-HCV antibodies but is also the most variable region of the entire viral genome, enabling emergent virus strains to escape from neutralising antibodies.
  • This genomic instability and antigenic variability have seriously hampered efforts to develop an HCV vaccine. In particular, **elevated titers of anti-HCV IgG occurring after an active infection do not confer effective immunity.
  • A characteristic feature of HCV infection, therefore, is repeated bouts of hepatic damage, the result of reactivation of a pre-existing infection or emergence of an endogenous, newly mutated strain
1948
Q

What is the incubation period hepatitis C infection? How is this reflected in antibodies?

A
  • The incubation period for HCV hepatitis ranges from 4 to 26 weeks, with a mean of 9 weeks.
  • In about 85% of individuals, the clinical course of the acute infection is asymptomatic and typically missed.
  • HCV RNA is detectable in blood from 1 to 3 weeks, coincident with elevations in serum transaminases.
  • In symptomatic acute HCV infection, anti-HCV antibodies are detected in only 50-70% of patients; in the remaining patients, the anti-HCV antibodies emerge after 3-6 weeks.
1949
Q

How does the clinical course of hepatitis C virus compare to hepatitis A or hepatitis B?

A
  • The clinical course of acute HCV hepatitis is milder than that of HBV; rare cases may be sever and indistinguishable from HAV or HBV hepatitis,
  • In contrast to HBV, chronic disease occurs in the majority of HCV-infected individuals (80-90%) and cirrhosis eventually occurs in as many as a 20% of individuals with chronic HCV infection.
1950
Q

The mechanisms that lead to the chronicity of HCV infection are not well understood, but it is clear that the virus has developed multiple strategies to evade host antiviral immunity. What are some of these?

A

HCV is able to actively inhibit the IFN-mediated cellular antiviral response at multiple steps, including toll-like receptor signalling in response to viral RNA recognition and signalling downstream of IFN-receptors that would otherwise have antiviral effects.

1951
Q

In more than 90% of individuals with chronic hepatitis C infection, circulating HCV RNA persists despite the presence of antibodies. What effect does this have of serum testing? How do the LFTs change?

A
  • In persons with chronic hepatitis, HCV RNA testing must be performed to asses viral replication and to confirm the diagnosis of HCV infection.
  • A clinical feature that is quite characteristic of chronic HCV infection is persistent elevations in serum aminotransferases. Their levels wax and wane but almost never become normal
  • Even rare patients with normal transaminases are as risk for developing permanent liver damage. Therefore, any individual with detectable HCV RNA in the serum needs close
    clinical follow-up.
1952
Q

A feature unique to hepatitis C infection is the association with the metabolic syndrome. Tell me more about this…

A

It is particular with HCV genotype 3.
Apparently, HCV can give rise to insulin resistance and NAFLD

1953
Q

HCV is potentially curable. How? What influences it?

A
  • Until recently, treatment has been based on combination of pegylated IFN-α and ribavirin and cure rates depended on the viral genotype; patients with genotype 2 or 3 infection generally have had the best responses.
  • Interestingly, host genotype also influences the response. Certain polymorphism in the IL-28B gene are associated with better response to IFN-α and ribavarin. IL-28B encodes interferon lambda, which is involved in resistance in HCV.
  • New drug targeting viral protease and polymerase have now been approved and in development. These will hopefully work similar antiretroviral therapy HAART for HIV infection.
1954
Q

What does hepatitis D virus need for life cycle?

A

Hepatitis D virus (HDV) is a unique RNA virus that is dependent for its life cycle on HBV.

1955
Q

What are the settings in which infection of hepatitis D virus arises?

A
  • Co-infection occurs following exposure to serum containing both HDV and HBV. The HBV must become established first to provide the HbsAg necessary for development of complete HDV virions. Co-infection of HBV and HDV results in acute hepatitis that in that is indistinguishable from acute hepatitis B. It is self-limited and is usually followed by clearance of both viruses
  • Superinfection occurs when a chronic carrier of HBV is exposed to a new inoculum of HDV. This results in disease 30 to 50 days later presenting either as severe acute hepatitis in a previously unrecognised HBV carrier or as an exacerbation of pre-existing chronic hepatitis B infection. Chronic HDV infection occurs in almost all of such patients. The superinfection may have two phases: an acute phase with active HDV replication and suppression of HBV with high transaminases levels, and a chronic phase in which HDV replication decreases, HBV replication increases, transferase levels fluctuate, and the disease progresses to cirrhosis and sometimes HCC.
1956
Q

What is the structure of the hepatitis D virus?

A
  • HDV is a double-shelled particle.
  • The external coat antigen of HBsAg surrounds an internal polypeptide assembly, designated delta antigen (HDAg), the only protein produced the virus.
  • Associated with HDAg is a small circular molecule of single-stranded RNA, whose length is smaller than the genome of any known animal virus. Replication of the virus is through RNA-directed RNA synthesis by host RNA polymerase
1957
Q

What are the serum levels during hepatitis D infection?

A
  • HDV RNA is detectable in the blood and liver just before and in the early days of acute symptomatic disease
  • IgM anti-HDV antibody is the most reliable indictor of recent HDV exposure, although its appearance is late and frequently short-lived
  • Nevertheless, acute co-infection by HDV and HBV is best indicated by detection of IgM against both HDAs and HBcAg (denoting new infection with hepatitis B).
  • With chronic delta hepatitis arising from HDV superinfection, HBsAg is present in serums dn anti-HDV antibodies (IgG and IgM) persist for months or longer
1958
Q

How is hepatitis E transmitted? Who is it most common in?

A
  • Hepatitis E virus is an enteric ally transmitted water-borne infection that occurs primarily in young to middle-aged adults.
  • HEV is a zoonotic disease with animal reservoirs, such as monkeys, cats, pigs and dogs.
  • Epidemics have been reported in Asia and the Indian subcontinent, sub-Saharan Africa, Middle East, China and Mexico, although sporadic cases are seen in industrialised nations, particularly in regions where pig farming is common.
1959
Q

What is the prognosis of hepatitis E? What population does it have a high mortality in?

A
  • In most cases the disease is self-limiting
  • HEV is not associated with chronic liver disease or persistent viraemia in immunocompromised patients.
  • A characteristic feature of HEV infection is the high mortality rate among pregnant women, approaching 20%
  • Chronic HEV infection does occur in patients with AIDS ad immunosuppressed transplant patients.
1960
Q

What is the structure of the hepatitis E virus?

A

HEV is an undeveloped, positive -stranded RNA virus in the Hepevirus genus. Virions are shed in stool during the acute illness

1961
Q

What are the serum levels with hepatitis E virus?

A
  • The average incubation period following exposure is 4-5 weeks
  • Before the onset of clinical illness, HEV RNA and HEV virions can be detected by PCR in tool and serum.
  • The onset of rising serum aminotransferases, clinical illness, and elevated IgM anti-HEV titers are virtually simultaneous
  • Symptoms resolve in 2-4 weeks, during which time the IgM is replaced with a persistent IgG anti-HEV antibodies
1962
Q

Several clinical syndromes may develop following exposure to hepatitis viruses. What are they?

A

1) Acute asymptomatic infection with recovery (serologic evidence only)
2) Acute symptomatic hepatitis with recovery, anicteric or icteric
3) Chronic hepatitis, with or without progression to cirrhosis
4) Acute liver failure with massive to sub-massive hepatic necrosis

1963
Q

What type of virus are all the different hepatitis viruses?

A

A - single stranded RNA
B - partially double stranded DNA
C - single stranded RNA
D - circular defective single stranded RNA
E - single stranded RNA

1964
Q

What viral family do the different hepatitis viruses belong to?

A

A - hepatovirus: related to picornavirus
B - hepadnavirus
C - flaviviridae
D - sub viral particle in Deltaviridae family
E - hepevirus

1965
Q

What is the route of transmission for the different hepatitises?

