Immunology Flashcards

1
Q

Aulus Cornelius Celsus defined the cardinal symptoms of inflammation in his 1st century BCE book De Medicina. What are they (4)?

A

redness (rubor), swelling (tumor), heat (calor) and pain (dolor).

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

Give the causes for each of Celsus’ 4 cardinal symptoms of inflammation

A
  • Redness – increase in local blood flow caused by vasodilator action of inflammatory mediators
  • Swelling – increase in vascular permeability, proteins and fluid leak from vasculature
  • Heat – as for redness
  • Pain – direct action on nociceptors (sensory neurones tuned to detect noxious stimuli)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Give 3 causes of inflammation

A

noxious stimulation
infection
autoimmune reaction

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

What is the triple response of Lewis

A

after drawing a pointed object across the skin:
within seconds- flush
30 to 60 secs after - flare
within minutes - wheal

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

What causes the flush response in Lewis’ triple response to injury

A

within seconds of injury, a band of redness that matches the size and shape of the stimulus appears due to local release of vasodilator substances, such as histamine, from cells disturbed by the stimulus causing dilation of capillaries

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

What happens during flare (Triple response of Lewis )

A

reddening spreads to the surrounding area because of an axon reflex (neurogenic inflammation)

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

What happens in neurogenic inflammation

A

stimulated branch of a sensory nerve fibre will send an action potential to the branching point, an action potential travels orthodromically to the spinal cord giving rise to the sensation of pain and antidromically along collateral branches resulting in the release of vasodilatory substances, which causes vasodilatation of the surrounding arterioles

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

What happens to the inflammatory flare response if you section a central axonal branch from the injured area

A

sectioning of the central branch will prevent perception of the stimulus, but the flare will continue to occur until the nerve degenerates

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

What happens during wheal (Triple response of Lewis )

A

localised swelling occurs within a few minutes of the stimulus due to histamine causing increased vascular permeability

leakage of plasma from the blood to the extracellular space results in swelling due to increased tonicity of plasma compared to normal ECF

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

Do you have to cut someone to replicate the triple response of Lewis?

A

no

responses can be largely replicated by injection of histamine (effects are mediated by H1 receptors)

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

Noxious stimulation of a sensory neuron sends APs to CNS and also to other fibre branches. What do these branches release

what process is this?

A

calcitonin gene related peptide (CGRP) and substance P (SP), which are able to directly cause vasodilatation and SP is a potent activator of mast cell degranulation

neurogenic inflammation

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

What does production of SP in neurogenic inflammation result in

give a piece of experimental evidence of this

A

vasodilation and degranulation of mast cells, which leads to the local production of histamine resulting in both vasodilatation and increased vascular permeability

mast cell deficient mice display no increased vascular permeability after intradermal injection of SP

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

What is D
dermatographic urticaria

what is the cause

how is it treated

A

a condition in which the triple response is exaggerated;

largely idiopathic

generally treated with H1-receptor antagonists, but it is also treated with the monoclonal antibody omalizumab, which recognises IgE and acts to decrease mast cell activation

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

How does bacterial/viral/fungal infection cause inflammation (3)

A

directly cause inflammation through release of toxins, which cause inflammation or by lysing host cells that liberate inflammatory factors, such as ATP.

also activate the innate and adaptive immune systems

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

Give an example of a bacteria directly causing inflammation through toxin release

A

alpha-haemolysin secreted by uropathogenic Escherichia coli induces Ca2+ oscillations in cells that cause synthesis of the proinflammatory cytokines interleukins IL-6 and IL-8

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

What are the two key components of the host’s immune response to pathogens

A

1) innate response – first line of defence, non-adaptive, developed early in evolution, present in some form in many multicellular organisms
2) adaptive response – second line of defence, physical basis of immunological “memory”, emerged later in evolution, only present in vertebrates

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

What are PAMPs

A

products of bacteria, viruses, fungi etc. that they are unable to readily change to avoid detection.

