Respiratory Block Flashcards

1
Q

Where are mast cells found?

A

Found everywhere but mostly on sites exposed to the external environment. Also very common to be found near blood vessels, nerves and glands.

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

What are some external stimuli that can activate the mast cell?

A

Drugs such as morphine and vancomycin (red man syndrome). Mechanical, UV light/heat, Allergen (IgE), stings and osmotic stimuli causing hypertonic saline (exercise induced asthma).

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

What are some internal stimuli that can activate mast cells?

A

Activated complement and neuropeptides

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

Explain the allergen-induced mast cell degranulation process.

A

Antigen-specific IgE must be produced by initial exposure to allergen. IgE is typically produced in atopic subjects which is heritable.

Once IgE antigen specific is formed it will cross-link to the FceR1 that can be further activated by antigen binding.

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

What does it mean to be an atopic patient?

A

Means they have the atopic triad - asthma, hay fever, eczema.

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

How do we think desensitisation develops against allergens?

A

Exposure to the allergens stimulate Treg cells to form which oppose auto-immunity and suppresses Th2 development.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What are the characteristics of FceR1?

A

It has no intrinsic kinase activity so it has ITAMs (immuno receptor tyrosine-based Activation Motifs) that act as a scaffold for other kinases to become activated.

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

What is the pathway of FceR1 activation by allergen binding?

A

Allergen binds to the receptor, which phosphorylates itself. Syk is recruited which is a tyrosine kinase that brings in Lyn. The Lyn phosphorylates the FceR1 itself. They both activate other tyrosine kinases. Eventually leading to PLC (phospholipase C) activation.

DAG (PKC) and IP3(Ca2+ mobilisation) leading to eventual degranulation.

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

What are the outcomes of FceR1 activation? Specifically MAPK and PLC produced.

A

Activation of MAPK and PLC will cause degranulation to occur, Arachidonic Acid (AA) mobilisation, cytokine gene transcription.

MAPK is only involved in AA mobilisation and cytokine gene transcription.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Is mast cell degranulation cytotoxic?

A

No

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

What are the three different stages of mast cell communication with the internal environment?

A

Immediate (preformed granule) - histamine, TNF-alpha, heparin and tryptase (30-45 seconds)

Rapid (Peaks 10-30minutes) - Cys-LTs and PGD2

Slow (Hours to days) - IL-4 (promote IgE), IL-5 (recruits eosinophils) and GM-CSF (promote eosinophil life and macrophage activation) that enforce the Th2 phenotype. Essential activation of cytokine genes for Th2.

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

What is the response to the immediately released mediators (Histamine on H1 receptors)?

A

Bronchospasm, Pain, Itch, mucus secretion, vasodilation (hypotension), increased vascular leak (permeability). These usually involve the H1 receptors.

It can also act elsewhere to increase gastric secretion (H2 receptors), increased wakefulness (CNS) and positive chronotropic and ionotropic (H2).

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

What is the outcome of delayed release of mast cells (cys-Leukotrienes)?

A

LTA4 is the precursor to LTC4. LTC4 form LTD4, LTE4 that act on CysLT1 receptors.

No known physiological roles other than inflammation (good target).

Causes hypotension in anaphylactic shock due to vasodilation and decreased CO.

Causes mucus, oedema and ASM (airway smooth muscle) shortening - asthma.

Oedema and mucus found in hayfever caused by this.

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

Where are Cys-Leukotrienes produced and their stimuli for release?

A

Eosinophils, mast cells and macrophages (inflammatory cells only).

Stimulated by allergens, C5a and platelet activating factor. Increase in cystolic calcium produced in infection, allergic reactions and other forms of inflammation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What is the overall response from activating Cys-Leukotriene receptors? What about LTB4 specifically?

A

Bronchoconstriction, vasoactive and leak vessels (tissue oedema).

LTB4 involves promoting inflammation by attracting leukocytes. Does not act on the smooth muscles.

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

What are the site of drug actions along the AA pathway?

A

Glucocorticoid steroids (PLA2), Aspirin (COX), Coxibs (COX-2) and Montelukast (Cys LT receptor antagonists).

The Cys-LT receptors prevent mucus production and bronchoconstriction.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

What is the response of delayed and protracted release of cytokines from the mast cell?

A

This response is prolonged in cytokines - IL, TNF, CSF and chemokines.

Usually comes on slow and induces gene expression changes (proinflammatory cell infiltration) leading to inflammatory cell infiltration.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

What are some endogenous and pharmocological inhibitors of mast cell activation (specifically disodium cromoglycate)?

A

Endogenous: PGE2,adrenalin and cortisol.

Pharmacological: Disodium cromoglycate (reduces mast cell degranulation in some patients as well as eosinophil activation). It also stimulates annexin-1 release that resolves inflammation.

Genereally used for allergic responses of mucosal surfaces (not orally active)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

How does omalizumab work to inhibit mast cell activation?

A

It is a humanised antibody to IgE. Prevents the IgE from cross-linking to FceR1 so mast cells are unarmed.

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

How are the use of aspirin and selective COX-2 inhibitors for asthma and anti-allergic agents?

A

Mixed roles of prostaglandins in allergies. No real benefits in asthma or hayfever. Sometimes aspirin can provoke asthmatic symptoms (but aspirin induced asthma can be treated with LTRA (leukotriene receptor antagonists)).

Blocking COX leads to increased Leukotriene production.

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

Are H1 receptor antagonists useful in colds or asthma?

A

Not useful - only to some extent.

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

What are the three classes of H1 competitive antagonists and their adverse effects and example?

A

Sedative - promethazine (sedative)

Non-sedative - Terfinadine (sudden ventricular arrhythmia)

Newer non-sedative - Loratidine (reduced risk of unwanted cardiac effects)

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

How many classes of hypersensitivity reactions are there and what do they generally involve?

A

Type I - IgE, mast cell and lipid mediators

Type II - IgG and IgM against cell bound or ECM antigen

Type III - IgM and IgG immune complex deposition

Delayed Type Hypersensitivity IV - CD4 mediated

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

What does Type I hypersensitivity involve and what does it cause?

A

Usually causes allergy through immune-mediated inflammation to harmless environmental antigens.

Found in atopic individual who have: High IgE levels, large numbers of eosinophils and large numbers of IL-4 secreting Th2 cells.

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

What are the general steps to Type I hypersensitivity after IgE mediated inflammatory response to antigens?

A

Sensitisation phase which is followed by;

Response phase: local (common) or systemic (rare). Locally it will cause rhinitis, bronchoconstriction and conjuctivitis. Systemically will cause anaphylaxis.

Responses have both immediate and late phase.

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

What contributes to the effector (allergic) mechanism?

A

Allergens, Th2 cells, IgE, FceR1, mast cells and eosinophils

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

What are the typical allergens and their characteristics that elicit Type I hypersensitivity?

A

Pollen, house dust mite, food, etc.

Should have repeated exposure, highly soluble (to pass through mucus), very stable and hard to degrade and introduced in low doses.

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

What is the mechanism of sensitisation in Type I hypersensitivity?

A

Low dose Antigen enters the mucosal route. It is taken up by APC and displayed to naive T cells. The APC produce IL-33 that indirectly releases IL-4 when stimulating the T-cell. This leads it to differentiate to Th2 cells (increasing in IL-4, IL-5 and IL-13)

Upon re-exposure the Th2 already present will promote IgE secretion proliferation and isotype switching.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

How does the Th2 differentiation work during the sensitisation phase?

A

The DC’s do not actually produce IL-4 to co-stimulate the naive T-cell. DCs produce IL-33 which activate basophils to directly secrete IL-4 onto the naive T-cell. This aids the differentiation to Th2 and productiong of IgE.

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

Why are basophils key for the initiation of allergen specific Th2 responses?

A

They can act as APC (express MHC I and II, PRR) and importantly secretes IL-4.

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

Why does Th2 differentiation lead to IgE isotype switching?

A

The activated Th2 cells can stimulate B-cells to isotype switch to IgE.

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

What is the response phase of Type I hypersensitivity?

A

Found in mucosal and epithelial tissue near blood vessels. Mast cells contain pre-formed granules, binds IgE to FceR1 and surface bound IgE is very stable.

Once allergen is bound it will result in granular exocytosis and the synthesis of lipid mediators, cytokines and chemokines.

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

What is the immediate phase after mast cell activation - wheal and flare?

A

Occur in seconds due to pre-formed granules that are rapidly metabolised.

It causes blood vessels to dilate and leak plasma. Localised swelling around the site of challenge (wheal). The blood vessels dilate further and engorge with blood (flare)

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

What is the late phase after mast cell activation?

A

Induced mediators released such as chemokines, cytokines and leukotrienes.

Involves cell infiltration and prolonged oedema and may/or may not cause smooth muscle contractions.

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

What are the different types of allergic reactions depending on where it occurs?

A

GI tract - Diarrhoea vomiting

Skin - swelling itching urticara

Airways - nasal blockage, coughing, phlegm and asthma

Blood vessels - increase tissue fluid and cell infiltration, anaphylatic shock

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

What are the roles of eosinophils in the allergic response?

A

Present in mucosal lining as protective role against parasites. Found LATE in allergic reactions. Produce toxic granule derived proteins and free radicals (which is usually for parasites - but will cause tissue remodeling). Eosinophils will also produce chemical mediators leading to epithelial cell activation and inflammatory cell recruitment and activation.

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

Why is there increased eosinophils during allergies (bypassed)?

A

Increased production in bone marrow due to IL-5 from Th2 and mast cells. The eosinphils are activated and infiltrate into tissue. There is decreased threshold for activation due to the sensitisation phase of IgE binding to FceR1.

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

What are the symptomatic treatment of allergies?

How do these work?

A

Adrenanine - Anaphylaxis (reforms tight endothelial junctions, relaxes smooth muscle as bronchodilator, increase HR)

Inhaled B-agonists - Asthma (Bronchodilators)

Antihistamines - hives and allergic rhinitis (Block histamine and reduces itching and decreases oedema)

Corticosteroids - provides broad non-antigen specific treatment of symptoms (suppresses chronic inflammation and broad immunosuppression by blocking gene transcription - multiple side effects and tolerance builds)

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

What is immunotherapy AKA desensitisation?

A

Administer increasing dose of allergen to induce T-cell tolerance.

Tolerance by: Anergy (decreased T cell proliferation), deviation of secreted cytokines, stimulate apoptosis and production of Treg cells.

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

What is Type IV hypersensitivity and what does it cause?

A

Cell mediated with heavy involvement of T cells and macrophages - mostly Th1 but sometimes CTL.

Generally caused by microbial infection, intradermal injection of protein antigens and contact with chemicals through the skin.

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

How does the sensitisation phase of delayed type hypersensitivity (DTH) work?

A

The antigen is taken up through the skin of mucosa that is taken up by APC and presented in the lymph node. This activates Th1 and CD8 T cells. The T cells leave the lymph nodes into the circulation and to the tissue of interest.

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

Why is there an accumulation of T cells and macrophages in DTH and what is its implications?

A

There is an accumulation because of antigen persistence. Leads to continuous release of the cytokines and chemokines. Means more infiltration, tissue destruction, adhesion molecules, monocyte production and ROS and RON.

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

What are three examples where persistent antigen can cause DTH?

A
  1. Contact sensitivity - poison ivy and TB tests will have central and effector memory cells triggered when re-exposed after previous sensitisation.
  2. M. Tuberculosis - Cannot clear organism
  3. Celiac Disease - Exposure to wheat products induce Th1 dependent immunopathology of intestinal wall.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

How long does DTH take to manifest?

A

Usually takes a few days because the cells involved are slower

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

What is the mechanism of contact sensitivity?

A

First exposure to antigen will be taken up by APC leading to production of memory T cells. When re-exposed to the antigen the memory T-cells will activate and cause excessive inflammation (a lot of IFN-gamma and recruit macrophages)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

How does M. Tuberculosis cause DTH?

A

Inhale TB that multiply in resisdent alveolar macrophages. This results in increase cytokine and chemokine production inducing monocyte influx. DCs migrate to lymph node that activates and recruits CD8, Th1 cells to the infection to further activate macrophages.

But after all of this the myocobacteria still persists leading to prolonged infection.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

What is the result of contracting M. Tuberculosis?

A

M. TB induces granuloma formation essentially walling off the pathogen.

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

How does celiac disease cause DTH?

A

Results from hypersensitivity to components of gluten (gliadins). But it requires exposure unlike the persistence of TB. The celiac disease results in damage to the small intestine.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

What kind of MHC molecule is found in patients with celiac disease?

A

>90% have HLA-DQ2

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

What is the mechanism of celiac disease and DTH?

A

The T cells recognise gliadin peptides through HLA-DQ2. Generally prefers to bind -ve AA side chains.

Unmodified gliadin proteins are +ve charged so cannot bind. But when tissue transglutaminase 2 will change gliadin proteins to -ve charged. Deamidated gliadin will bind to the HLA-DQ2 effectively now. Able to trigger immune response.

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

What is the definition of Asthma?

A

Chronic inflammation of airways associated with hyper-responsiveness leading to recurrent episodes of wheezing, breathlessness, chest tightness and coughing. Often reversible airflow limitations (not fixed airway obstruction).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

What are the host and environmental factors that can influence asthma?

A

Host: Genetic, atopy, airway hyper-responsiveness (narrow too early and too much), Gender and obesity.

Environmental Factors: Indoor allergens, outdoor allergens, occupational sensitisers, tobacco smokes, air pollution, respiratory infection and diet.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

What are the cells and mediators involved in asthma?

A

Allergen activate mast cells: to eosinophil and neutrophil leading to nerve activation.

Allergen activate macrophage/DC and Th2 cell.

Eventual responses lead to mucus hypersecretion, vasodilation (angiogenesis), plasma leak (oedema), bronchoconstriction and even subepithelial fibrosis.

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

What are the three primary responses in asthma?

A

Bronchospasm, airway mucosal oedema and mucus plugs (other mechanism to narrowing especially in smaller airways).

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

There are three contributors to narrowing of airways how do drugs target these mechanisms and do they act as preventers, relievers or controllers?

A

Airway smooth muscle contraction - reliever, controller and preventer

Bronchial oedema - Preventers

Mucus hypersecretion - Preventers

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

What is the impact of tidal stretch to the lumen size and muscles pulling on the airways?

A

Inflated airways the airway smooth muscles will open the airways the most. Deflated usually has very little pool and therefore is likely to have more resistance.

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

Where are the airway smooth muscles found?

A

They are arranged circumferentially around the airways that narrows the lumen when contracted. Found in the bronchioles.

There is no direct SNS but there is PS. The B-adrenoceptor comes from Adr.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

How does the load and velocity of contraction affect airway resistance?

A

Load decreases during expiration causing increase in airway resistance and also increases likelihood of airways collapse.

Unloaded muscle shorten faster

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

What is the contractile mechanism of ASM? How is calcium level controlled?

A

Similar to skeletal muscle contraction. It is important to regulate calcium levels. Increase in calcium usually done by PLC and IP3that releases calcium stores.

Plasma Ca2+ ATPase, SR ATPase taking it back up into internal stores.

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

What determines the level of airway smooth muscle cell contraction?

A

The balance between contraction (ACh, HA, LTC4, LTD4) and relaxation (PGE2, adrenaline, PGI2)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

How can calcium ion oscillation regulate smooth muscle tone?

A

Calcium will affect myosin light chain kinase. When the MLC is phosphorylated it becomes more calcium sensitive that can influence the contraction.

On the otherhand protein kinase A can dephosphorylate the MLC-phosphate.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

What are the other airway smooth muscle dysfunction other than contracting?

A

Not only constrict and relax it also contributes to wall volume in airway remodeling and inflammation.

  • Proliferation, migration, secretion of cytokines and secretion of ECM proteins.

Shows subepithelial collagen thickening, infiltration of inflammatory cells, increase mucosal vascularity and smooth muscle volume.

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

How to measure the severity of asthma?

A

FEV1 numbers

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

How do SABA (short acting B-agonists) work and their characteristics?

A

Salbutamol with rapid onset and B2 selectivity leading to bronchodilator. The adverse effects may be tremors, tachycardia and hypokaelamia.

Tolerance build up.

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

What is the mechanism of B2 agonists to reduce bronchioconstriction?

A

Increase in cAMP and PKA inhibits IP3 leading to reduced calcium levels. Reduced calcium levels means less MLCK (myosin light chain kinase) activation.

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

What are LABA and their characteristics?

A

Salmeterol are longer acting B2 agonists. The duration can be up to 12 hours. Usually used for prophylaxis and used in combination.

Monotherapy is associated with increase in mortality and morbidity.

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

How is B2-adrenoceptor activation regulated? Especially the desensitisation of these receptors?

A

Within seconds the receptors are phosphorylated that desensitises it by PKA.

Within minutes the receptors are sequestrated (internalised) - may be degraded or recycled to plasma membrane.

Within hours the receptors are down regulated by decreased mRNA stability and rate of transcription.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
68
Q

Are muscarinic antagonists good for asthma? How do they work?

A

Not generally used in asthma - more for COPD.

Less bronchodilation than B2 agonists.

Usually M3 receptors on ASM.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

What are the rough levels of evidence that we should use for clinical practice?

A

Level I - Systematic review of randomised clinical trials

Level IV - Case series

Randomised controlled trials are the best

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

What is PICOT and its use?

A

P - Population

I - Intervention

C - Comparator/Control

O - Outcome

T - timing

Used to help determine what therapy to commence on.

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

What is internal validity?

A

Extent of validity of results that come from the study. Did it answer the question proposed? How well is the study design and data collection?

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
72
Q

What is the point of randomisation in study designs?

A

Reduce confounding in studies. Try to make treatment groups identical in all aspects other than the intervention.