A

A - faecal-oral (contaminated food or water)
B - parenteral, sexual contact, perinatal
C - parenteral; intranasal cocaine use is a risk factor
D - parenteral
E - faecal-oral

1966
Q

What is the mean incubation period for the different hepatitis viruses?

A

A - 2 to 6 weeks
B - 2 to 26 weeks (mean 8 weeks)
C - 4 to 25 weeks (mean 9 weeks)
D - same as HBV
E - 4 to 5 weeks

1967
Q

What is the frequency of chronic liver disease in the different hepatitis viruses?

A

A - never
B - 5-10%
C - >80%
D - 10% (co-infection); 90-100% for superinfection
E - in immunocompromised hosts only

1968
Q

How do you diagnose the different hepatitis viruses?

A

A - detection of serum IgM antibodies
B - detection of HBsAg or antibody to HBcAg; PCR for HBV DNA
C - 3rd generation ELISA for antibody detection: PCR for HCV RNA
D - detection of IgM and IgG antibodies; HDV RNA serum; HDAg in liver
E - detection of serum IgM and IgG antibodies, PCR for HEV RNA

1969
Q

What is chronic hepatitis?

A

It is defined as symptomatic, biochemical, or serologic evidence of continuing or relapsing hepatic disease for more than 6 months.

1970
Q

A “carrier” is an individual who harbours and can transmit an organism, but has not manifest symptoms. In the case of the hepatotropic hepatitis virus this definition is somewhat confusing. What can it mean?

A

1) Individuals who carry one of the viruses but have no liver disease
2) Those who harbour one of the viruses and have non-progressive liver damage, but are essentially free of symptoms or disability

In both cases, particularly the latter, these individuals constitute reservoirs for inferction.

1971
Q

What are the serum levels of a “healthy carrier” of hepatitis B?

A

It is defined as an individual with HBsAg, without HBeAg, but with presence of anti-HBe; these patients have normal aminotransferases, low or undetectable serum HBV DNA, and a liver biopsy showing a lack of significant inflammation and necrosis.

1972
Q

What are the basic microscopic changes in acute and chronic hepatitis?

A
  • They both evoke a lymphoplasmacytic (mononuclear) infiltrate.
  • Portal inflammation in acute hepatitis in acute hepatitis is minimal or absent.
1973
Q

Most parenchymal injury in hepatitis is scattered throughout the hepatic lobule as “spotty necrosis” or lobular hepatitis. The hepatocyte injury may result in necrosis or apoptosis. What are the cellular changes in both of these?

A
  • In necrosis, the cytoplasm appears empty with only scattered wisps of cytoplasms remnants. Eventually there is rupture of cell membranes leading to “dropout” of hepatocytes, leaving collapsed sinusoidal collagen reticulin framework behind; scavenger macrophages mark sites of dropout
  • With apoptosis, hepatocytes think, becoming intensely eosinophilic and their nuclei become pyknotic and fragmented; effector T cells may be present in the immediate vicinity.
1974
Q

What are the cellular changes in severe acute hepatitis?

A
  • Confluent necrosis of hepatocytes is seen around central veins. In these areas there may be cellular debris, collapsed reticulin fibers, congestion/haemorrhage and variable inflammation.
  • With increasing severity, there is central-portal bridging necrosis, followed by, even worse, parenchymal collapse.
  • In some cases, massive hepatic necrosis and acute liver failure ensue.
  • In occasional cases, the injury is not severe enough to cause death, and the liver survives, although with abundant scarring, usually with replacement of areas of confluent necrosis. In such cases, some patients rapidly develop post-hepatitic cirrhosis
1975
Q

There is considerable morphologic overlap in hepatitis caused by various hepatotropic viruses. However subtle differences may be seen, what is an example of this?

A

The mononuclear infiltrate in hepatitis A may be especially rich is plasma cells

1976
Q

What are the defining histologic features of chronic viral hepatitis?

A
  • The defining histologic feature of chronic viral hepatitis is mononuclear portal infiltration. It may be mild to severe and variable from one portal tract to the next.
  • There is often interface hepatitis as well, in addition to lobular hepatitis, distinguished by its location at the interface between hepatocellular parenchyma and portal tract stroma
  • The hallmark of progressive chronic liver damage is scarring. At first, only portal tracts exhibit fibrosis, but in some patients, with time, fibrous septa - bands of dense scar - extend between portal tracts. In parallel with increasing scarring there is also increasing ductular reaction, reflecting stem cell activation
  • In the most severe cases, continued scarring and nodule formation leads to the development of cirrhosis.
1977
Q

Clinical assessment of chronic hepatitis often required liver biopsy in addition to clinical and serologic data. Why is this helpful?

A
  • Liver biopsy is helpful in confirming the clinical diagnosis, excluding common concomitant conditions (eg, fatty liver disease, haemochromatosis), assessing histologic features associated with an increased risk of malignancy (eg, small and large cell change), grading the extent of hepatocyte injury and inflammation, and staging the progression os scarring.
1978
Q

A greater range of histologic features distinguish one viral infection from another in chronic hepatitis. How?

A
  • In chronic hepatitis B, “ground glass” hepatocytes - cells with endoplasmic reticulum swollen by HBsAg - is a diagnostic hallmark. Immunostaining can confirm the presence of viral antigen
  • Chronic hepatitis C quite commonly shows lymphoid aggregates or fully formed lymphoid follicles. Often hepatitis C, particularly genotype 3, shows fatty change of scattered hepatocytes, although the infection may also cause systemic alterations leading to metabolic syndrome and, therefore, a superimposed NAFLD. Bile duct injury is also prominent in some individuals with hepatitis C infection.
1979
Q

How can you help remember the differences between the hepatitises?

A
  • The vowels (A &E) don’t cause chronic hepatitis only AcutE hepatitis, except HEV in immunocompromised and pregnant females
  • Only the consonants (B, C, D) have the potential to cause chronic disease (C for consonant and for chronic)
  • Hepatitis B can be transmitted by Blood, Birthing, and Bonking
  • Hepatitis C is the single virus that is more often Chronic that not
  • Hepatitis is endemic in equatorial regions and frequently epidemic
1980
Q

Why are some bacterial infections of the liver? How does bacterial get in there and what is this called?

A
  • Several bacteria can infect the liver directly, including Staphylococcus aureus is toxic shock syndrome, Salmonella typhi in typhoid fever, and Treponema pallidum in secondary or tertiary syphilis.
  • Bacteria may also proliferate in the biliary tree especially when outflow is compromised by partial or complete obstruction
  • The intrabiliary bacterial composition reflects the gut flora, and the acute inflammatory response within the intrahepatic biliary tree is called ascending cholangitis
1981
Q

How can bacteria cause liver abscesses? What are their symptoms and how are they treated?

A
  • Bacteria may give rise to abscesses by spreading from extrahepatic sites, through the vascular supply, or from adjacent infected tissues.
  • Liver abscesses are associated with fever and, in many instances, RUQ pain and tender hepatomegaly. Jaundice may result from extrahepatic biliary obstruction
  • Although antibiotic therapy may control smaller lesions, surgical drainage is often necessary for the larger lesions
1982
Q

Apart from bacteria and viruses, what else can infect the liver disease? How can you tell?

A
  • Funghi (eg, histoplasmosis) and mycobacteria can also infect the liver is disseminated diease, with hostology showing classical granulomas. Organisms are usually not visible histologically, even with special stains, although serologic studies and tissue or blood cultures can often identify the causative agent.
  • Parasitic and helminthic infections are major causes of morbidity worldwide, and the liver is frequently involved. These diseases include malaria, schistosomiasis, cryptosporidiosis, amebiases.
1983
Q

What is autoimmune hepatitis?