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

What are the best studied PRRs

A

Toll-like receptors (TLRs)

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

Why are they called Toll Like Receptors

A

the first toll gene was identified in Drosophila melanogaster as necessary for establishing the dorsal-ventral axis, the underdeveloped ventral portion of fruit fly larvae mutant for toll evoked the exclamation, “das ist ja toll!” (“that’s amazing!”) from Christiane Nüsslein-Volhard

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

How many TLRs are there

what do they do upon activation

A

~10

activation recruit various adaptor molecules and trigger signalling cascades that result in increased expression of pro-inflammatory cytokines and interferons

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

On which part of the cell are TLRs expressed

A

. Some TLRs are expressed at the plasma membrane like TLR4, which detects lipopolysaccharide, but others are expressed intracellularly in endosomes and generally detect DNA/RNA, e.g. TLR8 detects ssRNA.

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

Give 3 examples of sentinel cells

A

macrophages, dendritic cells and mast cells

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

What happens when TLRs on sentinel cells are activated by PAMPs

A

initiates production of a variety of proinflammatory mediators, which varies depending upon the cell type activated

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

Name some proinflammatory mediators released from sentinel cells upon activation of TLRs from PAMPs

A

prostaglandins (PGE2/PGI2 both cause vasodilatation),

histamine (vasodilatation and vascular permeability)

the cytokines tumour necrosis factor alpha (TNFα) and IL-1, which induce the production of further cytokines, vascular permeability and expression of adhesion molecules on the intimal surface of postcapillary venules.

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

Generally how do leukocytes leave the blood vessel to reach the site of infection

A

Leukocytes adhere to endothelial cell adhesion molecules, which enables them to migrate out of the vascular system and attack pathogens, a process aided by pathogen generated chemotaxins and host chemokines whose expression the pathogen has induced.

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

Components from which 4 systems does inflammatory exudate contain

A

complement system, coagulation system, fibrinolytic system and kinin system

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

How is factor XII activated

what is the product and its role?

A

Factor XII becomes activated upon contacting negatively charged substances such as collagen, producing Factor XIIa, which results in the production of thrombin, plasmin and bradykinin

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

Give an example of the coagulation, fibrinolytic and complement system interacting

A

Factor XII becomes activated upon contacting negatively charged substances such as collagen, producing Factor XIIa, which results in the production of thrombin, plasmin and bradykinin. Thrombin and plasmin hydrolyse the complement protein C3 to produce active C3a and C3b

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

What converts fibrinogen into fibrin

A

thrombin

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

What are the roles of C3a and b

A

C3a stimulates mast cells,

C3b attaches to pathogens aiding their destruction by white blood cells and cleaves C5 into C5a and C5b

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

Give 3 roles of C5a

A

activates mast cells, is chemoattractant for white blood cells and also activates them

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

Describe the roles of C5-9

A

form a membrane attack complex that can attach to bacterial membranes and induce lysis (one subunit of C5b, C6, C7, C8 and 12-18 C9 subunits)

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

Which cells are usually first to the site of injury

A

neutrophils

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

Give the 5 steps required for neutrophils to exit the vascular system

A

1) PRRs detect PAMPs causing release of TNFα and IL-1 inducing expression of adhesion molecules (e.g. selectins) on the endothelium
2) Neutrophils initially tether to and are captured by the endothelium via interaction between endothelial P-selectin and neutrophil expressed ligands, such as P-selectin glycoprotein 1 (PSGL1). involves interaction between neutrophil expressed integrins eg LFA1 and ICAM1 expressed by the endothelium.
3) Transmigration across the endothelium and basement membrane takes up to ∼15 minutes and requires both integrins and further adhesion molecules such as platelet/endothelial cell adhesion molecule 1 (PECAM1)
4) Numerous chemotaxins (e.g. C5a and the bacteria derived formyl-Met-Leu-Phe, fMLF) attract the neutrophils to the bacteria invader
5) Neutrophils eliminate pathogens via intracellular and extracellular mechanisms

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

What are ICAM1 and LFA1

A

Firm arrest and adhesion involves interaction between neutrophil expressed integrins such as lymphocyte-associated antigen 1 (LFA1) and intercellular adhesion molecule 1 (ICAM1) expressed by the endothelium.

