ICS Flashcards

1
Q

What is the main cell involved in acute inflammation and what are the 3 stages of migration?

What happens at the site of inflammation?

A
  • Margination, adhesion (selectins), emigration/diapedesis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Differences between an arterial and venous ulcer

A

Location
Arterial- usually on the tips of toes and lateral malleolus of ankle
Venous - usually on the medial malleolus and inner calf

Exudate
Arterial - punched out hole (deeper) with little exudate
Venous - less demarcated punched out hole appearance (shallower) with a lot of exudate

Colour
Arterial - pale cool skin (or yellowish-grey base)
Venous - Reddish base

Skin changes
Arterial - hair loss, thickened toenails, weak distal pulse of affected limb
Venous - Varicose veins, itchy skin, eczematous (stasis dermatitis)

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

What is virchow’s triad and what are its components?

A

The formation of a thrombus is dependent on any one of Virchow’s triad being present

1) Abnormal blood flow (stasis/decreased blood flow) - due to e.g. periods of immobility (long flights/being bed bound) –> Most common caused of DVT

2) Abnormal blood components (Hypercoagulability - excessively easy clotting of blood) –> alterations in the constitution of blood caused by smoking, sepsis (reaction to an infection), malignancy/cancer – can be due to genetics also - Gene mutations like in essential thrombocythemia (high number of platelets in blood)

3) Abnormal blood vessel wall (Endothelial injury) –> can be from NICOTINE (smoke), atheroma formation (Fatty substance build up in arteries), inflammatory response, surgery, direct trauma, etc.

atheroma = plaque

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

What are plaques that build up in arteries composed of?

A

Lipid, smooth muscle, macrophages + foam cells (macrophages that ingest LDLs), platelets, calcium, fibroblasts, T lymphocytes

Lipid, necrotic core, connective tissue, fibrous cap.

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

What is atherosclerosis?

A
  • The accumulation of fibrolipid plaques in systemic arteries. It narrows the arteries, reducing blood flow to important areas and thus cause illness e.g. MI of the heart

Take note: not really found in low pressure systems (pulmonary arteries). common in high pressure systems (aorta/systemic arteries)

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

Describe the formation of an atherosclerosis

Look into lectures about smooth muscle cap.

A

1) Fatty streak –> Precursor turns into plaque (late teenage/early 20s) (consists of lipid laden macrophages and T lymphocytes within the INTIMAL layer of the vessel wall)

2) Lipid accumulation –> Endothelial damage initiates an INFLAMMATORY response. This results in monocytes and macrophages being recruited to phagocytose LDLs to become foam cells, which contribute to plaque formation.

3) Platelet aggregation (due to damage of endothelial lining) –> Accumulated lipids lead to plaque protruding into the artery lumen, causing platelet aggregation

4) Smooth muscle cells contribute to the formation of the fibrous cap over the plaque by releasing fibroblast growth factor producing collagen and elastin. (stabilising the plaque - stable atheroma)
atheroma=plaque

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

What are some risk factors for atherosclerosis?

A

Smoking, diabetes, hypertension, obesity, hyperlipidemia increased age, males

(Also risk factors for MI)

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

Name a few possible complications of atherosclerosis

A
  • Cerebral infarction
  • Myocardial infarction
  • Emboli (when pieces of the atherosclerotic plaque break off and travel downstream to smaller vessels) –> can cause transient ischemic attacks or cerebral infarcts if in the carotid artery (carotid atheroma)
  • Aortic aneurysm (if atherosclerosis occurs in the aorta, it can weaken the wall of the aorta causing it to be less elastic and causing turbulent blood flow.
  • Gangrene (lack of blood flow and lack of oxygen to extremities can lead to necrosis)- body tissue dies (usually starts in the toes)
  • Intermitted claudication (muscle pain due to lack of oxygen)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

State 4-5 differences been apoptosis and necrosis

for point 4 need to check with lectures

A

Apoptosis –> Non inflammatory, programmed cell death
Necrosis –> Inflammatory, traumatic cell death (due to injury, disease initiated, etc)