A

A chronic, progressive hepatitis with all the features of autoimmune disease in general: genetic predisposition, association with other auto-immune diseases, presence of autoantibodies and therapeutic response to immunosuppression.

1984
Q

What are the genetic predispositions and triggers for autoimmune hepatitis?

A
  • Strong HLA-associations for autoimmune hepatitis support a genetic predisposition.
  • In Caucasians, there is a frequent association with DRB1* alleles.
  • Triggers for the immune reaction may include viral infections or drug or toxin exposures
1985
Q

What populations are more likely to have autoimmune hepatitis?

A
  • The annual incidence is highest among white northern Europeans but all ethnic groups are susceptible
  • There is a female predominance (78%)
1986
Q

Autoimmune hepatitis is classified into types 1 and 2, base on patterns of circulating antibodies. What are these types and how old are the people often affected?

A
  • Type 1, more common in middle-aged to older individuals, is characterised by the presence of ANA, anti-smooth muscle actin (SMA), anti-soluble liver antigen/liver-pancreas antigen (anti-SLA/LP) antibodies, and less commonly, anti-mitochondrial (AMA) antibodies.
  • In type 2, usually seen in children and teenagers, the main serologic markers are anti-liver kidney microsome 1( anti-LKM-1) antibodies, which are mostly directed against CYP2D6, and anti-liver cytosol-1 (ACL-1) antibodies.
1987
Q

Although autoimmune hepatitis shares patterns of injury with acute or chronic viral hepatitis, the time course of histologic progression differs. How?

A
  • In viral hepatitis, fibrosis typically follows many years of slowly accumulating parenchymal injury
  • Whereas in autoimmune hepatitis, there is an early phase of severe parenchymal destruction followed rapidly by scarring.
1988
Q

What are the typical morphologic features of autoimmune hepatitis?

A
  • Severe necroinflammatory activity indicated by extensive interface hepatitis or foci of confluent (perivenular or bridging necrosis) or parenchymal collapse
  • plasma cell predominance in the mononuclear inflammatory infiltrates
  • Hepatocyte “rosettes” in areas of marked activity
1989
Q

Autoimmune hepatitis may be rapidly progressive or indolent, both giving rise eventually to liver failure. Clinical evolution correlates with a limited number of histologic patterns, any of which may be seen at the time of initial diagnosis. What are these?

A
  • Very severe hepatocyte injury with widespread confluent necrosis, but little scarring; this is often seen as symptomatic acute hepatitis and represents the first sign of disease
  • A mix of marked inflammation and some degree of scarring, seen in early or later stage disease
  • Burned-out cirrhosis, with little necroinflammatory activity, that has been preceded, presumably, by years of subclinical diseease
1990
Q

What is the clinical course and prognosis of autoimmune hepatitis?

A
  • An acute appearance of clinical illness is common (40%) and a fulminant presentation with hepatic encephalopathy within 8 weeks of disease onset may also occur.
  • The mortality of patients with severe untreated autoimmune hepatitis is approximately 40% within 6 months of diagnosis and cirrhosis develops in at least 40% of survivors.
  • In general, prognosis is better in adults than in children, possibly due to delay in diagnosis in the paediatric population.
  • Hence, diagnosis and intervention are imperative
1991
Q

What is the treatment of autoimmune hepatitis?

A
  • Immunosuppressive therapy is usually successful, leading to remissions in 80% of patients that permits long term survival.
  • In end stage disease, liver transplantation is indicated.
  • Ten-year survival rate after liver transplant is 75%, but recurrence in the transplanted organ may affect 20% of transplanted patients.
1992
Q

As the major drug metabolising and detoxifying organ in the body, the liver is subject to injury from an enormous array of therapeutic and environmental agents. How may this injury occur? How is the diagnosis made?

A
  • Injury may result from direct toxicity, occur through hepatic conversion of a xenobiotic to an active toxin, or be proceed by immune mechanisms, such as by the drug or a metabolite acting as a hapten to convert a cellular protein into an immunogen.
  • A diagnosis of drug- or toxin-induced liver injury may be made on the basis of a temporal association of liver damage with drug or toxin exposure, recovery (usually) upon removal of the inciting agent, and exclusion of other potential causes.
1993
Q

What are some morphologic findings with cholestatic and cholestatic hepatitis and what drugs cause them?

A
  • Cholestatic - bland hepatocellular cholestasis, without inflammation - contraceptive and anabolic steroids, antibiotics, HAART
  • Cholestatic hepatitis - cholestasis with lobular necroinflammatory activity; may show bile duct destruction - antibiotics, phenothiazines, statins
1994
Q

What are some agents that cause hepatocellular necrosis and their morphologic findings?

A
  • Methyldopa, phenytoin - spotty hepatocyte necrosis
  • Acetaminophen, halothane - massive necrosis
  • isoniazid - chronic hepatitis
1995
Q

What are some agents that cause fatty liver disease and what are their morphologic findings?

A
  • Ethanol, corticosteroids, methotrexate, total parenteral nutrition - large and small droplet fat
  • Valproate, tetracycline, aspirin (Reye syndrome), HAART - “microvesicular steatosis (diffuse small droplet fat)
  • Ethanol, amiodarone - steatohepatitis with Mallory-Denk bodies
1996
Q

What are some agents that cause hepatic granulomas or cirrhosis, what are their morphologic findings?

A

** Periportal and pericellular fibrosis** - alcohol, methotrexate, enalapril, vitamin A and other retinoids
Non-caseating epithelioid granulomas - sulfonamides, amiodarone, isoniazid
Fibrin-ring granulomas - allopurinol

1997
Q

What are some agents that cause hepatic vascular lesions and what are the morphologic findings?

A
  • Sinusoidal obstruction syndrome (veno-occlusive disease): obliteration of central veins - high-does chemotherapy, bush teas
  • Budd-Chiari syndrome - oral contraceptives
  • Peliosis hepatis: blood-filled cavities, not lined by endothelial cells - anabolic steroids, tamoxifen
1998
Q

Drug toxic reactions may be classified as predictable (intrinsic) or unpredictable (idiosyncratic). Why does this mean? Does it have any correlation to how long it takes for the injury to develop?

A
  • Predictable reactions affect al people in a dose-dependent reaction
  • Unpredictable reactions depend on idiosyncrasies of the host, particularly the propensity to mount an immune response to the antigenic stimulus or the rate at which the agent can be metabolised.

Both classes of injury may be immediate or take weeks to months to develop.

1999
Q

A classic, predictable hepatotoxin is acetaminophen. How does it cause liver injury?

A
  • The toxic agent is not acetaminophen itself but rather a toxic metabolite produced by the cytochrome P450 system in acinus zone 3 hepatocytes.
  • As these hepatocytes die, the zone 2 hepatocytes take over this metabolic function, in turn becoming injured.
  • In severe overdoses, the zone of injury extends tot the periportal hepatocytes, resulting in acute hepatic failure.
2000
Q

Excessive alcohol consumption is the leading cause of liver disease in most Western countries. There are three distinctive, albeit overlapping forms of alcoholic liver injury. What are they?

A

1) Hepatocellular steatosis or fatty change
2) Alcoholic (or steato-) hepatitis
3) Steatofibrosis (patterns of scarring typical for all fatty liver diseases including alcohol) up to and including cirrhosis in the late stages of disease

2001
Q

Where in the liver does alcohol liver disease begin and progress?

A

All changes in alcoholic liver disease begin in acinus zone 3 and extend outward toward the portal tracts with increasing severity of injury

2002
Q

What are the morphologic changes in hepatic steatosis (fatty liver)? Is it reversible?