Reduces selection bias (investigators may assign subjects with bias)

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

What is stratified randomisation?

A

Break up key confounders and randomise people into those groups to make it more even. Makes the composition of groups even more similar and therefore reduces potential of confounding.

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

What is the purpose of Table 1 in studies?

A

Shows the base line characteristics of trials and more important should show that groups are evenly matched. The parameters are confounders.

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

What is the point of blinding in studies and its importance?

A

Non-awareness of intervention allocated. This will reduce information bias. Knowing what intervention you get can influence the outcomes.

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

What is objective outcome ascertainment?

A

Outcomes must be strict, standardised and have objective criteria. Multi-centre studies need to have a centralised process.

  • Reduce information bias.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
77
Q

What is the intention-to-treat analysis and the point of it?

A

Assume subjects remain in the groups they were randomised in. This is to reduce selection bias.

Since drop outs selects for healthier group in drug group. Misrepresentation of new drug being better than placebo.

It always gives underestimate of the treatment effect - gives us more confidence of the results.

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

How to interpret statistical analysis?

A

P value and confidence interval (this gives precision)

P value - gives probability that chance gave us the results. Conventionally set it as p < 0.05 significant.

Confidence interval: 95% chance the true value lies within this particular interval.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
79
Q

What is the number needed to treat?

A

Number of people needed to undergo treatment to prevent one outcome.

Marker of intervention efficiency.

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

How to calculate number needed to treat?

A

NNT = 1 / (absolute risk or rate reduction)

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

What affects the NNT (number needed to treat)?

A

Affected by relative effect (often constant) and the underlying likelihood of outcome.

It is important to work out what the chance of the outcome is of occuring.

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

What is meant by external validity?

A

How similar is the PICOT in the study to my patient? Should I use the particular intervention for my case specifically?

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

What are the main functions of the lungs?

A

Oxygen the pulmonary arterial blood, remove carbon dioxide from blood and maintain acid-base balance.

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

What is the metabolic requirement at rest and during exercise?

A

At rest (BMR) - 250mL/min O2 and 200mL/min CO2

During exercise - >4000mL/min O2 and >4000mL/min CO2

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

What carries oxygen in the blood?

A

4 oxygen molecules per Hb molecule

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

How is CO2 transported around in the plasma?

A
  1. Dissolved - 10%
  2. Attached to proteins (include Hb) - 30%
  3. Bicarbonate - 60%
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
87
Q

Where does the exchange of oxygen and carbon dioxide occur over?

A

Alveolar and capillary membrane (AC membrane)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
88
Q

What is the alveolar-capillar membrane composed of and its characteristics?

A

Very thin layered with two cells (alveolar and endothelial). Has a large surface area. These two qualities make it ideal for gas exchange.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
89
Q

What is the rate of diffusion across the AC membrane driven by?

A

Fick’s Law

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
90
Q

How long does it take for the oxygen to diffuse into the venous blood?

A

0.75 seconds that is normal for rest. During exercise there is only 0.25 seconds available.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
91
Q

When is gas exchange the most procifient?

A

When there is ventilation to perfusion matching.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
92
Q

What is the anatomy of the airways briefly?

A

Upper airway - nose, mouth, pharynx and larynx

Trachae and bronchi.

Multiple branching of bronchi into bronchioles eventually terminating into alveoli.

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

What are the respiratory muslces involved in inspiration and expiration?

A

Inspiratory - diaphragm, external intercostals and sternocleidomastoids

Expiratory - Internal intercostals and abdominal muscles

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

How do our lungs inflate with air?

A

Inspiration via negative pressure created. Contraction of diaphragm increases longitudinal and lateral dimensions of the thorax (negative intra-pleural pressure).

Contraction of the external intercostals increases the AP diameter of the thorax (negative intra-pleural pressure).

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

Is inspiration and expiration generally passive or require use of muscles?

A

Inspiration requires muscle to create the negative pressure but the expiration is generally passive because of the lung recoil to push the air back out.

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

What makes up the work of breathing?

A
  1. Resistive - the airflow through bronchi
  2. Elastic - the expansion of lungs and chest wall (lungs have some degree of stiffness and want to collapse down).
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
97
Q

What are the different pressure levels of gas found in the blood and the pH level normally?

A

PO2 - 100mmHg

PCO2 - 40mmHg

pH - 7.4

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

What can you notice when you breath through an obstructed airway?

A

Increased sensation of breathing, increased use of respiratory muscles, active exhalation, longer inspiration, longer expiration and reduced maximum ventilation.

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

Why do we feel an increased sense of breathing?

A

There may be an increase in load (such as stiff lungs, narrow airways, chest wall and diaphragm that makes it harder to breath).

There could be a larger drive to breath (usually by receptors or even when we purposely hyperventilate).

There is a larger work required of our body. Such as when we exercise but we perceive it as normal.

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

What is increased in the sensation of breathing usually referred to as by people? When is it perceived as a sensation or symptom?

A

It is breathlessness. Sensation when it is normal for it to occur whereas symptom when it is inappropriate.

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

What is the work of breathing usually like at rest? What does it involve?

A

Normally very low at around 3% of total oxygen usage. Involves stimulation of phrenic nerve for diaphragm and external intercostal muscles through intercostal nerves. Increasing volume of thorax will lead to generation of negative intra-pulmonary pressure.

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

How is the work of breathing affected when there is an airway obstruction?

A

Respiratory muscles need to generate higher pressures to overcome the obstruction to air flow (resistive work of breathing). It also makes expiration become active.

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

What are the important consequences of increased work of breathing?

A
  1. Recruiment of accessory muscles (scalene and sternocleomastoid muscles).
  2. Increased oxygen consumption by respiratory muscles.
  3. Risk of respiratory muscle fatigue if the airway obstruction is severe.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
104
Q

Respiratory muscle fatigue is a common cause of ventilatory failure - what do we define as ventilatory failure?

A

PaO2 <60mmHg and PaCO2 > 50mmHg

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

What are the different lung pressures during breathing?

A

Intra-alveolar pressure < Patmospheric during inspiration

Intra-alveolar pressure > Patm during expiration

Intra-alveolar pressure = Patm at the end of inspiration and expiration

Intra-pleural pressure is ALWAYS < intra-alveolar pressure because of the elastic recoil of lungs and chest wall.

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

Explain how we do a spirometry and what we can get out of it, FEV1 and FEV1/FVC?

A

Spirogram involves breathing in TLC then exhaling as quickly and much out as possible. The graph will increase significantly during FEV1 (70-80% of FVC) until it stabilises to give FVC.

Airflow obstruction will generally give a more linear curve meaning that exhalation is much longer.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
107
Q

How else can we measure prolonged inspiration and expiration? How does the test work? How do we interpret it?

A

Use of flow-volume loops. Involves taking a breath in to TLC then blow out as quickly as possible - once it has been exhaled the patient needs to breath in through the tube again to TLC.

Different locations of obstruction will lead to different flow-volume loops.

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

What are the summary of mechanical effects of airflow obstruction?

A
  1. Increased sensation of breathing
  2. Increased respiratory muscle effort
  3. Active exhalation
  4. Prolonged inspiration and exhalation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
109
Q

If the respiratory muscles become fatigued or if metabolic demand exceed ventilatory capacity during exercise - what are the changes to PO2 and PCO2?

A

There will always be a decrease in PaO2 and increase in PaCO2

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

What is ventilation perfusion matching?

A

Gas exchange is most efficient when this matching is 1 for individual AC units (alveolar capillary unit)

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

V/Q scan in normal patient compared to those with COPD?

A

Normal people have even distribution and COPD has mismatch between the two.

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

What are the consequences of V/Q mismatch?

A

Not all oxygen binds to Hb molecules - reduce PaO2so that some blood returning to the LA is not fully oxygenated.

Usually as a result from less ventilation than perfusion.

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

What is meant by a shunt?

A

Extremely low form of V/Q unit where there is no ventilation at all and V/Q = 0.

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

What happens to compensate for low V/Q units?

A

Usually results in vasoconstriction of blood around the areas with low ventilation to reduce hypoxaemic effects.

Sometimes the vasoconstriction may elevate the pulmonary artery pressure.

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

What happens with V/Q mismatch when there is a high V/Q unit?

A

Usually results in wasted ventilation because there will be more ventilation of the units than required to oxygenate the blood.

  • Leads to increase in physiological dead space
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
116
Q

How can we generally measure the integrity of A-C membrane?

A

Measure the CO2 levels.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
117
Q

Describe asthma and the two ways it causes airway obstruction.

A
  1. Bronchial smooth muscle constriction which is responsive to B agonists, anticholinergics, methyl-xanthines.
  2. Airway inflammation: responsive to corticosteroids, leukotriene antagonists and mast cell stabilisers.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
118
Q

What is COPD and how does it obstruct airways and its consequences?

A

COPD is smoking related disease that causes inflammation of bronchial mucosa, loss of elastic support of small airways and destruction of the alveolar-capillary membrane.

  1. Collapse of small airways -> airflow obstruction.
  2. Impaired gas exchange -> because of VQ mismatch and loss of A/C membrane
  3. Reduced capillary bed -> pulmonary hypertension
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
119
Q

How is AC membrane integrity used to differentiate COPD and asthma?

A

Typically COPD has a lack of AC membrane integrity.

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

How can we quantify the Alveolar-arterial gradient for oxygen?

A

PAO2 = PiO2 – PACO2 / RQ

PaCO2 = PACO2 and RQ = 0.8 and PiO2 = 150 at sea level.

Then for gradient you need PAO2 - PaO2

Normal levels < 15-30.

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

What is the difference between the conducting and respiratory parts of the respiratory system?

A

The conducting part is what carries the air along whereas the respiratory part is where gas exchange actually occurs.

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

What is the nasopharynx?

A

This is the nose area.

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

What are the roles of the sinuses, olfactory epithelium and turbinates in the nasopharynx?

A

The sinuses is used for phonation and conditioning air. Olfactory epithelium is for the sense of smell. Turbinates warm and moisten the air so that the air will not dehydrate the airways.

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

What is the epiglottis and larynx and its functions?

A

The epiglottis and larynx divert food away from the airways. Larynx is involved in phonation (speech).

The larynx is the uppermost part of the trachae.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
125
Q

What is the respiratory epithelium?

A

Lines the airways and much of the nasopharynx (lines conducting part instead of respiratory part)

Pseudostratified epithelium with ciliated cells, secretory cells (goblet and deeper glands) of mucus and sensory cells to initiate coughing to expel irritants.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
126
Q
A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
127
Q

Why do we have mucus secreting cells and ciliated cells?

A

This is to protect against air that have dust particles, fungal and bacterial spores.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
128
Q

How do goblet cells and cilated cells aid in the protective mechanisms?

A

The mucus layer traps particles that are then swept up by the cilia to the epiglottis. Ends up in the stomach for sterilisation.

Smokers who have damaged cilia cells rely heavily on coughing to shift the mucus upwards of the lungs.

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

What is the trachae and its structures?

A

Long tube reinforced with hyaline cartilage. The cartilage is in C-shaped formation with smooth muscle bridging it.

The trachae has three layers - mucosa, submucosa and adventitia.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
130
Q

What is the difference between the mucosa and submucosa layers and what are found in each layers?

A

Mucosa is much thinner than the submucosa layer. Submucosa is much broader with connective tissue and glands that secrete mucus.

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

What is found in the adventitia layer of the trachae?

A

It contains cartilage and outer layer of connective tissue.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
132
Q

What is the bronchi and its structures that differentiate it from the trachae?

A

The trachae splits into five different bronchi that lead to different lobes in the lungs (3 - right and 2 -left).

Initialy looks like trachae but with thinner walls. As you progress down the airways cartilage rings become cartilage plates instead (lose C-shaped rings).

Smooth muscle, glands and lymphoid nodules still present.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
133
Q

How to differentiate a bronchiole from a bronchi and what structures does it have?

A

Bronchioles no longer have the cartilage. Over the length of the bronchiole they being to lose goblet cells and ciliated columnar cells and gain Clara cells.

Still have radial connective tissue, smooth muscle and ciliated cells extend further down than goblet cells (lose the goblets earlier).

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

What are clara cells and what do that do?

A

These are columnar to cuboidal cells with short microvilli (instead of cilia). Generally found deeper in bronchioles.

They secrete surfactants to destroy surface tension (causes alveoli to collapses). They may also neutralised toxin.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
135
Q

What are terminal bronchioles?

A

Last level of the conducting system. No golbet cells. Clara cells plus cuboidal epithelium with some cilia. There is one or two layers of smooth muscles and they give rise to respiratory bronchioles (first site of gas exchange).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
136
Q

What are respiratory bronchioles and what do they do?

A

First respiratory structures to appear where alveoli will appear intermittently. Alveoli are thin walled pouches. The epithelium of the respiratory bronchiole are made up of cuboidal to squamous cells.

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

What are the alveolus for and what are their structures?

A

Surface for gas exchange. Lined with simple squamous epithelium. The wall contains many pulmonary capillaries (deoxygenated blood within them). Also the individual alveoli are connected by pores so that they become connected.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
138
Q

What is the interalveolar septum and what is it made of and function?

A

This contains reticular fibres and elastin fibres between the alveoli. They prevent the alveoli from collapse during breathing when negative pressure is generated. Positive air pressure helps it stay open.

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

What are the pulmonary capillaries surround the alveoli and its structure?

A

They form dense anastomising network of vessels for exchange of gases. The pulmonary epithelium is very thin.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
140
Q

What are type I pneumocytes?

A

These are present in alveoli made up of simple squamous epithelium. It provides about 95% of the surface area of alveoli. It provides the exchange surface. The basal lamina is prominent because it limits the gas exchange process.

  • There are tight junctions between the pneumocytes to limit ECF leakage.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
141
Q

What are type II pneumocytes and their role?

A

More numerous than type I but only found on 5% of the surface area. These are cuboidal cells in the angles between alveoli. They consist of short microvilli and lamellar bodies.

  • The lamellar bodies secrete surfactants to prevent collapse from surface tension.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
142
Q

How are the pneumocytes replaced? Are they replaced?

A

There is turnover of pneumocytes that involves Type I dying and being replaced by stem cells.

The Type II can divide and give rise to new type I and II as required (stem cells).

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

What is the blood-gas barrier?

A

This is where the exchange of air takes place across the Type I pneumocyte and endothelial of the capillaries.

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

What contributes to the blood-gas barrier?

A

Contains mucus, surfactants, type I pneumocytes, basal lamina, connective tissue, basal lamina, endothelial cell then finallay plasma.

The thinnest barrier is when there is essentially no connective tissue between the two basal laminas.

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

What are intra-alveolar macrophages?

A

These are macrophages that exist in the alveoli and essentially outside of the body.

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

What are the outcomes for intra-alveolar macrophages when they have ingested particles?

A

They can migrate up the airways until they are carried away by ciliated cells. Or some macrophages end up in the interalveolar septum with the particles they ingested (return to body and eventually die and release their contents).

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

What is the significance of the visceral pleura having blood and lymphatic vessels?

A

There is abundant lymphatic systems in the pleura to drain fluids. But there are also vessels opened on the surface of the pleura (releases fluid into the space). The consequence of this is that tumour cells can use this pathway to metastasise.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
148
Q

Why is there a lag phase between the innoculation of virus until we see growth and how much growth do we observe from viruses?

A

The lag phase is when the virus components are being synthesised and eventually assembled together to form viable viruses. The number of viruses produced are enormous.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
149
Q

What are the steps involved in viral replication?

A

Attachment, penetration, uncoating, genome replication, RNA synthesis, protein synthesis, assembly and release of viral particles.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
150
Q

How do viruses attach to the host cells?

A

The viral attachment protein must bind to a receptor found on the cell surface of the host. This could be proteins or glycoproteins expressed on the surface (these receptors are just used for normal cell processes)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
151
Q

Why would some viruses use two attachments to bind to the cell surface? Give an example of one.

A

HIV viruses uses two different cell receptors. gp120 binds to CD4 as primary receptor. This induces conformational change in the glycoproteins - gp41 now binds to CCR-5 as a secondary receptor.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
152
Q

How do viruses penetrate into the host cells?

A

This is achieved via two ways.

  1. Lipid envelope viruses fuse with the host cell membrane and then releases the virus nucleocapsid directly into cytoplasm.
  2. Other viruses enter the cell via endocytosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
153
Q

What is the process of virus uncoating once it has penetrated the cell?

A

It just refers to the release of viral genome from its protective capsid.

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

What is an example of a virus that enters the cell via the endocytosis pathway?

A

Togavirus does this - it triggers endosome release when the pH is low.

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

What happens during the amplification phase of the viral genome and proteins?

A

Nucleic acid replication will produce new viral genomes. DNA replicate in nucleus and RNA based virus replicate in cytoplasm.

mRNA is produced and used to produce proteins.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
156
Q

What machinery is available in the host cell for transcription, translation and nucleic acid replication?

A

DNA dependent DNA polymerase, DNA dependent RNA polymerase and ribosomes, tRNA and AA.

  • no RNA-dependent RNA polymerase present for RNA invading viruses.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
157
Q

What is meant by plus sense strand of virus? And what is meant by negative?

A

Means the strand of genetic material is the same as mRNA. Negative sense means that it is the complementary base sequence.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
158
Q

What is the characteristic of Polio virus genome?

A

Linear, single stranded, plus sense RNA.

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

What are the two possible outcomes of plus sense RNA viruses, what pathway does it typically go with?

A

The RNA can be replicated to produce more mRNA but that would require RNA-dependent RNA polymerase.

Another pathway is that it will be directly translated into proteins. This protein is then autocleaved by itself to produce structural coat proteins, proteases and RNA POLYMERASE.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
160
Q

What happens when the +RNA viruses are translated and produce RNA polymerase?