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

Is it faster for a neutrophil to migrate across the vascular endothelium transcellularly or paracellularly

A

transmigration can occur either paracellularly (between endothelial cells), or transcellularly (through endothelial cells, which takes longer ∼30 minutes).

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

Give some strategies used by neutrophils to eliminate pathogens (3)

A

phagocytosis and killing via reactive oxygen species and antibacterial proteins (e.g. cathepsin, lysozyme and defensins)

degranulation to release antibacterial proteins into the extracellular fluid

neutrophil extracellular traps (NETs), which are composed of a core DNA element alongside enzymes that immobilise pathogens, thus preventing spreading and facilitating phagocytosis, as well as likely also directly killing some pathogens

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

What predisposes neonates to infection

A

Neutrophil dysfunction is thought to be important in the development of sepsis in newborns and neonatal neutrophils fail to form NETs, which likely contributes to newborns being predisposed to infection

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

What TLRs/ receptors do mast cells express

what does activation of these trigger? (5)

A

receptors for C3a, C5a and IgE,

stimulation of these receptors results in release of histamine, heparin, leukotrienes, nerve growth factor and, unusually, pre-formed packets of cytokines.

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

What is 2-(1H-imidazol-4-yl)ethanamine

A

chemical named for what is usually called histamine, a name resulting from it being an amine occurring in tissues (histos = tissue)

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

Where is histamine made and found?

A

Histamine is formed from the amino acid histidine by histidine decarboxylase

at the cellular level histamine is found in 4 cell types: mast cells, basophils, enterochromaffin-like cells (ECL) in the gut and histaminergic neurones in the brain. In mast cells and basophils, histamine is packaged in acidic granules with a high molecular weight heparin called macroheparin

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

Name 3 substances that evoke histamine release from mast cells

A

C3a/C5a, SP, and IgE

trigger increased [Ca2+]i which leads to degranulation

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

How do C3a and C5a affect mast cells

A

C3a and C5a receptors are Gi-coupled; the βγ subunits activate PLCβ and induce intracellular Ca2+ release via IP3

this leads to degranulation and histamine release

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

How does SP affect mast cells

A

SP primarily induces mast cell degranulation, not via neurokinin 1 receptors, but rather Mas-related gene X2 (MrgX2) receptors, which in human mast cells appear to be Gq-coupled resulting in PLCβ/IP3 mediated Ca2+ release

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

How does IgE affect mast cells

A

Allergen-induced cross linking of IgE with its high affinity receptor FcεRI induces phosphorylation of the adaptor protein linker for activation of T cells (LAT) that causes activation of PLCγ/IP3 mediated Ca2+ release.

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

True or false

all histamine receptors are GPCRs

A
true
There are four histamine receptors that are GPCRs (H1-4)
H1= Gq/11
H2= Gs
H3=Gi
H4=Gi
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

What are the effects of H1 receptors

A

H1 = Gq/11 -> PLCβ -> IP3 + DAG/PKC (important in inflammation)

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

How do H2 receptors work

what are they important for

A

Gs coupled so increases AC, increasing cAMP/PKA

important for gastric secretion

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

How do H3 and H4 receptors work

what are they important for

A

both decrease AC so decrease cAMP/PKA

H3 is an important inhibitory autoreceptor in the CNS

H4 is important for chemotaxis/cytokine release

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

Give the 7 key effects histamine has in the body

give the receptor important for each effect

A
  • smooth muscle contraction (H1) in the ileum, bronchioles and uterus
  • blood vessel dilation, largely mediated via endothelial release of nitric oxide (H1)
  • itching evoked by activation of distinct sensory nerves (pruritoceptors, H1)
  • triple response of Lewis (H1)
  • increased heart rate (H2)
  • gastric acid secretion (H2)
  • brain neurotransmission (predominantly H1-3, but also H4)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

How is histamine metabolised

A

2 possible enzymes:

histaminase -oxidatively deaminates histamine to produce imidazole acetaldehyde,

histamine N-methyltransferase -catalyses the transfer of a methyl group on to the nitrogen of the imidazole ring to produce NT-methylhistamine

both products are inactive at histamine receptors.