A
  • After even moderate intake of alcohol, lipid droplets accumulate in hepatocytes increasing with amount and chronicity of alcohol intake
  • The lipid begins as small droplets that coalesce into large droplets which distend the hepatocyte and push the nucleus aside.
  • Macroscopically, the fatty liver in individuals with chronic alcoholism is a large (as heavy as 4-6kg), soft organ that is yellow and greasy
  • Fatty change is completely reversible if there is abstention from further intake of alcohol
2003
Q

What are the three morphologic characteristics that occur in alcoholic (steato-) hepatitis?

A

1) Hepatocyte swelling and necrosis: single or scattered foci of cells undergo swelling (ballooning) and necrosis. The swelling results from the accumulation of fat and water, as well as proteins that are normally exported.
2) Mallory-Denk bodies: these are usually present as clumped, amorphous, eosinophilic material in ballooned hepatocytes. They are made up of tangled skeins of intermediate filaments such as keratins 8 and 18 in complex with other proteins such as ubiquitin. These inclusions are characteristic but not specific feature of ALD, they are also in NAFLD, Wilsons’s disease and chronic biliary tract diseases
3) Neutrophilic reaction: neutrophils permeate the hepatic lobule and accumulate around degenerating hepatocytes, particularly those having Mallory-Denk bodies. They may be more or less admixed with mononuclear cells

2004
Q

Alcoholic hepatitis is often accompanied by prominent activation of sinusoidal stellate cells and portal fibroblasts, giving rise to fibrosis. What are the morphological changes that occur with this?

A
  • Fibrosis begins with sclerosis of central veins
  • Perisinusoidal scar then accumulates in the space of Disse of the centrilobular region, spreading outward, encircling individual or small cluster of hepatocytes in a chicken wire fence pattern
  • These webs of scar eventually link to portal tracts and then begin to condense into central-portal fibrous septa. With developing nodularity, cirrhosis becomes established.
  • When alcohol use continues without interruption over the long therm, the continual subdivision of established nodules by new webs of, perisinusoidal scarring leads to a classic micronodular or Laennec cirrhosis.
2005
Q

How many grams of alcohol cause what levels of damage?

A
  • Short-term ingestion of as much as 80 grams of alcohol over one to several days generally produces mild, reversible hepatic steatosis.
  • Daily intake of 80 grams or more of ethanol generates significant risk for severe hepatic injury
  • Daily ingestion of 160 grams or more for 10 to 20 years is associated more consistently with severe injury.
2006
Q

Only 10-15% of alcoholics, however, develop cirrhosis. Thus, other factors must influence the development and severity of alcoholic liver disease. What are these?

A
  • Gender. Women seem to be more susceptible to hepatic injury than men, although the majority of patients are men.
  • Ethnic and genetic differences. In the US, cirrhosis rates are higher for African American drinkers than for white American drinkers. ALDH2, a variant of aldehyde-dehydrogenase, has a very low activity. Individuals homozygous for ALDH2 do not tolerate alcohol
  • Co-morbid conditions - iron overload and infections with HCV and HBV synergise with alcohol, leading to increased severity of liver disease
2007
Q

What are the detrimental effects of alcohol and its bypasses on hepatocellular function?

A
  • Exposure to alcohol causes steatosis, dysfunction of mitochondrial and cellular membranes, hypoxia, and oxidative stress
  • At milimolar concentrations, alcohol directly affects micro tubular and mitochondrial function.
2008
Q

How does alcohol cause hepatocellular steatosis?

A

Hepatocellular steatosis results from
1) shunting of normal substrates away from catabolism and toward lipid biosynthesis, as a result of increased generation of reduced nicotanamide adenine dinucleotide (NADH) by the two major enzymes of alcohol metabolism, alcohol dehydrogenase and acetaldehyde dehydrogenase.
2) impaired assembly and secretion of lipoproteins
3) increased peripheral catabolism of fat, thus releasing free fatty acids into the circulation

2009
Q

The causes of alcoholic hepatitis are uncertain, but what are some factors that likely play important roles?

A
  • Acetaldehyde induces lipid peroxidation and acetaldehyde-protein adduct formation, further disrupting cytoskeletal and membrane function
  • Cytochrome P-450 metabolism produces reactive oxygen species that react with cellular proteins, damage membranes, and alter hepatocellular function.
  • Alcohol impairs hepatic metabolism of methionine, which decreases glutathione levels, thereby sensitising the liver to oxidative injury.
  • The induction of cytochrome P450 enzymes in the liver by alcohol increases alcohol catabolism in the endoplasmic reticulum and enhances the conversion of other drugs (acetaminophen) to toxic metabolites
  • It causes the release of bacterial endotoxin from the gut into the portal circulation, inducing inflammatory responses in the liver, due to the activation of NF-κB, and release of TNF, IL-6, and TGF-α.
  • It stimulates the release of endothelins from sinusoidal endothelial cells, causing vasoconstriction and contraction of activated, myofibroblastic stellate cells, leading to a decrease in hepatic sinusoidal perfusion.
2010
Q

What are the clinical features of alcoholic liver disease?

A
  • Hepatic steatosis may cause hepatomegaly, with mild elevation of serum bilirubin and alkaline phosphatase levels. Severe hepatic dysfunction is unusual
  • In contrast, alcoholic hepatitis tends to appear acutely, usually following a bout of heavy drinking. Symptoms and laboratory manifestations may range from minimal to those that mimic acute liver failure.
  • Between these two extremes are the non-specific symptoms of malaise, anorexia, weight loss, upper abdominal discomfort, and tender hepatomegaly, and the bloods showing hyperbilirubinaemia, elevated serum aminotransferase and alkaline phosphatase, and often a neutrophilic leukocytosis.
2011
Q

How do the bloods in alcoholic liver disease differ from other chronic liver diseases?

A

In contrast to other chronic liver diseases, where serum ALT tends to be higher than serum AST, serum AST levels tend to be higher than serum ALT levels in a 2:1 ratio or higher in alcoholic liver disease

2012
Q

What is the clinical course of alcoholic liver disease?

A
  • The outlook is unpredictable; each bout of hepatitis incurs about a 10-20% risk of death.
  • With repeated bouts, cirrhosis develops in about 1/3rd of patients within a few years
  • Alcoholic hepatitis also may be superimposed on established cirrhosis.
  • With proper nutrition and total cessation of alcohol consumption, the alcoholic hepatitis may clear slowly.
  • However, in some patients, the hepatitis persists, despite abstinence, and progresses to cirrhosis.
2013
Q

In end-stage alcohol liver disease, what are the proximate causes of death?

A

1) hepatic coma
2) massive GI haemorrhage
3) intercurrent infection (to which these patents are pre-disposed)
4) hepatorenal syndrome following a bout of alcoholic hepatitis
5) HCC

2014
Q

A distinct group of liver diseases is attributable to disorder ot metabolism, either acquired or inherited. What are some of these?

A
  • The most common acquired metabolic disorder is NAFLD
  • Among inherited metabolic disease, haemochromatosis, Wilson disease, and α1-antitrypsin deficiency are most prominent
  • Also included among liver metabolic disease is neonatal hepatitis, a broad disease category encompassing rare inherited diseases and neonatal infections.
2015
Q

What is NAFLD?

A

NAFLD represents a spectrum of disorders that have in common the presence of hepatic steatosis (fatty liver) in individuals who do not consume alcohol or do so in very small quantities (less than 20g of ethanol/week)

2016
Q

What are eh features included in the WHO criteria for metabolic syndrome?

A
  • One of
    Diabetes mellitus OR
    Impaired glucose tolerance OR
    Impaired fasting glucose OR
    Insulin resistance
  • An two of:
    BP >140/90 mmHG
    Dyslipidaemia
    Central obesity
    Microalbuminuria
2017
Q

What are the effects of NAFLD on HCV, HBV and HCC?

A
  • NAFLD contributes to the progression of other liver diseases such as HCV and HBV infection
  • Increasingly, NAFLD is found to increase the risk for HCC, although, unlike in chronic viral hepatitis and ALD, it may often do so in the absence of significant scarring
2018
Q

Current available data suggests a two-factor model for the pathogenesis of NAFLD. What are these factors?

