TI Flashcards

1
Q

What is the definition of anaemia?

A

Anaemia is a condition in which the number of red blood cells (and consequently their oxygen-carrying capacity) is insufficient to meet the body’s physiologic needs

Insufficient oxygen carrying capacity is due to reduced haemoglobin concentration as seen with insufficient RBC

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

About haemoglobin…

A

Haemoglobin contains iron and transports oxygen. It is a metalloprotien within RBCs.

Reduction in haemoglobin = anaemia (reduction in oxygen carrying capacity)

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

About the production of blood cells and where they are…

A

Blood cells are made within bone marrow. As a child this is mostly within most of the long bones and it is being produced all the time. When we are older it is more predominantly in our pelvis, femur, sternum – the bones have become a bit dried out. We are continually producing RBCs and platelets etc

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

How are haemoglobin levels used to diagnose anaemia?

A

Every lab has its own normal range, and the normal range will vary throughout life and also between males and females.

We usually use g/L however some use g/dL and this is why sometimes values may seen to be out by a factor of 10 (eg 7 actually means 70).

From 6 months to 5 years there is a lower normal levels because the child is transitioning from breast milk to using other nutrients, but by adolescence it reaches around the normal range.

Women that aren’t pregnant have slightly lower levels than men, this is because they are menstruating. When women are pregnant it will drop even further – this is because their physiological volume increases so there is a decrease in concentration.

Men however, should stay at around 130 g/L.

Depending on how low you drop tells you how severe the anaemia is

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

What is normal erythropoiesis?

A

Maturation of RBCs required Vitain B12 and folic acid for DNA synthesis and iron for haemoglobin synthesis.

  • Vitamins
  • Cytokines (erythropoietin)
  • Healthy bone marrow environment
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What are the mechanisms of action of anaemia?

A
  • Failure of production: hypoproliferation, reticulocytopenic
  • Ineffective erythropoiesis
  • Decreased survival: blood loss, haemolysis, reticulocytosis

For example, when you might have been stabbed you lose a lot of blood which could make you anaemic despite having healthy bone marrow

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

What deficiencies can cause anaemia?

A
  • Iron deficiency
  • Vitamin B12 deficiency
  • Folate deficiency

Reduced concentration of Hb means that we can’t get the building blocks from food sources

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

About iron…

A
  • Essential for O2 transport
  • Most abundant trace element in body
  • Daily requirement for iron for erythropoiesis varied depending on gender and physiological needs
  • Daily requirement depends on TBC production amount, gender, age and physiological needs
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What are the daily iron requirements?

A

Recommended intake assumes 75% of iron is from heme iron sources (meats, seafood). Non-heme iron absorption is lower for those consuming vegetarian diets, for whom iron requirement is approximately 2-fold greater.

Daily dietary iron requirements differ at various stages of development, between men and women, and between pregnant and nonpregnant women. The data reported in this table assume an average dietary iron absorption of 10%.

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

What food are rich in iron?

A
  • Meats: Liver, Liverwurst, Beef, Lamb, Ham, Turkey, Chicken, Veal, Pork, Dried beef
  • Seafood: Shrimp, Dried cod, Mackerel, Sardines, Oysters, Haddock, Clams, Scallops, Tuna
  • Vegetables: Spinach, Beet greens, Dandelion greens, Sweet potatoes, Peas, Broccoli, String beans,Collards, Kale, Chard
  • Breads & Cereals: White bread (enriched), Whole wheat bread, Enriched macaroni, Wheat products, Bran cereals, Corn meal, Oat cereal, Cream of Wheat, Rye bread, Enriched rice
  • Fruits: Prunes, Watermelon, Dried apricots, Dried peaches, Strawberries, Prune juice, Raisins, Dates, Figs
  • Other Foods: Eggs, Dried peas, Dried beans, Instant breakfast, Corn syrup, Maple Syrup, Lentils, Molasses
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What is the distribution of iron in adults?

A

• Iron is an essential component of cytochromes, oxygen-binding molecules (ie haemoglobin and myoglobin), and many enzymes
• Dietary iron is absorbed predominantly in the duodenum
• Fe+++ ions circulate bound to plasma transferrin and accumulate within cells in the form of ferritin. Stored iron can be mobilized for reuse.
• Adult men normally have 35 to 45 mg of iron per kilogram of body weight. Premenopausal women have lower iron stores as a result of their recurrent blood loss through menstruation.
• More than two thirds of the body’s iron content is incorporated into haemoglobin in developing erythroid precursors and mature red cells.
• Most of the remaining body iron is found in hepatocytes and reticuloendothelial macrophages, which serve as storage deposits.
• Reticuloendothelial macrophages ingest senescent red cells, catabolise haemoglobin to scavenge iron, and load the iron onto transferrin for reuse.
• Iron metabolism is unusual in that it is controlled by absorption rather than excretion. Iron is only lost through blood loss or loss of cells as they slough.
• Men and nonmenstruating women lose about 1 mg of iron per day. Menstruating women lose from 0.6 to 2.5 percent more per day.
An average 60-kg woman might lose an extra 10 mg of iron per menstruation cycle, but the loss could be more than 42 mg per cycle depending on how heavily she menstruates

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

How is iron metabolised?

A

> 1 stable form of iron: Ferric states (3+) and ferrous states (2+).

Most iron is in the body as circulating Hb. Hb: 4 haem groups, 4 globin chains able to bind 4 O2.

The remainder is as storage and transport proteins: ferritin and haemosiderin. These are found in the cells of the liver, spleen and bone marrow.

Iron is absorbed form the duodenum via enterocytes into the plasma and binds to transferring and then transported to bone marrow to make RBCs. Excess absorption of iron is stored as ferritin

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

How is iron absorbed?

A

Iron absorption is regulated by GI mucosal cells mechanism: max absorption in the duodenum and proximal jejunum via ferroportin recetpors.

The amount absorbed depends on the type ingested: heme, ferrous (red meat, used to contain haemoglobin) > than non heme, ferric forms which is bound to other substances. Heme iron makes up 10-20% of dietary iron.

Other foods, GI acidity, state of iron storage levels and bone marrow activity affect absorption. Vitamin C helps absorption while milk/dairy decreases absorption, especially in children

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

How is iron regulated by Hepcidin?

A

“The iron regulatory hormone hepcidin and its receptor and iron channel ferroportin control the dietary absorption, storage and tissue distribution of iron…

Hepcidin cause ferroportin internalisation and degradation, thereby decreasing iron transfer into blood plasma from the duodenum, from macrophages involved in recycling senescent erythrocytes, and form iron-storing hepatocytes.

Hepcidin is feedback regulated by iron concentrations in plasma and the liver and by erythropoietic demand for iron.”

You can’t excrete it but there is a regulatory mechanism

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

How is iron transported and stored?

A
  • Iron transported from enterocytes and then either into plasma or stored as ferritin
  • Once attached to transferrin binds to transferrin receptors on RBC precursors
  • A state of iron deficiency will see reduced ferritin stores and then increased transferrin

Transferrin will go up as you are trying to get whatever iron there is into RBCs as quickly as possible

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

What various iron studies are there?

A
  • Serum Fe: hugely variable during the day
  • Ferritin: primary storage protein and providing reserve, water soluble
  • Transferring saturation: ratio of serum iron and total iron binding capacity – revealing % of transferring binding sites that have been occupied by iron
  • Transferrin/Transferrin receptors: made by liver, production inversely proportional to Fe stores. Vital for Fe transport. Uptake of Fe from protein needs transferrin to be attached to the cell via the transferrin receptor

Some are useful, and some are not. We can test for the serum iron which gives us a number of the amount of iron in the serum. This doesn’t tell us how much is in the RBCs or how much it goes up however, and it is very different throughout the day and in response to meals – so it is not very useful.

Ferritin is relatively easy to do. It is very reliable for iron deficiency. If it is low we now that we don’t have enough stored iron.

Transferrin saturation can be measured. We can see how much there is and if it goes up. We can also test for saturation

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

What is total iron binding capacity?

A

This is a measurement of the capacity of transferring to bind iron. It is an indirect measurement of transferrin – a transport protein that carried iron. TIBC is technically easier to measure in the laboratory than transferrin levels directly.

In IDA, TIBC is high…
There is more transferrin produced, aiming to transport more iron to tissues in need

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

What are the causes of iron deficiency?

A

¬ NOT ENOUGH IN: poor diet, malabsorption, increased physiological needs
¬ LOSING TOO MUCH: blood loss, menstruation, GI tract loss, parasites

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

What iron deficiency investigations might be done?

A
  • FBC: Hb, MCB, MCH, reticulocyte count
  • Iron studies: ferritin, transferrin saturation
  • Blood film
  • BMAT (bone marrow aspiration trephine) and iron stores – but this is rarely done now
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

What are the stages in the development of IDA?

A

• Before anaemia develops, iron deficiency occurs in several stages.
• Serum ferritin is the most sensitive laboratory indicators of mild iron deficiency. Stainable iron in tissue stores is equally sensitive, but is not performed in clinical practice.
• The percentage saturation of transferrin with iron and free erythrocyte protoporphyrin values do not become abnormal until tissue stores are depleted of iron.
• A decrease in the haemoglobin concentration occurs when iron is unavailable for haem synthesis.
MCV and MCH do not become abnormal for several months after tissue stores are depleted of iron

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

What might the laboratory results of IDA be?

A

It shows red cells that are in general much smaller than a neutrophil, with marked anisocytosis (variation of the red cell size) and hypochromia (area of central pallor of red cells that is larger than normal, indicating a low MCHC).

The RBCs stain blue as the Hb is stained

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

What is the prevalence of iron deficiency anaemia?

A

World’s most common nutritional deficiency
o 2% in adult men (70 years old
o 10% in caucasion, non-Hispanic women
o 19% in African-American women
o Common cause of referral
o Excessive menstrual losses 1st cause in premenopausal women

Blood loss from the GI tract is the most common cause of IDA in adult men and postmenopausal women.

Iron deficiency is the most common nutritional deficiency as well as the most common cause of anaemia throughout the world and a common cause of referral.
Premenopausal women with excessive menstrual losses are particularly at risk of developing iron deficiency anaemia (IDA)

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

What are the signs and symptoms of iron deficiency anaemia?

A

Symptoms:

  • Fatigue
  • Lethargy
  • Dizziness

Signs:

  • Pallor of mucous membranes
  • Bounding pulse
  • Systolic flow murmurs
  • Smooth tongue
  • Koilonychias
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

What is microcytic anaemia?

A

low Hb and high MCV with normal MCHC

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

What are the two types of microcytic anaemia?

A

Magaloblastic: Low reticulocyte count:

  • Vitamin B12/folic acid deficiency
  • Drug-related
  • (Interference with B12/FA metabolism)

Nonmegaloblastic:

  • alcoholism ++
  • hypothyroidism
  • liver disease
  • myelodysplastic syndromes
  • reticulocytosis (haemolysis)

Vitamin B12 = cobalamin and folic acid are both are important for the final maturation of RBC and for the synthesis of DNA. We also need both for thymidine triphosphate synthesis.

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

What is folate needed for?

A

folate is necessary for DNA synthesis: adenosine, guanine and thymidine synthesis

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

What are causes of folate deficiency?

A

Folate comes from most foods with 60-90% lost in cooking. It is absorbed in the jejunum and the body has enough stores usually for 3-5 months

INCREASED DEMAND: pregnancy/breastfeeding, infancy and growth spurt, haemolysis and rapid cell turnover e.g. SCD, disseminated cancer, urinary losses e.g. heart failure

DECREASED INTAKE: poor diet elderly, chronic alcohol intake

DECREASED ABSORPTION: medication, coeliac, jejunal resection, tropical sprue

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

About vitamin B12…

A

This is an essential co-factor for methylation in DNA and cell metabolism. There is intracellular conversion to 2 active coenzymes necessary for the homeostasis of methylmalonic acid (MMA) and homocysteine.

• Foods containing vitamin B12: animal sources – fish, meat, dairy
• UK intake recommendations are 1.5mcg/day
• EU: 1mcg/day and USA: 2.4mcg/day
• The average western intake is 5-30mcg/day
• Body (liver) storage: 1-5mg so many years for deficiency.
• Absorption of vitamin B12 in the terminal ileum requires the presence of intrinsic factor.
IF is made in parietal cells in the stomach. Transcobalamin II and transcobalamin I transport vitamin B12 to tissues

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

Why might there be an increased requirement for vitamin B12?

A
  • haemolysis
  • HIV
  • pregnancy
  • growth spurts
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

What are the clinical consequences of folate and vitamin B12 deficiency?

A

Brain: cognition, depression, psychosis
Neurology: myelopathy, sensory changes, ataxia, spasticity (SACDC)
Iron deficiency is the most common nutritional deficiency as well as the most common cause of anaemia throughout the world and a common cause of referral.
Premenopausal women with excessive menstrual losses are particularly at risk of developing iron deficiency anaemia (IDA).
Infertility
Cardiac cardiomyopathy
Tongue: glossitis, taste impairment
Blood: pancytopenia

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

What is pernicious anaemia?

A
  • Autoimmune disorder
  • Lack of IF
  • Lack of B12 absorption
  • Gastric parietal cell antibodies
  • IF antibodies
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

What are the treatments for pernicious anaemia?

A
  • Treat the underlying cuase
  • Iron: diet, oral, parenteral iron supplementation, stopping the bleeding
  • Folic acid: oral supplements
  • B12: Oral vs intramuscular treatment
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

About normal haumatopoeisis…

A
  • Blood cell production
  • Bone marrow
  • Long bones
  • Maturation occurs in bone marrow
  • Mature cells within peripheral blood

The purple tubes have EDTA in to stop the blood from clotting. As it doesn’t clot, if the blood is left for long enough it will segment out into its components

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

What is a full blood count?

A
Red blood cell results
•	Hb: concentration of Haemoglobin
•	Hct: percentage of blood volume as RBC
•	MCV: average size of RBC
•	MCH (mean cell Hb): Average haemoglobin content of RBC
•	RDW (red cell derivation width): range of deviation around RBC size
•	Reticulocyte count
•	Blood film

Hct: haematocrit (% of blood volume that is RBC) – usually around 50-52%

Not just RBC results…
• WHITE BLOOD CELL RESULTS: total WBC and differential neutrophils, lymphocytes, monocytes, basophils, eosinophils
• PLATELET RESULTS: platelet count and size

OTHERS: warning flags

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

What does a blood film show?

A

Confirming numbers
Morphology – are the cells ‘normal’
Are there cells present that shouldn’t be?

o	PURPLE: clotting
o	PINK: EDTA cross matching in blood transfusions
o	BLACK: ESR
o	RUST: clots biochemistry
o	GREEN: lithium pagrin

Tube colour does matter!!

In a lab, around 1600 FBC samples are processed every day!

Peroxide stain picks up myeloperoxidase in neutrophils

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

What are tear drop poikilocytosis?

A
  • These are seen in myelofibrosis or bone marrow infiltration with malignant disease
  • This suggests there is something else sitting inside the bone marrow
  • This wouldn’t be picked up in a numbers count
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

What is haemostasis?

A

This is a protective process evolved in order to maintain a stable physiology.

It forms a scaffold like structure so that the body can go on to heal

Haemostasis does more than just preventing blood loss. It also works in the context of someone who has an infection

Life preserving process designed to maintain blood flow:

  • Respond to tissue injury
  • Curtail blood loss
  • Restore vascular integrity and promote healing
  • Limit infection
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

What is disseminated intravascular coagulation (DIC)?

A

Coagulation is being activated by infection and blocking off the bacteria from getting to more vital places in the body. For example, the young child’s hand has become infarcted in trying to prevent the infection from getting to the liver or heart for example, and losing the hand as a result of this may not be as bad as the otherwise possible consequences.

Our biggest killer now is us (war, violence), rather than infection

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

What are the four key components of haemostats?

A
  • endothelium
  • platelets
  • coagulation
  • fibrinolysis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

What makes a blood clot?

A
  • Fibrin mesh
  • Platelets
  • Red blood cells

Platelets are the main features of a clot and they are very complex. Fibrin makes the mesh which is the proteins that come out of solution and is the final end point to strengthen the clot. Without this, people would bleed

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

What is the overview of haemostasis?

A

When you cut yourself, the first thing that happens is that you lose blood. Vasoconstriction occurs which reduces the size of the cut and therefore the amount of blood lost. There is lots of smooth muscle around the blood vessels to allow for this.

Next we get a platelet plug. The platelets recognise the damage to the endothelium and stick there to limit blood loss. This isn’t a very strong plug however, and it would only work for a little while. Fibrin comes out to strengthen this and maintain the clot by forming a mesh

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

What are the three phases of haemostasis?

A

Primary Haemostasis:

  • Vasoconstriction (immediate)
  • Platelet adhesion (within seconds)
  • Platelet aggregation and contraction (within minutes)

Secondary Haemostasis:

  • Activation of coagulation factors (within seconds)
  • Formation of fibrin (within minutes)

Fibrinolysis:

  • Activation of fibrinolysis (within minutes)
  • Lysis of the plug (within hours)

There are several different aspects to haemostasis. There is a fibrinolytic mechanism that occurs almost straight away in order to modify the clot and make sure it only stays at the site of injury

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

What is VWF?

A

In the lumen there are lots of components that are very important for haemostasis; platelets, VWF etc.

VWF is very important and people without it bleed. It is the anchor for the platelets to adhere to at the site of injury.

In the subendothelium there are the initiators for when you injur yourself; collagen and tissue factor

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

What is primary haemostasis?

A

In a damaged vessel there are two crucial molecules exposed, forget tissue factor.

Primary haemostasis involves collage. It interacts with VWF in the blood and then VWF unravels and acts as the anchor for platelets to adhere to that damaged area. The platelets adhering transform from small and round into star shaped cells, releasing their components that they are carrying, which makes more platelets and VWF coming along. This provides a nice phospholipid surface for the fibrin

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

What happens to VWF activity under shear stress?

A

VWF in the endothelium us released when you damage the surrounding area. It is vital for platelets to adhere. VWF unravels when it gets released, allowing the platelets to identify there is a problem and stick to it. You then form a platelet clot

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

What does platelet aggregation do?

A

Platelet aggregation prevents excessive blood loss at the site of injury

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

What is the role of platelets in haemostasis?

A

The things inside are exposed to the blood. Tissue factor and collagen are the important ones. Collagen causes the platelets to adhere and release substances to cause more platelets to migrate to the site. A platelet plug then forms and we get primary haemostasis

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

What is the process of haemostasis?

A

¥ When a vessel wall is damaged, various signalling molecules are expressed / exposed, including tissue factor and collagen
¥ The TF leads to the production of a small local amount of thrombin, which is the initiation step of the coagulation process
¥ The exposed signalling molecules attract circulating platelets, which attach themselves to the exposed sub-endothelial tissue (mechanism to be discussed later): this is adhesion
¥ These platelets become activated – principally through the presence of the thrombin – and release further attractant chemicals, which attract more platelets: this is secretion
¥ These new platelets bind to the adhered platelets (mechanism to be discussed later) and themselves become activated: this is aggregation
¥ Through the conformational changes inherent in activation, the loose platelet plug contracts to form a dense, adherent plug: this is contraction
¥ Together these steps comprise primary haemostasis, which may well be sufficient to achieve haemostasis if the injury is relatively minor
¥ The activated platelets also present a substantial area of negatively-charged phospholipid membrane at the site of the injury, upon which the subsequent processes of coagulation (secondary haemostasis) can occur, if needed

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

What is an overview of haemostatic plug formation?

A

The fibrin mesh binds and stabilises the platelet plug and other cells.

In coagulation fibrin forms, there are lots of clotting factors that circulate in the plasma. Most of them are produced in the liver and are complex. In the last 20-30 years we have learnt the crystalline structures of compositions of them

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

What are coagulation factors?

A

There are 13 coagulation factors and we don’t really need to learn them all. A haematologist will refer to them as their numbers. We need so many because the more you have, the more easily you can achieve regulation – ie we don’t need to turn everything off or everything on. This about having lots of light switches, you would be able to alter the light more specifically

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

What does it mean that blood has an intrinsic ability to clot?

A

Blood has an intrinsic ability to clot. It can clot without any obvious provoking factors. We call this contact activation – surface contact. The easiest way to think of it is if you cut yourself and go and clean it and wrap it up in the process made a mess of blood on bench and the blood on the bench has clotted and this is the intrinsic ability – there was no tissue factor and it clots because of a foreign surface. Factor 12 and some others can recognise a foreign, non physiological surface. These in turn can cause a clot to occur. Factor 12 activates 11 activates 9 joins up with common area does 10 then thrombin and then fibrin

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

What does FVII deficiency cause?

A

FVII deficiency causes bleeding: without factors we get bleeding. For example, without factor 7 we have a bleeding disorder and cant drive forward coagulation.

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

What does FXII deficiency do?

A

FXII deficiency is not associated with bleeding: without 12 there is no bleeding problem. This shows that the role it plays physiologically in forming clots isn’t quite the same

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

What is the revised waterfall hypothesis?

A

Each reaction requires:

  • Ca2+
  • Phospholipid
  • ± specific co-factors

In reality, these two limbs of the cascade, intrinsic and extrinsic, have a much more complex interaction

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

What is initiation of the cascade of events in haemolysis?

A
  • TF is outside the lumen
  • Formation of TF-FVIIa complex
  • Recruitment of FX and formation of thrombin

When there is bleeding from the blood vessel the tissue factor is not usually exposed to the blood, but damage leads to 7a coming into contact with tissue factor. This leads to production of prothrombin into thrombin, and then into fibrin

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

What is the cell based model of coagulation?

A

¥ Initiation of coagulation occurs when sub-endothelial tissue is exposed to the circulation at a site of injury. These tissues express tissue factor at their surface, which binds to endogenous activated FVII
¥ This complex binds small amounts of FX and FV to the exposed endothelial surface, which produce small quantities of thrombin
¥ The thrombin activates platelets that are attracted to the site by the process, as well as other plasma-borne clotting factors
¥ The activated factors (among them FVIII and FIX) enable the binding of activated FX and FV to the surface of platelets whose activation has produce conformational changes in their surface membranes to expose the ‘reaction sites’ necessary for continuation of the process
¥ This leads to the ‘thrombin burst’ that is necessary for the large-scale production of fibrin and so the development of an effective clot
¥ These three stages are called the initiation, amplification and propagation phases of coagulation
¥ Secondary haemostasis this time
¥ Tissue factor exposed to circulating blood
¥ Forms complies with factor seven
¥ Make a little bit of thrombin
¥ Very imprint regulator
¥ Need to make a lot
¥ Seven a cant do it alone
¥ Got to activate lots of other factors to do this
¥ Then make a huge amount of thrombin – thrombin burst and then get fibrin
¥ Thee is an initiator, form a small amount and then need to make more, amplification, the fed elopement of more and more sactivzted substances to PROPAGATE and produce a huge amount

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

Whats the main function of fibrinolysis?

A

The process of clot dissolution:
Main function
- Clot limiting mechanism
- Repair and healing mechanism

Series of tightly regulated enzymatic steps
- Feedback potentiation and inhibition

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

What are the main key players of fibrinolysis?

A
  • Plasminogen
  • Tissue plasminogen activator (t-PA) and urokinase (u-PA)
  • Plasminogen activator inhibitor -1 and -2
  • Alpha2-plasmin inhibitor
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

How does fibrinolysis occur?

A

First component of haemostasis and the process of removing the clot. If a clot formed and never went away you couldn’t replace it with normal tissue. Fibrinolysis is also important for limiting where the clot forms. It involves a lot of different steps. The main players are plasminogen, TPA which activated plasminogen, and things that turn it off.

The precursor wants to chew it up and is activated by TPA. It becomes active and will remove the clot. We can measure D-dimers in the lab to see if someone is going through a lot of the process. If they are then they may have a clot. This proves that it starts the moment that you are forming a clot.

If someone gets a clot it would be good if we could make this process a bit stronger. We can do this using TPA and similar substances to form plasminogen to be activated into plasminogen so it can chew up the clot

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

What happens in chronic venous insufficiency?

A
  • Atrophic changes
  • Hyperpigmentation
  • Ulceration
  • Infection

This can happen if you don’t get a DVT. There is damage to the blood vessels and they can become secondarily infected

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

Why might we get bleeding?

A

Normal haemostasis: a state of equilibrium

All the processes work in a fine balance. Blood is in a state of equilibrium between the processes

The imbalance can go the other way and we get bleeding. This would happen if you don’t have enough platelets or clotting factors.

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

What is ecchymosis?

A

Easy bruising: ecchymosis
This occurs in virtually all bleeding disorders and often in normal. If mild it may just result in easy bruising and heavy periods, or could be much more significant bleeding

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

What are methods of laboratory evaluation of bleeding?

A

There are different aspects for evaluating someone’s haemostasis. A FBC is interested in platelet number. It is not very specific but can get some degree of suspicion.

Fundamental tests are the coagulation tests. These aim to activate and produce fibrin. We can also do very specialised tests. We can look at if they have VWF which is the most common bleeding disorder. Global tests aren’t in routine diagnostic use but more at research level need standardising

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

What are the principles of clotting tests?

A

Incubate plasma with reagents necessary for coagulation

  • Phospholipid, co-factors
  • Trigger or activator
  • Calcium

Measure time taken to form fibrin clot.

We need to take blood from the patient and not allow it to clot until we get to the lab. We want to isolate the coagulation factors from the sample by administering triggering factors

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

What is prothrombin time (PT)?

A

rothrombin time is one of the coagulation screen tests. It is trying to look at extrinsic path and see how long it takes to occur. 7 tf to fibrin. This is called the prothrombin time or PT.

PT takes from 9-13 seconds and each lab has a normal range. If someone is lacking one of the factors it will take longer so might have an abnormal PT

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

What is APTT?

A

Activated partial thromboplastin time.

APTT is measuring the intrinsic path. We have to add something to activate factor 12. It is a foreign substance and we used to add things like sand and then glass beads. Now we add agents that we know have the right ionic charge. APTT is the slower way to make a clot, taking 32-45 seconds. If prolonged then we must wonder what is missing. We know know what though, because this is only a screening test

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

What is Thrombin Time (TT)?

A

The third assay is thrombin time. This is trying to mimic the last reaction. We wan to make a clot so we add thrombin and make sure that the end point is right

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

How is haemostasis testing done on a blood sample?

A
  • Accuracy of haemostasis laboratory tests depends on the quality of the specimen submitted.
  • Blood is anticoagulated with 3.2% (0.109M) sodium citrate
  • Most tubes contain 0.3mL anticoagulant and require 2.7mLs of blood
  • Under filling the tube yields grossly inaccurate results

We use citrates in the blood. Blood is collected in a tube containing citrate. The proportions have to be right, not too little or too much compared to the volume of blood. If the tube is only half filled for example, it cant be measured – it has to be completely full to have the right proportions

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

What are possible pre-analytical errors?

A

Problems with blue-top tube

  • Partial fill tubes
  • Vacuum leak and citrate evaporation

Problems with phlebotomy

  • Heparin contamination
  • Wrong label
  • Slow fill
  • Underfill
  • Vigorous shaking
  • Difficult venipuncture

Biological effects
- Hct >55 or

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

What is manual coagulation?