Apoptosis –> Cell membrane remains intact
Necrosis –> Disrupted cell membrane (loss of membrane integrity)

Apoptosis –> Cell shrinks (nucleus condenses)
Necrosis –> Cell swells (and bursts)

Apoptosis –> Chromoatin is unaltered. Pyknosis (condensation of chromatin) and Karyorrhexis (fragmentation of the nucleus)

Necrosis –> Chromatin is altered - degrades and fragments(could mutate). Pyknosis, karyorrhexis and karyolysis occurs (dissolution of cytoplasm)

Apoptosis –> Energy dependent process (and therefore controlled)
Necrosis –> Energy independent

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

How does a cell decide to apoptose?

Why does this not work in cancer cells?

What happens in HIV?

A
  • Via the amount of DNA damage within the cell. p53 is a protein that can detect DNA damage and can then trigger apoptosis.
  • In cancer cells, there is a mutation of the p53 protein so it can no longer detect DNA damage and induce apoptosis.
  • In HIV, the HIV virus can induce apoptosis in CD4 helper cells which reduce their numbers, resulting in an immunodeficient state.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What is atherosclerosis?

A
  • The accumulation of fibrolipid plaques in systemic arteries. It narrows the arteries, reducing blood flow to important areas and thus cause illness e.g. MI of the heart

Take note: not really found in low pressure systems (pulmonary arteries). common in high pressure systems (aorta/systemic arteries)

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

Describe the apoptosis intrinsic pathway.

A

Occurs due to internal stimuli (DNA damage, biochemical stress, lack of growth factors)

  • Pathway modulated by the molecules Bcl-2 (inhibits Bax) and Bax (promotes cytochrome C release)
  • Cytochrome C activates caspases - the executioners of apoptosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Describe the apoptosis cytotoxic pathway

A

Cytotoxic T cells release granzyme B and perforin which activate caspases for apoptosis. (perforin creates pores or channels in the target’s cell membrane for granzyme B to enter the cell and activate caspases)

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

Define hypertrophy and hyperplasia

A

Hypertrophy –> Increase in size of an organ caused by an increase in the size of its constituent cells (without an increase in number) - skeletal muscle in atheletes

Hyperplasia –> An increase in the size of an organ due to an increase in the number of its constituent cells - BPH

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

What is the hayflick limit and why is it a thing?

A

Hayflick limit - The limit to which a human cell can divide.

There is a limit because at each cell division, the telomere region at the end of chromosomes shortens and eventually becomes so short that it is not possible for chromosomes to divide and replicate. (telomere length appears to be paternally inherited)

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

Definition of carcinogenesis and neoplasm

A

Carcinogenesis –> The transformation of normal cells into neoplastic cells (cancer cell) through permanent mutation leading to uncontrolled/abnormal growth (possibly into a tumour subsequently)

Neoplasm –> The Autonomous, abnormal, persistent new growth of cells

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

What does a neoplasm consist of?

A

Stroma and neoplastic cells

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

Carcinoma definition

A

Malignant epithelial neoplasm

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

What are the characteristics of a neoplastic cell?

A

Autocrine growth stimulation - due to overexpression of growth hormone and mutation of tumour suppression genes. - very fast growth

They can evade apoptosis

They have telomerase - Prevents telomere shortening with each replication

Sustained angiogenesis - They have their own blood supply

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

Difference between benign and malignant tumour (behavioural classification of neoplams- benign, borderline, malignant)

A

Malignant - Endophytic, poorly circumscribed, ulceration, hyperchromic nuclei
Benign - Exophytic, well circumscribed, rare ulceration, normal shaped nuclei

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

What are tumours called when they are non/glandular benign/malignant?