A

1) Insulin resistance gives rise to hepatic steatosis
2) Hepatocellular oxidative injury resulting in liver cell necrosis and the inflammatory reactions to it

2019
Q

How does insulin resistance lead to NAFLD?

A
  • In individuals with established insulin resistance and metabolic syndrome, the visceral adipose tissue not only increases, but also becomes dysfunctional, with reduced production of the lipid hormone, adiponectin, and increased production of inflammatory cytokines such as TNF-α and IL-6
  • These changes in turn promote hepatocyte apoptosis. Fat laden cells are highly sensitive to lipid peroxidation products generated by oxidative stress which can damage mitochondrial and plasma membranes, causing apoptosis
  • Diminished autophagy also contributes to mitochondrial injury and formation of Mallory-Denk bodies. Kupffer cell production of TNF-α and TGF-β activate stellate cells directly leading to deposition of scar tissue.
  • Stellate cell activation also occurs through the hedgehog signalling pathway in part through natural killer T-cell activation. In fact, the level of hedgehog pathway activity correlates with stage of fibrosis in NAFLD
2020
Q

What is the difference between NAFLD and non-alcoholic steatohepatitis?

A
  • NAFLD is clinical features of liver injury, such as elevated serum transaminases
  • NASH (non-alcoholic steatohepatitis) is reserved for histologic features of hepatocyte injury
2021
Q

What are the morphological changes in non-alcoholic steatohepatitis? How is this different to alcohol hepatitis?

A
  • Pathologic steatosis is defined as involving more than 5% of hepatocytes. Small medium, and large droplets of fat, predominantly triglycerides, accumulate within hepatocytes just as they do in alcoholic steatosis.
  • At the most clinically benign end of the spectrum, there is no appreciable hepatic inflammation, hepatocyte death, or scarring, despite persistent elevation of serum liver enzymes
  • NASH almost completely overlaps in its histologic features with alcoholic hepatitis. In NASH, compared with alcoholic hepatitis, mononuclear cells may be more prominent than neutrophils and Mallory-Denk bodies are often less prominent.
2022
Q

How does the morphology of NAFLD compare with alcoholic liver disease?

A

Steatofibrosis in NAFLD shows precisely the same features and progression as it does in alcoholic liver disease, although portal fibrosis may be more prominent.

2023
Q

Does NAFLD cause cirrhosis?

A

Cirrhosis may develop, is often subclinical for years, and, when established, the steatosis or steatohepatitis may be reduced or absent

2024
Q

How does paediatric NAFLD compare to adult disease?

A

Typically children show more diffuse steatosis, portal rather than central fibrosis, and portal and parenchymal mononuclear infiltration, rather than parenchymal neutrophils

2025
Q

What are the clinical features of NAFLD? How do you diagnose it?

A
  • Individuals with simple steatosis are generally asymptomatic
  • Clinical presentation is often related to other signs and symptoms of the metabolic syndrome, in particular insulin resistance or diabetes mellitus
  • Imaging studies may reveal fat accumulation in the liver
  • However, liver biopsy is the most reliable diagnostic tool for NAFLD and NASH, and for assessment of scarring.
  • Serum AST and ALT are elevated in about 90% of patients with NASH
2026
Q

What is the prognosis and outcome of NAFLD?

A
2027
Q

What is haemochromatosis? What are the causes?

A
  • It is caused by excessive iron absorption, most of which is deposited in parenchymal organs such as the liver and pancreas, followed by heart, joints and endocrine organs.
  • When haemochromatosis results from an inherited disorder, it is referred to as hereditary haemochromatosis, of which there are many forms, some more likely than others to lead to overwhelming iron overload
  • When accumulation occurs as a consequence of transfusions or other causes, it is called secondary haemochromatosis.
2028
Q

What is the normal iron level? How much is stored in the liver? Where? How does this change in haemochromatosis?

A
  • The total body iron pool ranges from 2-6g in normal adults
  • About 0.5g is stored in the liver, 98% of which is in hepatocytes
  • In the most severe forms of haemochromatosis, total iron accumulation may exceed 50g, more than one third of which accumulates in the liver.
2029
Q

What are the characteristic features of severe iron overload in the body?

A
  • Fully developed cases exhibit 1) micronodular cirrhosis in all patients; 2) diabetes mellitus in 75-80% of patients; and 3) abnormal skin pigmentation in 75-80% of patients
  • Iron accumulation in hereditary forms is lifelong but the injury caused by excessive iron is slow and progressive; hence symptoms usually first appear in the fourth-fifth decades of life in men and later in women since menstrual bleeding counterbalances the accumulation until menopause
  • Because many women do not accumulate clinically relevant amounts of iron within their lifetime, hereditary haemochromatosis affects more males than females (ratio of 5 to 7:1)
2030
Q

What is the basis of the pathogenesis of haemochromatosis? What level of iron leads to the manifestations of haemochromatosis?

A
  • Because there is no regulated iron excretion from the body, the total body content of iron is tightly regulated by intestinal absorption
  • In hereditary haemochromatosis, regulation of intestinal absorption of dietary iron is abnormal, leading to net iron accumulation of 0.5-1g/year.
  • The disease manifests itself typically after 20g of stored iron have accumulated
2031
Q

Excessive iron appears to be directly toxic to tissues. What are the mechanisms of liver injury? Is it reversible?

A

1) Lipid peroxidation via iron-catalysed free radical reactions
2) Stimulation of collagen formation by activation of hepatic stellate cells
3) Interaction of reactive oxygen species and iron itself with DNA, leading to lethal cell injury and predisposition to HCC.

The actions of iron are reversible in cells that are not fatally injured, and removal of excess iron with therapy promotes recovery of tissue function

2032
Q

What is the main regulator of iron absorption? What regulates its levels? What does it do?

A
  • The main regulator of iron absorption is the protein hepcidin, encoded by the HAMP gene and secreted by the liver. It is a hepatocellular protein with bactericidal activities.
  • Transcription of hepcidin in increased by inflammatory cytokines and iron, and decreased by iron deficiency, hypoxia, and ineffective erythropoiesis.
  • Hepcidin binds to the cellular iron efflux channel ferroportin, causing its internalisation and proteolysis, thereby inhibiting the release of iron from intestinal cells and macrophages.
  • Therefore, hepcidin lowers plasma iron levels. Conversely, a deficiency in hepcidin causes iron overload
2033
Q

Some proteins involved in iron metabolism do so by regulating hepcidin levels. What are some of these? How are these relevant in haemochromatosis?

A

1) Hemojuvelin (HJV), which is expressed in the liver, heart and skeletal muscle
2) Transferrin receptor 2 (TFR2), which is highly expressed in hepatocytes, where it mediates the uptake of transferring-bound iron
3) HFE, the product of the haemochromatosis gene

Decreased hepcidin synthesis caused by mutations in hepcidin, HJV, TFR2, and HFE has a central role in the pathogenesis haemochromatosis.

2034
Q

What is the most common mutation is the adult and the juvenile form of haemochromatosis? What does it do?

A
  • The adult form of haemochromatosis is almost always caused by mutations of HFE.
  • The HFE gene encodes an HLA class I-like molecule that governs intestinal absorption of dietary iron by regulating hepcidin synthesis
  • The most common HFE mutation is a cysteine-to-tyrosine substitution at amino acid 282.
  • Mutations of HAMP and HJV cause severe juvenile haemochromatosis
2035
Q

Is adult or juvenile haemochromatosis more severe?

A

Adult haemochromatosis is generally a milder disease than the juvenile form

2036
Q

What are the morphological characteristics of severe haemochromatosis?