A

The RNA polymerase can now enter the through the other pathway. It will allow the RNA to replicate to form new plus strands. It will produce negative and positive strands.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
161
Q

How would -RNA viruses translate if there are no RNA polymerase in the host cells?

A

The -RNA viruses carry RNA polymerase with it.

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

Each virus must produce mRNA to translate into proteins. There are different types of viruses with different approaches to these (Baltimore classification)

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

Do DNA viruses need to carry their DNA-polymerase?

A

The DNA viruses do not need it because the host cells have the enzymes already.

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

Do RNA viruses need to bring along its own RNA polymerase?

A

Negative strands - must carry their own polymerase

Positive strands - do not need to carry it

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
165
Q

What is the translation and packaging process involved for viral proteins?

A

Translation of the proteins occur on the ribosomes in cytoplasm. After the polypeptide is formed it is cleaved by virus-coded proteases.

Glycosylation of envelope glycoproteins occur in RER and golgi vesicles -> allows them to be deposited on cell membrane.

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

Explain the assembly and release of viruses from the host cell.

A

For non-enveloped animal viruses: All have icosahedral structure. It can simply spontaneously assemble with capsid proteins around the nucleic acid genome. Final cleaving will need it to produce mature infectious virions. It will accumulate in the cell and eventually lyse.

For enveloped viruses: Release occurs by budding from the cell surface. Patches of viral envelope glycoproteins accumulate on membrane. Capsid proteins and nucleic acid condense directly adjacent to cell membrane. The nucleocapsid then bulges out and forms a new enveloped virion.

Another pathway for enveloped viruses is to use secretory pathway to exit the cells. Enclosed in Golgi derived vesicles that are then released through fusion of membrane.

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

What changes can occur to the host cell when the virus infects it?

A
  • Lytic infection (overwhelming the cell - enteroviruses, reoviruses): Fast release and death of cell.
  • Chronic inflammation (Hep C): Slow release without cell death.
  • Latent infection (herpesviruses): No harm to cell and can emerge later as lytic infection.
  • Transformation to tumour cells (oncogenic retroviruses)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
168
Q

What is cytopathic effects (CPE)?

A

This is the morphological changes that can be seen due to infection through a light microscope.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
169
Q

What are inclusion bodies?

A

Inclusions are accumulated areas of viral proteins at the site of viral assembly.

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

Why do some viruses cause cell transformation - especially oncogenic transformations?

A

Viruses encode for oncogenes that is associated with tumour production. These oncogenes were meant to promote growth promoting properties but their expression can lead to uncontrolled proliferation. Other viruses can affect tumours because of the cellular version of the oncogene is activated.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
171
Q

What are some ways the viral genetics can change?

A

Mutation (errors in RNA replication due to the lack of proof reading - or two viruses infect the same cell), recombination (exchange of stretches of nucleic acid between genomes of SIMILAR sequences especially DNA viruses), reassortment (swapping of segments for viruses that have segmented genomes - with many different properties of viruses).

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

What can halt the infectious process?

A

Antibody blocks the uptake and/or neutralises progeny virus. Killing of the infected cells by cytotoxic T cells, NK cells or antibody mediated processes. The interferons released will initiated a large number of anti-viral mechanisms. Blocking the replication cycle by using specific antiviral drugs.

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

What are the uses of nucleoside analogs e.g. Acyclovir?

A

It is used for herpesvirus infections. Works by attaching the nucleoside analogue to the extending DNA strand. But it does not have the 3’ OH group so it can no longer extend the chain further.

Acyclovir is an guanosine analogue. It is specific for Herpes because the nucleoside analogue must be phosphorylated three times before it can be attached. The virus’ thymidine kinase is required for this step - giving specificity for infected cells.

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

What are the general principles of viral pathogenesis?

A

Must gain entry to body, multiply, spread, target appropriate organ.

To maintain nature viruses it must be shed into environment, taken up by arthropod (insect) vectors or needle and passed congenitally.

Viral replication can be local or systemic. It will either be cleared or persist.

The virus infection location will be determined by whether or not they have the receptor for the viruses.

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

What are the different routes of entry for viruses?

A

Conjuctiva, respiratory, parenteral inoculation, alimentary (food and digestive) tract, urogenital tract and skin.

Enters through epithelial cells with mucosa. Other areas have dying cells and keratinisation making it a bad environment for viruses.

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

How do viruses infect the respiratory tract and what is the significance of the respiratory tract and viral infections?

A

Most important site of entry. Usually from aerosal inhalation or transmission of infected nasal secretions. The smaller the virus the further down it will lodge in the airways.

Viruses will attach to specific receptors on epithelial cells. It can either localise or spread away.

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

What are the barriers to infection found in the respiratory tract?

A

Mucus, cilia, alveolar macrophages, temperature gradient and IgA.

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

The different site of viral replication in the respiratory tract will cause different syndromes. What are they? And the likely viruses?

A

Upper respiratory tract infection - Rhinovirus (better in cooler temperatures), coronavirus and adenovirus.

Pharyngitis - Adenovirus

Influenza-like illness - influenza virus, RSV (respiratory synctial virus).

Croup (larynx and trachae) - Parainfluenza

Brochiolitis - RSV and parainfluenza 3

Pneomonia - RSV, parainfluenza 3, influenza virus and adenovirus.

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

What is the histological morphology of RSV?

A

Respiratory synctial viruses in the lungs are that the infect virus cells will fuse together to form a synctial (multinucleated).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
180
Q

How do measles virus infect children, what do they infect, what happens after infection and why is it so contagious?

A

Primary replication in URT epithelial cells. Infects local macrophages, lymphocytes and DCs through receptors. They then drain to the lymph nodes and enter the circulation. Eventually returns to the epithelial cells on the lungs and mouth.

Rash is due to the immuno response not the viral infection.

Transient immunosuppression since it affects immune cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
181
Q

What are the barriers to infection found in the alimentary tract?

A

Constant movement of contents, mucus, stomach acidity, intestinal alkalinity, proteolytic enzymes from pancreas, lipolysis of bile acid, IgA and scavenging macrophages.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
182
Q

What are the properties of viruses that infect the intestinal tract?

A

They must be acid and bile resistant (therefore have no envelope).

If viruses do not have receptors for the epithelial cells it must enter via a breach of the epithelial surface (HIV - through abrasions of rectal route).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
183
Q

What is an example of virus that causes diarrhoea and how?

A

Rotavirus - destroys walls of the stomach or other mechanisms.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
184
Q

What are some viruses that enter through the intestinal tract but cause systemic infections?

A

Poliovirus and Hepatitis A

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
185
Q

How does the virus infect the alimentary tract (entry)?

A

M cells ingest the virus and delivers the antigens to underlying lymphoid tissue by transcytosis. M cells are found between enterocytes with tight junctions.

Other viruses can infect and destroy M cells. This creates a gap for viruses to enter the intestinal tract.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
186
Q

How do rotaviruses cause intestinal tract infections?

A

They have triple shelled capsid and infect and destroy epithelial cells of the intestinal vili and the M cells then cause inflammion and diarrhoea.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
187
Q

Why does diarrhoea occur (mechanism)?

A

Destruction of enterocytes decreases absorption. Also secretion from the virus NSP4 proteins from infected cells make the diarrhoea even worse. NSP secretes fluids from the remaining cells.

Usually death through dehydration.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
188
Q

How do intestinal tract infections lead to systemic diseases?

A

Enters the body then replicates at the primary site. From here it can move into the blood stream (viraemia) and replicate in lymph nodes such as Peyer’s Patch. It will then reach the secondary viremia target tissue causing disease.

Moves to meninges, CNS, skin and muscles.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
189
Q

Which of the two viruses are more likely to cause systemic disease through the intestinal tract, picornavirus and enterovirus?

A

Picornavirus

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
190
Q

What are the other routes of human infections?

A

Transcutaneous route (bypassing skin) - minor trauma, injection via needles or blood products or insect/animal bites.

Genital tract.

Conjuctiva (very rare route of entry).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
191
Q

How can the virus spread within the body itself?

A

Local spread around epithelial surfaces. It can invade the subepithelial layer then invade the lymphatic system. Spread through the blood - also known as viremia. It is also able to spread neurally.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
192
Q

What is the pathway for the viruses to move from the epithelium to the blood?

A

Epithelium to lymph nodes to epithelium. This is also so that the viruses will face the lymphatic/immune system first. To trigger the immune system earlier.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
193
Q

What is meant by viremia?

A

This is when the viruses are free in the plasma.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
194
Q

What is meant by primary and secondary phase of viremia?

A

These phases refer to the growth phases. Secondary phase is when it infects other organs that can replicate enormous amounts before re-entering the blood stream again. The goal is to make enough to escape the immune system.

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

How are viruses neutralised typically?

A

Neutralised by developing antibody response that is then removed by the macrophages (1-2 weeks).

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

Describe the typical course of viremia and its timeline in relation to the immune system response? Also note where the primary and secondary viremia is.

A

Invades epithelial cells and multiply. Moves to lymph nodes then multiples. It becomes primary viremia when it moves into the blood initially. Moves to other organs and replicates there. Returns to blood (secondary viremia) and moves to the skin and multiplies.

Typically takes about 6 days for it to leave the skin so it would have avoided the immune system altogether.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
197
Q

Viruses are able to infect the fetus, what are the characteristics needed for this to occur?

A

The virus must be able to cross the placenta. The fetus may die from cytocidal viruses such as small pox.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
198
Q

What are the outcomes of viruses infecting the fetus?

A

It can cause death or abortion if it is cytocidal. It may not kill the fetus but will cause abnormal development - especially rubella and CMV (cytomegalovirus).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
199
Q

Can babies be infected during birth?

A

Yes! There can be many other infected sites (even faeces) that the baby may come into contact during birth.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
200
Q

What is the importance of viremia and its impact on a fetus?

A

The viremia may cause the fetus to develop an immunological tolerance to the virus (will no longer recognise it to be foreign in later life).

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

What is the result of congenital rubella syndrome?

A

Rubella slows down the rate of cell division leading to impaired growth and organ development in the first trimester.

Microcephaly, heart defects, cataracts and deafness.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
202
Q

What determines where the virus will infect?

A

The right receptor for the virus, temperature, pH, ability to replicate in macrophages and lymphocytes (to other tissues), polarised release (depends if viruses leave apically or basolateral - more systemic) and the presence of activating enzymes.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
203
Q
A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
204
Q

How does the virus cause viral-induced damage to the tissues and organs?

A

Death of cells directly from viral replication (cytocidal virus), death due to the toxins released by the viruses, initiation of apoptosis and loss of function of the cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
205
Q

What are the consequences of the immune response against viruses?

A

Immunopathology - virus elicits immune response, symptoms may be fever (IL-1 and TNF) and enlarged lymph nodes.

Antibody-mediated pathology - antibody dependent enhancement of infection as well as antigen-antibody complexes (cannot clear the viruses).

T cell mediated pathology - CD4 T cell mediated responses causes some viral rashes (measles) because it induces cytokines to recruit eosinophils. Kidney damage due to deposition of Ag-Ab complex.

CD8 T cell mediated responses contributes to liver damage.

Autoimmunity - molecular mimicry can affect myelin basic protein or heart muscles. Polyclonal B cell activation.

Immunosuppression - HIV replicates in CD4 T cells amd kills them. Measles temporarily immunosuppresses - stops T cell proliferation by infected DC - suppressing IL-12. This will lead to susceptibility to secondary infections.

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

What is the interplay between cells of innate immune system to viruses?

A

Virus infects cells and are taken up by macrophages/DCs. They will both release IFN alpha/B (type I IFN), macrophages will produce pro-inflammatory cytokines and IL-12.

Combination of both IL-12 and IFN alpha/B to stimulate NK.

NK cells leads to lysis of virus-infected cells and are the dominant sourceo f IFN-gamma (T cells as well).

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

What are the outcomes of type I IFN?

A

Inhibit viral replication, activate NK cells, enhance MHC I expression and are produced by virus-infected macrophages, DC and tissue cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
208
Q

What are the outcomes of Type 2 IFN?

A

Inhibits viral replication, activates macrophages, enhances MHC Class I and class II expression and produced by NK cells (and T cells).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
209
Q

How have viruses evolved to evade immune attacks?

A
  1. Not being recognised by the immune system
  2. Interfering with functioning of particular immune mechanisms.

Large complex DNA viruses have acquired many other strategies to evade the immune system (Herpesviruses and poxviruses).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
210
Q

What are the strategies viruses use to evade the immune system?

A

Antibody and T cell latency, antibody - antigenic variation, T cell priming by DC, Tc cell recognition, NK cell recognition, IFN, cytokines (blocking of cytokine production, apoptosis inhibition and complement.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
211
Q

How do viruses achieve antigenic variation?

A

Change in antigenic structure of viruses due to spontaneous mutation (errors in RNA replication - no proof reading) that are then selected by escape neutralisation.

Antigenic drift can occur within a single patient for long term infections such as HIV.

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

How can viruses prevent DCs from priming T cells?

A

Immature DC process the pathogen and becomes matured. It will then be able to interact with naive T cells to activate them.

  • Block cytokine induced maturation of DCs
  • Blocks signal transduction within immature DC
  • Blocks TLR from initiating signal transduction to maturation (the signal pathway binds to a homologue instead)

Block T cell stimulation by DCs.

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

How are virus antigen displayed on infected cells?

A

Usually virus antigens are expressed on MHC I. Achieved by viral proteins made in cytosol that enter a proteosome complex to be cleaved. It is taken up to the ER via transporter of antigenic peptides (TAP). It is processed with MHC to be expressed on the cell surface for recognition.

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

What are the mechanisms viruses can use to evade CD8 T cell recognition?

A

Viruses can cause the MHC I to be endocytosed back into the cytoplasm. HIV.

Antigenic variation in the virus displayed can allow viruses to escape the T cells. The T cells will raise its number for the wrong antigen. HIV.

Viruses can bind to cytosolic side of TAP and prevent peptides moving into ER - HSV. No receptors will be no the surface.

Viruses can bind to TAP on the luminal side to prevent peptide entering ER - CMV.

The peptide complex is retained in ER - adenovirus. (Anchors to ER so it cannot reach surface).

Infected cells will have virus disrupt the proteosome complex so fragmentation cannot occur. EBV.

Decrease in class I gene transcription - HIV, RSV and adenovirus.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
215
Q

What are the consequences of activating NK cells?

A

Kill virus-infected cells non-selectively. Recognises cell surface receptors that result from virus infections to send a killing signal.

The MHC I molecules send inhibitory signals to the NK cell. If the MHC I cell is absent or reduced the NK cell will not have inhibitory signals engaged and end up killing the target cell.

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

How is NK cell activation controlled?

A

Balance between activation and inhibitory stimulation.

MHC Class I molecule activate inhibitory receptor. Other ligands on cell activate activation receptor. Viruses that reduce MHC I expression can except CD8 T cell recognition but become more susceptible to NK killing.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
217
Q

What are the ligands that are found to stimulate activation receptors?

A

NKp44, NKp46, NKG2D receptors will bind to its ligand.

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

What are some ways viruses can affect NK activation?

A

Present the presentation of ligands to activate NKG2D by retaining it in the ER - Murine CMV. (Decrease activation ligand on surface).

Present MHC Class I-like molecule on the surface of infected cells so it delivers negative signal to NK BUT does not present peptides to CD8 T cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
219
Q

What are the roles of IFN in the anti-viral mechanism of the body?

A

Soluble factor released from virus-infected cells that inhibit viral replication in neighbouring cells.

Type I and II IFN stimulate signalling pathway to upregulate transcription/translation of certain cellular proteins. Especially PKR (protein kinase R) that has antiviral activity.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
220
Q

How does the upregulation of PKR provide anti-viral activity?

A

IFN will aid in the activation of PKR inactive state. But it requires autophosphorylation to become active where dsRNA is a cofactor. dsRNA is only found in the presence of RNA viruses during replication.

The PKR active state will cause elF2alpha to become inactivated. This will stop the initiation of translation to occur (both viral and cellular).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
221
Q

Why do we need dsRNA to activate PKR?

A

Simply because RNA needs to be present to allow the PKR to hook around and autophoshorylate.

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

How can viruses overcome the PKR that is posing a threat to viral replication?

A

Viruses produces small RNA that bind to PKR so it prevents dsRNA from binding - EBV, adenovirus.

Virus can encode proteins that bind to dsRNA preventing it from activating PKR - reovirus, vaccinia.

Virus encoded homologue of elF2 so it competes for PKR inhibition of the true elF2alpha (by phosphorylation) - vaccinia.

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

What are some genetic factors that can influence the susceptibility to viral infections?

A

Inherited defects such as Ig class, polymorphism in the genes controlling immune responses, IFN inducible genes and receptor genes (such as CCR5 for HIV as secondary receptors).

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

What are some non-genetic factors that influence susceptibility to viral infections?

A

Age - newborns and the elder are more susceptible, malnutrition - decrease resistance, hormones, dual infections - more severe disease.

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

What are the overall outcomes of viral infections?

A

Fatal, full recovery, recovery buut permanent damage (such as cancer), persistent infections (not completely cleared and can resurface to cause disease).

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

Where is the anatomical location that distinguishes between upper respiratory tract from the lower respiratory tract?

A

The epiglottis.

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

What part of the respiratory tract are the normal biota located?

A

URT - Has own normal biota

LRT - Does not have a normal biota

Also paranasal air sinuses and middle ear will be free of microbiota in the URT.

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

What is the prevalence of pathogens in the respiratory tract, those that are found in >50%?

A

Viridan streptococci (not a pathogen in respiratory tract but can cause endocarditis), Neisseria species, Corynebacterium species, Gram Negative anaerobes (majority), H. Influenza, C. albicans and Strept. pneumoniae (15-85% - large range of population it affects).