A

We have go the plasma we can do a manual clotting test/manual APTT. When the things are added and it is kept at 37*C we just see how long it takes to clot whilst tilting 3 times every 5 seconds

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

What is automated coagulation?

A

This machine has reduced human error in seeing the end point and it is much easier to measure

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

What are mixing studies for factor deficiency?

A
  • Clotting factor NR 50-150 IU/dL
  • APTT normal (24-34s) if factor level within range
  • 50:50 mix 32s/28s

If we have an abnormal clotting factor test and it takes a lot longer than it should it is usually due to a factor deficiency. We can mix their plasma with control plasma from healthy volunteers and this should give you a normal time. If its normal then we expect that their clotting factors are abnormal. If we mix them together, in theory we are replacing all the clotting factors from the patient with something right. If they are deficient and we mix it with normal it should replace the deficiency. We measure that, and whilst we might only get 50% of one of the clotting factors, it should be enough to correct the time. If it does correct it, then the patient was deficient in something.

Correction of a clotting test by mixing will investigate whether they are deficient in something

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

What are mixing studies for inhibitors?

A
  • Antibody in excess
  • Inhibits added factor
  • 50:50 mix 75s/30s

This wont always correct it. When mixed there may still be a long APTT. This would show the patient to have antibodies so while the clotting factors have been replaced, there are still antibodies which will now interact with the normal plasma that you have added, and so we will still get a low factor as the antibodies may inhibit normal function. Something is interfering with the coagulation

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

How are mixing studies done?

A
  • Mix patient and normal plasma in equal volumes (50:50 mix)
  • Repeat abnormal coagulation test
  • Test normalizes – factor deficiencies
  • Test remains abnormal – inhibitor (usually antibody)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
75
Q

What is D-dimer testing?

A

D-dimer testing
• A measure of the D-dimer, a fibrin degradation product
• Found elevated in the situation of enhanced fibrinolysis (thrombosis, DIC)
• Not specific for thrombosis also elevated as an acute phase reactant
• A negative result is useful if clinical suspicion of VTE is low

The D-dimer is a breakdown product of fibrin. It can be broken down into D-dimer form when it undergoes fibrinolysis. Someone who clots a lot will have a lot of D-dimer and this can be useful if we suspect DVT. If there is no raised D-dimer then it is likely that we are heading towards the wrong diagnosis

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

What is a haemocytometer neubauer chamber?

A

This is low technology which was used until the 1940s. It took a long time to count and was cumbersome, taking easily more than five minutes to do a single blood count.

It use, you put a drop into the counting chamber with a fine grid allowing counting under the microscope once the drop has been allowed to spread

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
77
Q

What is Advia 2120?

A

This is how the blood count is done now – the blood goes in, and a result comes out. This new technology has a high throughput and the blood goes through a capillary tube. The counting chamber applied an electrical field which is changed as the cells move through producing characteristic changes in current. This is then interpreted by a machine. We can also differentiate with different kinds of white cells

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

What is a normal WBC differential?

A

he most common white cells are neutrophils. This is reflected in the WBC count. We will see subtle differences in different labs, especially in neutrophils because there are ethnic variations. These variations do not indicate illness, they are just the normal ranges for different ethnicities. This may reflect on the normal range of the lab depending on where it is, even within a city.

Lymphocytes are the second most common, and have less variation between ethnicities – usually accepted as a global normal.

These cells do different things and are from different lineages within the development

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

About peripheral blood film…

A

If you have doubt about something and the results don’t quite fit, you can make a blood film. To do this you take a drop of blood and then take a slide and put a 40* angle onto it, pull back a bit so it spreads and then slide forward so you are spreading the blood thin enough to look at individual cells.

After this is done it needs to be fixed and stained to be looked at and investigated

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

What is Romanowsky Stain?

A

There are 2 main components:

  1. Azure B or Methylene Blue – basic dyes
  2. Eosin Y – acidic dye

Buffer: pH 6.8

To separate the white cells we will need two different stains. Basophillic stain (blue) and azurophilic stain (red). Some will stain blue and some will stain red allowing differentiation

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

What are normal neutrophils like on staining?

A

What you end up with is the staining of everything. Neutrophils are the most common cells. We can recognise that these are neutrophils because they have a segmented nucleus (polysegmented). Up to five segments is normal and this can be easily recognised. They become this way as they mature

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

What are lymphocytes like on staining?

A

They are different as they have a single round nucleus with a variable amount of cytoplasm. Some have more cytoplasm than others. When they become activated or reactive through infecition, they may change their morphology between

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

What is the role of lymphocytes?

A

The major role of the lymphocytes is adaptive immune response, the differentiation between self and non-self.
Eg viral attack, lymphocytes produce antibodies to destroy that virus, memory cells are produced

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

What are the divisions of the immune system?

A

The two basic divisions of the immune system are the adaptive and innate system. The innate system kills when and where it needs to. The two parts are integrated into one another and they don’t function independently but work as one

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

What are the two major types of lymphocytes?

A

There are three major types of lymphocytes:

  • T cells
  • B cells
  • Natural killer (NK) cells

These work together to initiate an immune response when you need it. They are naïve when you are born. The first virus you come across will activate a certain number of clones of the pre existing lymphocyte repertoire and want to kill it.

T cells are triggered directly and detect the virus/infected cells and kill it. B cells make antibodies which bind to organisms and coat cells that are affected by viruses. Natural killer cells mostly directly kill the infected cells and cancer cells – abnormal cells.

T cells are involved in cell-mediated immunity

  • CD4+ T-Helper cells
  • CD8+ Cytotoxic T-cells

B cells in humoural immunity ie antibody production

NK cells are part of the innate immune system attacking virally infected cells and tumour cells

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

When do the lymphocyte cells become abnormal?

A

These cells become abnormal when they are infected by viruses. Part of the virus is expressed on the surface of cells and they start looking different; altered self. This is how the immune system recognises and destroys cells. If you couldn’t do this, you would die of autoimmune disease and you wouldn’t be able to differentiate between your own cells. You also wouldn’t be able to absorb food etc as it would all see foreign. The immune system is programmed to tolerate, and it may switch and make an immune response when you need it to.

The nucleus of resting lymphocytes is roughly the same size as a red blood cell. The blue cytoplasm comes from antibodies staining blue

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

What is a large granular lymphocyte or natural killer (NK) cell?

A

A proportion of lymphocytes may look like this. They are large granular lymphocytes. It can be natural to see when fighting infection, for example in leukaemia there is a clinical expansion of these. Activated T cells into natural killer cells look like this

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

What are monocytes?

A

Monocytesa re the next most commonly seen WBC. They are part of the innate immune system and they are phagocytosing cells. These don’t have visible granules like the lymphocytes but the nucleus can be funny, irregular shapes. They are recognised by this characteristic as well as by their colour which is grey blue and no visible in cytoplasm and there are often vacuoles within the cytoplasm

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

What are eosinophils?

A

Eosinophils are also recognised quite easily. Eosinophils are recognised by the polynucleotide and granules. They also have granules but are more red than the neutrophils. There are very few basophilic granules. They are eosinophilic. These cells fight a specific corner that provides protection against parasites, especially in the gut. They also have involvement in allergic reactions. The gut reactions and allergies can show degranulation of eosinophils – antihistamines.

They are present in eczema, asthma, worms and GI parasites

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

What are basophils?

A

These are the rarest of the WBC in the blood. Sometimes none will be seen in a blood film. If there are too many it can be associated with certain malignancies. Blood is arguably the least important place for them to be – they go out into tissues to survey tissues and do their job and then they die. The typical life span of a basophil is less than one day – it goes where it is needed, fights infection and then dies

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

How do cells develop in the bone marrow?

A

Because they don’t live long we need good production of them in the bone marrow. We need millions produced every second to keep up with what you need. They start dividing in the bone marrow and are becoming more committed progenitors. The stem cell can be anything.

The reason we don’t run out of stem cells is because of asymmetric division. They go through a number of precursor stages in the bone marrow, and the whole process takes 7-10 days

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

What is the physiological response to most infections?

A

The physiological response to most infections is to increase neutrophils and push them out. Certain drugs can cause neutropenia, such as chemotherapy

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

What is toxic granulation?

A

when they become activated they change how they look

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

What are the steps of myloid maturation?

A
Myeloblast
 promyelocyte
 myelocyte
 metamyelocyte
 band form
 segmented neutrophil
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
95
Q

What is a hzypersegmened neutrophil?

A

six or more segments make the neutrophil hyperhsegmented.

this can be caused by a deficiency in vitamin B12, for example.

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

What is a Dohle Body?

A

These are rather non-specific. Some innate problems can be seen. They are a glomerulus of large blue granules, forming one large blue longitudinal shaped thing. They can be seen in conditions of infection or inflammation and are a good thing to know.

They usually go alongside toxic granulation as well. There are three tongs in this neutrophil. Body, granulation and not fully mature segmented neutrophil is a slightly earlier precursor which sometimes pushes out into the blood, sometimes in response to infection/inglammation

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

What are possible causes for neutrophilia?

A

Normal physiological reaction:

  • Post-operative
  • Pregnancy

Other causes:

  • Bacterial infection
  • Inflammation eg vasculitis, myocardial infarction
  • Carcinoma
  • Steroid treatment
  • Myeloproliferative disorders
  • Treatment with myeloid growth factors

This can be in response to stress, autoimmune disorders, steroids, pregnancy. Neutrophils try to go to the side of the blood vessels because they leave the blood vessels. In steroids they become more in the middle which is why there is an increase when blood is drawn.

Is this a normal response to stress of the system? If it is without any real reason then it is called myeloproliferative disorder

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

What are investigations of neutrophilia?

A
  • FBC and differential white cell count
  • Blood film examination
  • Bacterial culture screen for infection
  • Bone marrow examination and chromosome analysis for chronic myeloid leukaemia – Philadelphia chromosome: translocation between chromosomes 9 and 22, molecular analysis for BCR-ABL oncogene

The white cell count makes you want to do a blood film. We want to look at the chromosomes in the cells of blood vessels. We may see characteristic translocation between chromosomes driving this growth and we now have treatments for these things. We can recognise, detect and monitor the treatment

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

What is neutropenia?

A
  • Viral infection
  • Drug induced eg sulphonamides
  • Radiotherapy and chemotherapy
  • Part of a pancytopenia in bone marrow failure (aplastic anaemia) or infiltration eg leukaemia
  • Racial: ‘benign ethnic neutropenia’
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
100
Q

What is eosinophilia?

A

They are bi-lobed with big red granules. This could also be a malignant process. Part of the stem cell is destined to be an eosinophil. Are there lost of basophils for example?

  • Allergic diseases eg asthma, hayfever
  • Parasitic infections
  • Drug sensitivity
  • Myeloproliferative diseases eg chronic myeloid leukaemia
  • Hidgkin’s lymphoma

Allergic diseases will bring up eosinophils and the differential is very wide. It is important therefore not to jump to conclusions, but to report what you see and seek further information

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

What are investigations of eosinophilia?

A
  • FBC and differential white cell count
  • Blood film examination
  • Stool examination for ova and parasites
  • CLARIFICATION!
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
102
Q

What is monocytosis?

A

These are recognised as bigger cells with irregular shaped nuclei. They have pale grey blue cytoplasm and there is no prominent granulations but some have vacuoles.

  • TB
  • Acute and chronic monocytic and myelomonocytic leukaemia
  • Malaria (chronic, not so much acute)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
103
Q

What are the investigations of monocytosis?

A
  • FBC and differential white cell count
  • Blood film examination: for abnormal white blood cells and for malarial parasites
  • Bone marrow examination – leukaemia
  • TB cultures
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
104
Q

What is lymphocytosis?

A

Some are clear cut, big square monocytes. The morphology alone may not get you to know what something is, so you could have to use certain stains etc and send them away to find out with phenotyping etc. There could be something malignant in this picture…

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

What are the possible causes of lymphocytosis?

A

Normal physiological status:
• Lymphocytosis of childhood (1-6 years) normal: 5.5-8.5x10^9/L
Other causes:
• Bacterial infection
• Viral infections eg hepatitis, mumps, rubella, pertussis, glandular fever, (infectious mononucleosis)
• Leukaemias and lymphomas

Higher in early life and drops down and reaches normal range in teenage life

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

What are the investigations of lymphocytosis?

A
  • FBC and differential white cell count

* Blood film examination

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

What are atypical mononuclear cells?

A

This may be seen in glandular fever etc. All the glands produce these. They are not malignant, they are blasts – reactive T cells often look like this. They envelope the red cells. Peripheral basophilia is present which means there is a blue ring around the red cells. This may be how blood cells look after glandular fever for example, or anything that drives a strong T cell immune response

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

What is infectious mononucleosis?

A

Glandular Fever

Heterophile antibodies are antibodies which react against an antigen which is completed unrelated to the antigen which originally stimulated it eg human antibodies reacting against sheep or horse or bovine cells

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

What is clearview infectious mononucleosis?

A

This is the modern way of testing. There are antibodies that will detect antibodies. If they are in the serum moving upwards and we put on a secondary antibody that will recognise them, we will get a control line.

When we see the second line occurring this may show mononucleosis. This may not work in the first few days of infection because they take a while to develop – so a repeat should be suggested

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

What investigations might be done into lymphocytosis?

A

Mature lymphocytes: chronic lymphocytic leukaemia? Lymphoma?

Immunophenotyping to determine if lymphocytes are:

  • B cells: demonstrate clonality by light chain restriction
  • T cells: demonstrate clonality by T-cell receptor gene rearrangement studies
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
111
Q

What are blood group antigens?

A

Antigens are part of the surface of cells
• All blood cells have antigens

Reactions to blood usually occurs when the antibody in the plasma reacts with an antigen on the cells.

  • There are 26 known blood group systems
  • ABO and Rh are clinically most important
  • Antigens in transfused blood can stimulate a patient to produce an antibody but only if the patient lacks the antigen themselves
  • The frequency of antibody production is very low but increases the more transfusions that are given
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
112
Q

What are blood group antibodies?

A

Antibodies are protein molecules – immunoglobulins (Ig)
• Usually of the immunoglobulin classes: IgG and IgM
• Found in the plasma
• Produced by the immune system following exposure to a foreign antigen

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

How is antibody production stimulated?

A
  • Blood transfusion: ie blood carrying antigens foreign to the patient
  • Pregnancy: fetal antigen entering maternal circulation during pregnancy or at birth
  • Environmental factors: ie naturally acquired eg anti-A and anti-B
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
114
Q

What are example of in vivo and in vitro antibody-antigen reactions?

A
In vivo (in the body) – leads to destruction of the cell either:
Directly when the cell breaks up in the blood stream (intravascular)
Indirectly when liver and spleen remove antibody coated cells (extravascular)
In vitro (in the laboratory)
Reactions are normally seen as agglutination tests
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
115
Q

What is agglutination?

A
  • Agglutination is the clumping together of red cells into visible agglutinates by antigen-antibody reactions
  • Agglutination results from antibody cross-linking with the antigens
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
116
Q

What can agglutination identify?

A

As the antigen-antibody reaction is specific, agglutination can identify:
• The presence of a red cell antigen: ie blood grouping
• The presence of an antibody in the plasma: ie antibody screening/identification

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

What is the clinical significance of the ABO grouping system?

A
  • A and B antigens very common (55% UK)
  • Anti-A, anti-B or anti-A,B antibodies very common (97% UK)
  • High risk of A or B cells being transfused into someone with the antibody in a random situation
  • ABO antibodies can activate complement causing intravascular haemolysis

(Almost) all serious/fatal transfusion reactions caused by technical/clerical error are due to ABO incompatibility

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

What is blood grouping?

A

The patients red cells and plasma are both tested.

Test patient’s red cells with anti-A, anti-B and anti-D
• Agglutination shows that a particular antigen is on the red cells
• No agglutination shows the antigen is absent

Test patient’s plasma with A cells and B cells
• Agglutination shows that a particular antibody is in the plasma or serum
• No agglutination shows the antibody is absent

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

What is the Rh grouping system?

A

50+ antigens:
• Most important antigen is called D
• People with D antigen are RhD positive (85% of UK)
• People who do not produce any D antigen are RhD negative (15%)
• The other 4 main Rh antigens are known as C, c, E and e

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

What is Rh (D) typing?

A
  • Most important after ABO
  • Must be tested in duplicate (or tested each time and compared to historical result)
  • Patient/donor classified as RhD pos or RhD neg
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
121
Q

What is the clinical significance of Rh?

A

Rh antibodies are clinically significant. Second only to ABO.

Transfusion:
• D antigen is very immunogenic and anti-D is easily stimulated – PREVENTION!
• All Rh antibodies are capable of causing severe transfusion reaction – ANTIBODY DETECTION

Pregnancy:
• Rh antibodies are usually IgG and can cause haemolytic disease of the newborn
• Anti-D is still most common cause of severe HDN

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

What is laboratory testing of HDN?

A
  • Blood group and antibody screen at antenatal booking to identify pregnancies at risk of HDN – RhD negative women who may need anti-D prophylaxis
  • Blood group and antibody screen at 28 weeks
  • Atypical antibodies are quantified periodically to assess their potential effect on the fetus
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
123
Q

What is RAADP?

A
  • An injection of anti-D will bind to and remove any fetal RhD positive red cells in the circulation
  • 1500 iu of anti-D is given routinely at 28 weeks and a smaller dose (usually 500 iu) after delivery if baby RhD+
  • in some hospitals 2 smaller (500 iu) doses are given at 28 and 34 weeks instead of the 1 larger dose
  • anti-D is also given after any event that may cause a feta-maternal haemorrhage (bleed between mum and fetus) such as:
  • abdominal trauma
  • intrauterine death
  • spontaneous or therapeutic abortion
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
124
Q

What is antibody screening?

A
  • there are other clinically significant antibodies that can cause a haemolytic transfusion reaction
  • it is important that we screen for these antibodies so that if detected, antigen negative blood can be provided to avoid causing an immune reaction
  • patients serum is mixed with 3 selected screening cells, incubated for 15 minutes at 30^C and then centrifuged for 5 minutes
  • any clinically significant antibodies reacting at body temperature should be detected and then identified using panel of known phenotyped red cells
  • specific antigen negative blood can then be provided for these patients to avoid stimulating an immune response
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
125
Q

What must be do if an antibody is detected in an antibody screen?

A
  • identify the antibody

* assess its clinical significance: for transfusion, in pregnancy

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

How do we detect an antibody in an antibody screen?

A
  • compare pattern of reactions with each reagent cell of ID panel with the pattern fo antigens on the reagent cells
  • matching pattern will identify the antibody
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
127
Q

What is the zeta potential?

A

the potential difference existing between the surface of a solid particle immersed in a conducting liquid (e.g. water) and the bulk of the liquid.

IgM antibodies can span the gap between RBCs. IgG can not, because it is too small to overcome the ZETA potential (positive chage). LISS (low ionic strength saline) is negatively charged, so neutralises positive ZETA potential. Therefore, IgG can now span the gap

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

What is the indirect anti-globulin test (IAT)?

A
•	used to detect IgG antibodies
•	LISS counteracts Zeta potential
•	Results in agglutination
•	Used for:
•	Screening for antibodies
•	Identifying antibodies
Cross matching donor blood with recipient plasma when there are known antibodies or a previous history of antibodies
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
129
Q

What is an intermediate spin cross-match (ISX)?

A
  • Antibody screen is negative
  • Checking donor red cells against patients plasma: ABO check, incubate for 2-5 minutes (room temp), spin and read

ISX – basically just checking the ABO group. Therefore IgM antibodies (therefore no problem with ZETA potential, therefore no need to IAT).

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

What is a full indirect anti globulin test (IAT) cross match?

A
  • Antibody screen positive or patient has known antibody history
  • Select antigen negative donor red cells and incubate with patient serum for 15 minutes at 37^C
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
131
Q

How is donor blood tested?

A
  • Blood establishment: MHRA licensed manufacturer of blood and products
  • Donor selection: questionnaire; lifestyle, health, not previously transfused
  • Collection procedure arm cleansing/diversion pouch
  • Comprehensive testing of all products: Viral; HIV 1+2, hepatitis B, hepatitis C, syphilis, HTLV
  • Platelets: bacteria
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
132
Q

What are the relative risks of transfusion?

A
  • 1 in 910,000 for hepatitis B
  • 1 in 70 million for hepatitis C
  • 1 in 5 million for HIV infection
  • 1 in 23 million for HTLV infection

There are believed to have been 4 cases of infected with vCJD from blood transfusion

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

About fresh frozen plasma…

A

• FFP contains all clotting factors
- Given for coagulopathy with associated bleeding
- Requires clotting screens to monitor
• Only has 24 hour life after thawing

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

About platelets (donor)…

A
  • Adult pool of platelets fro 4 donors (suspended in plasma from 1 donor)
  • 85% of doses are apheresis units
  • platelets required to create clots to reduce bleeding
  • some drugs given to reduce efficacy of platelets (anti-platelet agents) so patient history is important
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
135
Q

What is cryoprecipitate?

A

Contains factor VIII, VWF and fibrinogen

  • 2 units usually given at one time
  • monitor fibrinogen levels by clotting screens
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
136
Q

How is donor blood regulated?

A
  • EU Blood Safety Directive
  • Blood Safety Quality Regulations
  • Better Blood Transfusion 3
  • MHRA inspections
  • CPA inspections
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
137
Q

What are methods of haemovigilance?

A

Serious hazards of transfusion (SHOT):

  • Voluntary reporting
  • Report all serious adverse events (SAE) and serious adverse reactions (SAR)

Serious adverse blood reactions and events (SABRE):

  • Mandatory reporting
  • Report all SAR and SAE where the root cause error was the quality system
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
138
Q

What sources maintain blood glucose levels?

A

Blood glucose levels are maintained from several sources:
• Dietary carbohydrate
• Glycogenolysis
• Gluconeogenesis

Livers role:
• After meals: stores glucose as glycogen
• During fasting: makes glucose available through glycogenolysis and gluconeogenesis

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

Why should glucose levels be regulated?

A
  • Brain and erythrocytes require continuous supply – avoid deficiency
  • High glucose and metabolites cause pathological changes to tissues; eg, micro/macro vascular diseases, neuropathy – avoid excess
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
140
Q

What is insulin?

A
  • Peptide hormone (51 amino acids)
  • Synthesised in -cells of pancreas as proinsulin; cleaved to insulin and C-peptide
  • Secretion is stimulated by rise in blood glucose
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
141
Q

What are glucose counter-regulatory hormones?

A

Glucagon
• Secreted by alpha-cells of pancreas in response to hypoglycaemia
• Stimulates glycogenolysis and gluconeogenesis

Adrenaline
• Increased glycogenolysis and lipolysis

Growth Hormone
• Increased glycogenolysis and lipolysis

Cortisol
• Increased gluconeogenesis, glycogen synthesis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
142
Q

What is diabetes mellitus?

A

Metabolic disorder characterised by chronic hyperglycaemia, glycosuria and associated abnormalities of lipid and protein metabolism.
• hyperglycaemia result of increased hepatic glucose production and decreased cellular glucose uptake
• blood glucose > ~10mmol/L exceeds renal threshold – glycosuria
• long term complication – micro-macrovascular disease

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

What is the prevalence of diabetes?

A
  • globally 415 million people currently have diabetes; estimated to double by 2030
  • estimated prevalence of diabetes in UK 2013 was 3.2 million (ages 17+)
  • number of people in UK with diabetes has more than doubled since 1996 (1.4 million)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
144
Q

What are the reasons for an increase in the prevalence of diabetes?

A
  • increased awareness
  • more complete recording
  • poor diet
  • low physical activities
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
145
Q

How is diabetes diagnosed?

A

In the presence of symptoms: (polyuria, polydipsia weight loss)
• random plasma glucose > 11.1 mmol/l (200 mg/dl) OR
• fasting plasma glucose > 7.0mmol/l (126 mg/dl) fasting is defined as no caloric intake for at least 8 hours OR
• 2-h plasma glucose > 11.1 mmol/l (200 mg/dl); 2hrs after 75g oral glucose tolerance test (OGTT)

In the absence of symptoms:
• test blood samples on 2 separate days

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

What is IGT (pre-diabetes) and IFG?

A

Impaired glucose tolerance (IGT)
• fasting plasma glucose 6.1-6.9 mmol/L**
• OTTG value of

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

What is the oral glucose tolerance test (OGTT)?

A

• OGTT should be carried out:
o In patients with IFG
o In unexplained glycosuria
o In clinical features of diabetes with normal plasma glucose values
o For the diagnosis of acromegaly
• 75g oral glucose and test after 2 hours
• blood samples collected at 0 and 120 mins after glucose
• subjects tested fasting after 3 days of normal diet containing at least 250g carbohydrate

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

What are the classifications of diabetes?

A

TYPE 1:
insulin secretion is deficient due to autoimmune destruction of beta-cells in pancreas by T-cells

TYPE 2:
insulin secretion is retained but there is target organ resistance to its actions

GESTATIONAL:
occurs for first time in pregnancy

SECONDARY:
chronic pancreatitis, pancreatic surgery, secretion of antagonists

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

About type 1 diabetes mellitus…

A
  • predominantly in children and young adults’ but ages also
  • sudden onset (days/weeks)
  • appearance of symptoms may be preceded by ‘prediabetic’ period of several months
  • commonest cause is autoimmune destruction of B-cells
  • interaction between genetic factors and environment
  • strong link with HLA genes within the MHC region on chromosome 6
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
150
Q

What is the pathogenesis of type 1 DM?

A

• HLA class II cell surface present as foreign as self antigens to T-lymphocytes to initiate autoimmune response
• Circulating autoantibodies to various-cell antigens:
o Glutamic acid decarboxylase
o Tyrosine-phosphatase-like molecule
o Islet auto-antigen
• The most commonly detected antibody associated with type 1 DM is the islet cell antibody
• More than 90% of newly diagnosed persons with type 1 DM have one or another of these antibodies
• Destruction of pancreatic beta-cell causes hyperglycaemia due to absolute deficiency of both insulin and amylin
• Amylin, a glucoregulatory peptide hormone co-secreted with insulin
lowers blood glucose by slowing gastric emptying, and suppressing glucagon output from pancreatic cells

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

What is the presentation of type 2 DM?

A
  • Slow onset (months/years)
  • Patients middle aged/elderly – prevalence increases with age
  • Strong familiar incidence
  • Pathogenesis uncertain – insulin resistance; beta cell dysfunction may be due to lifestyle factors – obesity, lack of exercise
  • Emergency presentation as hyper-osmolar, non-ketotic state (HONK)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
152
Q

What are metabolic complications of type 2 DM?

A
Hyper-osmolar non-ketotic coma (HONK)
•	Development of severe hyperglycaemia
•	Extreme dehydration
•	Increased plasma osmolality
•	No ketosis, minimal acidosis
•	Impaired consciousness
•	Death is untreated
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
153
Q

What are ways of biochemical monitoring DM?

A
  • Immediate: blood glucose
  • Few hours: urinary dipstick
  • 3-4 months: blood HbA1c (glycated Hb, covalent linkage of glucose to residue in Hb)
  • Others: orinary albumin (index of risk of progression to nephropathy), abnormalities in serum lipids common in T1 and T2 DM – increased risk of MI and stroke
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
154
Q

What are the aims of monitoring DM?