A

They are Epithelial tumours

Papilloma - non glandular benign
Carcinoma - non glandular malignant
Adenoma - glandular benign
Adenocarcinoma - glandular malignant

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

State some classes of carcinogens

A
  • agents capable of causing cancer

1) Chemicals - e.g. paints, dues, rubber, soot. (most require metabolic conversion from pro-carcinogen to ultimate carcinogens)
2) Viruses - e.g. HPV (cervical cancer), EBV (burkitt lymphoma) - causes 10-15% of cancer
3) Ionising + non-ionising radiation - UVB (or UVA) (skin cancer) - basal cell carcinoma, squamous cell carcinoma (increased risk of xeroderma pigmentosum)
4) Hormones, parasites, mycotoxins - mycotoxins can be found in food/fungi/mold (aflatoxin - hepatocellular carcinoma)
5) Asbestos (fibrous minerals that are microscopic and can be inhaled)

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

What are dendritic cells and what is their origination?

A

Dendritic cell - Antigen presenting cells that initiate the adaptive immune response.

They are mesenchymal in origin and not hematopoietic.

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

What are primary and secondary lymphoid organs (name some examples)?

A

Primary –> Where immune cells originate, mature or develop
- Bone marrow (all cells originate here including T cells) - B cells mature here
- Thymus - development and maturation of T cells (thymic tolerance)

Secondary –> Where immune responses are initiated and coordinated
- Lymph nodes - antigen presenting cells and T/B cell interactions –> filters lymphatic fluid allowing immune cells to encounter and respond to antigens
- Spleen - RBC recycling, bacteria killing - it has lymphocytes and macrophages which can respond to infections

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

What is thymic tolerance and where does it occur?

A

A critical process that occurs in the thymus where T cells are developed to recognise and respond to foreign antigens while remaining tolerant to the body’s own tissues thus preventing autoimmune reactions. (T cell selection)

Positive selection - If the T cells recognise the thymus Major histocompatibility complexes (MHCs 1 and 2), then they are selected for

Negative selection - If the T cells produce an immune response (by recognising self antigens as foreign, they are selected against.)

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

Cytokines secreted by T helper cells 1 and 2

A

1 - interferon gamma - activates NKC and marcophages

helper cell 2 - interleukin 4,5,,,10 - activates b cells to differentiate into plasma cells

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

Examples of Type 1,2,3,4 hypersensitivity reactions

A

1 - Anaphylaxis, hay fever, allergies
2- Blood transfusion reactions, graves, hashimoto’s thyroiditis, good pasture’s, guillain barre, lambert eaton

3 - Post strep glomerulonephritis, SLE, farmer’s lung, rheumatoid arthritis
4) TB, MS

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

What is a naive T cell?

A

A T cell that has not encountered an antigen or has not matured

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

3 differences between innate and adaptive immunity

A

Innate - non specific, rapid response with no memory
Adaptive- specific, slow response with memory involved

Innate - Neutrophils and macrophages primarily involved
Adaptive - T cells and B cells are primarily involved

Innate - Killing via complement system
Adaptive - Killing is antibody mediated

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

What are symptoms that may present with anaphylactic shock (an acute medical emergency)?

A
  • Severe hypotension
  • Tachycardia + dyspnoea
  • Pale
  • Cold extremities
  • Puffed up face + tongue
  • Itching
  • Urticaria (Hives)
  • Central cyanosis (lips are blue)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

What is immune tolerance?

A

A safeguard mechanism to prevent the production of autoreactive cells (where autoreactive B and T cells infiltrate and attack healthy tissues and organs - producing antibodies with high affinity for self antigens)

Split into

Central tolerance - which occurs in primary lymphoid organs. Thymus for T cells and bone marrow for B cells (where they mature)

Peripheral tolerance - occurs in secondary lymphoid organs e.g. Spleen (in the event faulty T and B cells evade central tolerance)

32
Q

Immunodeficiency can be acquired or inherited, give examples of both.

A

Inherited (genetic) - Defects in T cells e.g. SCID- severe combined immunodeficiency (caused by adenosine deaminase deficiency, where infants lack the enzyme required for T-cell survival)

Acquired- HIV (Unprotected sex, sharing injection drug equipment)

33
Q

What are covid 19 vaccines (pfizer and moderna) made of?

A

mRNA, (ACE 2 receptors)

34
Q

What are examples of active natural, passive natural, active artifical and passive artificial immunity?