A

1) Deposition of hemosiderin in the following organs (in decreasing order of severity) the liver, pancreas, myocardium, pituitary gland, adrenal gland, thyroid and parathyroid glands, joints and skin
2) Cirrhosis
3) Pancreatic fibrosis

2037
Q

What are the morphologic changes in the liver in haemochromatosis?

A
  • In the liver, iron becomes evident first as golden-yellow hemosiderin granules in the cytoplasm of peri-portal hepatocytes that stain with Prussian blue
  • With increasing iron load, there is progressive involvement of the rest of the lobule, along with bile duct epithelium and Kupffer cell pigmentation
  • Iron is a direct hepatotoxin and inflammation is characteristically absent.
  • In early stages of accumulation, the liver is typically slightly larger than normal, dense and chocolate brown
  • Fibrous septa develop slowly, leading ultimately to a small, shrunken liver with a micronodular pattern of cirrhosis.
  • The liver parenchyma in later stages is often dark brown to nearly back due to overwhelming iron accumulation
2038
Q

What are the morphological changes in the pancreas in haemochromatosis?

A
  • The pancreas becomes intensely pigmented, has diffuse interstitial fibrosis, and may exhibit some parenchymal atrophy
  • Hemosiderin is found in both the acinar and the islet cells, and sometimes in the interstitial fibrous stroma
2039
Q

What are the morphologic changes in the heart in haemochromatosis?

A
  • The heart is often enlarged and has hemosiderin granules within the myocardial fibers, producing a striking brown coloration to the myocardium
  • A delicate interstitial fibrosis may appear
2040
Q

What are the morphologic changes in the skin in haemochromatosis?

A

Although skin pigmentation is partially attributable to hemosiderin deposition in dermal macrophages and fibroblasts, most of the pigmentations results from increased epidermal melanin production, the mechanism of which is unknown

2041
Q

What are the morphologic changes in the synovial joints and testes in haemochromatosis?

A
  • With hemosiderin deposition in the synovial joint livings, an acute synovitis may develop. Excessive deposition of calcium pyrophosphate damages the articular cartilage, producing a disabling polyarthritis referred to as psuedogout
  • The testes may be small and atrophic, secondary to derangement in the hypothalamic-pituitary axis resulting in reduced gonadotropin and testosterone levels
2042
Q

What are the principal clinical manifestations of haemochromatosis?

A
  • The manifestations of classic haemochromatosis include hepatomegaly, abdominal pain, abnormal skin pigmentation (particularly in sun-exposed areas), deranged glucose homeostasis or diabetes mellitus due to destruction of pancreatic islet cells, cardiac dysfunction (arrhythmias, cardiomyopathy), and atypical arthritis
  • In some patients, the presenting complaint is hypogonadism (eg, amenorrhoea in the female, impotence and loss of libido in the male)
  • It is more often a disease of males and rarely becomes evident before age 40
2043
Q

What are the major causes of death from haemochromatosis? What is the treatment?

A
  • Death may result from cirrhosis or cardiac disease
  • A significant cause of death is HCC; the risk is 200-fold greater than in the general population
  • Treatment by regular phlebotomy steadily depletes tissue iron stores
  • Treatment for iron overload does not fully remove the cancer risk presumably because of DNA alterations that occur prior to the time of diagnosis and treatment initiation.
2044
Q

How do you screen for haemochromatosis?

A
  • Screening involves demonstration of very high levels of serum iron and ferritin, exclusion of secondary causes of iron overload, and liver biopsy if indicated
  • Screening of family members of probands is important.
2045
Q

What is neonatal haemochromatosis?

A
  • Neonatal haemochromatosis is a disease of unknown origin manifested by severe liver disease and extrahepatic hemosiderin deposition
  • It is not an inherited disease; liver injury, leading to hemosiderin accumulation occurs in utero, and might be related to maternal alloimmune injury to the fetal liver
  • Extrahepatic hemosiderin deposition, detected by buccal biopsy, needs to be documented for the correct diagnosis
  • There is no specific treatment, except for supportive care and liver transplantation in severe cases.
2046
Q

What are the most common causes of secondary (or acquired) haemochromatosis?

A
  • Disorders associated with ineffective erythropoiesis, such as severe forms of thalassaemia and myelodysplastic syndromes.
  • In these disorders, the excess iron results not only from transfusions, but also from increase absorption.
  • Transfusion alone, when given repeatedly over a period of years (eg, in patients with chronic haemolytic anaemias) can also lead to systemic hemosiderosis and parenchymal organ injury
2047
Q

What is Wilsons disease? How is it inherited?

A
  • Wilsons disease is an autosomal recessive disorder caused by mutation of the ATP7B gene
  • It results in impaired copper excretion into bile and failure to incorporate copper into ceruloplasmin
  • This disorder is marked by the accumulation of toxic levels of copper in many tissues and organs, principally the liver, brain and eye.
2048
Q

How is copper normally absorbed by the body?

A
  • Normally 40-60% of ingested copper (2-5mg/day) is absorbed in the duodenum and proximal small intestine, and is transported to the portal circulation complexed with albumin and histidine.
  • In the liver copper binds to an α2-globulin (apoceruloplasmin) to for ceruloplasmin, which is secreted into the blood
  • Excess copper is transported into the bile
  • Ceruloplasmin accounts for 90-95% of plasma copper.
  • Circulating ceruloplasmin is eventually desialylated, endocytosed by the liver, and degraded within lysosomes, after which the released copper is excreted into bile.
  • This degradation/excretion pathway is the primary route for copper elimination.
2049
Q

What is the normal total body copper volume?

A

The estimated total body copper is only 50-150mg

2050
Q

What is the genetic abnormality in Wilson disease?

A
  • Wilson disease results from mutations in the ATP7B gene, which encodes a transmembrane copper-transporting ATPase, expressed on the hepatocyte canalicular membrane
  • The overwhelming majority of patients are compound heterozygotes containing different mutations on each ATP7B allele.
  • The overall frequency of mutated alleles is 1:100, and the prevalence of the disease is approximately 1:30,000
2051
Q

What is the pathogenesis of Wilson disease?

A
  • Deficiency in the ATP7B protein causes a decrease in copper transport into bile, impairs its incorporation into ceruloplasmin, and inhibits ceruloplasmin secretion into the blood.
  • These changes copper accumulation in the liver and a decrease in circulating ceruloplasmin.
2052
Q

The accumulated copper in Wilson Disease cause toxin liver injury in three different mechanisms. What are these?

A

1) Promoting the formation of free radicals by the Fenton reaction
2) Binding to sulfhydryl groups of cellular proteins
3) Displacing other metals from hepatic metalloenzymes

2053
Q

Do the symptoms of Wilson disease often present suddenly or gradually?

A
  • Although there is a latent period of variable duration, there may be sudden onset of a severe systemic illness.
  • This is triggered by spillage of non-ceruloplasmin-bound copper from the liver into the circulation, causing haemolysis and pathologic changes at other sites such as the brain, corneas, kidneys, bones, joints and parathyroids.
  • Concomitantly, urinary excretion of copper markedly increases from is normal minuscule levels
2054
Q

What are the morphologic changes in the liver in Wilson disease?

A
  • The liver often bears the brunt of injury, but the disease may also present as a neurologic disorder
  • The hepatic changes are variable, ranging from relatively minor to massive damage.
  • Fatty change may be mild to moderate with focal hepatocyte necrosis.
  • An acute, fulminant hepatitis can mimic acute viral hepatitis.
  • Chronic hepatitis in Wilson disease exhibits modest to severe inflammation and hepatocyte necrosis, admixed with fatty change and features of steatohepatitis (hepatocyte ballooning with prominent Mallory-Denk bodies).
    *Eventually cirrhosis supervenes
2055
Q

How can you test for Wilson’s disease on staining? What is required for diagnosis?