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

What is the prevalence of the pathogens found in the respiratory tract, that are uncommon (1-10%)?

A

Strept. pyogenes (Group A) and meningococci

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

What is the prevalence of pathogens found in the respiratory tract, uncommon ones (<1%)?

A

Enterobacteria, Pseudomonas, C. Diphtheriae (Unique to humans - humans are the only carriers)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
231
Q

What are some pathogens that are found in the latent state, in the lungs and lymph nodes?

A

Lungs - P. jirovecii (carinii - causes PCP (pneumocystis pneumonia) this is an AIDs dependent infection) and M. Tuberculosis.

In lymph nodes and sensory nerves - CMV, HSV and EBV. After the primary infection with these viruses they become resident and when you become immunosuppressed it will resurface as blisters.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
232
Q

What is the relationship between the strict aerobe and strict anaerobe - in the URT?

A

The anaerobe outnumbers the aerobe by 10:1.

There are microenvironments in the URT that allow microbiota to grow.

233
Q

What are the typical URT syndrome?

A

Common cold, Pharyngitis/tonsilitis (sore throat syndrome), sinusitis, otitis media, epiglottis and Croup.

234
Q

What is the relationship between the syndrome we see and the causes (pathogens)?

A

The same syndrome can have different causes - each of these pathogens can cause many other syndromes.

235
Q

Are viruses are able to respond to antibiotics?

A

No they do not.

236
Q

Do you get the common cold from being exposed to cold weather?

A

FALSE

237
Q

What is the significance of having the common cold then being exposed to cold environments?

A

Secondary bacteria infection risk is increased when exposed to cold again.

238
Q

What are the frequent aetiological agents that cause of common cold?

A

Rhinovirus, parainfluenza, RSV, enterovirus (summer enteric virus more prevalent), coronavirus, human metapneumovirus (HMPV).

239
Q

If you have pharyngitis/tonsilitis with nasal involvement are you more likely to have bacterial or viral infection?

A

It is less likely going to be bacteria.

240
Q

What are the frequent aetiological agents of pharyngitis/tonsilitis with nasal involvement?

A

Adenovirus, enterovirus, parainfluenza and influenza - mostly viruses.

241
Q

What are the general causes (proportions too) that cause pharyngitis/tonsilitis without nasal involvement?

A

It could be either virus or bacteria (10-20%). With bacteria it is important to treat with antibiotics.

242
Q

What are the frequent aetiological agents that cause pharyngitis/tonsilitis without nasal involvement?

A

Adenovirus, enterovirus, reovirus, influenza, Strept. pyogenes (Rash is more likely to be bacteria), Strept groups C and G.

243
Q

If you treat pharyngitis/tonsilitis without nasal involvement with amoxicillin against bacteria and a rash forms, what is it indicative of?

A

The rash is a result of toxic amoxicillin due to active EBV (Epstein Barr Virus).

244
Q

What are the implications of getting sinusitis?

A

The infection can spread along the respiratory epithelial cells to the sterile sinus. Once the mucus of epithelial cells are damaged it becomes susceptible to secondary infection (from the normal biota present).

245
Q

What are the primary and secondary frequent aetiological agents that may cause sinusitis?

A

Primary - Viral (usually part of the common cold syndrome)

Secondary - H. Influenzae, Strept. pneumoniae

246
Q

What is otitis media and who is it common in and why?

A

Otitis media is the inflammation of the middle ear most commonly found in children. The reason why is because children’s Eustachian tube is much wider, shorter and straighter than adults. This means that it can be easily infected because it is not sterile.

Typically these begin with a virus infection that are not symptomatic - will eventually display symptoms from the secondary bacterial infection.

247
Q

What is the frequent aetiological agent causing epiglottitis?

A

It used to be H. Influenza B but there are many other pathogens that can cause it now.

248
Q

Why is it important to distinguish between Croup and Epiglottis?

A

The treatments are very different.

249
Q

What are the frequent aetiological pathogens that causes Croup?

A

Parainfluenza, influenza A and RSV.

250
Q

What is the pathogenesis of the common cold syndrome? In terms of viruses.

A

The virus infects and is absorbed by the cells in the nasal epithelium. Virus will replicate within that cell until it bursts. At this stage there will be a clear discharge of fluid from the lamina propria. The cell is damaged and the infection will spread.

At this stage the host immune system will kick in and begin to phagocytose the cell debris. Virus infection is about over and there will be overgrowth of the commensals - changing the fluid from clear to yellow (purulent). After a while the body will finally recover.

251
Q

What does the Eustachian tube connect?

A

The middle ear to the nasal pharynx.

252
Q

How do we diagnose URT infections? Especially whether or not the specific syndrom will need diagonostic testing?

A

This is mostly done clinically. However some laboratory diagnosis can be carried out, it varies which syndrome truly needs a diagnosis though.

Common cold syndrome - Not needed

Pharyngitis/Tonsilitis - If possible to test for Strept Group A found in URT (usually low in carrier numbers)

Sinusitis - Seldom necessary

Otitis media - Seldom necessary

Epiglotiss - Whenever possible

Croup - Seldom necessary

253
Q

Why do we not touch the epiglottitis?

A

Avoid touching an inflamed epiglottis as it could kill a person by suffocating them as you push on the epiglottis. Instead clinicians should take X-rays to check if it is swollen then take a blood culture to confirm the pathogens.

254
Q

Do enteroviruses cause rash and blisters?

A

Yes. Blisters (Herpangina) is caused by enteroviruses and rash can be caused by coxsackie virus.

255
Q

What are the treatment of URT infections?

A

It is mostly supportive treatment - to relieve symptoms such as pain. Typically use aspirin in adults and not children due to liver damage - but antiinflammatory drugs are immunosuppressive so it makes the patient more infectious. So it may be beneficial to use panadol instead.

Common cold - nothing available unless for specific patients

Pharyngitis/Tonsilitis - Only treat if it is bacterial to prevent downstream complications (especially delayed complications like autoimmunity).

Sinusitis - Only treat bacterial infections and if they are severe. (Use antibiotic against the specific agent).

Otitis media - Only treat if they are <2 y.o or prolonged and severe.

Epiglottitis - Essential to treat properly

Croup - Usually no treatment, can use inhaled steroids if severe.

256
Q

Why is it essential not to treat viral infections with antibiotics?

A

If you treat with antibiotics the secondary infection is generally very resistant and make it hard to treat.

257
Q

What are the LRT infections syndromes?

A

Acute bronchitis, Acute exacerbation of chronic bronchitis, bronchiolitis, pneumonia, lung abscess and empyema (pus in the cavity of the body).

258
Q

What are the frequent aetiological agents that causes acute bronchitis?

A

Usually it is the viral part of the URT infections.

259
Q

What are the frequent aetiological agents that cause acute exacerbation of bronchitis?

A

Usually pneumococcia and/or H. Influenzae

260
Q

What is the clinical characteristic of acute exacerbation of bronchitis?

A

The production of sputum.

261
Q

Who is bronchiolitis commonly found in?

A

Children.

262
Q

What is thought to be the mechanism of developing bronchiolitis?

A

It is thought to be an immune reaction between the antigen and pre-formed antibodies inherited from the mother. The complex will deposit on tissues and attract complements and start inflammation.

263
Q

Why does bronchiolitis cause asthma-like wheezing, is it on inspiration or expiration?

A

It is during expiration where the airways collapse. This is because of the already inflamed bronchioles.

264
Q

What is the frequent aetiological agent of bronchiolitis?

A

RSV (respiratory syncytial virus).

265
Q

What are the three different types of pneumonia?

A

Typical (acute bacterial), atypical and others (caused by fungi)

266
Q

What are the different characteristics and causes of typical and atypical pneumonia?

A

Typical pneumonia is generally localised to lobar infections. Whereas atypical pneumonia is patchy and have be diffuse. Typical pneumonia will have acute onset of the infection symptoms whereas atypical will be very gradual on and off.

Typical - inflammation of alveoli, caused by bacteria.

Atypical - inflammation of interstitial fluid, may be caused by viruses or bacteria.

267
Q

What are the frequent aetiological agents that cause typical (acute bacterial) pneumonia?

A

Pneumococci (mostly -from normal respiratory biota).

The other pathogens could be H. Influenzae, Staphylococcus, Klebs (acquired from hospital, Staph and Klebs), Legionella (immunocompromoised - older?), TB and Chlamydophila (intracellular bacteria that has a life cycle of replicating and infecting).

268
Q

What are the frequent aetiological agents that can cause atypical pneumonia?

A

Mycoplasma (no cell wall so has no fixed shape), Chlaymydia (usually in babies 1st month of life - acquired from the mother), M. Catarrhalis, influenza, RSV, adenovirus, etc

269
Q

What can cause ‘other’ pneumonias?

A

Histoplasma, Aspergillus and pneumocystis (Fungis)

270
Q

What are the frequent aetiological agents that can cause lung abscess?

A

Mixed anaerobes. Staph and Klebsiella.

271
Q

What are the frequent aetiological agents that causes empyema?

A

This is pus found in the pleural space.

Primary infection is Staph. Aureus and secondary to pneumonia.

272
Q

Why do we need to make specific diagnosis of Pneumonia?

A

To prescribe appropriate antibiotics.

There are a few must know pathogens: SARS, MERS, Influenza (some types such as swine flu is important), Legionella species (public health), Bioterrorism agents (anthrax and plague), community acquired MRSA (prognosis and treatment of methacillin resistant Staph Aureus).

There are also some ‘should know’ pathogens: Penicillin G resistant Staph. Pneumonia and Gram Negative Rods such as P. Aeruginosa.

273
Q

How can we make specific diagnosis of Pneumonia?

A

Clinical, radiological and laboratory tests.

The laboratory tests include types of specimens, microscopy, detectiong of antigens, nucleic acids, culture and serology.

274
Q

What are some clinical considerations to make while diagnosing Pneumonia?

A

Whether it is community or hospital acquired. How severe the disease is. Are there any underlying diseases such as COPD, AIDs, cystic fibrosis?

Any other risk factors that could contribute such as occupation (animals, hides, air conditioning, soil), travel or even homelessness.

275
Q

What are the type of specimens that you can attain for diagnostic testing for pneumonia?

A

Properly collected sputum (not spit - since the saliva organisms can cause pneumonia too).

The sputum sample will be checked for the presence of inflammatory cells (important to know it was collected properly), then gram stained to differentiate pneumonia causing agents.

Gram positive diplococci - Pneumococci (then do further testing)

Gram negative - Klebsiella

Other specimen types: Transtrachael aspirate, aspiration via tracheostomy (useless because it is easily contaminated), aspiration via bronchoscope, pleural tap (if effusion), lung biopsy (needle and open) or blood for culture and serology.

276
Q

What is serological diagnosis for pneumonia useful for?

A

Mycoplasma pneumonia, Legionella pneumophila, Chlamydophila and Chlamydia species and Coxiella burnetti (intracellular bacteriam different from Chlamydia because it does not have a life cycle - Typhus fever)

Used to look for specific IgM and rising titre.

Can be used to detect common antigen for common viruses, Bordetella, Legionella and pneumophila type 1 (from urine for legionella).

277
Q

Why are throat swabs generally useless and when is it useful?

A

It is generally useless because it contains the microbiota of the URT that can cause LRT infections. But the swabs are useful when looking for virus diagnosis, because the presence of virus higher up in the URT could lead to LRT infections.

278
Q

How can we treat the different pneumonias?

A

For community acquired pathogens we can use the best guess approach. We can assume it will be pneumococcus.

  • Administer Penicllin G/Amoxicillin with doxycycline/Macrolide (usually antagonistic but we get plenty in the lungs). Once you confirm pathogen you can remove one of the drugs.
  • Modify treatment if it is severe, know the cause of specific risk factors.

For hospital acquired pathogens: It will depend on the severity and risk group of the patients.

279
Q

What are the vaccines available to prevent pneumonia?

A

Influenza vaccine (to specific strains and has no effect on developing pneumonia). Pneumococcal vaccines are available as well as specialised vaccines.

280
Q

What is meant by systematic review?

A

This is literature review focused on a single question. It involves identification, appraisal, selection and synthesis of high quality evidence to answer the question.

Can reviewl both clinical trials and observational data.

This is the highest level of evidence (generally - sometimes randomised controlled trials can be better)

281
Q

What is meant by meta-analysis and its use?

A

This is a statistical analysis of all the combined data from multiple studies. The purpose of it is to increase power, resolve uncertainty, improve estimates of effect size (precision)

282
Q

What is generally the guideline to our question?

A

PICOT or a clear clinical/research question.

283
Q

Where can we source our evidence from?

A

MEDLINE, EMBASE, CENTRAL, CINAHL, DARE are the general big journal publications. Important to use standard terms when searching and include all relevant spellings (US vs UK). Once a search has been done remember to limit the papers acceptable (such as random controlled trials only).

Check the reference list sourced in the papers to find any other relevant papers.

Grey literature may be used as well for additional information.

284
Q

What are the inclusion and exclusion criteria to be used for paper selection?

A

Make sure the papers are relevant to the question we are answering (PICOT). But be careful not to let this introduce bias (selection).

285
Q

What is the selection process for studies?

A

Done by at least 2 people who read all the abstracts (but sometimes may not be representative of the full paper), apply inclusion/exclusion criteria, obtain full papers then assess for quality.

286
Q

What is the CONSORT checklist, flowchart and assessing risk of bias used for?

A

This is to outline the process needed to be done as well a checklist to improve the quality of report. The risk of bias table is used to assess the protocol used.

287
Q

How we do undertake meta-analysis to synthesise the studies?

A

This is generally done by statistical software such as STATA.

288
Q

What are the outcomes of meta-analysis and the different weighting of individual studies and heterogeneity of studies?

A

The outcomes give the weighted average effect size (which can be a relative measure - RR or OR, or absolute measure - mean difference).

Individual studies are weighted differently because of the different sample size and inverse variance (the more variant it is from the mean the less weighting it will have).

The impact of heterogeneity will impact on the effect sizes. This will require chi-square test to see how similar the studies are.

289
Q

What are the different components of the forest plot?

Test for heterogeneity? Line of Null effect? How to interpret the squares for effect size of study? Pooled Effect?

A

For heterogeneity tests we want P>0.05 because that would indicate low heterogeneity.

Line of null effect on the right uses ‘1’ value as ratio. Whereas the line on the left will indicate ‘0’ value as absolute changes.

Interpreting effect size of study - larger area of square gives relative weighting of contribution. The width indicates the confidence interval (line is used if sample size is small).

290
Q

Why is heterogeneity so important to our meta-analysis?

A

The validity of meta-analysis relies on whether or not the studies are similar enough to be pooled together.

This can be done through non-statistical methods as well such as comparing PICOT. This is not an objective method to assess it.

291
Q

How we report the systematic reviews? What guidelines do we use?

A

Generally use PRISMA checklist and flow chart to aid in our presentation of the review.

292
Q

What is the point sensitivity and specificity in terms of diagnostic testing?

A

It is to provide validity to whether or not to use the tests.

293
Q

What is meant by True positive, False positive, True negative and False negative?

A

True positive - Positive test and has disease

False positive - Positive test but has no disease

True negative - Negative test and has no disease

False negative - Negative test but has the disease

294
Q

What is the definition of sensitivity?

A

%people with the disease that test positive

Given by: TP/(TP + FN) *100

295
Q

What is the definition of specificity?

A

%people without the disease that test negative

Given by: TN/(TN+FP)*100

296
Q

What are the two measures to assess the quality of the diagnostic tests?

A

Positive predictive value - % positive tests that are truly positive

Given by: TP/(TP+FP)*100

Negative predictive value - % negative tests that are truly negative

Given by: TN/(TN+FN)*100

297
Q

Sensitivity and specificity are constant to the test, what does positive predictive value and negative predictive value rely on?

A

PPV and NPV are dependent on sensitivity and specificity as well as the underlying prevalence of disease.

298
Q

Do you want a high PPV or low PPV test to be used for screening tests?

A

You want a high PPV. If you have a low PPV the positive tests that come back will most likely be without disease.

299
Q

Why is glucocorticoids useful in asthma?

A

Overall decrease in inflammatory cell number and activation. This will decrease the probability and severity of asthma episode.

  • Reduces activation/recruitment of eosinophils, T cells.
  • Reduces cytokine production from macrophages, mast cells, SMC and fibroblasts.
300
Q

What is the very broad glucocorticoid mechanism in terms of transactivation and transrepression?

A

GC will enter the cell and bind to its receptor causing chaperone proteins to dissociate. Depending on whether or not the GCR/GC complex is a momer or dimer it will result in different responses.

Transrepression - monomer: It inhibits the activation of pro-inflammatory proteins (COX-2, IL-8, iCAM-1 and NOS-2). This is done by protein-protein interaction of the monomer with NFkB found on the gene. The GC will attract HDAC that will tighten up the DNA.

Transactivation - dimer: These will bind to the GRE (response element) to promote transcription of anti-inflammatory proteins (GILZ, MKP-1 and IkBa).

301
Q

What is the mechanism of action of transactivated protein (anti-inflammatory effectors)?

A

Increase in IkBa that will inhibit NFkB by forming the complex. IkBa is dissociated by phosphorylation by IKK.

GILZ - directly inhibits NFkB activity before it reaches the nucleus.

MKP-1 inactivates JNK-P to form JNK. This will reduce AP-1 and therefore not promote the pro-inflammatory gene expression.

302
Q

What is GC used to treat and when do we begin to use it?

A

Used to treat asthma when the patient requires >3 times/week of B2 agonists.

Also can be used for COPD - but the results are not as impressive as asthma.

303
Q

Why are GCs more effective in asthma than COPD?

A

The disease both have different inflammatory processes. The GCs target the component that is significant in asthma - eosinophils and mast cells. Whereas for COPD it is predominantly macrophages and neutrophils.