A

Active natural - Body encounters pathogen and produces memory cells against it (after the infection)

Passive natural - Maternal antibodies/immunoglobulins are passed onto feeding baby in breast milk

Active artificial - Vaccine mimics encountering pathogen and stimulates immunoglobulin production

Passive artificial - Antivenom (injection of immunoglobulin from another organism)

35
Q

What are the 4 main drug targets?

A

1) Receptors (Majority)
2) Enzymes
3) Transporters
4) Ion channels

36
Q

Where drugs target receptors, the ligands can either be agonists or antagonists. What is the difference in terms of affinity and efficacy with the receptor?

What is the most common type of receptor?

A

Agonists - full affinity (binds well to receptor) and full efficacy (fully activates the receptor)

Antagonists - full affinity and zero efficacy (inactivates the receptor)
(Can competitively or non competitively inhibit receptors)
(irreversible antagonist – won’t come off the receptor and will never become available for an agonist)

Most common receptors are G-protein coupled receptors.

37
Q

What is the definition of efficacy and potency?

A

Efficacy- The maximum effect a drug can produce, regardless of dose (the ability of a drug receptor complex to produce a maximum functional response)

Potency - How much of a drug is needed to elicit a response in the body

38
Q

Definition of pharmacokinetics and pharmacodynamics

A

The fate of a chemical substance administered to a living organism

((It examines how drugs are absorbed, distributed, metabolised and excreted in the body. ))(what the body does to the drug)

(Pharmacodynamics - what the drug does to the body)

39
Q

What are the symptoms of a cholinergic crisis, when does it happen? (also symptoms of organophosphate poisoning)

A

It happens when there is an increased level and duration of cholinergic stimulation (due to inhibition of acetylcholinesterase enzyme) - basically has an agonist effect. (antagonist effect with TCA drugs will have opposite effect of sludge)

SLUDGE
Salivation
Lacrimation
Urination
Defecation
GI distress
Emesis (vomiting)
—— These are all due to excessive parasympathetic nervous stimulation.

Can also lead to severe muscle weakness, paralysis and respiratory distress.

TREAT WITH ATROPINE (as it crosses BBB can cause delirium, confusion and hallucination)

40
Q

Noradrenergic pharmacology

A

Tyrosine –> DOPA –> Dopamine –> Noradrenaline –> Adrenaline

41
Q
A
42
Q

What does activation of Alpha 1, alpha 2 beta 1 and beta 2 receptors result in?

A

Alpha 1 - Blood vessels and sphincter –> vasoconstriction, bladder contraction and pupil dilation

(alpha 2 - negative feedback - suppresses noradrenaline (catecholamine) release)- slows heart rate

Beta 1 - Heart. –> Agonism increases inotropy (force of contraction of the heart) and increases BP and renin release.

Beta 2 - Lungs. –> Agonism causes bronchodilation (e.g. SABAs and LABAs)

43
Q

What are the 3 steps of the pain ladder?

Which medication are used for mild, moderate and severe pain?

A

First step - Mild pain –> Non-opioid analgesic with or without adjuvant therapy

Second step - mild to moderate pain –> Weak opioids (hydrocodone, codeine, tramadol) with/without non opioids, with/without adjuvant therapy

Third step - Moderate to severe pain –> Strong opioid (morphine, oxycodone), with/without non-opioid analgesics, with/without adjuvant therapy

Mild - Paracetamol, ibuprofen
Moderate - Naproxen, tramadol
Severe - Morphine

44
Q

What is the Rawlins and Thompson classification of an adverse drug reaction? (types of reaction)

What should you do if you notice an adverse drug reaction?