A
  • Prevent complication through tight control
  • Avoidance of hypoglycaemia
  • Self-monitoring: capillary blood measurement, urine analysis: glucose in urine gives indication of blood glucose concentration above renal threshold
  • Glycated Hb (HbA1c) – aim at
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
155
Q

What are possible long term complications of DM?

A
  • Occur in both type 1 and type 2 DM
  • Micro-vascular disease: retinopathy, nephropathy, neuropathy
  • Macro-vascular disease: related to atherosclerosis heart attack/stroke
  • Exact mechanisms of complications are unclear
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
156
Q

How might we reduce the CV risk in DM?

A
  • Attain normal weight and waist circumference
  • Eat food low in fat and salt
  • Exercise
  • Stop smoking
  • HbA1c
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
157
Q

What is hypoglycaemia?

A

• Defined as plasma glucose

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

What are the causes of hypoglycaemia?

A
  • Insulinoma
  • Drugs eg sulphonylureas, insulin, alcohol abuse
  • Inherited metabolic disorders eg glycogen storage diseases, galactosaemia, hereditary fructose intolerance
  • Endocrines disease; eg cortisol disorder
  • Others: severe liver disease, non-pancreatic tumours, postgastrectomy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
159
Q

What are the adaptations to falling glucose levels in fasting?

A

• Insulin levels fall, limiting glucose entry into non-cerebral tissues
• Hepatic gluconeogenesis stimulated
• Glycogen breakdown activated
• Fatty acid oxidation activated
• Release of counter regulatory hormone raising glucose
- Glucagon – (

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

What are the symptoms of hypoglycaemia?

A

Neurogenic (autonomic):

  • Triggered by falling glucose levels
  • Activated by ANS and mediated by sympathoadrenal release of catecholamines and Ach
  • Sign and symptoms include shakiness, anxiety, nervousness, palpitations, sweating, dry mouth, pallor and pupil dilation

Neuroglycopaenia:

  • Due to neuronal glucose deprivation
  • Sign and symptoms include confusion, difficulty speaking, ataxia, paresthesia, seizure, coma and death

Insulinoma:
- Tumour in the insulin secreting beta-cells of pancreas

Symptoms include:

  • Fasting hypoglycaemia, patients may present with behavioural changes
  • Inappropriate high insulin concentration at time when plasma glucose is low (
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
161
Q

What are examples of inherited metabolic diseases?

A
  • Glycogen storage disease type I (von Gierke’s disease); deficiency of G-6-Phosphatase: impaired glucose release from glycogen
  • Galactosaemia: deficiency of galactose-1-phosphate uridyl transferase: liver damage
  • Hereditary fructose intolerance: deficiency of fructose-1-phosphate adolase B: accumulation of fructose-1-phosphate in liver
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
162
Q

What is glycogen storage disease type Ia?

A
  • Autosomal recessive disorder (described in 1929)
  • Glucose synthesis from glycogen or by gluconeogenesis is blocked
  • Presents in early infancy; severe fasting hypoglycaemia as only source of glucose is dietary carbohydrate
  • Accumulation of glycogen causes hepatomegly; inability to produce glucose from lactose causes acidosis
  • Tx: uncooked cornstarch; a slow releasing glucose prep
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
163
Q

What is galactosaemia?

A
  • Defects in 3 enzymes can cause galactosaemia; most common is galactose-1-phosphate uridyl transferase deficiency
  • Autosomal recessive disorder (1st described in 1935)
  • Deficiency of G-1-PUT impairs conversion of galactose-1-phosphate to glucose-1-P gal-1-phosphate accumulates in liver – toxicity
  • Hypoglycaemia and vomiting/diarrhoea after starting milk feeds
  • Galactose excreted in urine. Tx – exclude galactose from diet
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
164
Q

What is hereditary fructose intolerance?

A
  • Autosomal recessive disorder (1st described in 1956)
  • Deficiency of fructose 1-phosphate aldolase B when ingested fructose accumulates – inhibits glycogenolysis at phosphorylase step
  • Severe hypoglycaemia and vomiting after ingesting fruit, sweetened foods
  • Fructose detected in urine
  • Avoid ingestion of fructose, sucrose
  • Benign fructose intolerance: this is due to absence of fructokinase
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
165
Q

What are the hypothalamic/pituitary connections?

A
  • These are neural connections between the hypothalamus and posterior (but not anterior) pituitary
  • Blood form the hypothalamus supplies the anterior pituitary
  • The secretion of anterior pituitary hormones is regulated by hypothalamic releasing factors
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
166
Q

What is the regulatory factor to growth hormone?

A

GR-RH (stimulatory) and somatostatin (inhibitory)

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

What is the regulatory factor to prolactin?

A

PIF (dopamine)

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

What is the regulatory factor to TSH?

A

TRH

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

What is the regulatory factor to ACTH?

A

corticotrophin (CRF)

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

What is the regulatory factor to LH?

A

GnRH

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

What is the regulatory factor to FSH?

A

GnRH

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

About thyroid hormones…

A

¥ Essential for normal growth and development
¥ Increase basal metabolic rate (BMR) and affect many metabolic processes
¥ Synthesized in thyroid via series of enzyme catalyzed reactions, beginning with uptake of iodine into gland
¥ Synthesis and release controlled by TSH
¥ T4 main hormone secreted by thyroid, T3 is more biologically active – mostly formed by peripheral conversion from T4
¥ Effects are mediated via activation of nuclear receptor

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

About thyroid hormones in blood…

A
  • Both T4 and T3 are extensively protein bound, principally to TBG
  • Free (non-protein bound) fraction physiologically active
  • Changes in protein binding affect total hormone concentrations
  • Measurement of free hormones is not affected by variation in TBG and so discriminates more reliably between normal and abnormal thyroid function
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
174
Q

What are possible causes of hyperthyroidism?

A
  • Graves disease: most common (TSH-R antibodies)
  • Toxic multinodular goiter
  • Toxic adenoma thyroiditis
  • Excessive TSH production (rare)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
175
Q

What is hyperthyroidism?

A

excessive production of thyroid hormones

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

What are the clinical features of hyperthyroidism?

A
  • weight loss
  • heat intolerance
  • palpitations
  • goitre
  • eye changes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
177
Q

What investigations might be done into hyperthyroidism?

A
  • Serum TSH concentraitons suppressed

- fT4 raised

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

What is hypothyroidism?

A

deficient production of thyroid hormones

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

What are possible causes of hypothyroidism?

A
  • primary: autoimmune thyroiditis, iodine deficiency, post-surgery/antithyroid drugs, congenital
  • secondary: pituitary or hypothalamic diorder
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
180
Q

What are the clinical features of hypothyroidism?

A
  • weight gain
  • cold intolerance
  • lack of energy
  • goitre
  • congenital - development abnormalities
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
181
Q

What investigations might be done into hypothyroidism?

A
  • serum TSH concentrations raised
  • fT4 reduced (primary)
  • reduction in both TSH+fT4 consistent with hypopituitarism
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
182
Q

What are treatment and monitoring options for hyperthyroidism?

A
  • anti-thyroid drugs (g carbimazole)
  • partial thyroidectomy
  • radioactive iodine
  • fT4 levels fall rapidly with successful treatment
  • TSH may remain suppressed for several months
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
183
Q

What are treatment and monitoring options for hypothyroidism?

A
  • Thyroxine replacement
  • fT4 may remain slightly raised
  • best monitored by TSH (elevated values indicate under/and suppressed values over replacement)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
184
Q

What are the zones of the adrenal cortex?

A
  • Aldosterone is the main mineralocorticoid synthesized in (outer) zona glomerulosa
  • Cortisol is the principal glucocorticoid produced by the zona fasciculate
  • Glucocorticoids and adrenal androgens are synthesized in zona fasciculate & zona reticularis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
185
Q

What are the biological effects of adrenal steroids?

A

Cortisol

- antagonizes effects of insulin by enhancing gluconeogenesis 
- promotes the central deposition of fat
- increases catabolism of proteins - weak mineralocorticoid effects
- anti-inflammatory

Aldosterone
- increases sodium reabsorption in renal DCT

Androgens
- virilizing effects when secreted in excess

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

How is adrenal steroid secretion controlled?

A
  • Synthesis of cortisol regulated by hypothalamic - pituitary - adrenal axis
  • Aldosterone synthesis is controlled by the renin-angiotensin system
  • Adrenal androgen synthesis is regulated by ACTH, not gonadotrophins
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
187
Q

What are examples of hyper functioning of the adrenal cortex?

A

Conn’s syndrome:
excess secretion of aldosterone due to tumour or hyperplasia of adrenal - sodium retention

Cushing’s syndrome:
excess cortisol secretion - causes:
-	pituitary tumour (Cushing’s disease)
-	ectopic ACTH production
-	adrenal tumour
-	exogenous administration of steroids
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
188
Q

What is cushings syndrome and what are the clinical features?

A

Due to excessive cortisol production by adrenal.

Clinical features include:
•	muscle weakness
•	menstrual disturbances
•	psychiatric disturbances
•	moon face, truncal obesity
•	striae, bruising
•	hirsutism, acne
•	hypertension
•	glucose intolerance
•	osteoporosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
189
Q

What are the diagnostic steps to cushion’s syndrome?

A
  1. Demonstration of excessive cortisol production
    Screening Tests:
    - Loss of cortisol circadian rhythm
    - Elevated 12am serum cortisol
    - Increased 24hr urine cortisol
    - Inadequate suppression of serum cortisol levels following low dose (1mg dexamethasone)
  2. Elucidation of cause
    - High dose dexamethasone suppression test
    - ACTH assay
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
190
Q

What are dynamic tests of endocrine function?

A

Dynamic tests are important tools in the investigation of endocrine function.

There are two types:

  • Stimulatory test may be used to investigate endocrine gland hypofunction
  • Suppression tests are used to investigate suspected hyperfunction
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
191
Q

What is the dexamethasone suppression test?

A

Dexamethasone

  • synthetic glucocorticoid
  • binds to GC receptors in pituitary to suppress
  • ACTH release (and cortisol secretion from adrenal)

Low dose dexamethasone suppression test

  • 1mg dexamethasone given at night. Serum cortisol is measured at 9am next morning
  • in normal subjects, cortisol
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
192
Q

What are the causes of andrenocortical insufficiency?

A
Causes:
 Primary adrenocortical failure –
	Addison’s disease due to :
	  autoimmune disease
	  tuberculosis
	  less common; metastases, haemochromatosis, 
             haemorrhage

Secondary to impaired ACTH release eg –
tumour, head trauma, surgery,
rapid withdrawal of steroids

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

What are the clinical features of adrenocortical insufficiency?

A
  • fatigue
  • weakness
  • hypoglycaemia
  • hypotension
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
194
Q

What is the short syncathen test?

A

Short synacthen test
- to assess the ability of the adrenal to produce cortisol in
response to ACTH

Serum cortisol is measured after giving synacthen (synthetic ACTH)
(normal response – cortisol increases by >200nmol/L over basal
level with peak of >550nmol/L).

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

What is the long syncathen test?

A

Long synacthen test
- 3 day stimulation with 1mg depot synacthen i.m. daily

In secondary adrenal hypofunction, serum cortisol levels increase
to at least 200nmol/L over baseline values

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

What is congenital adrenal hyperplasia?

A

¥ Group of inherited metabolic disorders of adrenal steroid hormone biosynthesis
¥ Clinical features depend on the position of enzyme defect in metabolic pathway
¥ 21-hydroxylase deficiency > 90% cases, prevalence 1:5000-12,000
Leads to impaired production of cortisol and aldosterone. Raised ACTH stimulates production of adrenal androgens

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

What are neonatal manifestations of CAH?

A

¥ Virilization of female infants
¥ Salt wasting and dehydration occurs during first 4 weeks of life, serum Na 2 days after birth
¥ Requires glucocorticoid and mineralocorticoid replacement
¥ Treatment monitored by measurement of either plasma 17a-hydroxyprogesterone or androstenedione

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

What is polycystic ovarian syndrome?

A

¥ Condition of excess androgen secretion and chronic anovulation
¥ Ovaries are the source of excess androgens and there are multiple ovarian cysts
¥ Associated with menstrual disturbances, hirsutism and infertility
¥ Many patients with PCOS are overweight and a link has been made with insulin resistance
¥ Increased serum LH:FSH ratio
¥ Decreased SHBG

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

What are possible causes of hypopituitarism?

A
  • Tumours eg pituitary adenoma
  • Trauma/ head injury
  • Hypothalamic disorders
  • Vascular disease
  • Infection – eg meningitis
  • Iatrogenic – eg therapeutic skull irradiation
  • Miscellaneous – eg sarcoidosis, haemochromatosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
200
Q

What is a combined pituitary function test?

A

Destructive lesions of the pituitary tend to present with pituitary hypofunction, thus stimulatory tests are used toassess the ability of the gland to secrete hormones.

In assessing patients with suspected anterior pituitary dysfunction, it is often convenient to test the capacity of the gland to secrete GH, ACTH, TSH and the gonadotrophins in a single procedure – the combined pituitary function test.

This involves measurement of anterior pituitary hormones following adminstration of TRH, GnRH and insulin

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

What is isolated growth hormone deficiency?

A
  • GH deficiency in children is a cause of growth retardation
  • The diagnosis depends on the demonstration of subnormal growth velocity and subnormal serum growth hormone concentrations
  • While a random growth hormone concentration >20mU/L excludes significant deficiency, a low level is not diagnostic
  • GH status can be measured by a number of stimulatory tests
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
202
Q

About pituitary tumours…

A

¥ Pituitary tumours may be functional, producing excess hormone secretion
¥ Prolactin, GH and ACTH secreting tumours are well recognised
¥ Gonadotrophin and TSH secreting tumours are rare
¥ Since the presence of the tumour may give rise to destructionof normal pituitary tissue, decreased production of other hormones should be investigated with the combined pituitary function test

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

What is acromegaly?

A

¥ Acromegaly is the result of excessive GH secretion by a pituitary tumour leading to increased growth of soft tissue/bone increase in size of hands, feet, jaw change in facial appearance
¥ GH concentration in a random serum sample usually raised
¥ Diagnosis confirmed biochemically by demonstrating failure of suppression of GH levels in response to oral glucose
¥ Treatment options – surgery, external irradiation, drugs
¥ Accompanying hypopituitarism requires appropriate treatment with cortisol, sex steroids and thyroxine

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

What are the major functions of the liver?

A

Carbohydrate metabolism
• Gluconeogenesis
• Glycogen synthesis and degredation

Lipid Metabolism
•	Fatty acid synthesis
•	Cholesterol synthesis and excretion
•	Lipoprotein
•	Ketogenesis
•	Vitamin D 25-OH
•	Bile acid synthesis and excretion

Protein metabolism
• Plasma protein
• Ammonia detoxification

Bilirubin metabolism

Hormones
• Conjugation of steroid hormones
• Degradation of peptide hormones

Xenobiotic metabolism

Storage
•	Vitamin A
•	Glycogen
•	B12
•	Iron
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
205
Q

About liver function tests…

A

¥ Liver has a considerable functional reserve, and therefore simple tests of liver function as bilirubin or albumin concentrations are insensitive indicators of disease
¥ Tests reflecting liver cell damage are more sensitive, e.g., hepatic enzymes in plasma
¥ True tests of liver function, e.g., bromsulphophthalein (BSP), indocyanin green, caffeine clearance and radio-labelled bile acid uptake and clearance tests are available for research purposes
Biochemical tests usually reflect the basic pathological processes common to many conditions, and rarely provide a precise diagnosis, However, they are cheap, non-invasive, and can direct other investigations, e.g., liver biopsy and imaging. They are useful in detecting the presence of a liver disease and in monitoring progress

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

What are the objectives of liver function tests?

A
  • Aid in establishing liver disease by showing abnormal values
  • Inform or help specific diagnosis by showing different patterns
  • Establish the severity of liver damage and provide prognostic insight
  • Monitor disease progression
  • Assess response to therapy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
207
Q

About bilirubin and urobilinogen (bile pigments)…

A

o Derived from the break down of haem of senescent PBCs, and to a lesser extent from myoglobin, cytochromes and some enzymes
o Being water insoluble, it is transported in the plasma to the liver by albumin. At this stage, bilirubin is unconjugated and can not pass in urine even if present at high level
o Unconjugated bilirubin is taken up by liver cells to be conjugated with glucuronic acid by the enzyme UDP-glucuronyl transferase to produce bilirubin diglucuronide which is water soluble and s called conjugated bilirubin which is then secreted into bile and eventually reaches the intestine where more of it is oxidised by bacteria to urobilinogen and stercobilin
o A small amount of urobilinogen and bilirunin are reabsorbed through entero-hepatic circulation to be secreted in urine

Total bilirubin reference range (RR): 3-17 micromol/l
Plasma bilirubin rises when significant liver damage occurs and causes yellowish discolouration of skin and mucous membranes (Jaundice) which can be clinically detected at bilirubin level of 50 umol/l

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

What is monitoring bilirubin used for?

A
  • Investigate liver diseases and assess subclinical hyperbilirubinaemia
  • Determine the need for exchange transfusion in neonatal jaundice
  • Assess surgical treatment of bile duct stricture
  • Determining correct doses of drugs, e.g., cytotoxic therapy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
209
Q

What is alkaline phosphatase?

A

Alkaline phosphatase (ALP): (RR: 30-130 U/I)

o A group of enzymes that hydrolyse phosphate esters at alkaline pH and are present in liver, bone, intestine and placenta. Plasma ALP increases in the presence of obstruction to the biliary passages or cholestasis. The block could be intrahepatic as in PBC, or due to a stone in the bile duct or pressure by a tumour, e.g., head of pancreas or lymph nodes
o ALP increases in liver and bone disease, with pregnancy, growing children and to enhance the specificity, ALP isoenzymes can be measured using electrophoresis or utilising different thermal stabilities of iso-enzymes, as liver isoenzyme activity drops by 40% after 15 min incubation at 56oC while bone isoenzyme activity decreases only by 15% or gGT is used

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

About alanine aminotransferase (ALT) and aspartate aminotransferase (AST)…

A

Alanine aminotransferase (ALT) and Aspartate aminotransferase (AST)
(RR: 0-40 U/l for both)
o Also called transaminases as they transfer an amino group from an amino acid to a keto acid. They increase in the plasma in presence of liver cell damage.
o They have wide tissue distribution, with AST present in liver, heart, skeletal muscle, kidney, brain, erythrocytes and lung. ALT has also a similar wide tissue distribution but activities are lower in extrahepatic tissues, and increased plasma ALT is more specific for liver disease (CK for muscle and troponins for heart)
o Aminotransferase activity above 10 times the upper limit of RR are most frequently due to hepatocellular damage, e.g., viral or drug induced hepatitis or autoimmune hepatitis
o Aminotransferase activity below 10 times the upper limit of RR, but higher than upper limit of RR are less specific but could occur in chronic liver diseases
o Aminotransferase activities within RR do not exclude liver disease that could be assessed by histological examination of a biopsy specimen
o The higher the AST/ALP ratio, the more likely the underlying condition to be a form of hepatitis. The lower the ratio, the more it is indicative of cholestatic disorder
o An AST/ALT ratio of more than 2 in a patient who clinically appears to have hepatitis strongly suggests that alcohol is involved

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

What is mitochondrial AST?

A

Accounts for 80% of total AST in liver cells and the ratio of mAST/total AST is a good marker for chronic alcohol consumption

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

What is gamma-glutamyl transferase?

A

o A microsomal enzyme that transfers glutamyl groups between peptides and aminoacids and is involved in the transport of peptides across plasma membranes.
o Plasma activity is mainly derived from liver, despite its wide distribution, and it has a poor specificity and high sensitivity for liver disease
o Its measurement helps in linking an elevated ALP to the liver and is elevated with chronic alcohol consumption and is more elevated in the presence of chronic liver disease with alcohol consumption

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

About albumin…

A

Albumin: (RR: 30-50 g/l)
o Synthesised exclusively by liver and its plasma half-life is 21 days. It maintains plasma oncotic pressure, binds calcium, fatty acids, bilirubin, some hormones and drugs.
o Its level tends to fall with progress of chronic liver disease and therefore plays a major prognostic role. It also falls with inadequate protein intake

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

What is prothrombin time (PT)?

A

Measures the rate at which prothrombin is converted to thrombin in the presence of thromboplastin, calcium, fibrinogen, and coagulation factors V, VII and X.

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

What is the international normalised ratio (INR)?

A

INR is the ratio of the PT of the patient to that obtained using a control preparation. Normal individuals have an INR

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

How can PT and INR be affected?

A

In the presence of severe acute or chronic liver damage, the synthesis of clotting proteins by the liver is impaired . Prothrombin half life is 6 hours and therefore, a prolonged INR which can only be improved by fresh plasma transfusion or providing the deficient clotting factors is a good indicator of severe liver damage, e.g., acute liver failure

Vitamin K is a fat soluble vitamin and requires bile salts from liver for absorption. It is also needed for the synthesis of clotting factors by the liver. In case of cholestasis, vitamin K absorption is defective and the synthesis of clotting factors is impaired causing an increased INR which can be corrected within 24 hours by an injection of vitamin K

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

What is the epidemiology of hepatocellular carcinoma?

A

Is relatively common in Southeast Asia, Sub-Saharan Africa, Japan, Pacific Islands and Mediterranean area. UK incidence ranges between 1-7/100 000. Incidence in some tribes in Mozambique is 107/100 000. Individuals who migrate from areas of high incidence to areas of low incidence of HCC have lower incidence compared with the population remaining in lands of origin. Therefore, environmental factors, rather than genetic or racial predisposition, play a key role in pathogenesis of HCC.

In recent years, the incidence of HCC seems to be rising in the West, as a result of improved treatment of alcoholic and other cirrhosis (e.g., after hepatitis B or C) and more prolonged survival. Incidence rates of about 10/100 000 have been reported

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

What is the clinical presentation of hepatocellular carcinoma?

A

In low-incidence populations, the tumour usually presents in a patient who is known to have had cirrhosis or haemochromatosis for some time. In high incidence areas, it appears in individuals who were apparently healthy. However, the majority of affected patients are found to have cirrhosis in non-tumour part of liver

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

What are the symptoms of hepatocellular carcinoma?

A

Usually runs a silent course until advanced where it presents with:
• Upper abdominal discomfort or pain, in the right hypochondrium or epigastrium, radiating to the right sub-scapular region or shoulder
• Weight loss and weakness
• Feeling of fullness in the upper abdomen after meals/ anorexia
• Generalised swelling of the abdomen (due to ascites)
• Nausea, vomiting, constipation
• Peritoneal haemorrhage (acute abdomen due to tumour rupture )
• Bone pain due to skeletal metastasis

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

What are the signs of hepatocellular carcinoma?

A
  • Early: Mild to moderate hepatomegaly, but later: Massive enlargemen, tenderness
  • Hepatic arterial bruit, ascites heard, splenomegaly, jaundice, wasting, fever
  • Stigmata of chronic liver disease: gynaecomastia, caput medusae, feminisation
  • Ectopic Hormonal Syndromes: erythrocytosis, hypercalcaemia, feminisation (dehydroepiandrosterone converted to oestradiol by the tumour)
  • Hypoglycaemia, hypercholesterolaemia, porphyria
  • Elevated a-fetoprotein, carcino-embryonic antigen (CEA), dysfibrinogenaemia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
221
Q

How is hepatocellular carcinoma diagnosed?

A

• Determination of a-fetoprotein: markedly elevated
• Biochemistry: similar to cirrhosis: markedly elevated alkaline phosphatase with normal or slightly elevated bilirubin and transaminases, low albumin and elevated IgG and IgM
Radiologic Investigations: isotope liver scanning (figures), hepatic angiography, ultrasonography (solid vs cysts), CT scans

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

What is the treatment of hepatocellular carcinoma?

A
  • Surgical Resection: partial hepatectomy, hemihepatectomy offer the only chance of cure of HCC. Tumour should be confined to one lobe and the rest of the liver free from cirrhosis or only have mild cirrhosis
  • Liver transplantation
  • Chemotherapy: 5-Flurouracil, cyclophosphamide, methotrexate, vincristine.
  • External irradiation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
223
Q

What is helicobacter pylori infection?

A

H. Pylori infection is the main cause of duodenal or gastric ulcers and plays an important role in gastric cancer.

It can be diagnosed by microscopical examination of biopsy specimens. Alternatively, biopsies could be tested for the presence of urease, which is specific for H. Pylori, in the presence of an indicator that changes colour with the release of ammonia in case of positive biopsies. Antibodies for H. Pylori can also be checked but C13 or C14 urea breath test of PCR are preferred

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

What are the pancreatic functions?

A

Exocrine:
• Alkaline secretion
• Enzymes: amylase, lipase, trypsin, chymotrypsin, carboxypeptidases, elastase, esterases, phospholipases
• Bile salts and lipases are essential for fat digestion and absorption and absorption of fat soluble vitamins A, D, E, K

Endocrine:
• Insulin
• Glucagon
• Somatostatin

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

What is acute pancreatitis and its aetiology?

A

Pain: upper abdominal, radiates to the back
Enzymes marked elevation of amylase and lipase in blood and urine

Aetiology
•	Alcohol
•	Gallstones
•	Post-operative
•	Hyperlipidaemia
•	Traumatic
•	Carcinoma
•	Drugs: steroids, azathioprine, diuretics
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
226
Q

What are the investigations of acute pancreatitis?

A
Biochemistry:
•	serum or urine amylase or lipase (high)
•	serum calcium (low)
•	falling arterial O2 tension
•	coagulopathy
Radiology and imaging (figures)
•	plain radiography
•	ultrasonography
•	computerized tomography (CT)
•	ERCP
•	PTC
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
227
Q

What are gallstones?

A

affects 10-20% of world population

  1. Cholesterol stones: >70% cholesterone, mucoprotein ± calcium
    • Supersaturation
    • Nucleation
    • GB motor dysfunction
  2. Pigment stones: calcium bilirubinate, cholesterol, calcium soaps and mucoprotein matrix
    • Bacterial beta-glucuronidase
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
228
Q

What is the natural history of gall stones?

A
  • Symptomless
  • Biliary obstruction
  • Cholangitis
  • Cholecystitis
  • Jaundice
  • Intestinal obstruction
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
229
Q

What investigations might be done into gall stones?

A
  • Polymorphonuclear leukocytosis
  • Increased alkaline phosphatase
  • Elevated bilirubin
  • Increased amylase

Abdominal radiograph: calcified gallstone
Ultrasonography: gallstones in GB

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

What is intrahepatic cholestasis?

A

Hepatocellular:

  • Viral or alcoholic hepatitis
  • Sex hormones and pregnancy

Biliary:

  • Intrahepatic atresia
  • Primary biliary cirrhosis
  • Sclerosin cholangitis
  • Malignancy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
231
Q

About carcinoma of the pancreas and what are the risk factors?

A

The incidence has doubled in UK over the past 50 years to 10/100 000/year. It is twice as high in men than in women, and over 80% of cases occur in the 60-80 y age group.