304
Q

What are some examples of inhaled GCS? How is it administered?

A

Budesonide and fluticasone propionate (combination with LABA).

Beclomethasone diproprionate

Start at an effective dose then eventually step down.

305
Q

What are some examples of systemic GCs (orally)? When is it implicated?

A

Prenisolone (oral administration).

A. Several days - used for acute exacerbation.

B. Chronically - Severe asthma only (because the adverse effects are significant)

306
Q

What are adverse effects of inhaled GCs?

A

Inhaled is generally well tolerated.

Dysphonia (change in voice), oral candidiasis (thrush - white on tongue), decrease serum cortisol. Mouthwash decreases absorption of GCs (locally).

307
Q

What are the adverse effects of oral GCS?

A

These are the GCS that have dose and indication limitations.

Osteoporosis, diabetes, muscle wasting, hypertension, growth suppresion (caution in children), suppression of HPA axis (requires tapering off if using chronically - to avoid withdrawal).

308
Q

Why is there suppression of the HPA axis and the need for tapering off GCS?

A

Cortisol and synthetic GC inhibit CRH by direct actions on the particular sites. This causes failure of the trophic hormones that causes the adrenal glands to undergo atrophy so it reduces the endogenous cortisol production. Acute stop in the synthetic GC could lead to acute adrenal crisis due to the reduced capacity of the adrenal glands.

Negative feedback to anterior pituitary and hypothalamus leading to reduced stimulation on adrenal glands.

309
Q

What are methylxanthines and PDE inhibitors? What are some examples and their rough mechanisms?

A

These reduce the degradation of cAMP therefore increases its levels (in bronchiole SMC - refer to myosin LC and IP3 receptors). This leads to muscle relaxing effects.

Theophylline - PDE inhibition/smooth muscle relaxant, HDAC2 activation and adenosine antagonism.

Roflumilast - selective PDEI because it reduces side effects compared with theophylline (approved for COPD).

310
Q

What are the dose-limiting effects of Theophylline?

A

Nausea, vomiting diarrhea, CNS stimulation (low safety margin), cardiostimulation (dysrhythmias).

Very low therapeutic index.

311
Q

What is the progression of treatment for asthma?

A

Intermittent - SABA (< daily)

Mild persistent - SABA + inhaled GCS (daily)

Moderate persistent - SABA + inhaled GCS + inhaled LABA (LABA always with GCS)

Severe Persistent - SABA + inhaled GCS (increase dose) + inhaled LABA + if needed theophylline, anti-leukotrienes, Oral GCS

Severe deteriorating - Add oral prenisolone on top of the other treatment.

312
Q

What are the risk factors that contribute to COPD and what are the major ones?

A

Major ones: Tobacco smok, lung growth development (start at lower functional point), age.

Others: exposure to particles in bad environment, air pollution, gender, respiratory infections (acceleration of decline), socioeconomic status, asthma/bronchial hyperreactivity, chronic bronchitis.

313
Q

What is the definition of COPD?

A

It is preventable and treatable disease with significant extrapulmonary effects. It is not fully reversible. Associated with airflow limitation that is progressive and abnormal inflammation of lungs against gases or noxious particles.

Usually see loss of plung parenchyma, small airways inflammation, fibrosis and thickening and pulmonary hypertension (could lead to cor pulmonale meaning it leads to RH failure).

314
Q

What are the gold standard measurements of FEV1 for classifying COPD?

A

Mild - >80% predicted FEV1

Moderate - between 50-80% predicted

Severe - 30-50% predicted

Very severe - <30% predicted

All of these are post-bronchodilators.

315
Q

How the lung function decline different in normal people compared to smokers?

A

The lung function decline due to age is constant. The smokers generally further the decline so they have a low starting point (less redundancy). After that it will decline as usual.

316
Q

Why do we use GCS in COPD?

A

Gives initial boost in lung function then after that the lung function will decline as usual.

317
Q

What is the alveolar capillary membrane composed of?

A

Layer of surfactant, type 1 alveolar cell, basement membrane and vascular endothelial cell.

318
Q

Why is the AC membrane so good for diffusion?

A

It is very thin (0.5 micron) and has a large surface area (50-100m squared).

319
Q

What are some processes or diseases that can cause AC membrane disruptions?

A

Inflammation, infection, fibrosis, emphysema, fluid, cancer etc.

320
Q

What is pneumotitis and its relation to pneumonia?

A

Pneumonia is a subtype of pneumotitis that is generally caused by infectious agents.

321
Q

What are the likely physiological effects of disrupting the AC membrane?

A
  1. Abnormal gas exchange
  2. Abnormal lung mechanics
  3. Pulmonary vascular complications
322
Q

How does the diffusion of gases over the alveolar work?

A

The diffusion process is passive because it is driven by the different in partial pressures of gases on either side of AC membrane.

323
Q

What happens to the blood gases when you under-ventilate the alveoli?

A

The PaO2 will go down and the PaCO2 will go up.

324
Q

What determines the different rates of diffusion?

A

This is given by the Fick’s Law.

V rate is proportional to (A.D.(P1-P2))/T

A - surface area

D - solubility of gas and molecular weight

T - Thickness

P - Partial pressures of gases

325
Q

What is the time course of oxygen diffusion to be fully saturated?

A

Usually oxygen diffusion is complete with 0.25 seconds. At rest we have 0.75 seconds for oxygen diffusion to occur. This allows time for redundancy when we need to increase our cardiac output (decrease transient time).

326
Q

When does the diffusion limitation of oxygen become apparent?

A

Usually occurs during exercise with less severe disease because the exercise highlights the limitation (less transient time). It is typically still fully oxygenated at rest.

However at rest it is highly unlikely to see diffusion limitation. It only occurs in severely abnormal AC membrane.

327
Q

What is the difference between the diffusion of CO2 compared to oxygen?

A

Diffusion of CO2 is similar to oxygen but it is 20 times faster. Therefore diffusion limitations of CO2 is extremely rare (severe abnormalities).

328
Q

In general what is implied by an elevated PaCO2 level?

A

It is due to inadequate ventilation.

329
Q

What are usually the different causes of changes in PaO2 and PaCO2?

A

Low PaO2 if:

  • Low PiO2
  • Low ventilation
  • Abnormal gas exchange: Low VQ (most common), shunt (extreme VQ mismatch), diffusion impairment (usually manifests during exercise).

High PaCO2 if:

  • Low ventilation.
330
Q

What are the mechanical effects of having disrupted AC membrane?

A

Usually feels like a tight band around your chest. This increases breathlessness, increased work of breathing (WOB), reduced lung volumes, altered pattern of breathing and reduced maximum ventilation.

331
Q

Disease of the AC membrane can cause increase in work of breathing, what else does this result in?

A

This will increase WOB, because greater forces are needed to overcome the stiffness (reduced compliance) of the lungs. This is known as elastic WOB.

Consequences of increased WOB, leads to accessory muscle recruitment, increased oxygen consumption by muscles, risk of respiratory muscle fatigue and ventilatory fati (O2 < 60mmHg and CO2 > 50mmHg).

332
Q

What happens to the FEV1, FVC and FEV1/FVC ratio in restrictive and obstructive lung diseases?

A

Restrictive (no change in airflow in the airways): Reduced FVC, reduced FEV1, normal FEV1/FVC.

Obstructive: Normal FVC, Reduced FEV1 and reduced FEV1/FVC.

333
Q

What contributes to the elastic properties of the lungs?

A
  1. Tissue composition
  2. Surface tension (which is reduced by surfactants). Reduced surface tension leads to increase in compliance. Usually not a problem unless it is in hyaline membranes in premature babies.
334
Q

What is meant by emphysema?

A

This is the loss of AC membrane that will cause diffusion problems. The loss of tissue makes the lungs more compliant (also loss of elastic tissue). So we usually see the barrel chest appearance.

335
Q

What are the different compliances of lungs and typically what are they caused by?

A

Increased in compliance - emphysema

Decreased in compliance - Pulmonary fibrosis

336
Q

Why is there an alter in pattern of breathing as a mechanical effect of AC membrane disruption?

A

For a particular minute ventilation a person will adopt a pattern of breathing to mimise WOB.

Stiffer lungs - Usually take rapid shallow breaths

Obstructive lungs - Will take slower longer breaths.

But essentially the same minute ventilation is achieved.

337
Q

Why and how does reduced maximum ventilation occur?

A

The maximum ventilation is only reduced in restrictive lung disease (e.g. 35 * FEV1).

In normal people the limitation is usually cardiac related.

Exercise usually worsens both hypoxia and pulmonary hypertension.

338
Q

Why does our pulmonary circulation tend to be low pressured?

A

This is to ensure that the fluid will not be exiting out of the capillary and into the interstitial fluid (eventually alveoli).

Pulmonary artery pressure - mean 15 mmHg

Capillary pressure - 12-8 mmHg

339
Q

What are some characteristics of the pulmonary circulation?

A

Thin walled RA and RV. Recives all right heart ouputs. And at rest the capillary volume is about 60-80mL. During exercise pulmonary artery pressure DOES NOT increase because of vessel dilation.

It is a reservoir for blood because blood pooling occurs during inspiration. Decrease VR, decrease CO, decrease systolic BP on inspiration.

340
Q

What are the abnormalities that can occur of the Pulmonary circulation?

A

Increase in fluid leaking out of capillaries and increase pressure on the pulmonary arteries.

341
Q

What is the interpretation of the clinical signs of JVP, central cyanosis and bilateral crepitations?

A

JVP - reflects RA pressure

Cyanosis - More than 4g/100mL of deoxygenated Hb

Crepitation - Implies fluid or fibrosis of the terminal lung units

342
Q

Typically what is suspected if a chest X-ray (CXR) showed cardiomegaly and fluid in lungs?

A

Usually implies fluid overload by heart failure.

343
Q

How can you tell that there is metabolic acidosis?

A

pH < 7.35 - 7.4 and there is no increase in PaCO2 levels. A better indication will be to look at the bicarbonate level. When acid is added bicarbonate will act as a buffer, we will see a reduced level.

344
Q

How does diarrhoea lead to metabolic acidosis?

A

The kidneys will excrete bicarbonate that will lead to metabolic acidosis because there is less buffer.

345
Q

What is the anion gap and how does it indicate metabolic acidosis?

A

Sum of positive ions (Na+ and K+) and sum of anion ions (Cl- and HCO3-). The difference between the cation and anion. The difference is usually about 15 mmol/L.

In metabolic acidosis the H+ will be buffered by HCO3-. This will make the anion gap >15mmol/L because of the buffer and the blood test not showing bicarbonate being produced.

The second type of metabolic acidosis is when the anion gap is normal. Because a reduction in bicarbonate also increases chlorine (normal <15 mmol/L gap).

346
Q

Explain the pathophysiological process of pulmonary oedema, impaired gas exchange, metabolic acidosis (from the tissue hypoxia) and tissue hypoxia.

A

Pulmonary oedema first from the heart failure, this will impair the gas exchange and therefore oxygen delivered to the tissues. The lack of oxygen to the tissues results in lactic acid being formed. Metabolic acidosis.

347
Q

What are the three different factors of the Starling Laws that tell us how pulmonary oedema occurs?

A
  1. Hydrostatic pressure inside and outside the capillary.
  2. Oncotic pressure inside and outside the capillary (usually retains water).
  3. Permeability of the capillary.
348
Q

What are the likely factors that will cause pulmonary oedema (increase in capillary fluid loss)?

A

Increase in capillary hydrostatic pressure (increase in pulmonary venous and left heart pressures) and increase in permeability of capillary. These are the most likely causes.

Other factors that will increase capillary fluid leaking. Decrease in oncotic pressure and reduction in lymphatic drainage.

349
Q

What conclusion can we draw from an CXR that shows pulmonary oedema without cardiacmegaly?

A

This implies that the cause of pulmonary oedema is most likely due to increase in capillary permeability.

350
Q

What are the hallmarks of respiratory alkalosis?

A

pH > 7.4 and there should be reduced PaCO2 levels.

351
Q

What happens to the interstitial fluid found in the lungs (due to passive fluid leaking from the capillary)?

A

The lymphatic flow usualyl drains the fluid 20mL/hour. The fluid moves into the interstitium but if the rates are mismatched with the drainage - fluid will fill up the interstitium. Eventually the fluid will move into the alveoli.

352
Q

What are the mechanical changes that occur from pulmonary oedema?

A

There will be decrease in compliance, decrease in lung volume (lungs will collapse), increase in airway resistance (fluid can press on the airways - clinically leads to wheezing as well as creps) and increase WOB (elastic and resistive - increased elastic work because of reduced compliance and only sometimes resistive work).

Hypoxia due to shunt or low V/Q units that leads to diffusion impairment.

Decrease PaO2, decrease CO2 and increase pH.

If it is very severe it will cause:

Increase PaCO2 and decrease pH.

Typically metabolic and respiratory acidosis. Metabolic acidosis generally occurs with poor cardiac output.

Finally may lead to pulmonary vascular resistance.

353
Q

What is the main difference between a bronchus and bronchioles?

A

Bronchus still has cartilage whereas bronchioles no longer have any.

354
Q

What is included in an acinus unit?

A

This is all the sites where gas exchange occur after the respiratory bronchioles.

Includes respiratory bronchioles, alveolar ducts, alveolar sacs and alveoli.

355
Q

What is a lobule and what does it contain?

A

Made up of 3-5 acinus units and usually includes the same parts of the system as well as the terminal bronchiole.

356
Q

What are the three characteristics of disease for obstructive and restrictive lung disease?

A

Chronic, diffuse and non-infectious.

357
Q

What are the different spirometry readings for obstructive and restrictive lung disease?

A

Obstructive - decrease FEV1, almost normal FVC

Restrictive - FEV1 decreases/normal (no real obstruction) but it is mostly decreased FVC for restrictive.

358
Q

What are the main causes for COPD compared to asthma?

A

COPD is caused by smoking that cause chronic injury to the small airways. Leading to emphysema and chronic bronchitis. (Overinflation and productive cough).

Asthma is caused by hyper-responsiveness triggered by allergens and infections. It is reversible obstruction.

359
Q

What is the definition of asthma?

A

Increased responsiveness of aiways to various innoculus stimuli leading to episodic bronchoconstriction that is at least partly reversible.

360
Q

What are the different types of asthma?

A

Atopic or allergic asthma - IgE levels and specific external allergens.

Non-allergic asthma - Normal IgE with non-specific triggers.

Others.

361
Q

Asthma involves the release of mediators by the mast cells, what happens in the immediate and late phase?

A

Acute/immediate response: increase permeability, increase mucus production and bronchospasm (can be targeted by B2 agonists).

Late response: Chemotaxis of eosinophils, mast cells, lymphocytes, macrophages leading to ongoing inflammation. Epithelial tissue damage. (NOT affected by bronchodilators).

Recruitment of neutrophils will destroy the underlying cells.

362
Q

What are some complications of asthma and complicates when severe chronic asthma is present?

A

Short term could lead to death, atelectasis (partial collapse of lungs), spontaneous pneumothorax.

In severe chronic asthma: Airway remodelling by fibrosis leading to irreversible obstruction. The chronic hypoxia leads to pulmonary hypertension and eventually cor pulmonale (RHF - Right heart failure).

363
Q

What are some signs of asthma?

A

Wheezing, SOB, coughing, anxiety (salbutamol or lack of oxygen to brain), hyperinflated lungs.

364
Q

What is meant by status asthmaticus?

A

Acute severe asthma that is not responding to bronchodilators.

365
Q

What is the definition of emphysema?

A

Abnormal, permanent enlargement of air spaces distal to the terminal bronchioles due to destruction of the alveolar wall WITHOUT fibrosis.

366
Q

What is Centriacinar (centrilobular) type of emphysema caused by?

A

Smoking. This is the enlargement of the respiratory bronchioles.

367
Q

What is the mechanism of smoking to cause centriacinar emphysema?

A

Tobacco produces ROS that can trigger neutrophil infiltration (nicotine from smokes can do this too). The ROS will also inactivate antiproteases (resulting in more protease). The neutrophils will release elastases on top of the existing proteases.

Both the proteases and ROS will cause tissue damage. Alveolar macrophages will also activate to increase elastase and metallo-proteinases.

Smoking results in an imbalance between proteases and anti-proteases in the lungs.

368
Q

How does emphysema cause airway obstruction?

A

Loss of elastic recoil because of the lack of supporting elastic tissue around the small airways will cause it to collapse. During expiration the flow of air out will cause the airway to collapse.

369
Q

What are the complications of emphysema?

A

Hypoxia due to airflow obstruction (main problem) or loss of surface area of your lungs (VERY RARE).

Could lead to pulmonary hypertension then cor pulmonale.

Pneumothorax.

370
Q

What is bullous emphysema and interstitial emphysema?

A

Bullous emphysema - large discs of air that could burst and cause pneumothorax.

Interstitial emphysema - air escaping into the soft tissue

NOT your normal emphysema.

371
Q

What is the clinical definition of chronic bronchitis?

A

Persistent productive cough of sputum for at least three months in the past two years.

372
Q

Where does chronic bronchitis generally occur in the respiratory tract?

A

Affects larger and smaller airways where as emphysema affects the distal tubules.

373
Q

What is the pathogenesis of chronic bronchitis?

A

Chronic irritation by inhaled substances, increased mucus production in the large airways (protective response to flush out the chemicals) and airway inflammation that could lead to scarring and narrowing of the smaller airways.

Mucus and the inflammation cause the obstruction in the airways.

374
Q

What does is the histological morphology of chronic bronchitis?

A

Excess mucus - hypertrophy of mucus secreting glands (Reid index > 0.4) and increased goblet cells.