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

What are the names of the 2 enzymes that can metabolise histamine

A

histaminase (also called diamine oxidase)

histamine N-methyltransferase (also called imidazole N-methyltransferase)

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

Give 3 allergic conditions which involve histamine

A

allergic rhinitis (hay fever)

urticaria (incl. dermatographism urticaria)

allergies such as nut allergy and penicillin allergy.

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

What responses are involved in the allergic reactions involving histamine (5)

A

include swelling, itchiness, nasal congestion, watery eyes and in non-human animals poor coat quality

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

Give 3 symptoms of severe anaphylaxis

how is it treated

A

throat swells
heart rate increases
blood pressure drops

rapid adrenaline treatment

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

What is mastocytosis

A

a group of conditions where too many mast cells are present leading to excessive allergic-type reactions when an attack is triggered by various factors (heat, cold, stress, infection, exercise, drugs etc.)

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

What causes mastocytosis in humans

A

Often results from a gain-of-function mutation in the receptor tyrosine kinase c-Kit/CD117 (D816V) causing enhanced mast cell proliferation and survival

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

What can cause mastocytosis in non humans

A

Mastocytosis resulting from mast cell tumours has also been described in dogs and cats, but, non-cancerous mastocytosis has also been described in dogs, but, where tested, there was no c-Kit mutation

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

What is the driver of symptoms in mastocytosis

A

histamine

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

What is the aim of treatment for pathophysiology associated with histamine

A

inhibition of mast cell degranulation

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

Name a mast cell stabiliser

A

Sodium cromoglycate

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

How does Sodium cromoglycate work

A

inhibits mast cell degranulation

MOA unclear but decreased Ca2+ influx is seen in mast cell activation in presence of sodium cromoglycate

perhaps Sodium cromoglycate inhibits an inward Cl- channel required to maintain a negative enough membrane potential to enable sustained influxes of extracellular Ca2

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

What can you use sodium cromoglycate to treat

A

mastocytosis

available as eye drops for the treatment of hay fever e.g. Opticrom and is occasionally used in the treatment of asthma

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

True or false

Raising [cAMP] inhibits mast cell degranulation

A

true
thus β2-adrenoceptor agonists (salbutamol and formoterol) and phosphodiesterase (PDE) inhibitors (theophylline) used in the treatment of asthma are partially efficacious through mast cell inhibition although their main therapeutic action is via bronchodilation

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

Name a monoclonal antibody used in allergic reactions

how does it work

A

omalizumab

recognises IgE, decreases mast cell degranulation because allergen bound IgE induces mast cell degranulation when bound to its receptor FcεRI.

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

Which drugs are classical antihistamines

A

H1-receptor antagonists

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

Name 2 β2-adrenoceptor agonists that are used to treat allergic reactions

name another drug which works via a similar mechanism

A

salbutamol
formoterol

phosphodiesterase (PDE) inhibitors (theophylline)

both types of drug work by increasing [cAMP] in mast cells to inhibit degranulation

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

name a first generation antihistamine

what was an issue with it

A

mepyramine

rapidly permeated the blood-brain barrier and caused drowsiness, and so were not suited for systemic use,

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

true or false

first generation antihistamines eg mepyramine are no longer in use

A

false
not suited for systemic use, but it is still used in topical creams for treating insect bites.
First generation drugs are also used in some cold/flu medications to aid sleeping

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

How did second generation antihistamines improve on first generation drugs

A

not cross the blood-brain barrier (terfenadine was the first)

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

What was the issue with the first second generation antihistamine to come to market

A

Terfenadine was a blockbuster drug in the treatment of hay fever ($440 million in 1996, the year before withdrawal), but there were increasing reports of people taking it developing torsade de points syndrome (also known as prolonged QT interval) and also reports of sudden cardiac death

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

How did terfenadine cause prolonged QT syndrome

A

inhibits KV11.1/hERG, which is important for repolarisation of the action potential

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

How is terfenadine metabolised

A

Terfenadine is a prodrug, metabolised by the 3A4 isoform of cytochrome p450 (CYP450) to fexofenadine, which is the active compound