Risk Factors:
• Tobacco: cigarette smoking is associated with PC, with a relative risk of 2-3 over non-smokers.
• Diet: Increased animal fat, combined with increased alcohol consumption, are associated with increased incidence of PC. The fish oil fatty acids (eicosapentaenoic and docosahexaenoic acids) seem to antagonise the carcinogenic effect. Vegetables and fruit are protective
• Coffee: prolonged intake of higher amounts of coffee might be linked to PC
• Chronic Pancreatitis
• Diabetes mellitus
• Gastric resection

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

What are the signs and symptoms of carcinoma of the pancreas?

A

Symptoms:

  • Pain: is the presenting feature in 50-80% of cases, usually dull aching or boring pain in the epigastrium and radiates to the back. It is aggravated by food intake and may be worst in recumbent position and eased by sitting.
  • Jaundice: is the 1st symptom in 10-30% of patients and the presenting symptom in 30-65%. It is progressive until releived by surgery or stenting. Pruritus is common.
  • Weight Loss: is the presenting feature in 90% of patients, usually rapid and progressive.
  • Other Symptoms: Fatigue, weakness, epigastric bloating, diarrhoea, steatorrhoea, or constipation. Nausea and vomiting occur in 25% of patients

Signs:

  • Jaundice
  • Hepatomegaly
  • a palpable gallbladder and epigastric tenderness
  • ascities
  • enlarged lymph nodes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
233
Q

What investigations might be done into carcinoma of the pancreas?

A
  • Imaging Techniques: CT scanning, ERCP, PTCA, US, angiography (Figures)
  • Blood Tests: ESR, CRP, Blood count, LFTs, enzymes: amylase, lipase, pancreatic elastase, tumour markers: DU-PAN 2, CEA, CA 19-9, CA 50
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
234
Q

What are treatments for carcinoma of the pancreas?

A
  • Pre-operative biliary drainage
  • Palliative surgery
  • Curative resection: prognosis and complications
  • Chemotherapy: 5-flurouracil, cyclophosphamide, methotrexate, vincristine
  • Radiotherapy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
235
Q

What biochemical tests are done in clinical medicine?

A
  • Screening (subclinical conditions)
  • Diagnosis (normal vs abnormal values)
  • Monitoring (course of disease)
  • Clinical management (treatment/response)
  • Prognosis (risk stratification)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
236
Q

What are cardiac markers?

A
  • Located in the myocardium
  • Released in response to cardiac overload
  • Released in response to cardiac injury
  • Released in response to cardiac failure
  • Can be measured in blood samples
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
237
Q

What is the classification of laboratory tests in cardiac disease?

A
  • Markers of risk factors for development of coronary artery disease
  • Genetic analysis for candidate genes of risk factors
  • Markers of cardiac tissue damage
  • Markers of myocardial function/overload
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
238
Q

What can biochemical makers of cardiac dysfunction/damage contribute to?

A
  • Rule in/out an acute MI
  • Confirm an old MI
  • Help to define therapy
  • Monitor success of therapy
  • Diagnosis of heart failure
  • Risk stratification of death
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
239
Q

What are the characteristics of the ideal cardiac marker?

A

Analytical characteristics

  • Measureable by cost-effective method
  • Simple to perform
  • Rapid turnaround time
  • Sufficient precision and accuracy
  • Reasonable cost

Clinical Characteristics

  • Early detection of disease
  • Sensitivity vs specificity
  • Validated decision limits
  • Selection of therapy
  • Risk stratification
  • Prognostic value
  • Ability to improve patient outcome
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
240
Q

Why is it important to define the type of IHD?

A

Stable angina vs acute myocardial infarction

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

What are some possible causes of chest pain?

A
  • Broken rib
  • Collapsed lung
  • Nerve infection (shingles)
  • ‘pulled’ muscle
  • infection
  • heart burn (hernia)
  • pericarditis
  • blood clot in the lungs (PE)
  • angina
  • myocardial infarction
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
242
Q

How might we assess IHD?

A
  • medical history
  • risk factors
  • presenting signs and symptoms
  • ECG
  • Biomarkers
  • Imaging/scans
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
243
Q

How does ECG and biomarkers define the type of acute coronary syndrome?

A

In general, an ST elevation MI is caused by complete obstruction of a coronary artery, and causes damage that involves the full thickness of the heart muscle.

A non-ST elevation MI is caused by partial obstruction of a coronary artery, and causes damage that does not involve the full thickness of the heart wall

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
244
Q

What is myocardial injury?

A
  • Irreversible injury typically requires 30 minutes of ischaemia
  • High risk that 80% of cardiac cells die within 3 hours and almost 100% by 6 hours
  • Cellular content leak out through membrane dependent on size and solubility
  • Concentration gradient from inside to outside important (high gradient improves detection of early damage)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
245
Q

What are markers of myocardial damage?

A
  • 7-36h peak after MI
  • heart muscle specific markers troponin-T and troponin-I
  • creatinine kinase (increase by 90% MIs, but less specific as also released from skeletal muscle)
  • heart specific isoforms of creatinine phosphokinase (CPK-MB)
  • myoglobin raised early but less specific for heart damage
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
246
Q

What are troponins?

A

The troponin complex is a component of the thin filaments in striated muscle complexed to actin.

There are three types of troponins:
o Troponin T (tropomyosin binding)
o Troponin I (inhibits actomyosin ATPase)
o Troponin C (calcium binding)

The troponins are three different proteins structurally unrelated with each other.

Cardiac troponin T and I differ significantly from troponin T and I found in skeletal muscle

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
247
Q

What are the advantages of cardiac troponin?

A

Advantages of cardiac troponin:

  • An index of cardiac damage
  • Blood levels related to severity of cardiac damage
  • Predicts major adverse cardiac events such as myocardial infarction
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
248
Q

What are some major causes of heart failure?

A
  • Coronary artery disease
  • Chronic hypertension
  • Cardiomyopathy
  • Heart valve disease
  • Arrhythmias – AF, VT
  • Infective endocarditis
  • Pulmonary hypertension – PE, COPD
  • Alcohol and drugs (eg cocaine)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
249
Q

What are signs and symptoms of congestive heart failure?

A
  • Shortness of breath
  • Swelling of feet and legs
  • Chronic lack of energy
  • Difficulty sleeping at night due to breathing problems
  • Swollen or tender abdomen with loss of appetite
  • Cough with frothy sputum
  • Increased urination at night
  • Confusion and/or impaired memory

Sensitivity and specificity of signs and symptoms of heart failure is relatively poor

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

What is the clinical utilisation of cardiac biomarker testing in heart failure?

A
  • Initial evaluation of heart failure
  • Screening for cardiac dysfunction
  • Guiding management of heart failure
  • Assessment of prognosis and survival

Natiuretic peptides as markers of cardiac damage/overload. An A,B,C of natriuretic peptides

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
251
Q

What are the advantages of N-terminal precursor forms over ANP or BNP?

A

Assays are now available for the active peptides and the N-terminal precursor forms of ANP and BNP.

Advantages of N-terminal precursor forms over ANP or BNP

  • Longer half life
  • Higher plasma concentrations
  • Less sensitive to rapid fluctuations
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
252
Q

When are plasma levels of ANP and BNP markedly raised?

A
  • Congestive heart failure
  • Aortic stenosis
  • Dilated cardiomyopathy
  • Hypertrophic cardiomyopathy
  • Myocardial infarction
  • Chronic renal failure
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
253
Q

What are possible causes for raised plasma natriuretic peptides in heart disease?

A
  • Haemodynamic
  • Structural
  • LVH
  • Ventricular dilation
  • Damage/remodelling after MI
  • Hypoxia/necrosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
254
Q

What are some conditions that are investigated for possible use of plasma ANP/BNP?

A
  • Assessment of severity of congestive heart failure
  • Screening for mild heart failure
  • Monitor response to treatment in congestive heart failure
  • Prognostic outcome/risk stratification
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
255
Q

What are candidate biomarkers in heart failure?

A

Inflammation

  • C-reactive protein
  • Tumour necrosis factor alpha
  • Interleukins 1, 6, 10 and 18

Oxidative Stress

  • Oxidised LDL
  • Myeloperoxidase

Extracellular-matrix Remodelling

  • MMP2, MMP3, MMP9
  • TIMP1
  • Collagen propeptides
  • Galectin-3

Neurohormones

  • Angiotensin II
  • Aldosterone
  • Arginine vasopressin
  • Endothelin-1

Myocyte Injury and Apoptosis

  • Troponins I and T
  • Myosin light-chain kinase I
  • Heart-type fatty-acid binding protein
  • Creatine kinase MB fraction

Myocyte Stress

  • BNP
  • NT-proBNP
  • MR-proANP
256
Q

What do calcium ions affect?

A
  • Neuronal and neuromuscular activity
  • Membrane permeability
  • Activity of many enzymes

The concentration of calcium in ECF is normally kept within closely defined limits

257
Q

Which hormones achieve the actions of calcium?

A
  • Parathyroid hormones

- Calcitriol (the active metabolite of vitamin D)

258
Q

About calcium ions distribution, function etc…

A
  • 1-2kg, 99% mineralized in bone mainly in form of hydroxyapatite
  • ~1% intracellular,
259
Q

About calcium in the body…

A
  • Dietary intake: 400-1200mg/day of which ~200mg is absorbed – increased requirement during pregnancy, lactation, growth
  • Intestinal absorption – in duodenum, stimulated by vitamin D
  • Calcium exchange in bone: ~99% of body calcium is in bone, ~5g in form that is readily exchangeable
  • Renal excretion: ~6mmol/day when renal function is normal, PTH increases reabsorption
  • Plasma: ~50% protein bound (mainly to albumin), ~50% ionized (active form ~1 mmol/L)

1000-12000mg per day. Only a fraction of that is absorbed.

About ½ is free [Ca2+] (physiologically active), half protein bound (mainly albumin)

260
Q

About normal calcium turnover…

A
  • Net gain of calcium from gut
  • Recycle through renal tubule multiple times per day
  • Small net loss of calcium in urine
  • Bone has a huge reservoir of calcium locked in mineralised form
  • Remodeling – can either be absorbed in the bone, during resorption the mineralized bone is digested/dissolved
261
Q

What is parathyroid hormone and how does it act on its target organs?

A

Parathyroid hormone – 84 aa peptide

  • Secreted by chief cells parathyroid gland. Stimulus for parathyroid hormone secretion is low [Ca++], negative feedback system
  • If high parathyroid hormone will not be stimulated, will be suppressed
  • [Ca++] levels monitored by chief cell membrane Ca receptor (GPCR)

Target Organs

  • Kidney: increases calcium reabsorption, decreases phosphate reabsorption, increases synthesis of calcitriol (active form of vitamin D)
  • Bone: increases bone resorption by increasing osteoclast activity, releasing Ca from mineralised bone (by dissolving/digesting bone)
  • Increases calcium absorption (indirect action via vitamin D)
262
Q

About vitamin D…

A

• Fat soluble ‘vitamin’ synthesised in skin in response to exposure to UV (‘sunshine vitamin’)
• Activated by 2 metabolic steps
- 25 hydroxylation in liver to form 25OH D, major circulating metabolite
- 1alpha hydroxylation of 25 OH D in kidney produces calcitriol (1alpha,25(OH)2D3), active hormonal metabolite
- calcitriol increases intestinal absorption of dietary calcium
• it is really a steroid hormone, not a vitamin

263
Q

What is calcitonin and how does it act on its target organs?

A

There are 32 amino acid peptides. They are secreted by C cells of thyroid. Stimulus for secretion is high [Ca++]

Target organs:

  • kidney: decreases calcium and phosphate reabsorption
  • bone: decreases bone resorption by inhibiting osteoclast activity
  • synthetic calcitonin used in treatment of Paget’s disease of bone/osteoporosis
264
Q

What are causes of hypercalcaemia?

A

Major causes:

  • cancer associated eg due to bone metastases
  • primary hyperparathyroidism

Less common causes include:

  • hyperthyroidism
  • excessive intake of vitamin D

In ambulatory patients = primary hyperparathyroidism (excess PTH)
In hospitalised patients = malignancy

265
Q

What are the clinical features of hypercalcaemia?

A

Kidney:

  • renal calcification/stones
  • increased urine volume (polyuria)

Neuromuscular and neurological effects

  • abdominal pain, constipation
  • anorexia, nausea, vomiting, mental changes

Heart:

  • cardiac arrhythmias
  • cardiac arrest
266
Q

What is primary hyperparathyroidism?

A
  • usually due to a benign adenoma
  • most common in post-menopausal women
  • often detected on screening – many patients asymptomatic
  • ~10% of patients present in clinical evidence of bone disease
  • 10-20% of patients present with kidney stones

Doesn’t lead to overt symptoms

267
Q

What types of serum biochemistry can be done?

A
  • serum calcium – modest to marked increase
  • serum phosphate – low or low normal
  • serum alkaline phosphatase raised in ~20% of cases
  • serum creatinine may be elevated in longstanding disease (kidney damage)
  • serum PTH concentration should be interpreted in relation to calcium

Calcium and phosphate handling by the kidney tend to be reciprocal. This is a marker of bone remodelling

268
Q

What is the hypercalcaemia of malignancy?

A

The most common cause of hypercalcaemia in hospitalised patients is malignant disease.

  • PTHrP, not quite ectopic source as is slightly different hormone
  • Do not have to do tests to distinguish between ectopic source and primary hyperparathyroidism
  • Regulation of delicate balance of osteoclasts and osteoblasts
  • Anything favouring osteoclast activity over osteoblast hypercalcaemia as bone breakdown leads to more free calcium
269
Q

What is the treatment of hypercalcaemia of malignancy?

A

• Rehydration/fluid therapy
• Inhibition of bone resorption
- Bisphosphonates (inhibits OCs)
- Calcitonin (inhibits OCs)

270
Q

What are the causes of hypocalcaemia?

A
  • Vitamin D deficiency and disordered vitamin D metabolism (eg renal failure)
  • Hypoparathyroidism/pseudohypoparathyroidism
  • Magnesium deficiency (Mg required for PTH secretion and action on target tissues)
  • Acute pancreatitis ? formation of insoluble calcium salts of fatty acids
  • Spurious – blood collected into EDTA tube

Pseudo = PTH is secreted but problem is with PTH receptor not PTH itself

271
Q

What are the clinical features of hypocalcaemia?

A
•	Numbness
•	Muscle cramps and spasms
•	Convulsions
•	ECG changes
•	Prolonged hypocalcaemia
-	Cataracts
-	Mental disturbances
272
Q

About rickets and osteomalacia…

A
  • Bone disease associated with vitamin D deficiency
  • Rickets – in children, failure of bone mineralisation and disordered cartilage formation
  • Osteomalacia – in adults, impaired bone mineralisation

Rickets occur in childhood, osteomalacia occurs in adulthood

273
Q

How does sunlight act as a source of vitamin D?

A
  • Adequate supplies of vitamin D3 can be synthesised with sufficient exposure to solar ultraviolet B radiation
  • Depends on latitude and season
  • Summer sunlight in cape town – 2500 IU vitamin D3 daily
  • Melanin, clothing or sunscreens that absorb UVB will reduce cutaneous production of vitamin D3

The balance between sufficient production of vitamin D and avoiding sun damage. Darker skinned people require more vitamin D in general

274
Q

How is vitamin D metabolised?

A

Synthesis from precursor in skin as well as diet = sources, can make up for one with the other
Intermediate undergoes final processing in kidney – inactive or active form.

Skin, dietary + metabolic (e.g. converting of intermediate enzyme problems) interruptions can all occur and lead to deficiency

275
Q

What are causes of vitamin D deficiency?

A

Deficient synthesis and supply of vitamin D

  • Inadequate intake
  • Limited sunlight exposure

Impaired absorption of vitamin D
- Malabsorption syndromes (eg coeliac disease)

276
Q

Who is at risk of rickets due to ‘nutritional’ deficiency?

A
  • Babies who are bottle fed on non-vitamin supplemented feeds
  • Children who have little sunlight exposure
  • Breast fed babies of mothers with marginal vitamin D status
277
Q

What are the manifestations of rickets?

A
  • Muscle weakness
  • Bowing deformity of long bones
  • Prominence of the costochondral junction (rachitic rosary)
  • On x-ray, generalised demineralisation of bone
278
Q

What are the features of osteomalacia?

A
•	Diffuse bone pain
•	Waddling gait, muscle weakness
•	On x-ray, stress fractures
•	Serum biochemistry:
-	Low/normal calcium
-	Hypophosphataemia
-	Raised alkaline phosphatase
-	Secondary hyperparathyroidism
279
Q

What is privation osteomalacia?

A

Diet vs sunlight as a source of vitamin D:
• Limited exposure to UV light leads to vitamin D deficiency
• Limited UV exposure is necessary but insufficient in itself to induce the biochemical, radiological and clinical expression of privational osteomalacia
• In the presence of limited UV exposure, dietary factors are the major determinants of privational osteomalacia
• Major dietary risk factors are low or absent meat intake and high intake of wholemeal cereals

280
Q

\what acquired/inherited abnormalities can occur in vitamin D metabolism?

A

Failure of 25 hydroxylation of vitamin D

  • Chronic liver disease
  • Anticonvulsant therapy

Failure of 1 hydroxylation of 25 OH-vitamin D

  • Chronic renal failure
  • Vitamin D dependent rickets type I

Others

  • Vitamin D resistant rickets
  • Vitamin D dependent rickets type II
281
Q

What is renal osteodystrophy?

A
  • Hypocalcaemia is common in patients with end stage renal disease – primarily due to decreased synthesis of calcitriol
  • Hypocalcaemia stimulated PTH secretion (secondary hyperparathyroidism) which also has detrimental effects on the skeleton
  • Increased [H+] (metabolism acidosis) may increase loss of mineral from bone
282
Q

About hypoparathyroidism…

A

• May be congenital or acquired (eg following thyroid surgery, autoimmune disease)
• Serum biochemistry
- Calcium decreased, phosphate increased, low PTH, alkaline phosphatase normal
• May be treated with vitamin D (or an analogue) with or without calcium supplements

283
Q

What are the causes of hypophosphataemia?

A
  • Primary hyperparathyroidism
  • Vitamin D deficiency
  • Persistent hypophosphataemia may be observed during intravenous nutrition
  • Severe hypophosphataemia (0.2-0.3mmol/L) may causes neuromuscular irritability, confusion and hyperventilation
284
Q

What are disorders associated with normal serum calcium concentration?

A
  • Paget’s disease of bone (generalised increase in bone turnover)
  • Osteoporosis
  • Medullary carcinoma of the thyroid (increased circulating concentrations of calcitonin)
285
Q

What are causes of osteoporosis?

A

• Aging, especially post menopausal
• Endocrine
- Premature ovarian failure/hypogonasism
- Thyrotoxicosis, Cushing’s, diabetes mellitus
• Drugs
- Glucocorticoids, alcoholism, prolonged heparin treatment
• Others
- Immobilisation, malabsorption of calcium, weightlessness (piezoelectricity)

286
Q

What are the two classifications of immunodeficiency?

A

Primary: defect in immune system
Secondary: caused by non-immune

287
Q

What are the clinical features of immunodeficiency?

A

• Recurrent infections (normal:

288
Q

What are the warning signs of primary immunodeficiency?

A

2 or more of the following:
• 8 or more new ear infections within 1 year
• 2 or more serious sinus infections within 1 year
• 2 or more months on antibiotics with little effect
• 2 or more pneumonias within 1 year
• failure of an infant to gain weight or grow normally
• recurrent, keep skin or organ abscesses
• persistent thrush (mouth/elsewhere on skin) after age 1
• need for intravenous antibiotics to clear infections
• 2 or more deep-seated infections
• a family history of primary immunodeficiency

289
Q

About primary immunodeficiency…

A
  • usually genetic
  • infrequent but can be life-threatening
  • adaptive immune system: T and B cells
  • innate immune system: phagocytes, complement

Frequency: 50% antibody, 30% T cell, 18% phagocytes, 2% complement

290
Q

What are major B lymphocyte disorders?

A
  • x-linked agammaglobulinaemia (Bruton’s disease)
  • common variable immunodeficiency (CVID)
  • selective IgA deficiency
  • IgG2 subclass deficiency
291
Q

What is x-linked agmmaglobulinaemia?

A
  • First described immunodeficiency (1952)
  • Bruton’s disease
  • Defect in btk gene (x chromosome)
  • Encodes Bruton’s tyrosine kinase
  • Block in B-cell development (stop at pre-B cells)
  • Recurrent severe bacterial infections

Investigations

  • All Igs absent/very low
  • B cells absent/low

Treatment

  • IVIg: 200-600mg/kg/month at 2-3 week intervals
  • Or subcutaneous Ig weekly
  • Prompt antibiotic therapy (URI/LRI)
292
Q

What is common variable immunodeficiency?

A
  • Commonest symptomatic Ab deficiency
  • Presentation: any age childhood to old age
  • Peak: early childhood/early adulthood
  • Recurrent bacterial infections (chest, sinuses)
  • Autoimmune problems
  • Usually missed (exclusion diagnosis late diagnosis complications (structural lung/sinus damage)
  • B cells are normal or low
  • One or more Igs low
  • T cells normal; CD4 T cells can be low
  • Treatment: IVIg; antibiotic prophylaxis
293
Q

What are other antibody deficiencies?

A

Selective IgA deficiency:

  • Most common: 1:400-1:800
  • Most cases are asymptomatic; some have respiratory infections/diarrhoea

Specific Ab deficiency with normal Igs:

  • HepB vaccination: 5% do not respond
  • Recurrent bacterial infections (URI/LRI)
294
Q

What are examples of primary immunodeficiencies?

A

Combined Immunodeficiencies:
- Severe combined immune deficiency (SCID)

Predominant T cell disorders:

  • DiGeorge Syndrome
  • Wiskott-Aldrich syndrome
  • Ataxia-telagiectasia
295
Q

What is SCID?

A
  • Involves both T and B cells
  • 50% x-linked

Presentation

  • well at birth; problems > 1st month
  • diarrhoea; weight loss; persistent candidiasis
  • severe bacterial/viral infections
  • failure to clear vaccines
  • unusual infections
296
Q

What are causes of SCID?

A
  • RAG-1/RAG-2 defect no T and B cells
  • Common cytokine receptor gamma-chain defect (IL-2R)
  • ADA (adenosine deaminase deficiency)
  • Bare lymphocyte syndrome (MHC I or MHC II)
297
Q

What investigations might be done into SCID?

A
  • Lymphocyte subsets: T, B, NK (% and numbers) low total lymphocyte count SCID sign!
  • Pattern: very low/absent T; normal/absent B cells
  • Igs low
  • T cell function decreased (proliferation, cytokines)
298
Q

What is the treatment of SCID?

A
  • Isolation to prevent further infections
  • Do not give live vaccines
  • Blood products from CMV-negative donors
  • IVIg replacement
  • Treat infections
  • Bone marrow/stem cell transplant

Outcome:
• Dependent on promptness of diagnosis
• Survival >80% (early diagnosis, good donor match, no infections pre-transplant)
• Survival

299
Q

What is DiGeorge Syndrome?

A
  • 22q11 deletion
  • complex array of deelopemntal defects
  • dysmorphic face: cleft palate, low-set ears, fish shaped mouth
  • hypocalcaemia, cardiac abnormalities
  • variable immunodeficiency (absent/reduced thymus affects T cell development)
300
Q

What is Wiskott-Aldrick Syndrome (WAS)?

A
  • x linked
  • defect in WASP (protein involved in actin polymerisation defect in signalling)
  • thrombocytopaemia, eczema, infections
  • progressive immunodeficiency
  • progressive decrease in T cells, decreased T cell proliferation
301
Q

What is ataxia-telangiectasia (AT)?

A
  • AR
  • Defect in cell cycle checkpoint cells very sensitive to ionising radiation
  • Progressive cerebellar ataxia
  • Typical telangiectasia (ear lobes, conjunctivae)
302
Q

What kinds of phagocyte defects are there?

A
  • Quantitative (decreased number)
  • Qualitative
  • Chronic granulomatous disease
  • Chediak-higashi syndrome
  • Leucocyte adhesion defects (LADs)
303
Q

How might phagocyte defects, such as chronic granulomatous disease (CGD), be diagnosed?

A
  • NBT test (nitroblue tetrazolium reduction)

- Flow cytometry assay dihydrorhodamine

304
Q

What is chediak-higashi syndrome?

A

Phagocyte defects

  • Rare genetic disease
  • Defect in LYST gene (regulates lysosome traffic)
  • Neutrophils have defective phagocytosis
  • Repetitive, severe infections

Diagnosis:

  • Decreased number neutrophils
  • Neutrophils have giant granules
305
Q

What is leucocyte adhesion deficiency (LAD)?

A
  • Defect in beta2-chain integrins (LFA-1, Mac-1)
  • Delayed umbilical cord separation diagnosis
  • Skin infections, intestinal and perianal ulcers
  • Decreased neutrophil chemotaxis
  • Decreased integrins on phagocytes (flow cytometry)
306
Q

What are complement deficiencies?

A
  • Recurrent infections (Neisseia – terminal complex C5, C6, C7, C8 and C9 deficiency)
  • Severe/fatal pyogenic infections (C3 deficiency)
  • SLE-like syndrome (C1q, C2, C4, deficiency)
  • Hereditary angioneurotic edema: failure to inactive complement (deficiency in C1 inhibitor); intermittent acute edema skin/mucosa vomiting, diarhhoea, airway obstruction

Investigations:

  • Complement function: CH50 (haemolysis)
  • Measure individual components
307
Q

What are the aims of treatment of PID?

A
  • Minimise/control infection
  • Replace defective/absent component of IS
  • Prompt treatment of infection
  • Prevention of infection: isolation, antibiotic prophyaxis, vaccination (not live vaccines!)
  • Nutrition
308
Q

What are causes/risk factors of secondary immunodeficiency?

A
  • Infections: viral, bacterial
  • Malignancy
  • Extremes of age
  • Nutrition
  • Chronic renal disease
  • Splenectomy

Infections:

  • Viral: HIV, CMV, EBV, measles, influenza
  • Chronic bacterial: TB, leprosy
  • Chronic parasitic: malaria, leishmaniasis

Malignancy:

  • Myeloma
  • Lymphoma (Hodgkin’s, non-Hodgkin’s)

Extremes of Age

Prematurity:
- Infants

309
Q

About bone marrow transplantation…

A
  • Reconstruction of full haematopoietic system by transfer of pluripotent stem cells
  • Used to replace defective, absent, or malignant cells
310
Q

What are the clinical indications in PID?

A
  • SCID
  • Combined ID (WAS, DiGoerge)
  • Phagocyte defects
  • Lymphomas, leukaemias, myeloma
311
Q

What are types of bone marrow transplantation?

A

By donor source:
• Allogeneic – genotypically matched individuals (sibling, matched unrelated donor (MUD))
• Autologous – patient is the source

By site of harvest:
• Puncture and aspiration of BM
• Peripheral stem cells (CD34+): from the blood
• Cord blood stem cells (CD34+): umbilical cord

312
Q

What complications can occur from bone marrow transplantation?

A
  • Infection (CMV, EBV, adenocirus)
  • Graft failure (increases with poor matching)
  • Graft vs host disease (GvHD)
313
Q

What is GvHD (graft vs host disease)?