Mild increase in lymphocytes, macrophages and plasma cells and oedema.

Peribronchial fibrosis in small airways.

There may or may not be squamous metaplasia.

375
Q

What are the complications associated with chronic bronchitis?

A

The most important one is superimposed infective exacerbations (usually the common respiratory pathogens will cause infective bronchitis).

Also hypoxia, pulmonary hypertension, cor pulmonale.

Squamous metaplasia, squamous dysplasia (change in size which is premalignant).

376
Q

Where do small airways disease affect and what is it typically caused by?

A

Affects the terminal bronchioles (very small airways) that is caused by cigarette smokes.

377
Q

What does small airway disease consist of?

A

Chronic inflammation, fibrosis and obstruction of terminal bronchioles (<2 mm).

378
Q

What is meant by COPD?

A

Chronic obstructive pulmonary disease.

It is an umbrella term that consists of emphysema, chronic bronchitis and small airways disease (there is some reversible bronchospasm - very minor component).

It is characterised by slow progression with superimposed infective exacerbations.

379
Q

What type of organisms cause most infective COPD exacerbations?

A

Bacterial bronchitis.

Also associated with increase in bronchospasm.

380
Q

What are the main clinical presentations of COPD, difference between predominant bronchitis and predominant emphysema?

A

Predominant bronchitis: Early copius sputum, 40-45 y.o, common infections, mild dyspnea late, increased airway resistance, normal elastic recoil and blue bloater.

Predominant emphysema: Late scanty sputum, 50-75 y.o, occasional infections, severe early dyspnea, normal airway resistance, low elastic recoil and pink puffer.

381
Q

How does smoking predispose you to pulmonary infection?

A

Inhibition of the muco-ciliary escalator leading to increased mucus production. Smoking also inhibits the leukocyte function and can do direct damage to the epithelial layer.

382
Q

What is bronchiectasis?

A

Irreversible, abnormal dilatation of bronchi/bronchioles.

383
Q

What is the pathogenesis of bronchiectasis?

A

Severe destructive inflammation of airways (or recurrent infection that may or may not lead to obstruction) causing loss of surrounding elastic tissue, and muscle outweigh the contraction of fibrous tissue. This leads to impaired clearance of organism and fluid.

384
Q

What can cause bronchiectasis?

A

Necrotising infections - Staph Aureus, Influenza or Aspergillus

Obstruction (+infection) could lead to fluid stasis and make it optimal for infections.

Cystic fibrosis, cilia disorders and other non-infectious inflammatory conditions.

385
Q

What are the clinical signs and symptoms of Bronchiectasis?

A

Dilated airways full of pus.

Severe productive cough that has foul smell, episodic fever, SOB and cyanosis, cor pulmonale, metastatic infection (to the brain) and amyloidosis (rare).

386
Q

What are the characteristic features of restrictive lung diseases?

A

Chronic, diffuse, non-infectious.

Inflammation and fibrosis of the inter-alveolar septa (interstitium) - this is usually very thin.

Diffuse reticulo-nodular and/or ground-glass patterns on CXR.

A specific cause or diagnosis is often not identified.

387
Q

What is idiopathic pulmonary fibrosis?

A

Cause is unknown but histologically it will show interstitial pneumonitis (UIP). This is interstitial inflammation that can have varying fibrosis stages (throughout the lungs). Survival years are usually 3 years as it will inevitably progress to end stage lung.

388
Q

What are some symptoms of seasonal influenza?

A

Fever/chills, cough, headache, muscle aches, fatigue and loss of appetite.

CXR will come up as normal.

Acute infection that can last 7 days or longer.

389
Q

How is the influenza virus spread?

A

Droplet infection from sneezing and coughing.

390
Q

What is the incubation and infectious period like for Influenza?

A

Incubation period 1-5 days. Infectious for 5-6 days.

391
Q

What is the broad pathogenesis of the Influenza virus?

A

Droplets of virus enters the respiratory tract. Virus binds to sialic acid-containing receptors on non-ciliated respiratory epithelium. In humans it is SA alpha 2-6 chains. Infection remains localised in the respiratory tract. Occurs mostly in the large airways (can be in upper and lower tract). It will cause tissue damage and an inflammatory response leading to fever, headache and muscle aches.

Pre-existing and developing immunity is sufficient to clear the virus in immunocompetent individuals. Later in infection the viruses will affect ciliated epithelium of trachae and bronchi. This is especially important because of a possibility of secondary bacterial infection (H. Influenzae, Staph. Aureus, Strep. Pneumonia) -> bacterial pneumonia is the main cause of death in elderly (rare for viral pneumonia).

392
Q

Who are at the greatest risk of death from Influenza?

A

Very young, very old and individuals with underlying chronic conditions such as heart, lung, renal or metabolic.

393
Q

What are the Influenza viruses, family, structure, genome?

A

Members of the Orthomyxoviridae family. They are enveloped viruses with a segmented genome of ssRNA of -ve sense (need to bring its own RNA polymerase).

394
Q

What are the three different types of Influenza that have no cross-reactivity and their significance?

A

Type A,B and C where A and B are the significant pathogen to humans.

Type A influenza viruses are able to infect other species.

395
Q

What are the key structures of a Type A influenza virus?

A

Hemagglutinin (HA) and Neuraminidase (NA) on the surface of the envelop. Right under the envelop is the M1 matrix that gives the virus rigidity.

M2 ion channel is also found here.

Contains 8 different gene segments in RNPs (ribonucleoproteins).

There are also 3 polymerase subunits found in the virus.

NS1 - Non-structural protein has anti-interferon activity

396
Q

What is the interaction of hemagluttinin and neuraminidase with sialic acid containing receptor?

A

HA acts as a gripper to bind with the receptors. NA acts to cleave the sialic acid off the receptor.

397
Q

Why are there different subtypes of the Type A Influenza virus?

A

Type A viruses have similar internal antigens. However they differ in their HA and NA.

HA - 16

NA - 9

Aquatic birds have all of the different subtypes because they have been hosts for a long time.

398
Q

What is the Replication cycle of the Influenza Virus?

A

HA binds to receptor (sialic acid linked to galactose) on surface respiratory epithelial cells. Virus taken up by endocytosis. Endosome becomes acidic - but this causes conformation change in virus’ HA allowing it to fuse with the endosome membrane.

The 8 Viral RNPs escape the endosome and enter the nucleus. Viral protein will be produced as well as new viral RNPs. These will come together to form the virus. HA and NA will go through glycosylation in ER and golgi then be expressed on the cell surface.

Newly formed viruses must be cleaved by tryptase (found with Clara cells - only in respiratory tract) to become infectious. Viral NA also cuts the sialic acid receptors found on the host cell so that they will not re-bind.

399
Q

Why is Influenza localised in the respiratory tract?

A

Only Clara cells have the tryptase to active the influenza virus.

400
Q

Why is it important that the virus is cleaved for it to become infectious?

A

The cleaving will reveal a hydrophobic fusion peptide that is important for the virus to escape the endosome.

401
Q

What is the role of the adaptive immune system against Influenza Virus, in particular CD8 T cells and antibodies?

A

CD8 T cells will kill virus-infected cells. Aid in recovery from Influenza. They are abe to recognise internal antigens which gives it cross-reactivity with different types of Influenza (A).

Antibody will help speed up the clearance of virus. It can act to prevent virus from binding or cleaving it self off from host cells. Can promote phagocytosis by Complement activation. Pre-existing antibody will neutralise input viruses.

402
Q

What is antigenic drift and why does it result in us needing seasonal Influenza vaccinations?

A

Antigenic drift leads to new subtypes of Influenza Virus by mutations due to the lack of proof reading in RNA-polymerase. The mutations can be advantageous, neutral or bad for the virus. Mutations in the HA and NA binding site can allow it to evade antibodies.

403
Q

How do neutralising antibodies bind to the HA receptor?

A

It binds through 5 epitopes found on the receptor. If all five of these sites mutate the majority of the population will have no pre-existing antibodies this may lead to an epidemic.

404
Q

Do the new influenza viruses replace the old ones?

A

Yes

405
Q

What do vaccines target in the virus replication cycle for immunity?

A

Aims to create antibodies against the HA and NA to block attachment and efficient release.

406
Q

What is the Influenza vaccine made of?

A

Contains three different Influenza viruses that represent the most recent strains. H1N1 and H3N2 and Influenza B.

These are chemically inactivated so that they do not induce cytotoxic T cell responses but will still create antibodies.

407
Q

What do antiviral drugs target?

A

The antiviral drugs inhibit the M2 ion channel so that the virus cannot escape the endosome. They also act as NA inhibitors to block efficient release.

408
Q

How do M2 ion channel blockers work to prevent viral RNP from escaping the endosomes?

A

When the virus is in the endosome the virus M2 ion channel delivers the H+ from the outside of the virus to the inside. Once you acidify the inside the virus it will be able to undergo conformational change to release itself.

This is blocked by the following drugs: Amantadine and rimantadine (not H5N1) not active against type B influenza.

409
Q

How do we use M2 ion channel blockers as durgs? What are their characteristics?

A

Orally administered daily. Usually used prophylatically to prevent widespread outbreaks.

However not readily used in the wider community because drug-resistant viruses easily arise.

410
Q

What are the examples of NA inhibitors? Names? Mode of administrations? Resistance?

A

Relenza and Tamiflu (H1N1 beginning to develop resistance).

Relenza - inhalation by mouth

Tamiflu - Orally as prodrug

411
Q

What is meant by antigenic shift?

A

Sudden appearance of new Influenza A virus of a new HA within the human population (sometimes NA).

The HA usually comes from bird species. Pandemics are rare because human virus receptors see alpha 2-6 galact whereas bird viruses see SA alpha 2-3 Gal.

A single amino acid change in receptor binding pocket of HA can lead to change in receptor binding specificity.

412
Q

What is one way to create new human subtypes of Influenza?

A

Swapping of gene segments when the viruses are co-infected in a single cell. This is shown in a pig example where it has both receptors for human and bird viruses.

413
Q

What is meant by pandemic?

A

Disease that is prevalent across a county or world

414
Q

What are the three possible abnormalities that can occur to the pulmonary circulation?

A

Increased fluid leakage from the capillaries, increased pressure in pulmonary arteries and obstruction in the arteries by an emboli.

415
Q

What is asbestosis?

A

A form of diffuse interstitial lung disease from the exposure to asbestos - this leads to progressive diffuse inflammation and fibrosis of the lung parenchyma causing disruption of the AC membrane.

416
Q

What are the consequences of asbestosis?

A

Gas exchange and mechanical defects leading to reduced PaCO2, Increasing A-a gradient, also decreased in lung volume (from the scarring - restrictive ventilatory defect), decrease in compliance and therefore increase in work of breathing.

417
Q

What are the clinical signs and symptoms of asbestosis?

A

Present with progressive exertional breathlessness and cough.

Clubbing, crepitations +/- cyanosis on examination.

418
Q

What are the likely causes of pulmonary artery hypertension?

A
  1. Destruction of pulmonary capillaries (inflammation and scarring will reduce the number of pulmonary capillary beds).
  2. Spasm of pulmonary arterioles.
419
Q

What is the normal pulse of a normal person after a brisk walk?

A

Should be <100, therefore >100 is abnormal.

420
Q

Do we expect the oxygen saturation to drop from 98% to 90% from a brisk walk?

A

No. In normal individuals the oxygen saturation does not drop at all during exercise unless it is an elite athlete.

421
Q

What do we expect to find if we suspect that airway disease is causing a decrease in oxygen saturation?

A

Would hear wheezing sounds

422
Q

What do we expect to find if we suspect that severe interstitial lung disease is causing a decrease in oxygen saturation?

A

Would expecet to hear crepitation.

423
Q

How do we diagnose pulmonary vessel issues?

A

This is typically down by exclusion.

424
Q

How do we check the V/Q within an individual?

A

Get them to inhale and swallow radioactive substances so that it stays in the lungs. It will show which regions in the lungs is being ventilated and what areas are being perfused.

425
Q

What is the effect of increase pulmonary artery pressures on the atrium and systemic vessels? How do they present themselves?

A

Increased RA pressure and systemic venous pressures. This will lead to peripheral oedema, ascites (accumulation of fluid in the peritoneal cavity) and pleural effusions.

Right ventriculation dilatation and hypertrophy will also lead to reduced cardiac output. Leads to breathlessness and lassitude.

426
Q

What are the main causes of increase in pulmonary artery pressures?

A

Most important one is increase in pulmonary vascular resistance. This is done by vasoconstriction, obstruction or obliteration.

Other ways involve increased LA pressure and increased pulmonary blood flow.

427
Q

What is acidosis and how can it occur? How do we identify each one? What are the compensation of each cause?

A

Acidosis is the reduction in pH levels. Can either be respiratory (CO2 retention) or metabolic (loss of HCO3 or addition of acid).

Respiratory - Lower pH, increased PaCO2

Metabolic - Lower pH, decreased HCO3

There are two compensation mechanisms:

  1. Bicarbonate retention for respiratory acidosis (Days because renal is slow).
  2. Hyperventilation (low PaCO2 - just below normal) for metabolic acidosis (seconds)
428
Q

What is alkalosis and how can it occur? How do we identify each one? What are the compensation of each cause?

A

Alkalosis is increase in pH by respiratory (hypoventilation - low CO2 levels) or metabolic causes (increased HCO3 or loss of acid).

Respiratory - High pH, Low CO2

Metabolic - High pH, High HCO3

Compensation by two mechanisms:

  1. Bicarbonate excretion for respiratory alkalosis (days)
  2. Mild hypoventilation for metabolic alkalosis (conflict of interest with reduced oxygen circulation leading to potential hypoxia)
429
Q

How much does HCO3 increase by for every 10 above the normal PaCO2 levels?

A

3.5 HCO3 for every 10mmHg.

430
Q

What are the normal pH, PaCO2 and HCO3- levels?

A

pH=7.40

PaCO2 = 40mmHg

HCO3- = 25

431
Q

How do we see compensated acidosis or alkalosis in terms of its pH?

A

It will never fully compensate to the normal pH.

Acidosis will most likely be found in the lower normal ranges.

Alkalosis compensation will most likely be found in the upper normal ranges.

432
Q

What can inadequate lung function lead to?

A

Hypoxaemia, hypercapnoea (and respiratory acidosis).

433
Q

What are the controllers of the respiratory system?

A

Respiratory centre and peripheral chemoreceptors.

The central controller includes the brain stem (pons and medulla) and cortex.

It is important that the respiratory rate is not only driven involuntarily. The cortex allows us to voluntarily control our breathing such as hyperventilate (hypocapnoea).

Brain stem outputs via phrenic nerves.

434
Q

What are the sensors involved with the respiratory system and what do they detect?

A

Central chemoceptors - on medulla and responds to CSF H+ that indirectly measures PaCO2. It does not respond to PaO2.

Peripheral chemoceptors - found in carotid and aortic bodies that respond to low PaO2, pH and increased PaCO2 that result in increased ventilation.

Lung and other receptors - pulmonary stretch, joint and muscle receptors, painful stimuli, upper airway receptors and J receptors.

435
Q

What is the ventilatory response to increase in carbon dioxide?

A

Increased ventilation linearly. There are varying sensitivity responses from different people.

436
Q

What is the ventilatory response to hypoxia (decrease in O2 levels)?

A

As the O2 levels drop in the artery it will only trigger an increase in ventilation if it drops below 50-60mmHg.

PaO2 of this level typically correlates to 90% oxygen saturation.

437
Q

What is the ventilatory response to exercise?

A

Ventilation will increase with work to maintain PaO2 and PaCO2. Once you are beyond the anaerobic threshold you will increase ventilation more.

Ventilation is matched to oxygen consumption and CO2/H+ production.

438
Q

What is hypoventilation?

A

This is when the rate of alveolar ventilation is not meeting metabolic requirements for oxygen consumption and carbon dioxide production. This leads to reduction in PaO2 and increase in PaCO2.

If it is acute it will lead to respiratory acidosis.

If chronic, there is compensatory metabolic alkalosis.

439
Q

What are the causes of hypoventilation?

A

Reduced respiratory centre activity from brain stem (injury/stroke). This will reduce the drive to breath or it can be induced by using drugs to suppress activity.

Neuromuscular disease (nerve paralysis or muscle weakness).

Chest wall deformity (gross)

Obesity (gross)

Sleep disordered breathing

As a consequence of respiratory muscle fatigue secondary to severe lung disease.

440
Q

What are examples of sleep disordered breathing?

A
  1. Obstructive sleep apnoea
  2. Central sleep apnoea
  3. Obesity hypoventilation syndrome
441
Q

What is obstructive sleep apnoea (OSA)?

A

Transient obstruction of airways during sleep that prevents breathing and eventually disrupts sleep.

Occurs in people who snore (but not all snores have OSA).

442
Q

Why does obstruction occur during sleep (OSA)?

A

Airway muscles relax when you enter REM (deep sleep), throat is already narrowed (obesity, tonsils) and tongue falls backwards to further narrow.

443
Q

What is the cycle of event for OSA? What is the result of this?

A

Snoring in light sleep, completion obstuction (apnoea) in deep sleep, reduced blood O2 and increased CO2, brain wakes up to lighter sleep (arousal), muscles contract and aiways open so that breathing occurs again, moves back into deep sleep then the cycle repeats.

This leads to very fragmented sleep and sleep deprivation -> maybe symptom is fatigue.

444
Q

How do we use polysomnograms to monitor obstructive sleep apnoea?

A

Air flow at mouth can measure the apnoeas and arousals.

Oxygen saturation is measured as well (generally there is a lag between desaturation and arousal).

Chest and abdominal wall movement may show paradoxic movement when the upper airway is obstructed. This can be used to distinguish between obstructive and central sleep apnoea.

445
Q

How is OSA managed?