A
  • All adverse drug reactions should be reported by MHRA yellow card scheme. (ALL ADRs should be reported for black triangle medicines)

Type ABCDEFG

Type A- Augmented
- Expected (seen in clinical trials)
(e.g. Bradycardia with beta blockers)

Bizarre -
- Unpredictable response
(Anaphylaxis with penicillins, bleeding with anticoagulants)

Chronic -
- ADRs that continue after the drug has been stopped
- Related to cumulative dose.
(Heart failure with pioglitazone)

Delayed -
ADRs that becomes apparent some time after use of the drug. (Has the patient used the drug in the past that could be an issue now?)
(Leucopenia with chemotherapy)

End of use - (withdrawal) –>
- ADR develops after the drug has been stopped
- Is the patient withdrawing from the drug?
(Insomnia after stopping benzodiazepine)

Failure of treatment
- Unexpected treatment failure (due to drug-drug interaction or drug-food interactions)
- Poor compliance with administration instructions
(Failure of DOAC due to enzyme inducers like carbamazepine)

Genetic
- Drug causes irreversible damage to genome
(Phocomelia in children of women taking thalidomide)

45
Q

What are some steroid side effects?

Check in lectures

A

CUSHINGOIND MAP

Cataract, Ulcer, striae (abdominal), hypertension, increased infection risk, necrosis of bone, growth restriction, osteoporosis, myopathy, pancreatitis.

46
Q

Examples of chemical carcinogens

A

Most chemical carcinogens require metabolic conversion from pro-carcinogen to ultimate carcinogens. (depending on where these enzymes are, the carcinogens can be spread?)

Polycyclic aromatic hydrocarbons
- Lung cancer (smoking) and skin cancer (mineral oils)

Aromatic amines
- bladder cancer (rubber/dye workers)

Nitrosamines
- Gut cancer (processed meat)

Alkylating agents
- leukemia (chemotherapy)

47
Q

5 places that human papillomavirus can cause cancer

A

Human papillomavirus (HPV) - Squamous cell carcinomas of the (CAPN’H) - Cervix, anus, penis, neck and head

48
Q

What is a neoplasm?

A

Also known as a tumour. A LESION resulting from the autonomous abnormal growth of cells that persist after the initiating stimulus has been removed.

Neoplasia - the autonomous, abnormal, persistent new growth of cells.

(A new growth) (25% of the population develops neoplasms)

49
Q

What are the 2 types of genetic drivers of carcinogenesis? Give examples of each

A

Proto-oncogenes - genes which promote cell growth and survival, promoting carcinogenesis e.g. myc.

Tumour suppressor gene - genes which inhibit cell growth and proliferation, playing a role in inhibiting carcinogenesis. e.g. BRCA, P53

50
Q

Describe the 5 step process of metastasis (spread of tumour)

A

5 step process
1) The tumour breaches the basement membrane (requires enzymes e.g. proteases and cell mobility)
2) Intravasation
3) Survival in the circulating system- A) Aggregate with platelets B)Shed surface antigens which lymphocytes pickup or C) Adhere to other tumour cells to resist the immune response
4) Extravasation
5) Growth at metastatic site

51
Q
A
52
Q

What are the commonest causes cancer deaths in males and females?

A

Males - Lung (commonest), prostate, bowel

Females - Lung, breast, bowel

53
Q

What are some problems with tumour hypoxia?

A
  • Hypoxia is a prominent feature of malignant tumours
  • When the blood supply is unable to keep up with the growing tumour cells, hypoxic tumour cells can adapt to low oxygen.

Tumour hypoxia is a poor patient prognosis
- It stimulates new vessel growth
- Suppresses immune system
- Resistant to radio and chemotherapy
- Increased tumour hypoxia after therapy

54
Q

What are the 3 traditional types of vaccines? Explain them

A

Whole-killed (inactivated) - Uses the killed version of the germ causing the disease (must be grown in-vitro so is expensive) - polio, influenza, hep A

Live attenuated - Uses a weakened form of the pathogen causing the disease - measles, mumps, rubella

Toxoid - Uses an inactivated toxin made by the germ causing the disease - diphtheria, tetanus

55
Q

Describe live-attenuated vaccines. Advantages + limitations. Examples.