A
  • Transplanted immunocompetent T cells of donor respond against recipient
  • Acute (rare now improved matching)
  • Chronic (main causes of death in BMT)
  • Graft vs tumour (leukaemia)

Prevention:

  • T cell depletion
  • Immunosuppressive drugs
314
Q

How can gene therapy be used in immunodeficiency?

A
  • Transfer of defective genes into cells
  • Used in ADA-SCID, X-linked SCID suspended: acute leukaemia I some patients due to insertion of retroviral vector into known oncogenes
  • Development of self-inactivating lentiviral vectors to reduce risk of insertional mutagenesis (some success for ADA-SCID)
315
Q

What is the difference between congenital and inborn?

A
  • Congenital (present at birth)

* Inborn (transmitted through the gametes)

316
Q

What is alkaptonuria?

A
  • AR
  • Urine turns black on standing (alkalinisation)
  • Black ochrontic pigmentation of cartilage and collagenous tissue
  • Arthritis
  • (homogentisic acid oxidase def.)
317
Q

What is cystinuria?

A
  • AR
  • 1:7,000
  • defective transport of cysteine and dibasic aa’s through epithelial cells of renal tubule and intestinal tract
  • cysteine has low solubility – formation of calculi in renal tract
  • COLA or COAL
  • Mutations of SLC3A1 aa transporter gene (Chr 2p) and SLC7A9 (Chr 19)
318
Q

What is albinism?

A

• Tyrosinase negative
- Type 1a – complete lack of enzyme activity due to production of inactive tyrosinase
- Type 1b – reduced activity of tyrosinase
• Tyrosinase positive
- Type II:
- AR, biosynthesis of melanin reduced in skin, hair and eyes
- Most individuals do acquire a small amount of pigment with age

319
Q

What is pentosuria?

A
  • Excrete 1-4g pentose sugar L-xylulose daily (reducing sugar)
  • Benign
  • Almost exclusively Ashkenazi Jews of Polish-Russian extraction (1:2,500 births)
320
Q

What is the one gene-one enzyme concept?

A

Beadle and Tatum 1945 (Nobel prize 1958)

  • All biochemical processes in all organisms are under genetic control
  • These biochemical processes are resolvable into a series of stepwise reactions
  • Each biochemical reaction is under the ultimate control of a different single gene
  • Mutation of a single gene results in an alteration in the ability of the cell to carry out a single primary chemical reaction
321
Q

What is the molecule disease concept?

A

Pauling et al 1949, Ingram 1956

  • Work on haemoglobin in sickle cell disease
  • Direct evidence that human gene mutations actually produce an alteration in the primary structure of proteins
  • Inborn errors of metabolism are caused by mutations in genes which then produce abnormal proteins whose functional activities are altered
322
Q

What is autosomal recessive inheritance?

A
  • Both parents carry a mutation affecting the same gene
  • 1 in 4 risk each pregnancy
  • consanguinity increases risk of autosomal recessive conditions
  • examples: cystic fibrosis, sickle cell disease
323
Q

What is autosomal dominant inheritance?

A
  • rare in IEMs

- examples: Huntingdon disease, Marfan’s, familial hypercholesterolaemia

324
Q

What is x-linked inheritance?

A
  • characterised by carrier females passing on condition to their affected sons
  • no male to male transmission
  • female carriers may manifest condition – lyonisation (random inactivation of one fo the x chromosomes)
  • x-linked dominant: fragile x
  • x-linked recessive: haemophilia, Fabry’s disease
325
Q

What is codominant inheritance?

A
  • two different versions (alleles) of a gene are expressed, and each version makes a slightly different protein. Both alleles influence the genetic trait or determine the characteristics of the genetic condition
  • example: ABO blood group, alpha1AT
326
Q

What is mitochondrial inheritance?

A
  • mitochondrial DNA
  • inherited exclusively from mother
    o only the egg contributes mitochondria to the developing embryo
    o only females can pass on mitochrondrial mutations to their children
    o fathers do NOT pass these disorders to their daughters or sons
  • affects both male and female offspring
  • distribution of affected mitochondria determine presentation
  • high energy requiring organs more frequently affected
  • current debate on three parent babies
327
Q

What are inborn errors of metabolism (IEMs)?

A
  • inborn errors of metabolism or inherited metabolic disease (IMD)
  • individually rare (1:10,000 – 1:500,000)
  • collectively present sizeable problem
  • important to recognise in sick neonate
  • screening programmes
328
Q

What is the frequency of IEMs?

A
  • genetic disease individually rare but cumulatively frequent and account for approximately 42% of deaths within the first year of life
  • they make a significant contribution to the 1% of children of school age with physical handicap and the 0.3% with severe learning difficulties
329
Q

What is the presentation of IEMs?

A
Neonate to adult
•	neonatal presentation often acute
-	maple syrup urine disease
-	tyrosinaemia
-	OTC (urea cycle defect)
•	Adult
-	Wilson’s
-	Haemochromatosis
Neonatal Presentation
Often caused by defects in energy metabolism
-	Amino acid metabolism
-	Organic acid metabolism
-	Carbohydrate metabolism
-	Urea cycle defects
-	Respiratory chain defects
330
Q

What is the criteria for screening for IEMs?

A
  • Condition should be an important health problem
  • Natural history of the condition should be understood
  • There should be a recognisable latent or early symptomatic stage
  • There should be a test that is easy to perform and interpret, acceptable, accurate, reliable, sensitive and specific
  • There should be an accepted treatment recognised for the disease
  • Treatment should be more effective if started early
  • There should be a policy on who should be treated
  • Diagnosis and treatment should be cost effective
  • Case-finding should be a continuous process
331
Q

What are the WHO criteria for a good screening test?

A

¥ condition screened for should be an important one
¥ there should be an acceptable treatment for patients with the disease
¥ facilities for diagnosis and treatment should be available
¥ there should be a recognised latent or early symptomatic stage
¥ there should be a suitable test or examination which has few false positives -specificity - and few false negatives - sensitivity
¥ test or examination should be acceptable to the population
¥ cost, including diagnosis and subsequent treatment, should be economically balanced in relation to expenditure on medical care as a whole

332
Q

What is newborn blood spot screening?

A
¥	Initial National programme
Ð	PKU
Ð	Congenital hypothyroidism
¥	Extended to include
Ð	Cystic fibrosis
Ð	MCADD
Ð	Haemoglobinopathies
¥	From 2015, the screening in England expanded to include four additional conditions
Ð	Maple syrup urine disease (MSUD)
Ð	Homocystinuria (pyridoxine unresponsive) (HCU)
Ð	Isovaleric acidaemia (IVA)
Ð	Glutaric aciduria type 1 (GA1)
333
Q

How are blood spots taken for screening?

A
  • Samples should be taken on day 5 (day of birth is day 0)
  • All four circles on card need to be completely filled with a single drop of blood which soaks through to the back of the Guthrie card
334
Q

About neonates with IEMs…

A

¥ Most are born at term with normal birthweight and no abnormal features
¥ Symptoms frequently in the first week of life when starting full milk feeds
¥ clues for IEMs
Ð Consanguinity
Ð FH of similar illness in sibs or unexplained deaths
Ð Infant who was well at birth but starts to deteriorate for no obvious reason

335
Q

What is the classic presentation of IEMs?

A

¥ Full term pregnancy
¥ Symtoms - can be very non-specific
Ð Poor feeding, Lethargy, Vomiting, Hypotonia, Fits
¥ or Specific
Ð Abnormal smell (sweet, musty, cabbage-like)
Ð Cataracts
Ð Hyperventilation 2 to metabolic acidosis
Ð Hyponatraemia and ambiguous genitalia
Ð Neurological dysfunction with respiratory alkalosis

336
Q

What might be other clinical scenarios of IEMs?

A
¥	Biochemical abnormalities
Ð	Hypoglycaemia
Ð	Hyperammonaemia
Ð	Unexplained metabolic acidosis / ketoacidosis
Ð	Lactic acidosis
¥	Clinical
Ð	Cognitive decline
Ð	Epileptic encephalopathy
Ð	Floppy baby
Ð	Exercise intolerant
Ð	Cardiomyopathy
Ð	Dysmorphic features
Ð	SUDI
Ð	Fetal hydrops
337
Q

What are first line laboratory investigations of IEMs?

A
  • Blood gas analysis
  • Blood glucose
  • Plasma ammonia
  • Liver function tests
  • Urinary ketones
  • Urine reducing substances
338
Q

What are second line investigations of IEMs?

A
Ð	Plasma & urine amino acids
Ð	Urinary organic acids
Ð	Urinary orotic acid
Ð	Blood acyl carnitines
Ð	Blood lactate and pyruvate
Ð	Urinary glycosaminoglycans
Ð	Plasma very long chain fatty acids
339
Q

What are confirmatory investigations of IEMs?

A
¥	Enzymology
Ð	Red cell galactose-1-phosphate uridyl transferase
Ð	Lysosomal enzyme screening
¥	Biopsy (muscle, liver)
¥	Fibroblast studies
¥	Complementation studies
¥	Mutation analysis
340
Q

What are possible metabolic causes for acute liver disease in neonates?

A
  • Classical galactosaemia
  • Hereditary fructose intolerance
  • An organic acidaemia
  • Tyrosinaemia type 1
341
Q

How much hydrogen ions are produced in normal metabolic processes?

A

40-80 mol hydrogen ions in 24 hours which are excreted in urine

hydrogen ion concentration of ECF maintained within narrow limits of 36-44nmol/l (pH 7.35-7.45)

342
Q

What is produced in aerobic respiration?

A

25,000 mmol/24 hours CO2 which produce carbonic acid H2CO3 and are normally secreted by the lungs

343
Q

What do the kidneys do in acid-base balance?

A

The kidneys are important in acid base balance as they reabsorb all the filtered bicarbonate and synthesise additional bicarbonate to add to blood. They also produce ammonia which is an important mechanism for H+ excretion and also use phosphate to excrete H+.

344
Q

What happens to CO2 in the capillary beds?

A

In the capillary beds, CO2 produced by tissue respiration diffuses into RBCs and forms carbonic acid (Figure) in the presence of carbonate hydratase. Carbonic acid dissociates to form H+ and bicarbonate HCO3-. H+ are buffered by deoxygenated haemoglobin while HCO3- diffuse outside RBCs in exchange for chloride ions (Cl-) (chloride shift) which maintains electrochemical neutrality. The reverse process happens in the alveolar capillaries and the produced CO2 is excreted into the alveoli.

345
Q

What does hydrogen ion homeostasis depend on?

A

Hydrogen ion homeostasis depends on buffering in the tissues and blood stream, acid excretion by the kidneys and expiration of carbon dioxide through the lungs. The blood hydrogen ion concentration [H+] is directly proportional to the partial pressure of carbon dioxide (pCO2) and inversely proportional to the concentration of bicarbonate [HCO3-]

346
Q

What is the Henderson-Hasselbalch equation say?

A

pH = pKa + [HCO3-] / pCO2

347
Q

What is metabolic (non-respiratory) acidosis?

A

Results from increased production or decreased removal of hydrogen ions (H+) [other than those that are derived from carbonic acid (or CO2)] or both or due to excessive loss of bicarbonate from the body

348
Q

What are causes of metabolic (non-respiratory) acidosis?

A

♣ Increased acid production in diabetic or alcoholic ketoacidosis, lactic acidosis, poisoning by ethanol, methanol, ethylene glycol (antifreeze) or salicylates, or intake of excess acid (parenteral amino acid administration
♣ Decreased H+ excretion as in renal failure, renal tubular acidosis (type 1 or 4), carbonic anhydrase inhibitors (acetazolamide)
♣ Loss of bicarbonate in diarrhoea, ileostomy, gastrointestinal drainage or fistulae (pancreatic, biliary or intestinal), renal tubular acidosis (type 2)

349
Q

What happens to excess hydrogen ions?

A

Excess H+ are buffered by bicarbonate to form carbon dioxide which is lost in expired air. This minimises the rise in H+ concentration (or drop in pH) of the plasma at the expense of a drop in plasma bicarbonate (alkali reserve)
H+ + HCO3- HCO3 CO2 + H2O

Low plasma bicarbonate concentration is a marker for the presence, and a measure of the severity, of metabolic acidosis

350
Q

What is combination effected by?

A

Compensation is effected by hyperventilation which is caused by excess H+ stimulating the respiratory centre. This removes excess CO2 and lowers the excess pCO2 which minimises the increase in H+
[H+ ] = K x pCO2 / [HCO3-]
N.B: Respiratory compensation minimises the rise in [H+ ] but does not completely normalise it as it is the elevated [H+ ] that stimulates the hyperventilation

351
Q

What happens if the cause of acidosis is not corrected?

A

If the cause of acidosis is not corrected, a new steady state is reached with increased [H+], low [HCO3-] and low pCO2

Respiratory compensation is limited if respiratory function is compromised, e.g., COPD, asthma, heart failure

If kidney function is normal, excess H+ can be excreted by the kidney, albeit at a slower rate as compared to the very rapid respiratory component of compensation

352
Q

What is the clinical presentation of metabolic acidosis?

A
  • Increased [H+] causes hyperventilation, hyperkalaemia with ECG changes, increased adrenaline, decreased myocardial contractility and CNS depression
  • Increased pCO2 causes peripheral vasodilatation, headache, bounding pulse, papilloedema, flapping tremors, drowsiness and coma
353
Q

How can complete correction of metabolic acidosis be achieved?

A

Complete correction of metabolic acidosis is achieved by treating the cause, e.g, insulin and fluids for diabetic ketoacidosis or removal of ethylene glycol in poisoning. In severe conditions with [H+ ] of 100 nmol/l or above, iv bicarbonate (1.26%, 150 mmol/l) is given in small volumes and the effect on arterial [H+ ] regularly checked. Careful monitoring of hyperkalaemia and treatment if necessary using glucose and insulin or dialysis

Summary of changes: [H+] elevated (RR 35-46 nmol/l), pH decreased (RR 7.36-7.44), pCO2 decreased (RR 4.5-6 kPa) and [HCO3-] much reduced (RR 22-30 mmol/l)

354
Q

What is respiratory acidosis characterised by?

A

Is characterised by increased pCO2 , increased [H +], decrease in pH and a compensatory increase in bicarbonates:
CO2 + H2O H2CO3 H+ + HCO3-

355
Q

What are causes of respiratory acidosis?

A
  • Airway obstruction as in chronic obstructive airway disease, bronchitis, emphysema, asthma
  • Respiratory centre depression as with anaesthesia, morphine, sedatives, cerebral trauma
  • Neuromuscular diseases: poliomyelitis, motor neurone disease, tetanus, botulism, neurotoxins, Guillain-Barré syndrome
  • Pulmonary diseases: pneumonia, respiratory distress syndrome, pulmonary fibrosis
  • Thoracic wall diseases: kyphoscoliosis, flail chest
356
Q

How can respiratory acidosis be corrected?

A

Respiratory acidosis can be corrected by means that restore pCO2 to normal. If elevated pCO2 persists compensation occurs through increased renal H+ excretion

357
Q

What is a summary of the biochemical changes in respiratory acidosis?

A
  • Acute respiratory acidosis: [H+] elevated (RR 35-46 nmol/l), pH decreased (RR 7.36-7.44), pCO2 increased (RR 4.5-6 kPa) and [HCO3-] slightly increased (RR 22- 30 mmol/l)
  • Chronic respiratory acidosis: [H+] slightly elevated or high-normal (RR 35-46 nmol/l), pH slightly decreased or low-normal (RR 7.36-7.44), pCO2 increased (RR 4.5-6 kPa) and [HCO3-] increased (RR 22-30 mmol/l)
358
Q

What is the management of respiratory acidosis?

A

Management aims at lowering pCO2 and combating hypoxaemia by improving alveolar ventilation. Bronchodilators, antibiotics, physiotherapy and artificial ventilation are useful. Oxygen at high concentration may be safely used in acute respiratory failure. However, this might be dangerous in chronic respiratory failure as the respiratory centre becomes insensitive to CO2 and hypoxia becomes the main stimulus to respiration.

359
Q

What is metabolic (non-respiratory) alkalosis?

A

Is characterised by an increase in extracellular fluid bicarbonate concentration with a consequent reduction in [H+] . Normally, increased plasma bicarbonate leads to incomplete renal tubular reabsorption of bicarbonate and increased excretion in urine.

360
Q

What are the causes of metabolic (non-respiratory) alkalosis?

A
  • ECF volume depletion and hypochloraemia as in gastric aspiration, repeated vomiting with pyloric stenosis, chloride-losing diarrhoea, loop and thiazide diuretics (but not potassium-sparing diuretics). Loss of sodium and water results in the contraction of ECF volume which stimulates renal sodium retention through stimulating renin-angiotensin-aldosterone system and increased excretion of potassium and hydrogen ions.
  • Alkali intake as in chronic antacid intake, over-treating acidosis
  • Potassium depletion due to reduced intake, excessive loss, mineralocorticoid excess as in Cushing syndrome, Conn’s syndrome, Barrter’s syndrome, secondary hyperaldosteronism, drugs like carbenoxolone. Potassium depletion and the loss of intracellular potassium results in intracellular shift of H+

This shift causes extracellular alkalosis. Potassium depletion also causes more hydrogen ions to be exchanged (lost in urine) for the reabsorbed sodium ions in the distal convoluted tubules. In addition, potassium depletion stimulates ammonia formation by the kidney and thus increase acid secretion.

361
Q

What are the biochemical changes in metabolic alkalosis?

A

decreased [H+] (RR 35-46 nmol/l), increased pH (RR 7.36-7.44), increased pCO2 (RR 4.5-6 kPa) and much increased [HCO3-] (RR 22-30 mmol/l

362
Q

What is the compensatory change in metabolic alkalosis?

A

The expected compensatory change is an increase in pCO2 which would increase the pCO2 / [HCO3-] and thus [H+]. A low arterial blood [H+] inhibits the respiratory centre, causes hypoventilation and an increase in pCO2. However, this mechanism is self –limiting as the increased CO2 stimulates respiration and thus it is mainly useful in acute conditions. In more chronic conditions, the respiratory centre becomes less sensitive to CO2. Should hypoventilation lead to significant hypoxaemia, the low pO2 would be a powerful stimulus of respiration and prevent further compensation

363
Q

What is the management of metabolic alkalosis?

A

0.9% (w/v) sodium chloride solution (isotonic saline) is used to expand the contracted ECF and correct hypochloraemia. The corrected hypovolaemia will improve renal perfusion and enhance the excretion of excess bicarbonate. In the presence of potassium depletion, correction of the underlying cause and potassium replacement should be initiated

364
Q

What is respiratory alkalosis?

A

The common feature of this condition is a fall in pCO2 which reduces the ratio of pCO2 to bicarbonate concentration thus reduces [H+].

365
Q

What are the main causes of respiratory alkalosis?

A
  • Hypoxia and increased respiratory drive as occurs in high altitude, anaemia, pulmonary oedema or pulmonary embolism
    • Other causes of increased respiratory drive like cerebral trauma, infection or tumours, respiratory stimulants like salicylates, liver failure, septicaemia, primary or voluntary hyperventilation and mechanical ventilation
366
Q

What are the biochemical features of respiratory alkalosis?

A

In acute respiratory alkalosis: decreased [H+] (RR 35-46 nmol/l), increased pH (RR 7.36-7.44), decreased pCO2 (RR 4.5-6 kPa) and a slight decrease in [HCO3-] (RR 22-30 mmol/l).

In chronic respiratory alkalosis, slightly decreased or low-normal [H+] (RR 35- 46 nmol/l), slightly increased or high-normal pH (RR 7.36-7.44), decreased pCO2 (RR 4.5-6 kPa) and decreased [HCO3-] (RR 22-30 mmol/l)

367
Q

What is the management of respiratory alkalosis?

A

treat the underlying cause and increase inspired pCO2

368
Q

About water balance…

A
  • Water accounts for 60% of body weight in men and 55% in women, 2/3 in intracellular fluid (ICF) and 1/3 in extracellular fluid (ECF), with only 8% in plasma
  • Water distribution is determined by the osmotic contents of these compartments, which are kept equal, except in the kidney. Changes in solute content of a compartment results in water shift, which restores isotonicity.
369
Q

What is osmolality due to?

A
  • Osmolality of ECF is mainly due to sodium (135mmol/l) and its associated anions chloride and bicarbonate, with glucose and urea also contributing. Osmolality of the ICF is mainly determined by potassium (110 mmol/l) and its associated anions phosphates, proteins and sulphates.
  • Osmotic effect of plasma proteins (colloid osmotic pressure or oncotic pressure) is important in determining water distribution between these compartments.
  • Changes in body water content independent of the amount of solute will alter osmolality, which is normally 280-295 mmol/kg of ECF water. Loss of water, as in water deprivation, would increase osmolality and cause water movement from ICF to ECF, thus a slight increase in ECF osmolality occurs. This stimulates thirst centre in the hypothalamus, which promotes a desire to drink, and stimulate the hypothalamic osmoreceptors, which cause the release of ADH (anti-diuretic hormone or vasopressin). This peptide hormone is synthesised in the hypothalamus and transported to the posterior pituitary to be secreted into the blood. It renders renal collecting ducts permeable to water , thus permitting water reabsorption and urine concentration.
370
Q

What happens is ECF osmolality drops?

A

If ECF osmolality falls, there will be no sensation of thirst, vasopressin secretion is inhibited and urine will be dilute, allowing water loss and restoration of normal ECF osmolality. In Diabetes Insipidus, either ADH is not produced by hypothalamus (Cranial DI) or the kidney receptors are resistant to its action (Nephrogenic DI). Very large volume of dilute urine is produced
• Vasopressin can also be stimulated by a decrease in plasma or blood volume of 8-10%, even if osmolality is decreased

371
Q

What are stimulating factors of vasopressin secretion?

A
  • Increased ECF osmolality
  • Severe hypovolaemia
  • Stress
  • Pain
  • Nausea
  • Exercise
  • Drugs: nicotine, morphine, sulphonylureas, carbamazepine, clofibrate, vincristine
372
Q

What are inhibiting factors of vasopressin secretion?

A
  • Decreased ECF osmolality
  • Hypervolaemia
  • Alcohol
373
Q

About sodium distribution…

A

The body of an adult man contains about 300mmol of sodium, 70% of it is freely exchangeable and 30% is complexed in bone.

ECF sodium concentration is 135-145mmol/l, while ICF sodium is 4-10mmol/l. This gradient is maintained by the activity of the Na+/K+-ATPase, which uses the energy of ATP to pump sodium into the outside of the cells in exchange for potassium, both against their own concentration gradient.

Western diet supplies 100-200mmol of sodium in 24h, while the obligatory sodium loss via kidney, skin and guy is less then 10mmol/24h. Excess sodium might contribute to hypertension.

374
Q

About potassium distribution…

A

¥ Potassium is the predominant intracellular cation, 90% of body potassium is freely exchangeable, while the rest is bound in RBCs, bone and brain, and only 2% (50-60mmol) is located in ECF. Plasma potassium concentration is not an accurate index of total body potassium and its reference range is 3.0 - 5.5 mmol/l
¥ Potassium tends to diffuse down its concentration gradient from cells into ECF, but this is efficiently opposed by the pumping action of Na+/K+-ATPase, which transports potassium into cells, in exchange for sodium, against concentration gradient.

375
Q

What does ECF volume depend on?

A

ECF volume depends upon total body sodium content, since water intake and loss is regulated to maintain a constant ECF osmolality, which in turn depends mainly on sodium concentration. Facing a variable intake, sodium balance is maintained by regulation of its renal excretion. Normally 70% of filtered sodium is actively reabsorbed in the proximal convoluted tubules, with further reabsorption in the Loop of Henle, thus less than 5% of filtered sodium reaches the distal convoluted tubules. Fine control of distal tubule sodium excretion depends on the activity of aldosterone, secreted by adrenal cortex in response to activation of renin-angiotensin aldosterone system.

376
Q

What is glomerular filtration?

A

The fluid flitered by the golmerular capillary filtration membrane is similar to blood plasma, with the exception of proteins, as it is virtually protein free. The hydrostatic pressure within the glomerular capillaries is the major force that drives water and solutes out of blood capillaries and into the Bowman’s capsule. It is opposed by oncotic pressure (due to plasma proteins) of the capillary blood.

377
Q

What is proximal tubular transport?

A

In the proximal tubule, 70% of filtered sodium, chloride and water, as well as >90% of potassium, glucose, bicarbonate, calcium, phosphate, amino acids are reabsorbed back to blood, mostly by active transport.

Glucose re-absorption has a maximum capacity, called renal threshold. If the amount of filtered glucose exceeds the renal threshold (because blood glucose is higher than 10mmol/l as occurs in diabetes mellitus), not all glucose can be reabsorbed back to blood and glucose will appear in urine.

378
Q

About the loop of henle and distal tubular transport…

A

The loop of Henle and distal and collecting ducts concentrate urine and conserve water under the influence of ADH. They also finely adjust the composition of urine under the influence of aldosterone, which stimulates further sodium reabsorption in exchange for potassium or hydrogen ions.

Hydrogen ions (acids) are excreted in the distal tubule, which thus contributes to the regulation of acid-base balance
Waste products like creatinine, urea, uric acid and organic acids are excreted through filtration at the glomerulous and secretion by tubules
379
Q

What are the different renal function tests?

A
  • plasma creatinine and creatinine clearance
  • blood urea nitrogen
  • urine analysis
380
Q

What is plasma creatinine and creatinine clearance test?

A

Creatinine is a metabolite of skeletal muscles and is excreted by the kidneys. In renal damage, creatinine concentration in the blood is increased above the upper reference level of 140umol/l. Elevated creatinine level indicates a decreased GFR (glomerular filtration rate). Creatinine clearance is calculated from creatinine level in blood and urine and volume of urine per minute. Monitoring creatinine clearance allows monitoring the progress of kidney disease and response to therapy estimating the time at which dialysis or renal transplantation would be required.

381
Q

What is the blood urea nitrogen test?

A

Reflects glomerular filtration and urine concentrating ability. Its level increases as GFR decreases. It also rises in states of dehydration or hypovolaemia as it is reabsorbed back to blood. It is also sensitive to protein intake. The normal reference range in the plasma is 3-7 mmol/l

382
Q

What is the urine analysis test?

A

Physical properties of urine, e.g., colour, aspect, odour, pH, volume, specific gravity; chemical properties as presence of albumin, glucose, acetone, nitrates, as well as microscopic picture e.g., pus cells, red cells, crystals, casts, epithelial cells, parasites ova are useful in diagnosis and management of renal diseases

383
Q

What is atrial natriuretic peptide (ANP)?

A

Secreted by cardiac atria in response to atrial stretch following a rise in atrial pressure. ANP acts directly by inhibiting distal tubular sodium reabsorption and through inhibiting renin-angiotensin-aldosterone system. ANP also antagonises the pressor effects of noradrenaline and angiotensin II and causing vasodilatation.

384
Q

What are causes of water depletion?

A
  1. increased loss from the kidneys as in renal disease, diabetes insipidus (DI), diabetes mellitus, diuretics
  2. increased loss from skin (sweating), lungs (hyperventilation), gut (diarrhoea or vomiting)
  3. decreased intake as in unconsciousness, old age, dementia or infancy
385
Q

What is the clinical presentation of water depletion?