A

Usual Nasal CPAP that provide positive pressure of air to keep airways open.

Other options could be manidibular advancement splint, surgery or lie on the side (prevent tongue falling back).

The goal of the management plan is to keep the airways open during sleep.

446
Q

How do we manage central sleep apnoea?

A

Generally need to manage underlying heart failure may/may not use CPAP. There are several different forms of central sleep apnoea.

Not as common as OSA.

447
Q

What is obesity hypoventilation? How is it managed?

A

Usually presents as ventilatory failure with/without RHF. This is very sensitive to oxygen.

It is managed with BiPAP and weight reduction.

448
Q

What are the consequences of chronic severe sleep apnoea?

A

The hypoventilation during sleep may cause a reset in set point of the respiratory centre leading to day-time hypoventilation.

Patients will develop chronic hypoxia, chronic hypercapnoea and a compensated respiratory acidosis.

449
Q

Why do we not give a large amount of supplemental oxygen to people with chronic hypoxia?

A

Their respiratory drive becomes dependent on low oxygen levels. When you shock them with high levels of oxygen it could lead them to stop breathing.

450
Q

What is pneumonia?

A

Inflammation of the lungs

451
Q

What is the main difference in the distinction between typical c.f. atypical cause of acute inflammation of the lungs?

A

Atypical pneumonia are caused by uncommon bacteria that result in a different presentation of the pneumonia. Usually has no consolidation or alveolar exudate.

452
Q

What are the two main patterns of acute bacterial pneumonia?

A

Acute bronchopneumonia (most common) and acute lobar pneumonia.

453
Q

What are the five different groups of lung defence failures that can lead to infections?

A
  1. Loss of cough reflex.
  2. Impairment of the mucus-cilliary functions.
  3. Accumulation of secretions.
  4. Abnormal functions of alveolar macrophages.
  5. Pulmonary oedema.
454
Q

What is the main difference between bronchopneumonia and lobar pneumonia?

A

Bronchopneumonia shows patchy distribution of inflammation that can be found across different lobes. (Usually infection at the terminal bronchiole and bilateral - tends to be low virulence organisms).

Lobar pneumonia has its entire lobe inflamed. (When inflammatory oedema spreads across the whole lobe - organism of high virulence, 80% caused by Strep. Pneumonia and 20% caused by Haemophilus Influenza).

Lobar pneumonia is more likely to cause inflammation in the pleura.

455
Q

What can we see histologically in an acute bronchopneumonia?

A

The air space is filled with lots of neutrophils, pus, vasodilation and congestion. This will come up as consolidation in the lungs.

456
Q

What is the meant by consolidation?

A

Where normal air space becomes full of fluid or non-compressible substances.

457
Q

Based on the histology of acute bronchopneumonia what are the clinical signs and symptoms we may observe?

A

Air space filled with fluids will lead to poor gas exchange. The patient may present as being hypoxic, SOB, with pleuridic chest pains, cough and purulent sputum.

458
Q

How will consolidation show up in acute bronchopneumonia macroscopically?

A

It will show up as pale and solid areas across multiple lobes.

459
Q

How will consolidation show up in acute lobar pneumonia macroscopically?

A

There will be solid and pale consolidation of the entire lobe. Also known as grey hepatization. The step before the consolidation is known as red hepatization because of the presence of RBC (haemorrhage) before its broken down and taken over by neutrophils (pus).

460
Q

How will lobar pneumonia present itself in a CXR (bacterial)?

A

There will be consolidation areas seen as pale opaque areas in the CXR.

461
Q

What are lung abscess?

A

This is a collection of pus found in the lungs.

462
Q

What are the causes of lung abscess?

A

Aspiration (movement of foreign bodies into the airways) are usually mixed with anaerobic bacteria (bacteroides).

Obstruction of the bronchial tree.

Haematogenous seeding of the lung from an extra-pulmonary infection (infection originating from the blood).

Certain types of bacterial pneumonia - Strep. Pyogenes, Staph. Aureus and Klebsiella pneumonia.

Acute bronchopneumonia in debilitated hosts (usually post operation where pathogens are introduced into the patient).

463
Q

What are the main characteristics of viral pneumonia?

A

It does NOT produce consolidation (No pus formation or heavy exudate, but oedema can occur. Viral pneumonia causes lymphocytes to infiltrate instead.)

Usually causes bronchiolitis and alveolar septum inflammation.

Viral pneumonia is cytopathic causing epithelial cell death leading to secondary bacterial infections or severe pulmonary oedema.

464
Q

What is the histological appearance of viral pneumonia?

A

Lumen of the bronchioles show eosinophils not neutrophils. The cells surround the bronchioles are lymphocytes. Important to know that viral pneumonia does not have pus.

465
Q

What is bronchiectasis?

A

This is defined as the permanent irreversible dilatation of cartilage containing airways.

Dilatation is a consequence of scar tissue deposition around the bronchi, weakening of bronchial wall (by inflammation) and pooling of lung secretions due to the dilations leading to optimal environment for infections.

466
Q

What is the main driver/cause of bronchiectasis?

A

Chronic bronchial infection

467
Q

What are the complicaitons of bronchiectasis?

A

Copius offensive sputum production, poor drainage of secretions lead to recurrent bacterial pneumonia and abscesses, rupture of vessels in bronchial walls leads to haemoptysis (dilation), pulmonary fibrosis could lead to right ventricular failure (cor pulmonale), cerebral abscesses (associated with lung abscesses) and amyloidosis.

468
Q

What is the definition of breathlessness?

A

Breathlessness arises when there is a recognition by the subject of an inappropriate relationship between respiratory work and total body work.

Up to the individual to determine whether or not the breathlessness is occurin inappropriately.

469
Q

What contributes to the respiratory work?

A

It is the same as ventilatory work that involves stretching work (elastic) and airflow work (resistive).

Generally resistive forces contribute a small proportion of work because we usually have laminar airflow. Becomes significant when there is airway obstruction.

470
Q

What is the rough mechanism to the sensation of breathlessness?

A

Load and drive feed into the respiratory work. This will lead to appropriate or inappropriate change in work. This will then be perceived to be either normal or abnormal sense of breathing.

471
Q

What are two ways to approach diagnosis of dyspnoea?

A

Can either approach it as either increase in load or drive.

Can approach the dyspnoea in terms of systems (use associated features from patient presentation to help identify system).

472
Q

What are the systems that may cause dyspnoea?

A

Cardiac, respiratory, muscle weakness, metabolic, anaemia and psychogenic.

473
Q

What are the respiratory causes of dyspnoea?

A

1) AIRWAYS DISEASE
a) Upper airways - tumour, foreign body, angioneurotic odema, CROUP

b) Lower airways - asthma, COPD, bronchiolitis

2) ALVEOLAR DISEASE
Pneumonia, lung collapse, pulmonary odema, pulmonary fibrosis

3) PULMONARY VASCULAR DISEASE
Pulmonary embolism, vasculitis, primary pulmonary hypertension

4) PLEURAL and CHEST WALL DISEASE

Pleural effusion, pneumothorax, chest wall deformity

5) RESPIRATORY MUSCLE DISEASE

Respiratory muscle weakness, phrenic nerve palsy

474
Q

What are the investigations you may order for respiratory-related dyspnoea?

A

Lung function tests - spirometry and flow volume loop.

CXR - Alveolar and parenchymal defects.

CT scan - check for COPD.

Wells Score - Cumulative score to test likelihood of event from risk factors.

475
Q

What is MUD?

A

Medically undiagnosed dyspnoea.

476
Q

What are the different types of MUD?

A

Is worsen dyspnoea a new clinical manifestitation on top of what we already know?

Disease or deconditioned (obese patients)?

At maximum effort is the breathlessness sensation or symptom? (Elite athletes)

Psychogenic or disease?

477
Q

What may cause breathlessness in Young athletes?

A

Might be normal sensation on effort, anxiety or disease.

Atopic asthma and exercise induced bronchoconstriction can be diseases that lead to dyspnoea. There is also vocal cord dysfunction (Laryngeal Dysfunction) not to be confused with asthma.

478
Q

What is the best way to determine whether dyspnoea is a disease or de-conditioning?

A

Best test is to use incremental exercise test to check maximum.

Look for anaerobic threshold and total capacity.

Can also attempt three month aerobic training problem then re-measure exercise capacity.

479
Q

How to tell psychogenic vs disease cause of dyspnoea?

A

Take a good medical history and the absence of particular symptoms will increase suspicion of MUD.

480
Q

What can a wheeze or stridor tell us about a particular disease?

A

It is localised to the airways.

481
Q

What can crepitations tell us about the disease?

A

Suggests terminal lung units such as the alveoli being filled with fluid along with air. It may suggest late inspiratory crepitations (when lungs are stiff and scarred - interstitial lung disease).

482
Q

What bacteria causes Tuberculosis and what type of bacteria is it?

A

Myocbacterium. Tuberculosis is an acid fast bacteria (thick waxy cell wall layer). Recently multiple drug resistant strains have risen.

483
Q

What are the characteristics of TB?

A

TB is characterised by granulomatous inflammation (collection of macrophages), type IV hypersensitivity and inability to clear foreign bodies completely leading it to be walled off in a granuloma.

It resembles caseous necrosis.

484
Q

What does a tuberculosis granuloma look like histologically?

A

Macrophages surround the granuloma with a caseous necrotic core.

485
Q

What is meant by Primary TB?

A

This is the first time the patient is exposed to TB.

Self-limiting illness.

Two areas where the granulomatous inflammation can be found - peripherally (usually middle) and hilar lymph nodes.

Ghons focus = peripheral area of granuloma

Ghon complex = peripheral + involved node.

Most patient’s immune system can control the infection and healing by fibrosis, often with calcification.

486
Q

What is secondary TB and its characteristics?

A

Patient had previous exposure to TB and is re-exposed.

Affects apical areas of upper lobe, tubercles and caseations.

Caseation progressive spreads through the lungs, erodes blood vessels (haemoptysis), cavitation into bronchial tree, pleural inflammation and fibrosis and lung scarring.

Lung scarring from the host response as defence.

487
Q

What is meant by miliary TB?

A

This type of TB has numerous small granulomas in the lungs.

It is more common in secondary TB and patients who are immunocompromised.

488
Q

Do we need to improve work place conditions to help with global health?

A

Yes a bad stress job is as bad as being out of work.

489
Q

How is TB spread?

A

Through droplets via coughing, sneezing or spitting.

490
Q

What are the main drivers for the prevalence of TB?

A

Vast majority of TB is in those of the poorest segments of society. It is associated to biological, social and economical factors rather than just performance of TB programs.

491
Q

How do we approach treating the TB issue?

A

Biomedical approaches and programs (DOTS) decreases incidence of TB. But we need to tackle the underlying factors that make these groups of people more vulnerable to TB such as social status.

492
Q

What is the relationship between the social-economic gradient with the risk of contracting TB?

A

The poorer you are the more likely you wll contract TB.

493
Q

What are the typical risk factors that contribute to the causal pathway of poverty to TB risk?

A

Exposure to infectious droplets, host defences (HIV), malnutrition, indoor airpollution, alcohol abuse, depression and stress (affects cell-mediated immunity).

  • Malnutrition, smoking, HIV, diabetes, alcohol and air pollution.
  • Malnutrition and air pollution are direct markers of poverty. Smoking is consistently higher in lower social economical groups.
494
Q

What are our responses to the TB epidemics by the WHO?

A

Higher quality DOTS, address the needs of the poor and vulnerable populations, strengthen health system based on primary health care, engage all care providers, empower peopl with TB through partnership and promote research.

495
Q

What is a venous thrombi?

A

Thrombus forms in any veins (superficial or deep) but majority is formed in the deep veins of the lower limb.

496
Q

What is the venous system of the lower limb like?

A

Anterior and posterior tibial veins join to the form the popliteal veins to femoral veins and eventually into the IVC.

497
Q

How are venous thrombus formed and where?

A

They form around the venous valves (a little upstream). They are able to continue propagating against the flow of blood.

Venous thrombus are typically occlusive, red (high RBC from stasis).

498
Q

What does a venous thrombus look histologically?

A

The venous thrombus will have layers of red and pink.

Red - erythrocytes

Pink - Platelets and fibrin

499
Q

What are some factors that can predispose to venous thrombosis?

A

Virchow’s Triad

  • Change in vessel wall (from trauma such as IV catheterisation or bacterial infection of the vessel wall).
  • Changes in blood hypercoagulable states.
  • Changes in blood flow: stasis, turbulence, prolonged immobilisation, dehydration, hypotension, congestive cardiac failure and polycythaemia.
500
Q

What are the primary (genetic) hypercoagulable states?

A

COMMON

Factor V Leiden - point mutation in factor V prevents activated Protein C from binding to the cleavage site. Natural anticoagulant will not work.

Prothrombin III deficiency leading to circulating levels of prothrombin.

UNCOMMON

Antithrombin III deficiency, Protein C deficiency and Protein S deficiency.

501
Q

What are the secondary hypercoagulative states caused by?

A

Surgery, massive trauma and burns, malignancy, obesity, smoking, hyper oestrogenic states (pregnancy or OCP contraceptives), nephrotic syndrome, antiphospholipid antibody syndrome.

502
Q

What can happen to a venous thrombus?

A

Lysis and resolution, organisation of the thrombus to form scar tissue, recanalisation (new blood vessels that resupply blood to the area) and embolism.

503
Q

What is pulmonary thromboembolus and where does it come from?

A

Arises from deep vein thrombus in the lower legs (pelvic and abdominal less commonly). The thromboemboli travel to the right side of the heart and enter the pulmonary arterial circulation via the pulmonary artery.

504
Q

What happens when the pulmonary thromboembolus is lodged in the bifurcation of the pulmonary artery (before it splits to left and right)?

A

The venous thrombus will be mainly red with pale areas, coiled shape (reflects shaped of the vein of origin) and occlusive.

Can cause sudden death (there will be electrical activity but lack of cardiac output because of the occlusion).

This is known saddle thromboembolus.

505
Q

What happens when the pulmonary thromboembolus is found in the right or left pulmonary artery?

A

It could lead to sudden death, dysphnoea, chest pain and circulatory failure mimicking myocardial infarction.

For clinical signs similar to MI we need to think about pulmonary embolus and aortic dissection.

506
Q

What happens when a pulmonary thromboembolus in the smaller pulmonary arteries?

A

Could lead to pulmonary infarct that is wedge shaped, sharply demarcated, pleural base, red (reperfusion from bilateral blood supplies), occluded artery at the apex.

Clinically: dyspnoea, haemoptysis (area of haemorrhage into the infarct), pleuritic chest pain (because infarct will be based around the pleura).

507
Q

What should you suspect if you have a patient with risk of DVT and pulmonary thromboembolus and they develop tachycardia or SOB?

A

Must consider the chance of pulmonary embolus.

508
Q

What is the most common cancer found in Australia is it the leading cause of death?

A

Basal cell carcinoma and squamous cell carcinoma are the most commonly cancers found in Australia but they rarely lead to death.

509
Q

What is meant by neoplasia and how does it arise? What is a neoplastic tissue compromised of?

A

Neoplasia is excessive unregulated cell division.

Begins with multistep mutations in a single cell. Affecting the cell cycle, cell death and DNA repair of the cell. As you mutate you also acquire different features.

Neoplasia is made up of the neoplastic cells and a reactive stroma. It can be either benign or malignant.

510
Q

What is meant by the vague term ‘tumour’?

A

This is a lesion of mass of any kind but typically these days we refer it as neoplastic lesion.

511
Q

What are the hallmark features of cancer tissue?

A

Resisting cell death, inducing angiogenesis, sustaining proliferating signaling, enabling replicative immortality, evading growth suppressors and activating invasion and metastasis.

An emerging hallmark is also the ability to evade the immune system.

512
Q

What is the leading cancer in Australia c.f. to what is the leading cause of death of cancer?

A

Leading cancer is prostate and breast for males/females. Lung is third for common.

But leading death is by lung cancer then the prostate/breast cancer.

513
Q

What is the difference between adult and paediatric cancers?

A

Most of paediatric cancers are referred to as blastoma because it resembles cells from the embryo.

E.g. Certain leukaemia, neuroblastoma, Wilm’s tumour (kidney), certain lymphomas, retinoblastoma and certain bone cancers (osteosarcoma).

514
Q

What is the difference between benign and malignant cancers?

A

Benign: Locally expansive, slow growth, well circumscribed (most of the times), well differentiated cells and cannot metastasise.

Malignant: Locally invasive, locally destructive, often poorly circumscribed, sometimes necrosis because of outgrowing the blood supply, variable differentiation, potential to metastasise (through lymphatics, blood vessels and transcoelomic - body cavities). Also induces desmoplasia in stroma as they grow.

515
Q

What are uncertain malignant potential/borderline?

A

This is another group of malignant cancer that histologically look like benign and malignant tumours.

516
Q

What vessels can the cancer travel and spread using?

A

Cancers tend to grow in lymph nodes and they like to grow in the first lymph node they are drained to. The tumour can enter the draining veins and enter the systemic circulation. Or it can translocate by transcoelomic spread (migrating along the pleural space - body cavity).

517
Q

What are the common sites for cancer metastasis?

A

Hilum lymph nodes, brain, liver, bone and lungs.

518
Q

What are the histopathological features of neoplasia?

A

Cytological atypia: Larger nuclei, pleomorphic nuclei (vary in size and shape), coarser nuclear chromatin, hyperchromatic nuclei (takes up more H&E), larger more prominent nucleoli and may see more mitotic activity.

Architectural disorganisation.

Benign neoplastic cells show less atypia than malignant cells.

519
Q

What is a desmoplastic stroma?

A

This is a stroma that has more fibroblasts therefore making it stiffer to touch.

520
Q

Where do necrotic tumours occur in?