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

Who is at enhanced risk of death from terfenadine

what else can make this drug extra dangerous

A

people with in conditions of diminished p450 3A4 activity (3A4 form of CYP450 metabolises terfenadine to fexofenadine, the active compound)

p450 3A4 is inhibited by many drugs, as well as bergamottin, a component of grapefruit juice

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

How do 3rd generation antihistamines improve on the previous 2 generations

name 2 3rd gen drugs

A

non-drowsy and lack cardiac side-effects

fexofenadine and loratadine

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

name a 1st, 2nd, and 3rd generation antihistamine

A

1: mepyramine
2: terfenadine
3: fexofenadine (also loratadine)

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

What are the main uses for fexofenadine and loratadine

A

treating hay fever, allergy and urticaria

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

Why do we think that histamine is not involved in the underlying pathology of hypersensitivity reactions in cattle

A

antihistamines are of little use in treating hypersensitivity reactions in cattle

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

How is adrenaline administered in anaphylaxis

can you use NA instead?

why might you inject a corticosteroid

A

administered i.m. to counteract systemic vasodilatation and reduction in tissue perfusion. also reduces bronchospasm

no: noradrenaline is such a potent vasoconstrictor it causes reflex bradycardia and is thus unsuitable

corticosteroid hydrocortisone is often co-administered for its anti-inflammatory effects

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

What is a potential treatment for mastocytosis in humans (other than of inhibitors of mast cell degranulation and histamine receptor antagonists)?

A

imatinib

only efficacious in patients not presenting with the common D816V c-Kit mutation.

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

Which drugs are used to control rare cases of mastocytosis in dogs

A

A combination of H1R and H2R antagonists

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

What is atopic dermatitis

how can it be treated

A

a relatively common allergic condition in dogs (~10%)

although H1-receptor antagonists can be used they are often inefficacious. Oclacitinib is a beneficial treatment, which works by inhibiting Janus kinases (JAK, greatest specificity for JAK1)

A canine monoclonal antibody against IL-31 has also been developed

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

How is oclacintinib involved in inflammation treatment

A

inhibits JAK

JAKs are common to many cytokine signalling pathways associated with allergic skin diseases and thus oclactinib lowers pro-inflammatory signalling and can be used as an alternative to corticosteroids and ciclosporin

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

Which cells are responsible for gastric acid secretion

A

parietal cells

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

What do parietal cells do

A

are responsible for secreting HCl into the stomach to aid digestion and kill pathogens

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

Where are mucous secreting cells in the stomach and what is their role

A

in the gastric mucosa

secrete HCO3- ions which are trapped in mucous creating a protective barrier (pH6-7)

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

What can disturbances in parietal and mucus secreting cells in the stomach lead to

A

can contribute to the pathogenesis and pain associated with peptic ulcers and gastro-oesophageal reflux disease.

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

What do G cells do

A

secrete gastrin on to ECL cells where it binds to CCK2 receptors to produce an increase in ECL [Ca2+], which causes ECL cells to release histamine.

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

What does histamine secreted from gastric ECL cells do

A

acts at Gs-coupled H2-receptors on parietal cells causing an increasing in cAMP and PKA activity. PKA phosphorylates proteins involved in trafficking of K+/H+ pumps to the apical membrane resulting in enhanced K+/H+ pump activity and thus increased H+ release

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

How does somatostatin affect gastric pH

A

it is a negative regulator of H+ release and has both direct and indirect mechanisms of inhibition

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

What are somatostatin’s direct and indirect methods of inhibiting H+ release into the stomach

A

activation of Gi-coupled SST2R on parietal cells counteracts the effects of histamine (direct inhibition),

activation of SST2R on G and ECL cells reduces gastrin and histamine release respectively (indirect inhibition).