A
  • Thirst
  • dry mouth
  • difficult swallowing
  • weakness
  • confusion
  • weight loss
  • low urine volume (except in DI)

Loss of water from ECF increases osmolality and causes movement of water from ICF to ECF, thus moderating the increased osmolality. Thirst centre is stimulated and vasopressin (ADH) secreted. Urine becomes concentrated and of low volume, except in renal disease or DI. Brain dehydration, haemorrhage or oedema may occur.

386
Q

What are causes of water excess (SAIDH)?

A
  1. Increased intake as in compulsive water drinking, excessive parenteral fluid, water absorption during bladder irrigation
  2. Decreased excretion: renal failure, cortisol deficiency, ectopic ADH secretion (bronchial or prostate carcinoma) or inappropriate ADH secretion (pneumonia, TB, head injury, brain tumours, pain, drugs as chlorpropamide, oxytocin). Clinical picture: Confusion, headache, behavioural disturbances, convulsions, muscle twitching, coma
387
Q

What are causes of sodium excess?

A
  1. increased intake as in excessive parenteral administration, absorption from saline emetics
  2. decreased excretion as in renal failure, primary aldosterone excess as in Cushing’s syndrome, secondary aldosterone excess as in heart failure, liver cirrhosis
388
Q

What are the clinical features of sodium excess?

A
  • shortness of breath (dyspnoea)
  • peripheral oedema
  • venous congestion
  • hypertension
  • weight gain
  • pulmonary oedema

These symptoms and signs are due to expansion of ECF volume.

389
Q

What is the management of sodium excess?

A

treat the cause, diuretics, control sodium intake

390
Q

How does the kidney achieve potassium homeostasis?

A

In the kidneys, filtered potassium is almost completely reabsorbed in the proximal tubules. Some active secretion occurs in the most distal part of the distal convoluted tubules, but potassium excretion is mainly a passive process that, together with hydrogen ions, counterbalance the active sodium reabsorption. Aldosterone stimulates potassium excretion directly at the distal tubule, and indirectly by increasing active sodium reabsorption in distal tubules and collecting ducts. Aldosterone secretion from the adrenal cortex is stimulated by renin-angiotensin system and by hyperkalaemia.

Both potassium and hydrogen ions can neutralise the membrane potential generated by active sodium reabsorption. In acidosis (a lot of hydrogen ions compete with potassium), potassium excretion decreases and its blood level rises (hyperkalaemia). In alkalosis (low hydrogen ion concentration), potassium excretion increases and its blood level decreases (hypokalaemia). Potassium depletion causes alkalosis.

391
Q

How does the gut achieve potassium homeostasis?

A

Potassium is excreted in gastric juice, and much of this, along with dietary potassium, is reabsorbed in the small intestine. In colon and rectum, potassium is secreted in exchange for sodium, partly under the control of aldosterone. Considerable amounts of potassium can be lost in stools in patients with chronic diarrhoea or fistulae, or in patients with persistent vomiting or nasogastric aspiration

The cellular uptake of potassium is stimulated by insulin, in exchange for sodium co-transported with glucose. Aggressive insulin treatment of high blood glucose as in non-ketotic hyperglycaemic coma might induce hypokalaemia. Conditions of rapid cell death or membrane damage, as in leukemia, lymphoma or crush accidents cause hyperkalaemia.

392
Q

What are the cause of hypokalaemia?

A

Normal dietary intake is 60-200 mmol/24h, prevalent in many foods.

Causes of hypokalaemia:

  1. decreased intake: parenteral or oral
  2. transcellular movement: alkalosis, insulin therapy, rapid cell proliferation, b-adrenergic agonists
  3. increased renal excretion as with diuretics, acute renal failure, aldosterone excess, Cushing’s syndrome, liquorice
  4. increased extra-renal excretion as in diarrhoea, purgative abuse, rectal adenoma, vomiting, gastric aspiration, entero-cutaneous fistulae, sweating
393
Q

What are the clinical features of hypokalaemia?

A
  • Neuromuscular: hypotonia and weakness, constipation, ileus, confusion, depression.
  • Cardiac: arrhythmia, ECG changes, digoxin toxicity.
  • Renal: polyuria and polydipsia.
  • Metabolic: alkalosis
394
Q

What is the management of hypokalaemia?

A

Plasma concentration of 3 mmol/l implies a deficit of 300 mmol, almost entirely from ICF. Replacement should be given with care, particularly when the iv route is used, with a rate of 20 mmol/h, and a total of 140 mmol/24h

395
Q

What are the causes of hyperkalaemia?

A
  1. haemolysed or delayed separation sample
  2. increased intake in parenteral infusion, or orally especially with potassium-sparing diuretics, transfusion of stored blood
  3. transcellular movement as in tissue damage, catabolic states, insulin lack, acidosis
  4. decreased excretion as in renal failure, angiotensin converting enzyme inhibitors, aldosterone deficiency as in Addison’s disease and adrenalectomy
396
Q

What are the clinical features of hyperkalaemia?

A

Can kill without warning through ventricular fibrillation and cardiac arrest. ECG changes may precede ventricular fibrillation

397
Q

What is the management of hyperkalaemia?

A
  • calcium gluconate: 10 ml of 10% solution iv over 1 minute, and repeated as necessary.
  • iv glucose and insulin
  • dialysis
  • restriction of intake
  • oral ion exchange resin
398
Q

What is hypersensitivity?

A

An inappropriate immune response to non-infectious antigens that results in tissue damage and disease

399
Q

What are the types of hypersensitivity?

A

1 - immediate hypersensitivity
2 - cytotoxic hypersensitivity
3 - serum sickness and Arthurs reaction
4 - delayed-type hypersensitivity, contact dermatitis

400
Q

What is an example of the immediate hypersensitivity reaction occurring?

A
  • Prick on the skin and then inject the allergen
  • If the person has mast cells specific IgE to that allergen then they get a local inflammatory response
  • They get a wheal, with oedema into the extracellular spaces and redness

Examples of diseases associated with Type-I Hypersensitivity

  • Allergen test/skin prick test
  • Infantile eczema
  • Asthma
  • Hayfever
401
Q

What is systemic anaphylaxis?

A
  • Swelling of mast cells in tissues
  • When it becomes systemic it can be life threatening
  • Anaphylaxis
  • Epi-pens for this reason
402
Q

What are type II hypersensitivity reactions?

A

Respond to altered components of human cells

  • cytotoxic hypersensitivity
  • due to antigens on surface of cells such as red cells
  • could be a drug such as penicillin
  • immune response recognised altered allergen on surface and announces immune response against it
  • killing of the cells that have encountered it and haemolytic anaemia
403
Q

What happens in cytotoxic hypersensitivity?

A
  • produce IgG

- when exposed to the thing again the cells are killed and degraded

404
Q

What is the special case of a type II response?

A
  • involved IgG antibodies directed at cell-surface receptors
  • these antibodies disrupt the normal functions of the receptor by either uncontrollable activation or blocking receptor function
  • this has been renamed to type V
405
Q

What are examples of type II HS?

A

GRAVES DISEASE: production og autoimmune Ab which stimulates thyroxin receptor uncontrollably, get lots of thyroxin

MYASTHENIA GRAVIS: relating to production of autoantibodies which block receptor for Ach causing paralysis as ACh can’t stimulate the muscular cells

406
Q

What is haemolytic disease of the new born (Type II HS)?

A

This relates to rhesus. If the mother is R- and the baby is R+ they can get an immune response against the antigen.

If this happens in the first pregnancy there wold be no reaction to the foetus until after birth when the chorion has broken, introducing it to the mother. The second baby gets crossing of the Ab across the placenta and get haemolysis of red cells and can get haemolytic disease of the newborn.

This can be prevented by giving an antigen immediately after birth and this will mop up all the foetal red cells preventing the mother becoming sensitised to the antigen.

407
Q

What is type III HS - serum sickness/Arthurs reaction?

A

Specific IgG binds to soluble antigen forming immune complexes in the circulation. Excess of one or another leads to development of immune complexes.

408
Q

What causes the Arthurs reaction following diphtheria/tetanus vaccination?

A

A tetanus booster can cause this – it is a strong local reaction relating to development of Arthurs reaction.

  • Activate mast cells to release inflammatory mediators
  • Inflammatory cells invade the site, and blood vessel permeability and blood flow are increased
  • Platelets also accumulate, leading to occlusion of the small blood vessels, haemorrhage, and the appearance of purpura.
409
Q

What is serum sickness?

A
  • caused by large intravenous doses of soluble antigens (e.g. drugs)
  • IgG antibodies produced form small immune complexes with the antigen in excess.
  • immune complexes deposited in tissues e.g. blood vessel walls.
  • tissue damage is caused by complement activation and the subsequent inflammatory responses

this is an example of T3 hypersensitivity if we inject a large amount of immunoglobulin into the circulation. If it hangs around then we get an immune response against it, and if it is cleared away then we get an immune response at the second injection. Complement is stimulated and we get inflammation.

Serum sickness following antivenom

  • most antivenom created from horses
  • snake bite get horse protein injected into you and can get serum sickness to that protein in circulation leading to local vasculitis
410
Q

What is Farmer’s lung?

A
  • type 3 hypersensitivity example
  • exposed to moulds in hay
  • sensitisation in lungs development of immune complexes
  • interstitial pneumonitis

Bagazosis – sugar cane reaction

411
Q

What determines the pathology observed in type III hypersensitivity reactions?

A

Antigen dose and route of delivery determine the pathology observed in type III hypersensitivity reactions

Type of pathology develops following depends on dose and route of exposure. Eg antivenom can get vasculitis, nephritis, arthritis

412
Q

What is the time course of Type IV (delayed) hypersensitivity?

A
  • first inject antigen to skin
  • get cells picking up antigen and presenting to TH1 cells
  • these attract inflammatory cells causing inflammatory reaction

Example – Mantoux test

  • intracutaneous injection of tuberculin
  • after 2-3 days measure size of reaction
  • someone exposed/latent TB may have a stronger reaction
413
Q

What is an example of contact dermatitis?

A

shoes or watch could cause a local delayed response. Could be TH2 or TH1

414
Q

What is type I allergy?

A
  • defined as “disease following a response by the immune system to an otherwise innocuous antigen”
  • a major health concern around the world
  • ~40% of the population in Europe have allergies to one or more common environmental allergens

This is extremely common with around 40% of those living in Europe having some form of allergy. Worldwide it is a growing problem. Disease follows response by the immune system to an innocuous allergen

415
Q

What is IgE?

A
  • First line of defence against worms
  • Binds FcεR1 receptor on mast cells
  • Pre-arms mast cells to react when in the presence of antigen
  • It binds to high affinity receptors on mast cells and other cell types
  • Makes it react to an allergen on exposure
416
Q

What is the simple model of allergen-specific IgE production?

A
  • Allergen exposure
  • Ag presenting cells take it up and present to T cells
  • Can lead to production of cytokines
417
Q

What is the complicated model of allergen specific IgE production?

A
What causes Allergic Sensitisation?
¥	Exposure to Allergen is critical, this includes:
Ð	Nature of the allergen
Ð	Dosage of Allergen (high vs. low)
Ð	Timing in life eg children vs adults
Ð	Location of Priming
¥	Role of pro-allergic dendritic cells and cytokines 
¥	Genetic predisposition to Allergy

Children are more likely to develop allergy than adults in later life

418
Q

What is the hygiene hypothesis?

A
  • Effect of birth order and sibship on hay fever and atopy
  • Reduced prevalence of atopy, asthma and hay fever among children living on farms
  • Living in a more aseptic environment leading to development of allergy?
  • Having older siblings makes you less likely to have allergy? They go to school and bring back the allergens?
  • Children on farms protected against allergy – wide variety of pathogens etc that programmes immune response to be more regulatory and less vulnerable to allergens
419
Q

About common allergens…

A

¥ Common allergens
Ð House dust mite, pollens, cockroach etc
¥ Allergens are named systematically:
Ð After the source organism and the order they were discovered
Ð e.g. Der p1 comes from Dermatophagoides pteronyssinus
Ð Not all proteins are allergenic
¥ One possibility is the allergens have common functionality
Ð Der p1 is a protease
¥ Allergens are normally received in small doses
Ð 1mg per year of ragweed pollen may be enough to sensitise
¥ High dose exposure (eg Fel d 1) may lead to tolerance

They tend to be proteins but there are no specific characteristics that define an antigen

420
Q

What is filaggrin and atopic dermatitis?

A
  • Filaggrin links skin integrity and allergy
  • When it is defective atopic dermatitis is greater
  • This is due to the access for allergens
421
Q

What makes dendritic cells pro-allergic?

A

Not known but one candidate protein is TSLP. This may switch DC to a ‘pro-allergic’ state.

422
Q

What are the effector mechanisms of the allergic immune response?

A
  • Primed for allergic response when first exposed to allergen
  • Ig lines surface of mast cells
  • When exposed again get immediate hypersensitivity response
  • Cross links leading to degranulation and the release of cytokines and various other inflammatory mediators
423
Q

What is mast cell activation?

A
  • Mast cell activation requires cross linking on surface on mast cells
  • Particular allergen
  • Only that one can unlink them on the surface of mast cells
424
Q

What are weal and flare?

A
  • Not just immediate response – early response
  • Also late phase response comes afterwards
  • Immediate hypersensitivity response
  • Delayed response after several hours – late phase response
  • Charaterised by recruitment of cells such as T cells, eosinophils
  • Activation of those cells in the tissues leads to a more prolonged allergic response
425
Q

What is the immune response to allergens?

A
  • primary allergen response - allergen presented by antigen representing cells eg dendritic cells to cells and can get production of IgE – important in lining surface of mast cells for 2nd exposure
  • Aggregation of cells types and activation I the tissues causes late phase response on 2nd exposure on recruitment of other cell types
426
Q

What effector mediators are produced by mast cells in the early phase?

A

HISTAMINE: increases vascular permeability, causes smooth muscle contraction

LEUKOTRIENES: increases vascular permeability, cause smooth muscle contraction, stimulates mucus secretion

PROSTAGLANDINS: chemoattractants for T cells, eosinophils and basophils

427
Q

What effector mediators are produced by mast cells in the late phase?

A

CYTOKINES

IL-4, IL-13: promotes Th2, promotes IgE
TNF-alpha: promotes tissue inflammation

428
Q

What effects does mast cell activation have on the GI tract?

A
  • increased fluid secretion, increased peristalsis

- expulsion of GI tract contents (diarrhoea and vomiting)

429
Q

What effects does mast cell activation have on airways?

A
  • decreased diameter, increased mucus secretion
  • congestion and blockage of airways (wheezing, coughing, phlegm)
  • swelling and mucus secretion in nasal passages
430
Q

What effects does mast cell activation have on blood vessels?

A
  • increased blood flow, increased permeability
  • increased fluid in tissues causing increased flow of lymph to lymph nodes, increased cells and protein in tissues
  • increased effector response in tissues
431
Q

What do eosinophils do?

A

located in the tissues
recruited to the sites of allergic reactions
express FceRI upon activation

432
Q

What are the two effector functions of eosinophils?

A
  1. Release highly toxic granule proteins and free radicals upon
    activation to kill microorganisms/parasites and cause tissue
    damage in allergic reactions.
  2. Synthesise and release prostaglandins, leukotrienes and
    cytokines in order to amplify the inflammatory response by
    activating epithelial cells and recruiting leukocytes.
433
Q

What is the late phase of the IgE-mediated allergic response?

A

¥ Late-phase reaction is dependent on allergen dose
¥ Continued synthesis and release of inflammatory mediators
¥ Chronic allergic inflammation caused by Th2 cells i.e. a type IV hypersensitivity reaction

The second phase of allergic response is T cell mediated
¥ Mostly consisting of allergen specific Th2 cells
¥ These cells recruit other cells by cytokine release
¥ Potentiate further responses

434
Q

What is the summary of how allergy develops?

A

¥ There is a key difference between sensitisation to allergen and reaction to allergen.
¥ Individuals must be sensitised to an allergen before they can react.
¥ Sensitisation requires presentation of allergen to T cells by DC and the priming of Cognate B cells to produce IgE
¥ The Reaction to allergen occurs when the individual is re-exposed to allergen and it binds preformed IgE on mast cells

435
Q

What is asthma?

A
¥	“A State of reversible bronchial hyper-reactivity resulting from a persistent inflammatory process in response to a number of stimuli in a genetically susceptible individual”. 
¥	Atopic and Non-Atopic
Ð	Non Atopic includes
¥	Occupational
¥	Exercise induced
¥	Nocturnal Asthma
¥	Post-bronchiolitic Wheeze
436
Q

What are the characteristics of allergic asthma?

A
•	episodes of wheezy breathing
•	narrowing of the airways
•	rapid changes in airway 
•	obstruction
•	reaction times and severity vary 
 slight wheeziness to asthma 
•	attack
•	common allergens causing 
•	asthma include
-	pollen
-	HDM
-	plants 
-	some foods
437
Q

What is the accuse response in allergic asthma?

A
  • occurs within seconds of allergen exposure
  • results in airway obstruction and breathing difficulties
  • caused by allergen-induced mast cell degranulation in the submucosa of the airways
438
Q

What is the chronic response in allergic asthma?

A
  • chronic inflammation of the airways
  • caused by activation of eosinophils, neutrophils, T cells and other leukocytes
  • mediators released by these cells cause airway remodelling, permanent narrowing of the airways, and further tissue damage
439
Q

What contributes to airway obstruction in chronic asthma?

A

contraction and mucus contribute to blocking the airway.

440
Q

What is the way of treating allergy in clinic?

A

Blockage of effector pathways:
• inhibit effects of mediators on specific receptors
anti-histamine (block the histamine H1 receptor)
• inhibit mast cell degranulation
non-steroidal anti-inflammatory (e.g. chromoglycate)
• inhibit synthesis of specific mediators
lipoxygenase inhibitors

  • Steroids – Act directly on DNA to increase transcription of anti-inflammatory mediators (e.g. IL-10) and decrease transcription of pro-inflammatory mediators
  • Bronchodilators – Reverse acute effect of allergy on airways
  • Immunotherapy – Reverses the sensitisation to allergen by means of tolerising exposure

Preventing allergy is the gold of allergy research. There are promising studies of food allergies.

441
Q

What are organ-specific autoimmune diseases?

A
  • Graves disease – TSH receptors in thyroid
  • Type 1 diabetes – insulin producing cells of the pancreas
  • Graves opthalmopathy, fibroblasts in the eye may express TSH leading to inflammation.
  • Autoimmune disease against tsh receptors, causes hyperthyroisism – graves
  • Agonistic antibody against TSHR- hyperthyroidism
  • T1 diabetes – get T cell destruction
442
Q

What is HLA B27-associated spondyloarthripathies?

A
  • Ankylosing spondylitis, undifferentiated spondyloarthropathy, reactive arthritis, psoriatic arthritis, urethritis, iritis
  • Spectrum of severity and HLA B27 association
  • Associated with bowel inflammation

HLA B27 – molecule involved in the attachment of antigens to molecules. It is associated with a number of diseases.

443
Q

What is systemic lupus evythmatosus (SLE)?

A
  • Multisystem disease
  • Characterised by autoantibodies to nuclear antigens eg double stranded DNA
  • Relapse and remission
444
Q

What are symptoms of SLE?

A
  • alopecia
  • discoid lesions
  • mucus membrane ulceration
  • raynauds phenomenon
  • proteinuria
  • anemia, leukopenia
  • butterfly rash
  • pericarditis
  • arthritis
  • anti-nuclear bodies
445
Q

What is autoimmunity?

A

The immune system has various regulatory controls to prevent it from attacking self proteins and cells.

Failure of these controls will result in immune attack of host components – known as autoimmunity.

446
Q

What is immune tolerance?

A

Immune system does not attack self proteins or cells – it is tolerant to them.

447
Q

What are the two types of tolerance?

A

¥ Central tolerance – destroy self-reactive T or B cells before they enter the circulation
¥ Peripheral tolerance – destroy or control any self reactive T or B cells which do enter the circulation

448
Q

How does central tolerance involve B cells?

A

If immature B cells in the bone marrow encounter antigens in a form which can crosslink their IgM, apoptosis is triggered.

Stromal cells in the bone marrow which B cells encounter before going out – then they die.

449
Q

What are T cells?

A

T cells recognise antigens that are presented to them by MHC proteins.

  • Thymus cells
  • Have to go through the thymus after bone marrow
  • Doesn’t just recognise a large protein, recognises part of it; a peptide
  • Recognises it in the context of MHC proteins

T cells need to be able to recognise foreign peptides that are bound to self-MHC

  • Binds to peptide and the MHC molecule itself
  • CD8 binds to side of MHC
  • In CD4 binds in similar way but different
  • CD4 binds to side of MHCclass II molecules
450
Q

What happens in T cell receptor and MHC binding?

A

need to select for T cell receptors which are capable of binding self MHC.

BUT

  • If binding to self MHC is too weak, may not be enough to allow signalling when binding to MHC with foreign peptides bound in groove.
  • If binding to self MHC is too strong, may allow signalling irrespective of whether self for foreign peptide is bound in groove.
451
Q

What is the T cell selection in the thymus?

A

Is it useless?
Doesn’t bind to any self-MHC at all
Death by neglect

Is it dangerous?
Binds self MHC too strongly
Apoptosis triggered – negative selection

Is it useful?
Binds self MHC weakly
Signal to survive – positive selection

452
Q

How can a T cell developing in the thymus encounter MHC bearing peptides expressed in other parts of the body?

A

A specialised transcription factor allows thymic expression of genes that are expressed in peripheral tissues.

453
Q

What is an autoimmune regulator (AIRE)?

A
  • Promotes self tolerance by allowing the thymic expression of genes from other tissues
  • Mutations in AIRE result in multi-organ autoimmunity
454
Q

What happens to auto reactive T cells that survive central tolerance control?

A

B cells and autoimmune IgM, no T cell help and so no class switch.

455
Q

What is ignorance in peripheral tolerance?

A
  • antigen may be present in too low a concentration to reach the threshold for T cell receptor triggering
  • immunologically privileged sites e.g. eye, brain
456
Q

What is anergy in peripheral tolerance?

A
  • Naïve T cells need costimulatory signals in order to become activated
  • Most cells lack costimulatory proteins and MHC class II
  • If a naïve T cell sees its MHC/peptide ligand without appropriate costimulatory protein it becomes anergic – ie less likely to be stimulated in future even if co-stimuation is then present
457
Q

What is regulation in peripheral tolerance?

A

A subset of helper T cells known as Treg (T regulatory cells) inhibit other T cells.

  • Treg express transcription factor FOXP3
  • Mutation in FOXP3 leads to severe and fatal autoimmune disorder – Immune dysregulation, Polyendocrinopathy, Enteropathy X-linked (IPEX) syndrome
458
Q

What is the major histocompatibility complex?

A
  • Each copy of chromosome 6 carries 3 different MHC class I and 3 different MHC class II genes
  • High levels f genetic variation (polymorphism)
  • MHC is associated with more disease than other region of the genome
459
Q

What are the endocrine factors involved in autoimmune disease?

A
  • SLE is >10 times more common in females than males
  • MS is approximately 10 times more common in females than males
  • Diabetes is equally common in females and males
  • Ankylosing spondylitis is approximately 3 times more common in males than females

This shows there is an endocrine factor predisposing one gender over the other.

460
Q

What are the environmental factors involved in autoimmune disease?

A
  • Hygiene hypothesis: NOD mice and SPF conditions. Migration and T1D, MS and SLE
  • Smoking and rheumatoid arthritis – 13 pairs of identical twins where 1 of each pair smoked and 1 of each pair had RA, in 12/13 cases the twin with RA was the smoker

SPF = specific pathogen free

461
Q

What might trigger a breakdown of self tolerance?

A
  • Loss of/problem with regulatory cells
  • Release of sequestrated antigen
  • Modification of self
  • Molecular mimicry
462
Q

What is citrullination?

A

Modification of self…

  • Citrillin is an amino acid, not coded for by DNA
  • Arginine can be converted to citrulline as a post-translational modification by peptidylarginine deiminase (PAD) enzymes
  • Citrullination may be increased by inflammation
  • Autoantibodies to citrullinated proteins seen in rheumatoid arthritis. Now used for clinical diagnosis.
463
Q

What is molecular mimicry - rheumatic fever?

A
  • Disease is triggered by infection with Streptococcus pyogenes
  • Antibodies to strep cell wall antigens may crossreact with cardiac muscle
464
Q

What is an example of antibodies in autoimmune pathology?

A

GRAVES DISEASE

  • Auto-antibodies bind Thyroid stimulating hormone (TSH) receptor and stimulate it, resulting in hyperthyroidism
  • Disease can be transferred with IgG antibodies

MYASTHENIA GRAVIS

  • Autoantibodies bind to acetylcholine receptor and block the ability of acetyl choline to bind
  • Also lead to receptor internalisation and degradation
  • Results in muscle weakness

IMMUNE COMPLEXES IN SLE AND VASCULITIS

  • Autoantibodies to soluble antigens form immune complexes
  • Deposited in tissue eg blood vessels, joints, renal glomerulus
  • Can lead to activation of complement and phagocytic cells
  • Immune complexes depositing in kidney can lead to renal failure
465
Q

How can autoimmune disease be mediated across the placenta?

A

IgG is the only antibody small enough to get across the placenta.

  • Patient with Graves disease makes anti-TSHR antibodies
  • Transfer of antibodies across placenta into foetus
  • Newborn infant also suffers from graves disease
  • Plasmapheresis removed maternal anti-TSHR antibodies and cures the disease
466
Q

What is the role of T cells in autoimmune pathology?

A
  • Direct killing by CD8+ CTL
  • Self-destruction induced by cytokines such as TNFalpha
  • Recruitment and activation of macrophages leading to bystander tissue destruction
  • Multiple sclerosis
  • Insulin dependent diabetes mellitus
467
Q

What are TH17 cells? (autoimmune disease)

A
  • Th17 cells are helper T cells that produce the cytokine IL-17
  • Implicated in autoimmune diseases including spondyloarthropathy, MS and diabetes
  • Highly inflammatory
  • Produce cytokines which are involved in the recruitment, migration and activation of immune cells
468
Q

What are therapeutic strategies in autoimmune disease?

A
  • Anti-inflammatories: NSAID, corticosteroids
  • T cell depletion (RA: anti-CD4, anti-CD20)
  • Therapeutic antibodies (nti-TNF; anti-VLA-4 (blocks adhesion))
  • Antigen specific therapies, indevelopemnt. Glaritamer acetate, increases T-regs
469
Q

What are autologous/syngenic donor/recipient relationships?

A

Donor and recipients are genetically identical, do not usually generate any immunological problems.

Skin transplant is an example of an autologous transplant in humans

470
Q

What is syngeneic transplant in humans?

A

This is between genetically identical twins

471
Q

What are allogeneic donor/recipient relationships?

A

Donors and recipients are from the same species but are genetically different. This is a more common type of transplant. Eg from one mouse to another non-genetically identical mouse or between non-identical humans such as siblings

472
Q

What are xenogeneic donor/recipient relationships?