A
  • Excess copper deposition can often be demonstrated by special stains (rhodamine stain for copper, orcein stain for copper-associated protein).
  • Because copper accumulates in chronic obstructive cholestasis and because histology cannot reliably distinguish Wilson disease from viral- and drug-induced hepatitis, demonstration of hepatic copper content in excess of 250μg per gram dry weight is most helpful for making a diagnosis
2056
Q

What are the morphologic changes in the brain in Wilson disease?

A

*Toxic injury to the brain primarily affects the basal ganglia, particularly the putamen, which shows atrophy and even cavitation.
* Nearly all patients with neurologic involvement develop eye lesions called Kayser-Fleisher rings, green to brown deposits of copper in Descemet membrane in the limbus of the cornea

2057
Q

What age does Wilson disease usually present?

A

The age at onset and the clinical presentation of Wilson disease are extremely variable (average age in 11.4 years), but the disorder usually manifests in affected individuals between 6 and 40 years of age.

2058
Q

How does Wilson disease present?

A
  • Initial presentation may be either with acute or chronic liver disease
  • Neurologic involvement presents as movement disorders (tremor, poor coordination, chorea, or choreoathetosis) or rigid dystonia (spastic dystonia, mask like faces, rigidity and gait disturbances); these symptoms may be confused with Parkinsonism
  • Patients may also have psychiatric symptoms such as depression, phobias, compulsive behaviour, and labile mood
  • Haemolytic anaemia may occur due to toxicity of copper to red cell membranes
2059
Q

What are the blood tests that show Wilson disease?

A
  • The biochemical diagnosis of Wilson disease is based on a decrease in serum ceruloplasmin, an increase in hepatic copper content (the most sensitive and accurate test), and increased urinary excretion of copper (the most specific screening test)
  • Serum copper levels are of no diagnostic value, since they may be low, normal or elevated, depending on the stage of evolution of the disease.
2060
Q

What is the treatment for Wilson disease?

A
  • Early recognition and long-term copper chelation therapy (with D-penicillamine or Trientine) or zinc-based therapy (which blocks uptake of copper in the gut) has dramatically altered the usual progressive downhill course
  • Individuals with hepatitis or unmanageable cirrhosis require liver transplantation, which can be curative
2061
Q

What is α1-antitrypsin deficiency? What does α1-antitrypsin usually do?

A
  • α1-Antitrypsin deficiency is an autosomal recessive disorder of protein folding marked by very low levels of circulating α1-Antitrypsin.
  • The major function of this protein is the inhibition of proteases, particularly neutrophil elastase, cathepsin G, and proteinase 3, which are normally released from neutrophils at sites of inflammation
  • α1AT deficiency leads to the development of pulmonary emphysema, because the activity of destructive proteases is not inhibited.
  • It also causes liver disease as a consequence of hepatocellular accumulation of the misfolded protein.
  • Cutaneous necrotising panniculitis also occurs in a minor subset of patients
2062
Q

What are the mutations involved in α1-antitrypsin?

A
  • α1AT is a small glycoprotein synthesised predominantly by hepatocytes
  • The gene is very polymorphic, and at least 75 α1AT forms have been identified.
  • The general notation is “Pi” for “protease inhibitor” and an alphabetic letter for the position on the the gel; two letters denote the genotype of an individual’s two alleles
  • The most common genotype is PiMM, occurring in 90% of individuals.
  • The most common clinically significant mutation is PiZ; homozygotes for PiZZ protein have circulating α1AT levels that are only 10% of normal.
2063
Q

What is the pathogenesis of α1antitrypsin deficiency?

A
  • With most allelic variants, the protein is synthesised and secreted normally.
  • Deficiency variants show a selective defect in migration of protein from endoplasmic reticulum to Golgi apparatus; this is particularly characteristic of the PiZ polypeptide.
  • The mutant polypeptide (α1AT-Z) is abnormally folded and polymerised, creating endoplasmic reticulum stress and triggering the unfolded protein response, a signalling cascade that may lead to apoptosis.
  • All individuals with the PiZZ genotype accumulate α1-AT in the endoplasmic reticulum of hepatocytes, but only 10-15% of PiZZ individuals develop overt clinical liver disease.
2064
Q

What are the morphologic characteristics of α1AT deficiency?

A
  • α1AT deficiency is characterised by the presence of round-to-oval cytoplasmic globular inclusions in hepatocytes, which on routine hematoxylin and eosin stains are acidophilic, but are strongly periodic acid-Schiff (PAS)-positive and diastase-resistant.
  • The globules are also present, but in diminished size and number in the PiMZ and PiSZ genotypes.
  • Periportal hepatocytes contain the mutant proteins in early and mild forms like the PiZZ variant.
  • However, the number of globule-containing hepatocytes in a patient’s liver is not correlated with the severity of pathologic findings.
2065
Q

What are the clinical features of α1antitrypsin deficiency?

A
  • Neonatal hepatitis with cholestatic jaundice appears in 10-20% of newborns with the deficiency
  • In adolescence, presenting symptoms may be related to hepatitis, cirrhosis or pulmonary disease.
  • Attacks of hepatitis may subside with apparent complete recovery, or they may become chronic and lead progressively to cirrhosis
  • Alternatively, the disease may remain silent until cirrhosis appears in middle to later life
  • HCC develops in 2-3% of PiZZ adults, usually, but not always in the setting of cirrhosis
2066
Q

What is the treatment for α1 antitrypsin liver disease?

A
  • The treat, indeed the cure, for severe hepatic disease is orthotropic liver transplantation.
2067
Q

Hepatic bile serves two major functions. What are they?

A

1) The emulsification of dietary fat in the lumen of the gut through the detergent of bile salts
2) The elimination of bilirubin, excess cholesterol, xenobiotics, and other waste products that are insufficiently water-soluble to be excreted into urine

2068
Q

What are the clinical features of bile retention and deposition?

A

Tissue deposition of bile becomes clinically evident as yellow discolouration of the skin and sclera (jaundice and icterus, respectively) due to retention of bilirubin, and as cholestasis, when there is systemic retention of not only bilirubin but also other solutes eliminated in bile

2069
Q

When does jaundice occur?

A

When there is bilirubin overproduction, hepatitis, or obstruction of the flow of bile, any of which can disturb the equilibrium between bilirubin production and clearance.

2070
Q

The metabolism of bilirubin by the liver consists of four separate but interrelated events. What are they?

A
  • Uptake from the circulation
  • Intracellular storage
  • Conjugation with glucuronic acid
  • Biliary excretion
2071
Q

What is bilirubin made from?

A
  • Bilirubin is the end product of heme degradation.
  • The majority of daily production (0.2-0.3gm, 85%) is derived from breakdown of senescent red cells by the mononuclear phagocytic system, especially in the spleen, liver, and bone marrow.
  • Most of the remainder (15%) of bilirubin is derived from the turnover of hepatic heme or hemoproteins (eg, the P450 cytochromes) and from premature destruction of red cell precursors in the bone marrow.
2072
Q

How is bilirubin formed from heme?

A
  • Intracellular heme oxygenase converts heme to biliverdin (step 1), which is immediately reduced to bilirubin by biliverdin reductase
  • Bilirubin thus formed outside the liver is released and bound to serum albumin (step 2).
  • Albumin binding is necessary to transport bilirubin because bilirubin is virtually insoluble in aqueous solution at physiologic pH.
  • Hepatic processing of bilirubin involves carrier-mediated uptake at the sinusoidal membrane (step 3), conjugation with one or two molecules of glucuronic acid by bilirubin uridine diphosphate (UDP) or glucuronyl transferase (UGT1A1) (step 4) in the endoplasmic reticulum, and excretion of the water-soluble, non-toxic bilirubin bilirubin glucoronides into bile.
  • Most bilirubin glucoronides are deconjugated in the gut lumen by bacterial β-glucoronides and degraded to colourless urobilinogens (step 5).
  • The urobilinogens and the residue of intact pigment are largely excretes in the faeces
2073
Q

What happens for the urobilinogens created in the liver?