A
  • Uses a weakened form of the pathogen that causes the disease (weakened via multiple mutations)
  • Better immune response so lower doses are required
  • Route of administration may be more favourable (oral)
  • Fewer doses are required to provide protection

However

  • Impossible to balance attenuation and immunogenicity
  • Possibility of the pathogen reverting to its virulent form (can cause the disease)
  • Transmissibility - Can be passed to non-vaccinated individuals.
  • May not be safe for immunocompromised individuals. (people with weakened immune system)

E.g. Measles, mumps, rubella, smallpox, chickenpox

56
Q

1 pro and 1 con of a viral vector vaccine

A

e.g. Oxford AstraZeneca

Pro
- Effective in delivering therapeutic genes into target cells

Con
- Pre-existing immunity in individuals due to previous exposure to the virus being used as a vector can limit the effectiveness of the therapy.

57
Q
A
58
Q

What are recombinant vaccines?
Examples?
1 advantage and 1 disadvantage

A

These vaccines are genetically engineered (using specific parts) of bacteria, yeast, insect or mammalian cells)

  • Avoids the problem of needing to grow the pathogen in vitro.
  • However it is difficult to find a protein that is protective and can generate a strong enough immune response.
  • e.g. Hepatitis B, HPV, SARS-Co-V2
59
Q

How do mRNA vaccines work? Advantages?

A

mRNA of the target foreign protein is synthesised in vitro (from a DNA template). It is complexed with lipid nanoparticles that stabilise and protect the mRNA from degradation and allows the mRNA to cross the plasma membrane. The mRNA is translated in the cytoplasm and the protein is presented on the surface of the cell with MHC, stimulating the immune response.

  • Avoids the need to grow the pathogen or viral vector
  • No live organism involved
  • mRNA is relatively cheap to produce

Con
- Requires extremely cold storage conditions

e.g. Pfizer, moderna

60
Q

Conjugate vaccine - describe

A

Haemophilus influenzae type b

uses polysaccharide antigens from the bacteria combined with a protein to stimulate the immune system.

61
Q

What are the 3 modes of action of complement proteins? What are they secreted by?

A

A group of 20 serum proteins secreted by the liver. They need to be activated to be functional.

1) Direct lysis
2) Attract more leukocytes to site of infection (chemoattract)
3) Coat invading organisms (opsonisation)

62
Q

Which are the complement proteins that allow for each of the steps of the complement cascade to occur (other than inflammation)

A

Lyse microbes directly - MAC (Membrane attack complex - final step of complement cascade)

Chemotaxis - C3a and C5a (IMPORTANT)

Opsonisation - C3b (inserts itself into the membrane of bacteria) - IMPORTANT

63
Q

What are the 2 regions of antibodies and what are their roles?

A

Fab regions (the two outgoing portions in a Y) – ANTIGEN RECOGNITION
- Variable in sequence
- Bind different antigens specifically (antigen binding sites)

Fc region (the single bottom port of the letter Y) - ANTIGEN ELIMINATION
- Constant in sequence
- Binds to complement Fc receptors on phagocytes, NK cells

64
Q

What are pattern recognition receptors (PRRs)?

Which cells are they expressed by?

A

They are proteins of the innate immune system which play a role in recognising molecular patterns associated with pathogens. These include pathogen associated molecular patterns (PAMPS) and damage associated molecular patterns (DAMPS)- released during tissue damage or stress.

PRRs are expressed in cells such as dendritic cells, macrophages, monocytes and neutrophils.

Upon binding to a specific ligand they initiate signalling cascades that lead to immune responses (inflammatory pathways, production of antibodies, recruitment of immune cells)

65
Q

What are some examples of PAMPS?
Name 2 membrane bound PRRs and 2 cytoplasmic sensor PRRs
(what do the receptors recognise?)

A

Pathogen associated molecular patterns –>
- Bacterial carbohydrates (Mannose)
- Nucleic acid (bacterial DNA)
- Bacterial peptides (flagellin, microtubules, peptidoglycan)

Above no need to know in depth.

Membrane bound PRRs
- Toll like receptors
- C type lectin receptors (recognise particularly mannose)

Cytoplasmic sensor PRRs
- Nod like receptors (- detects microbial infections or cellular stress - upon activation, they lead to the release of pro-inflammatory cytokines (interleukin-1)
- RIG-I-like receptors (primarily involved in detecting viral RNA. When activated, they trigger signalling pathways that lead to the production of interferons and other antiviral molecules)

66
Q

What do Toll like receptors (TLR) 2,4,5,7 and 9 detect?