A

Donor and recipient are different species. In humans the best animla transplant source are pigs, cows or maybe baboons.

Problems include the introductions of xenosis which is an infectious disease from animal and mismatch in MHC

473
Q

What are major histocompatibility antigens?

A
  • Histocompatibility = tissue compatibility
  • Immune responses to transplant are caused by genetic differences between the donor and the recipient
  • The most important are differences between the major histocompatibility antigens
  • Human transplants largely unsuccessful until identification of human MHC in 1967
  • In 1968 WHO nomenclature committee designated that human MHC proteins be names HLA (Human Leukocyte Antigen)
474
Q

What does MHC restrictive mean?

A
  • Described as MHC restrictive
  • Refers to the need for MHC to present antigens to T cells
  • The antibody on the B cell can bind to the antigen irrespective of any other proteins
  • MHC can only interact with TCR as part of MHC complex
475
Q

About T cells and MHC class I…

A

T cells recognise short peptide fragments that are presented to them by major histocompatibility (MHC) proteins.

All our somatic cells have HLA class I. Proteins within the cell are degraded and loaded onto MHC.

476
Q

What is MHC loading?

A
  • These are similar except MHC class II only present on the surface of WBC
  • They are not present in somatic cells that are not part of the immune system
  • They get their peptides to present in a different way
  • Will engulf a dying or dead cell
  • Will process the peptides particularly the MHC and will present on HLA to CD4 T cells
477
Q

About helper T cells and cytotoxic T cells…

A

Helper T cells are required to produce antibody and cytotoxic T cell responses.

HELPER T CELLS - information and support for other immune cells via cytokine production

CYTOTOXIC T CELLS - highly specific killer cells

478
Q

Why is it important to match HLA in the donor and the recipient?

A

In transplants, both the MHC protein and the peptide in its binding groove may be foreign

In transplants the MHC itself and the thing being presented can both be foreign. This is why it is so important to match HLA in the donor and the recipient. This make it more likely to be successful.

479
Q

About HLA mismatch and graft survival…

A

Usually we try to match 4/6 MHC class II loci, reduces likelihood of future transplants and problems with future transplants.

Clinicians try to match as many class II as passible. With an increasing number of mismatches get increasing amount of time going to be accepted by the host

480
Q

Live vs Dead donors…

A
  • Recipients will have a history of disease which will have resulted in a degree of inflammation
  • Organs from deceased donors are also likely to be in inflamed conditions due to ischaemia
  • Transplant success is less sensitive to MHC mismatch for live donors
  • Lots of inflammation aids in activation of t cells can can make it more likely the host immune system will break tolerance/direct an immune response against transplant
  • From deceased donor more likely to be inflamed, also doesn’t help as this will cause the WBC in the organ to have an activated phenotype
  • Not only the recipient immune system present – donor immune system in transplanted organ!
481
Q

What are the types of graft rejection?

A
  • hyperacute rejection
  • acute rejection
  • chronic rejection
482
Q

What is hyperacute rejection?

A

¥ Within a few hours of transplant – very rapid
¥ Most commonly seen for highly vascularised organs (e.g. kidney)
¥ Requires pre-existing antibodies, usually to ABO blood group antigens or MHC-I proteins
¥ (ABO antigens are expressed on endothelial cells of blood vessels)
¥ Antibodies to MHC can arise from pregnancy, blood transfusion or previous transplants
¥ Due to strong Ab response
¥ Usually because the recipient has seen the antigen (ABO or MHC) before in pregnancy or transfusion or another transplantation
¥ Not dissimilar to vaccination
¥ Immune system trying to reject transplanted tissue

483
Q

How can antibodies cause damage to transplanted tissue?

A

Recognition of Fc region leading to…

  • Complement activation
  • Antibody dependent cellular cytotoxicity (Fc receptors on NK cells)
  • Phagocytosis (Fc receptors on macrophages)

FAB region: hypervariable region responsible for binding the Ag
Fc: constant region that can be identified by complement pathway – serious for proteins that can opsonize the target cell

484
Q

What happens in hyperacute rejection?

A
  • Antibodies bind to endothelial cells
  • Complement fixation
  • Accumulation of innate immune cells
  • Endothelial damage, platelets accumulate, thrombi develop
  • Eg kidney transplantation
485
Q

What is acute rejection?

A

Inflammation results in activation of organs resident dendritic cells.

T cell response develops as a result of MHC mismatch.

Based upon the immune system within donor transplant – activated phenotype

486
Q

What happens if there is a large number of WBV in class I and class II HLA proteins in the transplant?

A

the recipient immune system can protect the MHC and TCR will bind to MHC and will identify them as foreign/non-self

487
Q

What is direct allorecognition of foreign MHC?

A

Inflammation results in activation or organ’s resident dendritic cells.

DC migrate to secondary lymphoid tissue where they encounter circulating effector T cells.

Macrophages and CTL increase inflammation and destroy transplant.

In this case eg kidney (last slides notes), the dendritic cells in the tissues WBC and get a kidney and all the cells come with it eg dendritic cells and macrophages.

DCs migrate and activate T cell immunity and will move from kidney into recipients lymph nodes.

T cells there will identify DCs. The immune system doesn’t recognize the DCs from donor. The T cells will then hunt down the cells in the donor kidney and destroy it.

  • kidney graft with dendritic cells
  • dendritic cells migrate to the spleen where they activate effector T cells
  • effector T cells migrate to graft via blood
  • graft destroyed by effector T cells
488
Q

What is chronic rejection?

A

¥ Can occur months or years after transplant
¥ Blood vessel walls thickened, lumina narrowed – loss of blood supply
¥ Correlates with presence of antibodies to MHC-I

This can occur even under immunosuppressant therapy. Antibodies bind to the endothelial cells and recruit immune effector cells, particularly cells of the inert immune system.

This causes damage to the endothelial cells apoptosis and internalized by cells of the recipient.

The recipient antigen presenting cells internalizes the donor cells and start to process the donor MHC molecules.

This is another case of MHC mismatch. Chronic rejection is when the donor cells die and are taken up by DCs. The foreign MHC is cut up unto peptides and presented to HLC class II. They are foreign to the immune system activating T cells to destroy the transplanted tissue.

489
Q

How can chronic rejection result form indirect allorecognition of foreign MHC?

A

¥ Donor-derived cells die
¥ Membrane fragments containing donor MHC are taken up by host DC
¥ Donor MHC is presented into peptides which are presented by host MHC
¥ T cell response is generated

Acute – donor WBC, host immune system detects MHC on those donor WBCs
Chronic – host DCs internalize donor cells and present MHC peptides to T cells (it is the peptide that is foreign) – this is indirect recognition

490
Q

What is hematopoietic stem cell transfer (HSCT)?

A

¥ Previously called bone marrow transplant, now renamed as source is often blood
¥ Often autologous – transfer of biological material from one part of a person, to another part
¥ Until 1980 only HLA identical siblings could be used as donors due to the risk of rejection or graft versus host disease

Stem cells are more often derived from peripheral blood now than bone marrow. It is a better way of getting more cells and it is easier.

HSCs can find their way to bone marrow after infusion and regenerate there.

They can be cryopreserved with little damage.

491
Q

What is graft vs host disease?

A

¥ When transplanted tissue is immune cells themselves, there is the risk of donor immune cells attacking the host – GVHD
¥ Can be lethal – best approach is prevention
¥ Removing T cells from transplant reduces GVHD

In this case the graft is attacking host instead of the other way round. If we remove the T cells from the recipient so wont be attacked, or from HST’s transplanted so no initial attack.

Prevent GCHD using immunotherapy. This can be beneficial….

492
Q

What can be beneficial of graft vs leukaemia?

A

¥ But sometimes mismatch and donor leukocytes can be benificial - removing original leukemia
¥ Graft versus leukemia response
¥ Development of GVL may prevent disease relapse

Patient could have residual leukemia, chemo may not have completely removed the leukemic cells, so new immune system sees leukocytes as foreign and will attack them, preventing patient leukemia reoccurring.

493
Q

What is immunosuppression?

A

¥ Essential to maintain non-autologous transplant
¥ Immunosuppressants for transplant can be -
Ð General immune inhibitors (e.g. corticosteroids)
Ð Cytotoxic – kill proliferating lymphocytes (e.g. mycophenolic acid, cyclophosphamide, methotrexate)
Ð Inhibit T cell activation (cyclosporin, tacrolimus, rapamycin)
• Immunosuppressives may need to be maintained indefinitely

Maintenance phase of immunosuppression to last potentially their whole life. If things go wrong then there can be a rescue phase – more powerful immunosuppressants

494
Q

What is cyclosporin?

A
  • Breakthrough drug for transplant
  • Blocks T cell proliferation and differentiation
  • Next generation therapies less toxic and effective at lower doses
  • Very toxic – making newer versions
495
Q

What are combination immunosuppression regimes?

A
  1. Steroids – eg prednisolone
  2. Cytotoxic – eg mycophenolate motefil
  3. Immunosuppressive specific for T cells – eg cyclosporine A, FK506

The best combination is being worked out and this is important for treating individuals

496
Q

What is immunosuppressive therapy monitoring?

A

¥ There is currently no immunosuppressive that will prevent transplant rejection whilst maintaining other immune responses
¥ Transplant patients more susceptible to infection and malignancy
Ð Immediate risk e.g. CMV
¥ Immunosuppressive drug toxicity can lead to organ failure eg cyclosporin nephrotoxicity in kidney transplant

The immune system is not meant to be inhibited indefinitely so can cause all sorts of problems. People on immunosuppressive therapy at greater risk of cancer. Also drug toxicity which can result in organ failure. New and better immunosuppressive therapy is vital to improving patients outcomes after transplantation

497
Q

What are the stages of lymphocyte development in the bone marrow?

A

1 - Haematopoietic stem cell
2 - common myeloid progenitor –> neutrophils, red cells, platelets etc
OR 2.1 - common lymphoid progenitor
3. T cell precursors and B cell precursors
4 - B cell precursors go on to become immature B cells

498
Q

What is the presentation of ALL?

A
  • usually non specific symptoms of bone marrow suppression

- symptoms of organ infiltration more often in advanced disease

499
Q

What is the epidemiology of ALL?

A
  • commonest leukaemia in children 40 y.o
500
Q

What is the investigations and diagnosis of ALL?

A
  • bone marrow morphology; infiltration by undifferentiated blast cells
  • immunophenotyping; B-cell surface markers (or T markers for T-ALL), light chain restriction, TdT positive
  • cytogenetics
501
Q

What is the treatment for ALL?

A

Chemotherapy

in children >90% cure

adults have a much lower survival because different cell or origin, different oncogene mutations, older patients do not tolerate intensive treatment

502
Q

What is Hodgkin’s lymphoma?

A

PRESENTATION: enlarged lymph nodes

EPIDEMIOLOGY: peak incense in young adults, possible association with Epstein Barr Virus (EBV) aka HHV4

HISTOPATHOLOGY: presence of large Reed-Sternberg cells is pathognomonic, these are malignant B-cells, but typically 99% of cells are reactive non-malignant cells

503
Q

What is the treatment of Hodgkin’s lymphoma?

A

chemotherapy +/- radiotherapy

  • 5 year survival ~50-90% depending on age, stage and histology
  • especially good results in young adults
504
Q

What are non-hodgkin’s lymphomas?

A
  • low grade
  • high grade
  • T-cell lymphomas
  • EBV (HHV4) driven lymphomas in immunosuppressed patients
505
Q

What happens with chromosome translocations and lymphoma?

A
  • Many lymphomas carry chromosome translocations involving the Ig heavy chain or light chain loci
  • Ig genes are highly expressed in B-cells
  • Each Ig gene has a powerful tissue specific enhancer near to the constant (C) segment
  • Most cases of follicular lymphoma carry t(14;18)(q32;31)
  • This juxtaposes the BCL-2 gene on chromosome 18 with the IgH locus on chromosome 14
  • Causes overexpression of BCL-2 protein
  • BCL-2 is an apoptosis inhibitor
  • Some cases of high grade lymphoma carry t(8;14)(q24;q32)
  • This juxtaposes the MYC gene on chromosome 18 with the IgH locus on chromosome 14
  • MYC is a powerful oncogene
  • Can also get MYC or BCL-2 translocations to one of the Ig light chain loci
506
Q

What is low grade NHL?

A

Presentation: enlarged lymph node(s)

Histology

  • Normal tissue architecture partially preserved
  • Normal cell of origin recognisable  used to name lymphoma – follicular lymphoma, mantle cell lymphoma etc
507
Q

What is the diagnosis of low grade NHL?

A
  • Histology
  • Immunocytochemistry
  • Cytogenetics
  • Light chain restriction
  • PCR  for clonal Ig gene rearrangement, for chromosome translocations (eg t(14;18) IgL Bcl-2)
508
Q

What is the treatment and prognosis of low grade NHL?

A

Treatment

  • Chemotherapy
  • Glucocorticoids (eg prednisolone)
  • Radiotherapy
  • Monoclonal Ab therapy – rituximab (anti-CD20)

Prognosis

  • Relatively indolent
  • Respond well to therapy
  • But hard to cure
509
Q

What is high grade NHL?

A

Presentation: enlarged lymph node(s)

Histology

  • Loss of normal tissue architecture
  • Normal cell of origin hard to determine
510
Q

What is the diagnosis of high grade NHL?

A
  • Histology
  • Immunocytochemistry
  • Cytogenetics
  • Light chain restriction
  • PCR  for clonal Ig gene rearrangement, for chromosome translocations
511
Q

What is the treatment and prognosis of high grade NHL?

A

Treatment

  • Chemotherapy
  • Glucocorticoids
  • Radiotherapy
  • Monoclonal Ab therapy  rituximab – anti CD20

Prognosis

  • Variable depending on type, stage and other factors
  • Overall long term survival
512
Q

About T-cell lymphomas…

A
•	Rare
•	Usually CD4 cells
•	Often present with skin infiltration eg
-	Sezary syndrome
-	Mycosis fungoides
513
Q

What is acute T-cell leukaemia/lymphoma?

A
  • Found in Japan, Caribbean and UK citizens of Caribbean origin
  • Associated with retrovirus HTLV-1 (human T-cell leukaemia/lymphoma virus 1) infection
514
Q

What are Epstein Barr Virus (EBV) driven lymphomas?

A
  • EBV or Human Herpes Virus 4 (HHV4) directly transforms B-lymphocytes in culture
  • Due to viral oncogene LMP-1
  • Over half of all normal individuals carry latent EBV infection
  • Do not develop lymphomas due to effective immune surveillance by cytotoxic T-cells
515
Q

What can happen with lymphomas in highly immunosuppressed individuals?

A
  • The endogenous latent EBV may transform B-cells
  • No longer eliminated by cytotoxic T-cells
  • Develop high grade lymphoma

Transplant patients on cyclosporine

  • Lymphoma usually regresses on withdrawal of immunosuppression
  • But patient may lose the graft

AIDS patient
- Lymphoma may regress on successful HAART

516
Q

What is CLL?

A

Presentation

  • Most often as incidental finding on fbc
  • Persistent infection(s); due to immunosuppression, low IgG, suppression of normal B cells
  • Lymph node enlargement
  • Symptoms of bone marrow suppression

Epidemiology
- 85% of cases >50yo

517
Q

What is the diagnosis of CLL?

A
  • FBC: lymphocytosis
  • Immunophenotyping: cell surface markers, light chain restriction
  • Cytogenetics
518
Q

What is paraproteinaemia?

A

Presence of a single monoclonal Ig in the serum

519
Q

What is the relation between paraproteins and disease?

A

Malignant
Multiple myeloma – high levels of paraprotein

Benign
Monoclonal gammopathy of undetermined significance (MGUS) – low levels of paraprotein.

Low levels of paraprotein occasionally seen in lymphoma or chronic lymphocytic leukaemia.

IgM paraprotein seen in waldenstrom’s macroglobulinaemia.

  • Monoclonal gammopathy of undetermined significance (MGUS) – low levels of paraprotein.
  • Low levels of paraprotein occasionally seen in lymphoma or chronic lymphocytic leukaemia.
  • IgM paraprotein seen in waldenstrom’s macroglobulinaemia.
520
Q

What are the three aspects of myeloma giving rise to different clinical features?

A
  1. Suppression of normal bone marrow, blood cell and immune cell function
  2. Bone resorption and release of calcium
  3. Pathological effects of the paraprotein
521
Q

What happens as a result of blood cell/immune suppression in myeloma?

A
  • Anaemia
  • Recurrent infections
  • Bleeding endency
522
Q

What happens as a result of bone resorption in myeloma?

A
  • Myeloma cells produce cytokines (esp. IL-6)
  • Which stimulate bone marrow stromal cells to release the cytokine RANKL
  • Which activates osteoclasts: lytic lesions of bone, bone pain, fractures
  • Calcium released form bone causes hypercalcaemia: multiple symptoms including mental disturbance
523
Q

What are the effects of paraprotein in myeloma?

A
  • Precipitates in kidney tubules cause renal failure
  • Deposited as amyloid in many tissues
  • 2% of cases develop hyperviscosity syndrome: increased viscosity of blood leading to, stroke, heart failure
524
Q

What is the diagnosis of myeloma?

A
  • serum electrophoresis for paraprotein
  • urine electrophoresis: Bence-Jones protein represents free monoclonal light chains
  • increased plasma cells in bone marrow
  • ESR (very high due to rouleaux formation)
  • Radiological investigation of skeleton for lytic lesion
525
Q

What is the treatment of myeloma?

A

Chemotherapy (not curative)

  • Cutotoxic drugs
  • Glucocorticoids
  • Thalidomide analogues
  • Bortezomib

Allogeneic bone marrow transplant
- Only available for a small number of younger patients (

526
Q

What is the definition of leukaemia?

A

The leukaemias are a group of diseases characterised by malignant overproduction of white blood cells or their immature precursors.

527
Q

What is the presentation of leukaemia?

A

• Varies between types of leukaemia
• But typically first presents with symptoms due to loss of normal blood cell production
- Commonest presentation is abnormal bruising (suppression of platelet production)
- Or repeated/abnormal infection (suppression of neutrophil production)
- Sometimes just anaemia (suppression of RBC production)

528
Q

What is the classification of leukaemia?

A

There are a few rare forms that don’t fit.

• Lymphoid- involving cells of the lymphocyte lineages
– commonly B-cell,
– more rarely T-cell
• Myeloid- involving any of the non-lymphocyte blood cell lineages
– commonly neutrophils or their precursors
– But can be erythroid, platelet, basophil lineages etc

529
Q

What is meant by acute and chronic leukaemia?

A

ACUTE: undifferentiated leukaemia, characterised by immature white cells (blast cells)

CHRONIC: differentiated leukaemia, characterised by mature WBCs

530
Q

What is the involvement of gene in leukaemia?

A

• Activation of oncogenes and inactivation of tumour suppressor genes
• May involve genes similar to other malignancies eg
- RAS
- MYC
- P45 – tumour suppressor gene
• But also some specific to the leukaemias
• Chromosome translocations to generate novel hybrid oncogenes eg
- BCR-ABL in CML
- PML-RARA in AML M4
- And many others

Numerical and chromosome changes eg

  • Monosomy: loss of normal genes, tumour suppressor genes
  • Trisomy: gene amplification
531
Q

What is leukaemia a clonal disease?

A

All the malignant cells derive from a single mutant stem cell

532
Q

What are the etiological risk factors of leukaemia?

A

Radiation

  • acute radiation accidents
  • atomic bomb survivors

Chemicals
- eg industrial exposure to benzene

Chemotherapy
- cancer chemotherapy with alkylating agents (eg busulphan) generates a future risk of leukaemia

Age
- the age related incidence is different in different types of leukaemia

Controversial

  • electric power lines
  • nuclear power stations
  • natural background radiation

Most cases are not associated with any identifiable precipitating cause

533
Q

What is the treatment of leukaemia?

A

• Chemotherapy with cytotoxic drugs
• Stem cell and bone marrow transplant (SCBMT)
• Disease specific agents
- Including oncogene targeted drugs

  • Combination of drugs used to kill leukaemic cells
  • Optimised for type and subtype of leukaemia
  • Cytotoxic drugs mostly target dividing cells
534
Q

What are the side effects of chemotherapy?

A

Kills normally dividing cells

  • GI epithelium  nausea and diarrhoea
  • Hair follicles  hair loss
  • Loss of fertility
  • Haemopoietic progenitors  bone marrow suppression

Haemopoiesis is reconstituted form the quiescent stem cell pool.

535
Q

What is SCBMT?

A
  • Give intense chemotherapy and totally body irradditation
  • Wipe out leukaemic cells AND normal stem cells
  • Reconstitute bone marrow by transplanted stem cells
536
Q

What are the problems with SCBMT?

A
  • Shortage of HLA (MHC) matched donors

- High mortality of the procedure for older or sicker patients

537
Q

What does a bone marrow smear of acute leukaemia look like?

A
  • bone marrow smear
  • all look like lymphoblasts
  • very suspiscious

Large numbers of myeloid blasts (AML) or lymphoblasts (ALL) in bone marrow – therefore “undifferentiated leukaemias’.

538
Q

What are the symptoms of acute leukaemia?

A

Typical symptoms due to bone marrow suppression

  • Thrombocytopenia  purpura (bruising), epistaxis (nosebleed), bleeding from gums
  • Neutropenia  recurrent infections
  • Anaemia  lassitude, weakness, shortness of breath
539
Q

What is the diagnosis of acute leukaemia?

A

Peripheral blood

  • Presence of blasts
  • Cytopenias

Bone marrow aspirate
- >30% blasts is diagnostic of acute leukaemia

Shortage of normal cells – cytopenia

540
Q

What is the pathophysiology of acute leukaemia?

A

If the cells don’t mature the blast cell pool will build up and up without stopping and wont die. There is a high turnover so they are being made very quickly.

Many of the chromosome translocations in acute leukaemias

  • involve genes for transcription factors
  • which control cell differentiation

Chromosome abnormalities also help determie prognosis and response to treatment.

541
Q

What is the classification of acute leukaemia?

A

French-American-British (FAB) classification

  • based on stage of differentiation arrest eg. AML M4 promyelocytic leukaemia
  • and predominant cell type eg AML M6 erythroleukaemia

(M0-M8)

Also some rare leukaemias that don’t fit the main classifications.

WHO classification

  • similar to FAB, but
  • acute leukaemias with specific chromosome translocations are classified separately
  • eg AML with a translocation between chromosomes 8 and 21
542
Q

What is the treatment of acute leukaemia?

A
  • chemotherapy
  • combinations of drugs
  • optimised for each type and sub-type of acute leukaemia
  • ALL- add chemo- or radio-therapy to CNS

ALL to CNS – BBB means in cytotoxic drugs don’t get to the cells and so brain may be a site where leukemic cells survive and can come back from.

Phase 1: remission induction  blast with chemotherapy, may get very ill and wipe out most of the blasts
Phase 2: consolidation therapy – to kill residual leukaemic stem cells  if leave at previous stage leukaemia may come back so send home with less powerful chemo agents oral for the next year and less likely to relapse.

543
Q

What are the outcomes of treatment of acute leukaemia?

A

Childhood ALL - >90% long term remission/cure

Adult ALL – poorer prognosis because disease is different

  • Different cell of origin
  • Different oncogene mutations

AML - >80% long term remission in young adults with aggressive treatment

Elderly unable to tolerate aggressive chemotherapy or SCMBT

The elderly you know may relapse because you can’t give really aggressive chemotherapy. Some may choose not to have chemo as it’s a horrible experience for just a few extra months of life.

544
Q

What are chronic leukaemia?

A

“Differentiated leukaemias” – increased numbers of differentiated cells

From patient with CLL. Too man WBC here and all are lymphocytes.

545
Q

What is chronic lymphoid leukaemia?

A
  • Large numbers of mature (clonal) lymphocytes in bone marrow and peripheral blood
  • Therefore also called chronic lymphocytic leukaemia
546
Q

What are the symptoms of chronic lymphoid leukaemia?

A
  • Recurrent infections – due to neutropenia, and suppression of normal lymphocyte function
  • Anaemia
  • Thrombocytopenia
  • Lymph node enlargement
  • Hepatosplenomegaly
547
Q

What is the treatment and outcome of chronic lymphoid leukaemia?

A
  • Controlled by regular chemotherapy to reduce cell numbers
  • A few patients may die in 12 years

As this is a disease of the elderly a lot of the time, then surviving of more than 12 years you are likely to die of something else first. These days many people live with cancer, rather than die of it.

548
Q

What are chronic leukaemia?

A

“differentiated leukaemias”

Increased numbers of differentiated cells.

  • CML
  • Far too many WBC of neutrophil lineage
  • Lots of mature cells but also immature cells
  • Doesn’t mean all the cells are differentiated and mature, but that they are not all arrested earlier on
549
Q

What are the symptoms of CML?

A

Symptoms

  • Anaemia
  • Night fever/night sweats
  • Splenomegaly

Infection is not a major symptom of CLL, because not short of neutrophils even though they’re not all working properly.

550
Q

What is the diagnosis of CML?

A
  • Very high white count (neutrophilia)
  • Left shift in blood and bone marrow
  • Presence of Philadelphia chromosome
  • BCR-ABL gene rearrangement
551
Q

What is the treatment and course of CML?

A

Controlled but not cured by chemotherapy.

  • Cytotoxic drugs
  • Interferon alpha – body normally makes interferons when you have a viral infection. If you give someone interferon alpha it makes them feel like they have the flu.
  • Imatinib

Course

  • Survival on treatment usually measured in years – 4-5 years
  • But eventually progresses to accelerated phase and then blast crisis

Treatment

  • Allogenic bone marrow or stem cell transplant curative
  • Autologous transplant sometimes tried
  • Most patients >50 years old. Do not tolerate transplant. Transplant only really used for younger and fitter patients
552
Q

What is a ‘blast crisis’?

A

resembles an acute leukaemia, but very refractory to treatment

553
Q

What is the Philadelphia chromosome?

A

95% of cases of CML have a detectable Philadelphia chromosome (Ph’)

  • Breakpoint on chr22 in middle of BCR gene
  • Breakpoint in chr9 in middle of ABL gene
  • after translocation get a gene with a bit of BCR and a bit of ABL – a chimaeric gene

Breakpoints occur within introns.

  • The ABL protein is a protein tyrosine kinase
  • But activity is tightly regulated
  • BCR-ABL protein has constitutive (unregulated) protein tyrosine kinase activity
  • Tyrosine kinases involved in cell signalling
  • ABL not cell surface protein but important signalling protien in cell
  • Tightly regulated
  • Most of the time not active and only stimulated when theres a need
  • BCR-ABL protein still has the catalytic domain but not regulated – active all the time independent of regulation
  • This causes problems
554
Q

What does unregulated BCR-ABL tyrosine kinase activity cause?

A
  • Proliferation of progenitor cells in the absence of growth factors
  • Decreased apoptosis
  • Decreased adhesion to bone marrow stroma
555
Q

What is Imatinib?

A

Inhibiton of BCR-ABL tyrosine kinase causes apoptosis of CML cells. Cells undergo apoptosis.

Entered clinical trials 1999. Approved by NICE 2003. This was very quick to be approved – notmally it takes >10 years. There was a large drive from patients etc that pushed it through to be approved.