A
  • The urobilinogens and the residue of intact bilirubin pigment are largely excreted in faeces
  • Approximately 20% of the urobilinogens formed are reabsorbed in the ileum and colon, returned to the liver, and re-excreted into bile.
  • A small amount of reabsorbed urobilinogen is excrete in the urine
2074
Q

Two thirds of the organic material in bile are bile salts, which are formed by the conjugation of bile acids with taurine or glycine. What are bile acids? What is their role?

A
  • Bile acids, the major catabolic products of cholesterol, are a family of water-soluble sterols with carboxylated side chains.
  • The primary human bile acids are colic acid and chenodeoxycholic acid.
  • Bile acids in bile salts are highly effective detergents.
  • Their primary physiologic role is to solubilise water-insoluble lipids secreted by hepatocytes into bile, and also to solubilise dietary lipids in the gut lumen.
2075
Q

What often happens to bile acids once they’ve done their job?

A

95% of secreted bile acids, conjugated or unconjugated, are reabsorbed from the gut lumen and recirculate to the liver (enterohepatic circulation), thus helping to maintain a large endogenous pool of bile acids for digestive and excretory purposes

2076
Q

There are two important pathophysiologic differences between unconjugated bilirubin and conjugated bilirubin. What are they?

A
  • Unconjugated bilirubin is virtually insoluble in water at physiologic pH and exists in thigh comp-lexes with serum albumin. This form cannot be excreted in the urine even when blood levels are high.
  • In contrast, conjugated bilirubin is water-soluble, non-toxic and only loosely bound to albumin. Because of its solubility and weak association with albumin, excess conjugated bilirubin in plasma can be excreted in urine
2077
Q

Normally, a very small amount of unconjugated bilirubin is present as an albumin-free anion in plasma. Why is this important and in what disease?

A
  • This fraction of unbound bilirubin may diffuse into tissues, particularly the brain in infants, and produce toxic injury.
  • The unbound plasma fraction may increase in severe haemolytic disease or when drugs displace bilirubin from albumin.
  • Hence, haemolytic disease of the newborn may lead to accumulation of unconjugated bilirubin in the brain, which can cause severe neurologic damage, referred to as kernicterus
2078
Q

When are normal bilirubin levels and at what level will you see jaundice?

A
  • Serum bilirubin levels in the normal adult vary between 0.3 and 1.2mg/dL, and the rate of systemic bilirubin production is equal to the rates of hepatic uptake, conjugation, and biliary excretion.
  • Jaundice becomes evident when the serum bilirubin levels rise above 2-2.5mg/dL
  • Levels as high as 30-40mg/dL can occur with severe disease
2079
Q

What are the causes of predominantly unconjugated hyperbilirubinaemia?

A

Excess production of bilirubin
- haemolytic anaemias
- resorption of blood from internal haemorrhage
- ineffective erythropoiesis (eg, pernicious anaemia, thalassaemia)
Reduced hepatic uptake
- drug interference with membrane carrier
- some cases of Gilbert syndrome
Impaired bilirubin conjugation
- physiologic jaundice of the newborn
- breast milk jaundice
- genetic deficiency of UGT1A1 activity
- Gilbert syndrome
- diffuse hepatocellular disease (eg, viral or drug-induced hepatitis, cirrhosis)

2080
Q

What are some causes of predominantly conjugated hyperbilirubinaemia?

A
  • Deficiency of canalicular membrane transporters
  • Impaired bile flow from duct obstruction or autoimmune cholangiopathies
2081
Q

What is neonatal jaundice?

A
  • Because the hepatic machinery for conjugating and excreting bilirubin does not fully mature until about 2 weeks of age, almost every newborn develops transient and mild unconjugated hyperbilirubinaemia, termed neonatal jaundice or physiological jaundice of the newborn
  • This may be exacerbated by breastfeeding, as a result of the presence of bilirubin-deconjugating enzymes in breast milk
2082
Q

Multiple genetic mutations can cause hereditary hyperbilirubinaemia. What are some examples?

A
  • The hepatic conjugating enzyme UGT1A1 is a product of the UGT1A1 gene, it is the only isoform responsible for bilirubin glucuronidation. Mutations of HGT1A1 cause hereditary unconjugated hyperbilirubinaemias.
  • Crigler-Najjar syndrome type 1 is caused by severe UGT1A1 deficiency and is fatal around the time of birth, while in Crigler-Najjar type 2 and Gilbert syndrome there is some UGT1A1 activity and the phenotypes are much milder.
  • In contrast, Dubin-Johnson syndrome and Rotor syndrome result from other defects that lead to conjugated hyperbilirubinaemia. Both are autosomal recessive disorders and innocuous.
2083
Q

What is cholestasis? What causes it?

A
  • Cholestasis is caused by impaired bile formation and bile low that gives rise to accumulation of bile pigment in the hepatic parenchyma.
  • It can be caused by extrahepatic or intraheptic obstruction of bile channels, or by defects in hepatocyte bile secretion.
2084
Q

What are the clinical and lab findings in cholestasis?

A
  • Patients may have jaundice, pruritus, skin xanthomas (focal accumulation of cholesterol), or symptoms related to intestinal malabsorption, including nutritional deficiencies of the fat-soluble vitamins A, D or K.
  • A characteristic laboratory finding is elevated serum alkaline phosphatase and γ-glutamyl transpeptidase (GGT), enzymes present on the apical (canalicular) membranes of hepatocytes and bile duct epithelial cells.
2085
Q

The morphological features of cholestasis depend on its severity, duration, and underlying cause. What are these features?

A
  • Common to both obstructive and non-obstructive cholestasis is the accumulation of bile pigment within the hepatic parenchyma
  • Elongated green-brown plugs of bile are visible in dilated bile canaliculi.
  • Rupture of canaliculi leads to extravasation of bile, which is quickly phagocytose by Kupffer cells.
  • Droplets of bile pigment also accumulated within hepatocytes, which take on a fine, foamy appearance, so called “feathery degeneration”
2086
Q

What are the causes of large bile duct obstruction?

A
  • The most common cause of bile duct obstruction in adults is extrahepatic cholelithiasis (gallstones) followed by malignancies of the biliary tree or head of the pacnreas, and strictures resulting from previous surgical procedures
  • Obstructive conditions in children include biliary atresia, cystic fibrosis, choledochal cysts and syndromes in which there are insufficient intrahepatic bile ducts.
2087
Q

Subtotal or intermittent obstruction of the large bile duct may promote ascending cholangitis. What is this? What bacteria cause it? How does it present?

A
  • Ascending cholangitis is a secondary bacterial infection of the biliary tree that aggravates the inflammatory injury
  • Enteric organisms such as coliforms and enterococci are common culprits.
  • Cholangitis usually presents with fever, chills, abdo pain, and jaundice.
2088
Q

What is the most severe form of cholangitis? How does it often present?

A
  • The most severe form of cholangitis is suppurative cholangitis, in which purulent bile fills and distends bile ducts.
  • Since sepsis rather than cholestasis tends to dominate this potentially grave process, prompt diagnostic evaluation and intervention are imperative.
2089
Q

What are the morphologic changes in acute biliary obstruction and ascending cholangitis?

A
  • Acute biliary obstruction, either intrahepatic or extrahepatic, causes distention of upstream bile ducts, which often become dilated
  • In addition, bile ductules proliferate at the portal-parenchymal interface, accompanied by stromal oedema and infiltrating neutrophils.
  • These labyrinthine ductules reabsorb secreted bile salts, serving to protect the downstream obstructed bile ducts from their toxic detergent action
  • The histologic hallmark of ascending cholangitis is the influx of these periductular neutrophils directly into the bile duct epithelium and lumen
2090
Q
A