Which are intracellular which are extracellular?

A

2- Peptidoglycans in gram positive bacteria (components of the cell wall of gram positive bacteria)

4- Lipopolysaccharides (components of outer membrane of gram negative bacteria)

5- Flagellin (for bacterial motility)

7 - Single stranded RNA from bacteria

9 - Non-methylated DNA

Intracellular - 3,7,8,9
Extracellular- 4 and 5

67
Q

What are the 3 activation pathways of the complement cascade?

A

Classical - Antibodies bind to antigens via FAB portion (antigen-antibody complex) –> (complement protein binds to fc portion of the antibodies –> triggers complement cascade (MAC-lysis, chemotaxis via inflammation, opsonisation which enhances phagocytosis)

Alternative - Complement protein C3b can bind directly to the antigen (of microbe) –> complement cascade

Lectin pathway - mannose-binding lectin to mannose (on the surface of bacteria –> complement cascade

All 3 pathways converge at the level of C3 activation, leading to the formation of the membrane attack complex (lyse microbes directly)- final stage of complement cascade

68
Q

What are the 3 antigen presenting cells?

A

Dendritic cells, macrophages and B cells

69
Q

Examples of Pattern recognition receptors mutations (2)

A

Non functioning mutations in the NOD2 gene can lead to Crohn’s disease

Gain of function mutations in TLR7 is associated with SLE

70
Q

Where are weak acid and weak base drugs absorbed best? Why?

A

Weak acids are better absorbed in the stomach. Because the stomach is an acidic environment, weak acids are more likely to remain in their non-ionised form (good lipid solubility) - thus well absorbed

Weak bases are better absorbed in the intestine because the small intestine has a more alkaline environment allowing weak bases to remain in their non-ionised form (good lipid solubility –> well absorbed)

71
Q

What are hallmarks of ageing (on a systemic, cellular and molecular level)

A

Systemic
- Nutritional dysregulation (reduced nutrient sensing can lead to metabolic dysfunction)

Cellular level
- Cellular senescence (cells lose the ability to divide and undergo functional changes–> senescent cells can accumulate in tissues over time and contribute to inflammation and tissue dysfunction) - due to telomere shortening and dna damage
- Stem cell exhaustion (stem cells undergo a decline in their ability to proliferate and differentiate, impairing tissue renewal)

Molecular level
- Telomere shortening - leads to cellular senescence (telomeres gradually shorten with aging)
- Compromised autophagy (a process responsible for the degradation and recycling of damaged cellular components –> reduced autophagy contributes to the accumulation of damaged proteins and organelles within cells.
- Loss of proteostasis –> process regulating proteins within the cell –> leads to accumulation of misfolded or aggregated proteins
- Epigenetic alteration – DNA methylation can affect gene expression and contribute to age related phenotypes
- Mitochondrial dysfunction - dysfunction (due to accumulation of ROS) can lead to increased oxidative stress

72
Q

What are the functions of M1,M2,M3 receptors and nicotinic receptors?

A

M1 receptor (CNS and periphery) - Increases gut motility and secretions in the gut, also CNS effects (cognition, learning, memory)

(M1,M4,M5 - brain + spinal cord (CNS))

M2 receptor (mostly in heart) - Reduced contractility and bradycardia

M3 receptor (mainly smooth muscles and glands) - vasodilation, bronchoconstriction, pupil constriction, salivary glands, bladder.

Nicotinic receptors - Primary role in synaptic transmission in the CNS and at neuromuscular junction (some vasodilatory effects)

73
Q

What are the 4 compartments that a drug can distribute through in the body?

A

Plasma, interstitial fluid, intracellular fluid and fat.

74
Q

How would you identify and avoid drug interactions?

Which patients would be at high risk of drug-drug interactions?

A
  • Obtain a complete drug history
  • Look out for high risk drugs with a narrow therapeutic index/window
  • Look out for new drugs

Patients may be at high risk of drug interactions due to:
- Polypharmacy
- Kidney or liver impairment
- Extremes of age

75
Q
A