Compared to previous treatments Imatinib

  • Remission is induced in more patients
  • With greater durability
  • And fewer side effects

But some patients become drug resistant

556
Q

Is Imatinib a cure?

A

Current trials

  • Patients in long term remission taken off therapy
  • But closely monitored by RT-PCR

In some cases the disease comes back. If the patient is put back on imatinib they will respond. Some patients a long time after will have no sign of relapse – so sometimes it is possibly a cure!!

557
Q

What are the types of pathogens?

A

Commensal non pathogen (in host)
Zoonotic non pathogen (in carrier)
commensal opportunist (in host)

558
Q

What is a commensal non pathogen?

A
  • in host
  • present but not capable of causing disease in the host
  • eg e.coli
559
Q

What is a zoonotic non pathogen?

A
  • in carrier
  • present but only capable of causing disease in another host
  • eg e.coli O157
560
Q

What is a commensal opportunist?

A
  • in host
  • present and capable of causing disease in host but only in certain circumstances
  • eg bacteroides fragilis
561
Q

Are all positive samples diagnostic of disease?

A

no

562
Q

Pathogen definition…

A

a microbe capable of causing a specific degree of host damage

563
Q

What makes a good sample for a test result?

A
  • Sterile sites must be free fro contamination eg skin flora in blood cultures
  • Non sterile sites require decontamination of normal flora eg faeces, mouth, skin
  • Samples with high volume or relatively low infected pathogen load require concentration (centrifugation, filtering) eg CSF, ascites, 24 hr urine
564
Q

What happens in a cultured sample?

A

1 - take culture

2 - PREPARATION PHASE
enrichment, purification, amplification

3 - IDENTIFICATION PHASE
molecular DNA/RNA, gross morphology (microscopy), chemical composition (HPLC MassSpec)

565
Q

What happens in a direct sample?

A

1- take sample

2 - PREPARATION PHASE
concentration, sample treatment

3 - IDENTIFICATION PHASE
molecular DNA/RNA, gross morphology (microscopy), chemical composition (HPLC MassSpec)

566
Q

What are the advantages of microscopy?

A
  • easy to perform
  • rapid screening
  • some parasites have specific morphology
  • specific immunofluorescence staining possible
567
Q

What are the disadvantages of microscopy?

A
  • not sensitive
  • screening sputum smears requires at least 10,000 orgs per ml to be visualised
  • general stains are not specific
  • labour intensive (expensive)
  • requires specialist interpretive expertise (more expensive)
568
Q

What is bacteriology?

A

this relies on the ability of the test system to be able to grow the pathogen

569
Q

What options of media are there for detecting pathogens?

A
  • non selective media e.g. blood agar
  • semi selective media eg MacConkey agar
  • selective growth temperatures e.g. campylobacter species
570
Q

What are examples of selective atmosphere?

A
  • aerobic culture
  • microaerophilic culture
  • anaerobic culture
571
Q

What is the campylobacter selective environment?

A

42^C

10% CO2

572
Q

What is done in quantification, identification, antibiogram of pathogens?

A
  • colony morphology, colour, haemolysis
  • colony count
  • colony identification
  • systematic identification
  • colony resistance to antibiotics
573
Q

What is antibiotic sensitivity plate testing?

A

where you put antibiotics on a plate and see where the bacteria will grow - if it grows near the drug then it is resistance. Can also be done for dosages.

574
Q

What is the virology process in classical culture and identification?

A

1 - culture
requires permissive cell lines
cytopathic effect
immunofluorescnet staining of culture

2 - direct antigen detection
eg influenza virus

575
Q

What is direct and serological ELISA?

A
  • Electron microscopy can see this virus but not identify it as ‘swine flu’
  • Culture takes 3-10 days
  • Rapid ELISA for fluA antigen = 15 mins
  • ELISA for flu antibody
576
Q

What are the advantages classical culture and identification?

A
  • Cheap, simple, reliable reagents
  • Sensitive – eg single organisms can be grown and identified
  • Validated specificity eg ‘gold standards’ with multiple parameters
  • Direct in vivo measurement of effectiveness of therapy eg antibiotic sensitivity
  • Easily archived eg epidemiology
577
Q

What are the disadvantages of classical culture and identification?

A
  • Some pathogens cannot be grown eg mycobacterium leprae
  • Some pathogens cannot be well differentiated by biochemistry alone
  • Slow: culture requires at least over night incubation, viral: 3-10 days, mycobacterial: 6-12 weeks
  • Some pathogens grow too slowly to aid rapid diagnosis eg mycobacterium tuberculosis
  • Labour intensive (expensive)
  • Requires specialist interpretive expertise (more expensive)
578
Q

What genes are suitable targets for PCR?

A
  • Constitutive
  • Virulence
  • Antibiotic resistane
  • Pathogenic phenotype
  • Repetitive
579
Q

What do you have to consider for if a molecular test for one gene is good enough?

A

Specificity
If the test unique to the genus/species/type?

Reliability
Is the target nonessential/transmissible?

Sensitivity
How many organisms does it take to suggest disease?
-	For every sample type
-	For every host type
-	For every epidemiological niche

Accuracy
Do we need to detect live organisms?
Is the detection system susceptible to genomic shifts/mutations?

Rapidity
Is the result generated going to be beneficial to the paitent?
Instant bedside? – diagnosis of paediatric meningitis
Same day? – transmission/quarantine
Next day? – antibiotic resistance
Next week? – chronic persistent infections

580
Q

What is multiple gene targeting?

A

microarrays

  • Ordered short oligonucleotide probes (40-70mer) attached to slides in defined spots
  • Each spot represents a single gene
  • Comparative genomic hybridisation (CGH) used mostly for DNA
581
Q

What are the advantages of microarrays?

A
  • Covers the whole genome
  • Strand dependent
  • Can be used for RNA and transcriptomics
  • Can look for microRNA
582
Q

What are molecular signatures?

A

Aim to detect a gene or gene products that are pathogen specific.

Single gene target (PCR) – PCR, qPCR
Multiple gene target (microarray) – microarray
Mass spectrometry (MALDI-TOF)

583
Q

Whats bio-signature profiling?

A

Mass spectrometry
MALDI-TOF (matrix assisted laser desorption ionisation-time-of-flight)

Compare against an archival database, 62,500 unique spectral profiles identifying 1,160 species and 233 genera.

584
Q

What are biomarkers of virulence?

A

Looking for selected genes or gene products that drive the disease process.

  • Latex agglutination test
  • Uses particles coated with specific antibody to cell wall antigens
  • CSF direct agglutination test
  • Neisseria meningitides Group B
  • Haemophilus influenza type b
  • Streptococcus pneumonia type 3

Specific cell wall antigens are predictive of invasiveness and virulence.

585
Q

What is Shiga toxin detection in E.coli O157?

A
  1. Enterohaemolysis
  2. Agglutination with anti-toxin antibodies
  3. PCR for the presence of the gene
586
Q

What are the advantages of biomarkers of virulence?

A
  • Good specificity
  • Good sensitivity
  • Easily automated
587
Q

What are the disadvantages of biomarkers of virulence?

A
  • Serological response is not rapid therefore not useful in acute infections
  • Single sera results are meaningless due to possible previous exposure
  • Some antibodies are cross-reactive
  • Virulence is only INFERRED by the presence of a biomarker ONLY in vivo testing of cultured pathogen infected into an animal model can prove virulence.
588
Q

What is direct sequencing?

A

Sequencing can show differences between SINGLE bases in strains or resistance mutations to antibiotics

589
Q

What are the advantages of molecular detection methods?

A
  • Rapid
  • Faster detection of pathogens that traditional techniques
  • Allows appropriate, timely antimicrobial therapy and infection control interventions
  • Increased sensitivity over culture and microscopy based techniques in POSITIVE samples
  • Can be automated and has potential for Point of Care testing
590
Q

What are the disadvantages of molecular detection methods?

A
  • Expensive
  • Does not screen for UNKNOWNS
  • Requires expertise
  • Labour intensive
  • Possibility of contamination
  • Require complex and efficient methods for extraction of nucleic acid
  • NEGATIVE samples may STILL need Gold Standard culture. Hospitalisation costs accounted for 95% of health-system costs among patients suspected of tuberculosis. In culture-negative patients, PCR tests do not significantly decrease time to TB exclusion
591
Q

What is bio-signature profiling?

A

following the host transcriptomic profile with microarrays

592
Q

What is the future for detecting pathogens?

A
  • bio-signature profiling
  • metabolic profiling
  • rapid intrinsic fluorescence
  • rapid point of care testing
  • lab on a chip
593
Q

What is rapid point of care testing?

A
  • Point of care (POC) testing
  • Lab on a chip
  • Nanotechnology
  • Microfluidics
  • Rapid sequencing of samples direct at the bedside…
  • DNA extraction, PCR and sequencing on site
594
Q

What are the queries of lab on a chip?

A
  • But who will do the analysis?
  • Where?
  • How will the testing be controlled?
  • Who will interpret the results?
  • When does the science become medicine?
595
Q

What is the polyclonal antibody response?

A

We can use a polyclonal mixture of antibodies that will bind to various epitopes. These are presented when challenged with an antigen.

In this example there are two epitopes. When they are injected into the mouse we get an immune response, one B cell with a particular receptor will bind to an epitope, and the other will bind to the other epitope. They will both then proliferate to make antibodies for those epitopes.

596
Q

How are monoclonal antibodies produced?

A

Monoclonal antibody technology was developed in 1975. A mouse is injected with an antigen. B cells are then isolated form the mouse after it has developed an immune response and these are fused using chemicals with myeloma cells from a B cell tumour.

We get a mixrue of cells (some original B cells, some myeloma cells and some hybridoma cells). We select the hybridoma cells on HAT medium. The medium stops myeloma cells being able to grow, B cells are not immortal so die quickly, and this means that in the medium the only cells that can survive and grow are the hybridoma cells.

When we have this population of hybridoma cells we can dilute them out to individual cells which then divide to clonus cells. Each will secrete an antibody with the same specificity which bind to one type of epitope. They are monoclonal antibodies.

  • Hybridomas can be stored indefinitely and grown to produce monoclonal antibody when required
  • Antibody genes can be cloned from the hybridomas which allows antibodies to be engineered for different applications
  • Polyclonal or monoclonal antibodies can be produced which bind to Fc regions of particular antibody classes eg to IgG’s, IgA’s etc. These ae called anti-isotypic antibodies.
597
Q

About the label in immunoassays?

A
  • Originally radioactive – radioimmunoassay (RIA)
  • Commonly now enzyme eg horseradish peroxidase, alkaline phosphatase (usually detected by coloured product) – enzyme linked immunosorbent assay (ELISA)
  • Other alternatives are luminescent

One antibody is labelled. Used to be radioactive but as radioactivity is dangerous we now use enzymes. When you give these enzymes to substrates a coloured product will be produced which is measured. The alternative is illuminescence.

598
Q

About solid phase immunoassays…

A

Direct
• Often used to quantify an antibody

Capture or switch
• Often used to quantify an antigen

Using these assays, the concentration of analyte (antibody or antigen) in the sample can be calculated by comparison to analyte standards of known concentration.

599
Q

About direct ELISA…

A
  • Antigen immobilised on solid support
  • Test antibody solution added, incubated and non-bound removed by washing
  • Bound antibody detected by incubation with labelled 2nd antibody (ie anti-immunoglobulin) and non-bound removed by washing

Uses:

  • Screen hybridoma supernatants
  • Detect exposure to infectious agent

Second Ab is the detecting Ab. This has a label bound to it. The mixture is then washed and we add a substrate for the enzyme. We then measure the colour formed and correlate it’s intensity to the amount of Ag formed on the plate. We can look for HIV in the blood using this assay.

600
Q

About capture ELISA…

A
  • Antigens mat be present in low concentration. Because antibodies have high affinity for antigen this technique can concentrate the antigen.
  • Need tow antibodies reacting with different epitopes on the antigen
  • One antibody immobilised on solid support
  • Test antigen solution added, incubated and non-bound removed by washing
  • Bound antigen detected by incubation with the other antibody, which has been labelled and non-bound removed by washing.

We coat the antigen with antibody. This can be low in concentration. The body has a high affinity for Ag and can concentrate it – we need two Abs to bind to different epitopes. One is immobilised on a solid support. We add solution containing Ag to detect – this will bind to the antibody. We wash the mixture to remove any non-bound antigen. The second antigen is linked to the enzyme. We add the substrate an get a colour formed from the product again.

601
Q

How can we measure cytokine secretion with immunoassays?

A

We can measure cytokine secretion from T cells. We coat the bottom of the well with antibody that binds specific cytokine. We add activated T cells which are secreting cytokines. Once secreted, they will bind specifically with antibody in wells. They are captured by those antibodies and we wash off the cells and the second labelled antibody is added. We then add the enzyme and detect the colour formation as before.

602
Q

what can SDS PAGE assays be used for?

A
  1. Can be used to detect antigens or antibodies
  2. Used to measure size of the protein being analysed
  3. Can be sued to calculate protein concentration
  4. May show is protein has been degraded
603
Q

About SDS PAGE assays…

A

SDS-PAGE is often combined with western blotting. This can be use to measure the size of a protein or for antibody/antigen detection. We can calculate the concentration of the protein we are looking at. We can also demonstrate whether the protein has been degraded in anyway.
We start with a mixture of antibodies. We take the mixture, mix it with SDS and boil it up. SDS will bind to the protein and give a negative charge. We load this onto a gel and run it in an electric current. Proteins are negatively charged and will migrate to the positively charged electrode. We take the gel and stain it with something that binds to the protein. This shows us that it is a mixture of proteins generally, but it would be better if we could specifically identify what we are interested in so we can do a western blot.

We transfer this to nitrocellular paper by an electric current. We then add antibodies specific to the antigen to detect. Then we take the nitrocellulose blot paper and incubate it with a substrate of enzyme. We get a single band corresponding to the protein we are trying to detect. At the same time, we run protein standards in the gel.

  • SDS PAGE/Western blotting often used alongside ELISA
  • In WB, protein concentration can be measured by comparing intensity of band we are detecting to band from a protein standard of known concentration
  • If protein is degraded it mat be more useful to use WB to calculate protein concentration, as some of degradation fragments mat contribute to signal in ELISA if both coating and detecting antibody are bale to bind to them.
604
Q

How are antibody-antigen interactions used in the purification of immune cell subsets?

A

antibody coated magnetic beads.

Purification of immune cell subsets. Couple them to particles that are paramagnetic. We then take the Abs and they will bind specifically to particular lymphocytes because WBCs have certain proteins on their surface.

We then pour them onto an iron wall mesh and apply a magnetic field. The Ab bound to cells will bind to iron mesh, and cells not bound to Ab are washed out.

Then we stop applying the magnetic field and elute the cells to get a population of a particular kind of cell. Used to purify particular subset base don protein it has on its cell surface.

605
Q

About flow cytometry and FACS…

A
  • Individual cells within a mixed population are tagged by treatment with monoclonal antibodies which bind to surface molecules and are labelled with fluorescent dyes
  • Mixed cells are then forced through a nozzle to form stream of single cells
  • Individual cells pass through a laser beam which scatters light and causes dye to fluoresce and provides information on bound antibody and cell surface protein
  • A cell sorter can separate specific subpopulations of cells

Fluorochromes fluoresce when a light is shone on them in different colours. When we combine cells from a mixed population with monoclonal antibodies they bind to a particular marker on the protein. They are then forced through a nozzle converting the mixture into a single stream of cells. We pass that laser through the stream and get scattering which gives us information about the cell. When the laser hits a fluorochrome will fluoresce in certain colours. This is measured by a computer giving information about the mixture of cells which is then plotted on a dot plot. Each spot corresponds to a single cell and gives information about the particular fluorescence.

Going up both the x and y axis corresponds to increasing fluorescence. This shows that it is a fixed population of B cells. Some don’t fluoresce at all – showing they are potentially not B cells but T cells instead. Part will fluoresce heavily for M and D (both on surface), some more for D and some more for M.

We can combine this with FACS and this means when the cells pass through the single stream we can pick up cells fluorescing one way or another. We can isolate a particular population of cells and make the machine just do cells of D or M for example.

606
Q

What are types of samples that are analysed by biomedical scientists?

A
  • Blood serum
  • Blood cells
  • Urine
  • Synovial fluid
  • Saliva
  • Mucus
  • Cerebrospinal fluid
607
Q

What types of disease are analysed?

A
  • Transplant compatibility
  • Immunodeficiency
  • Autoimmunity allergy
  • Malignancy
608
Q

About MHC genetics…

A
  • In humans known as HLA (Human Leucocyte Antigens)
  • The MHC is located on chromosome 6 and contains 3 MHC class I proteins and 3 MHC class II proteins
  • Highly polymorphic – 100s of different variants
609
Q

How is flow cytometry used in monitoring HIV infection?

A

¥ Lymphocyte subset estimations are performed using monoclonal antibodies to: CD3, CD4 and CD8 on whole blood and analysed by flow cytometry
¥ The percentages of cells in each subset is determined using a FACS machine
¥ The results are reported as percentages and absolute counts

610
Q

About using MHC-peptides for tracking T cell responses…

A

MHC-peptide complexes made which bind specifically to the T cell receptor of appropriate MHC-peptide specific T cells. A fluorochrome e.g. phycoerythrin added and visualisation is by flow cytometry.

By creating MHC complexes loaded with HIV antigen we can calculate for example what proportion of CD 8+ T cells will bind to the antigen.

Immunodeficiency – analysing cell function

611
Q

About neutrophil deficiencies…

A

¥ Neutrophils are found in acutely inflamed tissue
¥ Ingest pathogens and kill using reactive oxygen species
¥ Rapidly die after phagocytosis which generates pus
¥ Deficiency in neutrophil numbers – neutropenia – high rate of infection
¥ Deficiency in phagocyte function; chronic granulomatous disease – patients cannot form reactive oxygen species, succumb to bacterial and fungal infections

612
Q

How can we measure neutrophil function?

A

neutrophil oxidative burst assay

This test is based on the principle that nonfluorescent DHR (dihydrorhodamine) 123 when phagocytosed by normal activated neutrophils (after stimulation with PMA – phorbol myristate acetate) can be oxidized by reactive oxygen species (ROS), produced during the activated neutrophil respiratory oxidative burst, to rhodamine 123, a green fluorescent compound, which can be detected by flow cytometry.

613
Q

What is nephelometry?

A

• Nephelometry: an automated and rapid method used to measure serum immunoglobulin levels; it relies on the light-scattering properties of antigen-antibody complexes

Nephelometry measures light refraction

  • Often used to study the amount of antibody from different classes present in serum eg IgG, IgA etc
  • Here, serum is mixed with anti-isotope antibodies.

We shine a light onto complexes and the light will be scattered. We can measure the scattering with a machine and this will give an idea about the amount of antigen present We can study the amount of antibody in different populations. We can mix the serum with different antibodies which will bind specifically to IgG or IgA

614
Q

The basis of allergy…

A
  • IgE responses predominate

- Common allergens: house dust mites, cat, dog, trees, grasses, moulds, egg, milk, cod, soya, peanut, etc

615
Q

How can we diagnose allergy?

A

Skin Prick Test: In allergic individual, IgE binds to allergen and via the Fc region of the antibody binds to receptors on mast cells. This causes mast cells to degranulate causing the release of mediators (e.g histamine) which causes reddening and swelling of skin

Degranulation of mast cells results in release of mediators eg histamine causing reddening and raised skin.

¥ RAST (RadioAllergoSorbent Test) The suspected allergen is bound to an insoluble material and the patient’s serum is added. If the serum contains antibodies to the allergen, those antibodies will bind to the allergen. Radiolabeled anti-human IgE antibody is added where it binds to those IgE antibodies already bound to the insoluble material. The amount of radioactivity is proportional to the serum IgE for the allergen

¥ Often fluorescence is used instead of radioactivity e.g. ImmunoCap

Take suspected allergen and bind to solid face. Take serum from patient and add to antigen. If there are Abs specific to the antigen in the serum, it will bind.

Ag that are specific bind to IgE and will be radiolabelled. We can detect the amount of radioactivity. Radioactivity is dangerous and so we are replacing it with fluorescence (immunoCap).

616
Q

How can we identify antibodies?

A

¥ Autoimmune disease characterised by autoantibodies to nuclear antigens eg DNA, RNA
¥ Detection of autoantibodies is useful for diagnosis and monitoring disease activity

e.g. Systemic Lupus Erythematous (SLE) Autoimmune disease characterised by production of autoantibodies causing a range of symptoms, often dermatological

617
Q

What is SLE immunofluorescence?

A

¥ ELISA (Quantitative) and immunofluorescence (Qualitative)
¥ Performed in parallel to increase sensitivity
¥ Immunofluorescence: serum added to human cell line. Then probed with fluorochrome-labelled anti-immunoglobulin antibody. Visualised by fluorescence microscopy
¥ 98% of SLE patients are Anti-nuclear antibody positive,

618
Q

How can polyclonal antibodies be used in therapies?

A

¥ Intravenous immunoglobulin (IVIG) is a blood product purified from the serum of between 1000-15,000 people/batch

¥ Used to treat patients with antibody deficiencies at 200-400mg/Kg/3 weeks
¥ At high dose 2g/kg/4 weeks used as an immunomodulatory agent in a number of immune and inflammatory disorders
¥ Used in neurology, haematology, immunology, nephrology, rheumatology and dermatology
¥ Other example: rabies: after a bite from a suspected rabid animal, polyclonal antibodies isolated from the serum of individuals who have been immunised with the rabies vaccine injected into the wound site – Human Rabies Immunoglobulin (HRIG)

Deficiencies – given the Ab IV at low doses
Immunomodulatory ages at higher doses

619
Q

How can monoclonal antibodies be used in therapies?

A

¥ Approx. 45 monoclonal antibodies are licensed to treat disease: cancer, chronic inflammatory diseases, transplantation, infectious diseases and cardiovascular medicine
¥ Monoclonal antibodies can have their effects either by binding and blocking a process or by mediating immune responses such as initiation of complement or antibody-dependent cell mediated cytotoxicity (ADCC)
¥ Molecules such as toxins or radionuclides can be joined to monoclonal antibodies: antibody binds to cancer cell which is then killed by toxin or radioactivity

620
Q

How can antibodies protect against pathogens?

A

One way of action is to bind to something an block the process, or alternatively to mediate processes such as the complement pathway.

The two processes are shown above; left Ab bound to Ag on a pathogen, by binding it initiates the complement pathway leading to killing of the pathogen. This is called the classical pathway but there are several that have evolved in initiating complement.

The right picture shows initiating cytotoxicity (?) bound to pathogen, binds to Fc receptor on surface of immune cell and stimulates NK cell and when activated goes on to kill the pathogen by inducing pathogen to undergo apoptosis.
Can take these Abs and attach various molecules to them eg. toxins or radioisotopes. This can be used to kill cancer cells. This will target the protein produced on surface of cancer cells. Binds to the surface protein on cancer cell and the toxin or radioactivity kills the cancer cell.

621
Q

What is molecular epidemiology?

A

A resolved measure (diversity) of differences (variables) that determines:

a. Disease distribution in time and place
b. Disease transmission
c. Disease manifestation
d. Disease progression

622
Q

In what ways can we confirm outbreaks?

A

Confirming outbreaks
• Inside institutions
- Did patient A catch this pathogen from patient B?
- Do patients A, B and C from the same hospital ward have the same strain?
• In the community
- Who was the index case and what is the likely source?
• In the past
- What has driven the geographical spread of important strains?
• In the lab
- Is this an outbreak or a contaminant?

623
Q

In what ways do we identify disease risks?

A

• Shifts in virulence
- Is the incidence of annual infections increased from… last year?
- Are drug resistance strains on the rise? From where?
• Reservoirs of infection
- New infection or recrudescence?

It is important to know how quickly the mutations occur and are fixed. We also need to know if these patients are newly infected patients, or recrudescence where it is latent for years and suddenly comes back.

624
Q

What is single weighting of diversity?

A
  • Presence or absence – biochemical test, presence of O157 antigen, presence of verotoxin
  • Addictive weighting – combination of single tests
625
Q

What is additive weighting?

A

combination of single tests

626
Q

What is multiple weighting?

A

Genomic factors

  • Factoral – presence or absence of a gene base/s change in genome/gene relative to location in the genome
  • Functional – type of substitution (synonymous/non-synonymous)
  • Temporal – mutation rate (time since the last alteration)
627
Q

What is a spoligotyping dendrogram?

A

Spoligotyping dendrogram showing relatedness of pattern

We can use maths to define how different one strain is from another. Strains are all different. We can produce an evolutional tree of he patterns of change between the strains and see the differences between them. We can track back via the tree to find the original strain, and the length of the line tells us how different it is from the other.

628
Q

What is a silent mutation?

A

mutations that are intragenic (between genes) or synonymous (not altering coding

629
Q

What are non-synonymous mutations?

A

substitutions causing coding to be altered

630
Q

What are corruptive mutations?

A

deletions or insertions (disrupting coding frame), creation of STOP codons (truncation), corruption of STOP codons (elongation), corruption of CONTROL sequences (eg promoters)

631
Q

What is genetic drift?

A

when the DNA changes – want to predict this and we can measure it easily

632
Q

What is genetic shift?

A

not so easy to predict, the DNA has caused the protein to change, DNA changes into something functional

633
Q

What factors affect the speed of the molecular clock?

A

• Replication rate: a high division rate provides a higher mutation rate
• DNA or RNA polymerase proof reading fidelity: some species (eg HIV) have low fidelity promoting high mutation rate
• Selection pressure from the host or environment: high selection pressure removes ‘weak’ mutants and emphasises clusters, loss of selection pressure allows deletions
• Degree of redundancy in the genome: multiple copies of a single gene in the genome allow for mutations in one copy without compromising overall functionality, movement or recombination within genome may not effect phenotype
Transmission rate: high transmission rates relative to the mutation rate results in dissemination and single strain outbreaks

Some things divide slowly so huge changes and mutations; others divide really slowly.

Some hosts are not susceptible to pathogens. But this doesn’t mean that the organism can’t grow in the host – they can be infected just not affected

634
Q

How do we measure genome re-arrangements?

A

Restriction fragment length polymorphism (RFLP)

Stage 1
Cut genome with low frequency cut restriction enzyme

Stage 2
Separate fragments on a gel and visualise with a probe against repetitive element

Result is a profile of the number of SNPs at each locus

635
Q

What are epidemiological associations?

A

a. Transmission: hospital acquired infection
b. Reservoirs of infection: contact tracing
c. Spread or emergence of resistance

636
Q

What are drug resistant polymorphisms?

A

These are resistant to every drug – MDR TB is coming! It is currently mainly in Russia but is being brought into the UK.

637
Q

What does choosing the dos appropriate system require?

A

Molecular epidemiology offers a variety of methods to test questions involving disease transmission, strain virulence, pathogen evolution.

Choosing the most appropriate system requires:

  1. Knowing the most appropriate variable/s
  2. Quantitating variations and deriving diversity
  3. Generating identities or clusters
  4. Applying related data
    - Geographic location
    - Time of isolation
    - Incidence
    - Prevalence
    - Transmission rate
    - Severity of disease