A

They only occur in malignant tumours.

521
Q

What are the three different ways we classify the cell lineage of the tumours we find?

A

Epithelial - glandular, squamous, uroethelial and endocrine.

Mesenchymal - osteoblasts, endothelial cells, smooth muscle, skeletal and adipose tissue.

Others - melanocytes, myeloid cells, lymphoid cells and astrocytes.

522
Q

What is the histological characteristics of glandular tumour cells?

A

Formation of glandular lumina, formation of mucin (lots of mucus) and signet ring cells (mucus within the cells).

523
Q

What is the histological characteristics of squamous cell carcinoma?

A

Keratinization in the lumen, intercellular bridges (between the bridges are desmosomes) and eosinophilic cytoplasm.

It shows features of stratified sqamous epithelium - often have lots of eosinophilic cytoplasm and intercellular bridges.

524
Q

What is the differential diagnosis of lesion of mass?

A

Neoplastic or Non-neoplastic? If it is neoplastic, is it benign or malignant? Also what type is the neoplastic tissue?

If the tissue is malignant - is it primary or metastatic?

525
Q

What should we do when we suspect malignant tumour?

A

Must biopsy the tissue. They look at its cytological features, architecture (may or may not have necrosis), stroma and cell lineage.

526
Q

What are the general rules to cancer terminology?

A

Prefix - line of differentiation: Adeno (gladnular), squamous cell, leimyo (smooth muscle), osteo (osteoblastic).

Suffix: Benign (-oma) and malignant (epithelial - carcinoma and mesenchymal - sarcoma)

There are exceptions such as seminoma (malignant testicular cancer) and lymphoma.

Mixed: carinosarcoma

527
Q

What are the different degrees of differentiation? Why is it important to determine the degree?

A

Well differentiated: Resemble mature cells more, less cytologic atypia and the architecturally more organised.

Poorly differentiated: Poorly resemble to mature cell, more cytologic atypia, more mitotic activity and architecturally less organised.

Degree of differentiation is referred to as its grade (of malignant tumours). Well differentiated tumours tend to be less aggressive than poorly differentiated ones.

528
Q

What cells do neoplastic cells arise from?

A

It generally arises from one cell. It could be adult stem cells or it could be mature cells that acquire stem cell like qualitites leading to tumours.

529
Q

What are pre-malignant tumours (dysplasia)?

A

Invasive carcinomas arise from the progression of non-invasive precursor epithelial lesions. They are known as dysplasia and intraepithelial neoplasia.

530
Q

What is meant by dysplasia? Why do we grade these lesions?

A

Abnormal development, alteration in size, shape and organisation of cells. It is graded as mild, moderate or severe (1,2,3). Low or high can also be used.

High grade dysplasia is more likely to progress to malignancy.

531
Q

Describe the progression from normal epithelium to invasive neoplasm.

A

Epithelial are the most common cancer and it begins with intraepithelial neoplasia - can treat it before the cancer becomes invasive. Normal epithelium as simple columnar (can be stratified squamous) in the gut and there is the basement membrane. One of the stem cell in the epithelium develops multiple mutations and starts off in one cell. Mutations can arise from carcinogenic agents, microbes, nutrients (guts) or inherited. It then becomes multiple cells that proliferate. The cells become pleimorphic because of different nuclei shape and size, mitosis (dysplasia or intra epithelium neoplasia), but it still remains in the epithelium. When it becomes very severe it becomes in situ neoplasm. If one of these cells get mutated to express MMP will allow the cells to lose its cohesiveness and escape through and become invasive neoplasm.

532
Q

What are polyps?

A

These are glandular dysplastic lesions arising from lining epithelia.

533
Q

What are some examples of premalignant lesions?

A

Cervical intraepithelial neoplasia (CIN), vulvar intraepithelial neoplasia (VIN) and prostatic intraepithelial neoplasia (PIN). These are typically ideal for early screening so that we can prevent it before it goes malignant.

534
Q

How is hyperplasia and metaplasia different from neoplasia?

A

These are adaptive changes that are regulated. The change themselves are not premalignant (not change in genes). But in certain situations it can increase the risk of mutations to develop.

535
Q

The typical hallmarks of cancer are the same but what are four new emerging hallmarks?

A

Deregulating cellular energetics, avoiding immune system, genome instability and mutation and tumour-promoting inflammation.

536
Q

Why should we not look at the different mechanisms of cancer development in isolation? What does the cancer development affect overall?

A

Cancer development needs to be looked as an entire overview and collection of the different contributing mechanisms.

But essentially what happens is that there are changes in gene expressions that lead to the development of the hallmark capabilities.

537
Q

What is the very basic understanding of mutations in developing cancer?

A

Mutations in critical cell control factors and cancer in one cell first. Then the cell will proliferate but further mutations will occur. Typically requires multiple mutations before it reaches the stage of dangerous cell proliferation.

The cells are continuously evolving.

538
Q

Why are the genetic changes done in a particular order that will dictate biological outcomes and therapeutical treatment?

A

Most cancers are somatic mutations while 10% are germ-line mutations.

Methylation abnormalities, proto-oncogenes mutations, tumour suppressor gene mutations and additional mutations of gross chromosomal alterations.

539
Q

What are the four major classes of genes that are altered in cancer cells?

A

Proto-oncogenes (growth promoting), growth inhibiting tumour suppressor genes, genes that regulate program cell death (apoptosis) and genes involved in DNA repair.

540
Q

How well do our cells repair DNA damage and how is this achieved?

A

DNA repair is very good in cells mostof the times. Insufficient DNA repair will accelerate DNA mutations and eventually lead to oncogene and tumour suppressor gene mutation.

Accumulation of DNA damage - balance between DNA damage and DNA repair.

541
Q

There are single nucleotide polmorphisms found in cancers, where are the genomic locations that are essential in the risk of cancer development?

A

Coding region can affect the amino acid sequence. Regulatory sequence can affect the changes in amount of protein produced.

There are linked SNPs that are found outside the gene that have no effect on protein function and production.

542
Q

What are the types of mutations in cancer?

A
  • Errors in DNA replication that is not repaired: BRCA1 and BRCA2 or any damage to DNA repair genes.
  • Oncogenes, tumour suppresor genes and regulatory region changes.
  • Point mutations activate oncogenes and inactive TSG.
  • Amplication of oncogenes (via multiple copies).
543
Q

What is oncogene amplications in neuroblastom of N-MYC?

A

Certain genes on a chromosome will express double minutes (fragments of chromosomes that are also able to express N-MYC).

544
Q

How can gene translocation lead to the generation of oncogenic chimaeric molecules?

A

The translocation of chromosome 9 and 22 ABL and BCR genes.

ABL-BCR hybrid gene is oncogenic tyrosine kinase that contribuets to chronic myelogenous leukemia.

545
Q

What is the rate of neoplastic growth?

A

Neoplastic growth is very rapid can take 3 months for 10^9 cells (1g) to grow to 10^12 cells (1kg).

The rate of proliferation is dependent on the blood supply of tumour cells too. Eventually it could lead to vascularised tumour with central necrosis.

546
Q

Why are there high growth fraction tumours and low growth fraction tumours?

A

These are differences in the magnitude of tumour proliferation vs tumour death.

Fast: Leukaemia, lymphoma and small cell carcinoma

Slow: Breast, adenocarcinoma and colon.

547
Q

What is the general mechanism of cell proliferation and how is it different in tumour cells?

A

Growth factor binds to tyrosine kinase receptor to initiate signalling.

Activation will lead to activation of RAS that will activate two pathways.

  • RAF to MAPK to activation of transcription of MYC proteins (cell cycle progression)
  • PI3K to Akt to mTOR that activates transcription of MYC proteins (cell cycle progression).

What happens in cancer cells is that there is a mutation of one of the signalling molecules that leads it to be constitutively activated without any ligands needed.

548
Q

What are the oncogenes and tumour suppressor gene balance in the PI3K pathway of tyrosine kinase?

A

p110 is found as part of PI3K that is commonly mutated that drives Akt phosphorylation.

The TSG in this pathway is the PTEN that inhibits the PI3K. Mutation in cancer generally makes it inactive or complete deletion.

549
Q

What the typical mutations of the two opposing oncogense and tumour suppressor genes? Why does only 1 ongene allele need to be mutated whereas TSG need two alleles lost?

A
  1. Overactivity mutation of oncogenes.
  2. Underactivation mutation of TSG.

TSG - to lose the function must lose all the alleles. Whereas with function in order to get more it simply needs change in one allele.

550
Q

What are the mechanisms of the loss or inactivation of TSG expression?

A

Mutation within the gene to make it non-functional. The other mechanism is complete deletion of the TSG. An alternative regulation of the gene expression is by DNA methylation on the promoter region.

Arising is miRNA that are non-coding ssRNA that regulates the transcription and translation of normal genes. It can be either inhibitory or stimulatory.

551
Q

What is meant by the loss of heterozygosity?

A

LOH in a cell represents the loss of normal function of one allele of a gene in which the other allele was already inactivated. This is the general genetic feature involving tumour suppressors genes.

552
Q

What is the tumour suppressor gene p53’s function?

A

p53 is a transcription factor that regulates the expression of cell cycle factors. Generally initiates apoptosis, DNA repair, cell-cycle arrest and differentiation.

Cancer cells have defective p53 or are deleted.

553
Q

Why is the cell cycle so important in cancer? Where are the checkpoints found?

A

It regulates growth and mitotic phases. There are many cell cycle controls that could lead to increased proliferation.

End of G2, middle of M, end of G1 and during S.

Oncogenes inhibit the checkpoints (RAS), whereas the tumour suppressors p53 enhance the check points.

554
Q

How do tumour cells evade apoptosis?

A

Pathway to apoptosis: extrinsic (Fas) leading to caspases activation.

Intrinsic stress/radiation leads to DNA damage, p53 response (apoptosis) to produce BAX (pro apoptotic molecule) to act on the mitchondria. There is also the BCL-2 that acts as a key anti-apoptotic molecule. Once again a balance between the two the determine apoptosis.

Reduced CD95, inactivation of death induced signalling complex by FLICE protein, up-regulation of BCL-2, reduced levels of pro-apoptotic BAX from the loss of p53, loss of AFAX-1 and up-regulation of inhibitors of apoptosis.

555
Q

Why are cancer cells essentially immortal?

A

Cancer cells have good telomerases that extends the telomeres (act as buffer as cell replication shortens the chromosomes).

556
Q

How do metastasis of tumour cells occur?

A
  1. Detachment of tumour cells from each other
  2. Degradation of ECM
  3. Attachment to novel ECM components
  4. Migration of tumour cells

There will be alterations in the ECM of both primary and secondary locations.

Mutations occur in the following four genes: integrins (cell adhesion), catenins, cadherins and connexins.

557
Q

What is a tumour microenvironment?

A

There are other cells aside from the tumours cells themselves. These are endothelial cells (angiogenesis) it also needs factors from the endothelial cells as well as fibroblasts are very important for growth factors. Macrophages are also essential immune cells to sell signals to maintain and accelerate tumour growth.

558
Q

What are the factors that control angiogenesis?

A

VEGFs and VEGFRs these are popular therapeutic agents to starve the tumour of blood.

559
Q

Within the tumour itself there are genetic heterogeneity what is the signifiance of this?

A

This means that a particular chemotherapy may not be able to ablate the entire tumour because there may be some resistance.

560
Q

There are two types of cancer cells, cancer-initiating and fast replicating cancer cells, what do conventional cancer therapies c.f. with targeted cancer therapies differ?

A

The conventional cancer therapy will destroy the fast replicating cancer cells but may not necessarily kill the cancer-initiating one so that tumour relapse may occur.

Cancer stem cell specific therapy kills off the cancer-initiating cell so that the tumour eventually regresses.

561
Q

What are the clinical features of malignancy?

A

Local effect of the tumour, effect of the metastasises (local lymphadenopathy - enlargement of lymph nodes along the the lympathic pathway, bone pair or features related to hypercalcaemia, jaundice and seizures - altered electrical activity).

There is also weightloss, anorexia (TNFalpha and IL-1 that is produced by the tumour or tumour microenvironment).

562
Q

What is meant by paraneoplastic effect?

A

This is a syndrome as a consequence of the cancer secreting hormones/cytokines by the tumours of the body itself.

Cushing syndrome - ACTH leading to excess cortisol by small cell lung cancer (produces ACTH) and pancretic cancer.

Hypercalcaemia - Excess PTH-related protein that stimulates the breakdown of bone. This can be caused by squamous cell carcinoma of lung, breast carcinoma, renal cell carcinoma and adult T cell leukaemia.

563
Q

What are the clinical features of lung cancer?

A

Local effects of the primary tumour: cough, haemoptysis, wheeze, dyspnoea, pneumonia and Pancoat’s syndrome (cancer in the apical lobe that can affect subclavian vessels).

Effect of metastases - bone pair, hyper calcaemia, jaundice and seizures.

Weight loss, anorexia and paraneoplastic effects.

564
Q

What are the investigations that we can order for cancer?

A

Clinical history, examination, blood tests (where relevant), radiology and tissue sampling.

Blood tests: Hb levels (typical of colon cancer), liver function tests (liver metastases) and tumour markers. But tumour markers are non-specific and are not elevated in every case. Carcinoembryonic antigen (CEA) that can be produced by colon, pancreatic cancer and benign tumours. Alpha fetoprotein are elevated in testicular and primary hepatic cancer.

Radiology: CXR, CT, ultrasound - Staging of the tumour (how big and where is it to metastasised?)

Endoscopy (visualisation)

565
Q

How do we obtain tissues for sampling (biopsy)? Why do we take biopsy?

A

Biopsy of the sample tissue for diagnosis for histopathological diagnosis and other features to determine prognosis and management.

  • Look at cytological features, architecture, stroma
  • After surgery removal of the specimen it is sent for testing again to see the overall picture.

Cytology by fine needle aspiration (suck cells up without architecture intact) or exfoliative cytology (scrap onto slide)

566
Q

What are the tests we run after obtaining a sample of the tumour tissue?

A

Histopathology: H&E, Special Stains and immunohistochemistry

Molecular and cytogenic techniques including: In situ hybridisation, PCR and chromosomal arrangements.

567
Q

What else should we know about confirming that it is a metastatic cancer?

A

Specific tumour type, grade, stage, presence of lymphovascular invasion.

This is important for the prognosis and management of the malignancy.

568
Q

How do we classify lung carcinoma?

A

Non-small cell carcinoma: Sqaumous cell carcinoma, large cell (undifferentiated) carcinoma and adenocarcinoma.

Neuroendocrine carcinoma: Small cell carcinoma (very aggressive)

The listed cancers are the main types.

569
Q

What are the predisposing factors to lung carcinoma?

A

Tobacco smoking, asbestos and arsenic.

If you are a non-smoker you are most likely going to contract adenocarcinoma.

570
Q

What do lung carcinoma look like histologically and macroscopically?

A

Invasive small cell carcinoma in bronchus has a necrotic core.

Lung carcinoma are sometimes necrotic that may manifest as cavities. A wider spread of wide area are desmoplastic stroma.

Know the difference between squamous cell carcinoma and adenocarcinoma (part of lung carcinoma).

571
Q

What happens if the tumour type cannot be determined by H&E stain?

A

We can use immunohistochemistry to test for the presence of specific antigens so that we can identify the type of tumour.

LCA - for lymphoma

CAM5.2 - Carcinoma for epithelium

572
Q

What is referred to as the ‘stage’ of the cancer?

A

The stage incorporates the size or depth of the invasion as well as the location and extent of the metastases.

This is done by the TNM system by pathologists.

T - extent of primary tumour (size)

N - Regional lymph node metastases

M - Absence or presence of distal metastases.

X - cannot be assessed or unknown.

These are all grouped together to form a particular stage level.

573
Q

What other factors will influence the prognosis of lung carcinoma?

A

Vascular invasion seen in primary tumour, specific genetic alterations and others depending on what tumour it is (ulceration, patterns of inflammation).

574
Q

Give an example where we can use predictive factors to predict the likely response to certain therapies?

A

Screening for HER2 in breast cancer will tell us if we can use anti-HER2 drugs. Also the oestrogen and progesterone receptors in breast cancer can tell us a lot.

575
Q

What is the management of cancer?

A

Surgery with a detailed pathology report of the specimen, radiotherapy, chemotherapy and targeted therapy.

576
Q

Why is targeted therapy better than traditional chemotherapy drugs?

A

It is not a non-selective treatment so will not affect normal cell. Targeted therapies block growth of cancer cells by interfering with function of specific molecules. This method requires genotype/phenotype testing of the different tumours.

2 Main types:

  1. Small molecule that inhibits growth factor receptosr (tyrosine kinase).
  2. Monoclonal antibodies that target specific proteins or receptors.
577
Q

Usually lung cancers have at least one mutation in a critical pathway? What is this pathway and what kind of mutation is needed?

A

The growth factor activation process is very important. Activation of tyrosine kinase will increase proliferation, protein synthesis, cell cycle progression and cell death. There are complicated signalling pathways to reach the end points.

But mutations will generally result in continuous cell division, we can use a particular drug to inhibit the activity of that abnormal protein.

578
Q

What is the importance of EGFR mutations and targeted therapy of lung carcinoma?

A

Significant rate of EGFR mutations that contribute to non-small cell carcinomas. Involves increasing its activity leading to hyperactivation of downstream signaling pathways.

New targeted therapies target this mutation by inhibiting the EGFR tyrosine kinase. But tumour needs to be tested to see if this treatment will work or not. Resistance can develop as the tumours acquire new mutations.

579
Q

How does cancer cause death?

A

Once disseminated the patient will die from cachexia, secondary infection related to poor nutrition, effects of treatment and damage to vital organ or system by the tumours.