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

Which antagonists are used to decrease histamine release in the stomach

A

CCK2 receptor antagonist (eg proglumide)

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

What does proglumide do

A

CCK2 receptor antagonist (decreases histamine secretion)

however its use in treating peptic ulcers has been surpassed

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

What type of drug were the first success at treating peptic ulcers

give an example

A

H2-antagonists

cimetidine (Tagamet)

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

What is a problem with cimetidine

which drug was soon competing with it

A

inhibit CYP450

soon had competition from ranitidine

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

What was the first PPI introduced

A

omeprazole

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

Describe the activation and MOA of proton pump inhibitors

what are 3 drawbacks of PPIs

A

converted to their active form in the acidic environment of the parietal cell and form disulphide bonds with the K+/H+ pump.

However, PPIs are broken down by H+, have a slow onset to maximal effectiveness and predominantly metabolised by CYP2C19

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

What metabolises PPIs predominantly

what is the issue with this

A

CYP2C19

exhibits significant genetic polymorphism producing extensive and poor metabolisers, especially in Asians (1-2% in Europeans vs. ~20% in Asians).

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

name a K+/H+ pump blocker (not omeprazole)

what is the MOA

A

vonoprazan

a potassium-competitive acid blocker (P-CAB)

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

Why is vonoprazan an improvement on omeprazole (2)

A

has greater stability than PPIs in H+ and CYP2C19 is less vital in its metabolism

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

Give methods for treating peptic ulcers (8)

A
PPIs/ P-CABs
CCK2 inhibitors
H2 antagonists 
antacids
cholinergic blockade 
vagotomy 
eradication of Helicobacter pylori
stopping NSAID use
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
102
Q

Name an antacid

how do they help with gastric ulcer

A

Gaviscon

help neutralise pH in stomach

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

True or false

cholinergic blockage and vagotomy for peptic ulcer treatment essentially work in the same way

A

true

acetylcholine stimulation of muscarinic type 3 receptors (M3R) on parietal cells enhances acid release, which is then inhibited by atropine/vagotomy

both virtually obsolete

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

which antibiotic do you use to treat H pylori infection

A

clarithromycin

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

Does H pylori cause gastric ulcers in dogs

A

Helicobacter are not generally thought to be the cause although they are often present, e.g. H. heilmannii

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

Why do NSAIDS cause peptic ulcers

A

NSAIDs inhibit prostaglandin (PG) synthesis and PGE2 acts on ECL cell EP2/3R to inhibit gastric acid secretion, as well as enhancing mucin (EP4R) and bicarbonate secretion (EP1/2R).

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

when would you prescribe NSAIDs in combination with eg omeprazole

what can they be given instead of omeprazole

A

In patients at risk of developing peptic ulcers (e.g. the elderly) or those with gastric complications

misoprostol - a synthetic prostaglandin E 1 (PGE1) analogue

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

What is Arthrotec

A

a combination of the NSAID diclofenac and misoprostol that can be used in the chronic treatment of rheumatoid arthritis with misoprostol acting as a prophylactic against NSAID-induced ulceration

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

If you are giving a dog NSAIDs when ulcers have been induced through prior NSAID administration, which other drug should you give it

A

misoprostol

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

Etymology of bradykinin

A

originally identified as a slow (brady) contractor (kinin) of guinea pig ileum smooth muscle (slow being relative to tachykinins like SP)

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

How is bradykinin formed generally

A

by the action of kallikrein upon kininogens, which exist as high- and low-molecular weight forms

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

describe the steps involved in forming bradykinin

A

Hageman factor/Factor XII to become activated by contacting negatively charged surfaces, such as lipopolysaccharide, collagen or basement membrane.

Activated Hageman factor is a protease that converts plasma prekallikrein to kallikrein, which clips HMW-kininogen to bradykinin and LMW-kininogen in tissues to kallidin

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

How do you activate factor XII

A

by contacting a -ve surface

Hageman factor only contacts negatively charged surfaces, such as lipopolysaccharide, collagen or basement membrane when leaking out of leaky blood vessels during inflammation

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

How is bradykinin inactivated

A

clipped kininase II

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

What is kininase I activity mediated by

what does it form

A

via several peptidases, including serum carboxypeptidase,

results in the removal of the C-terminal arginine to form des-Arg-bradykinin, which is an agonist for bradykinin B1 receptors

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

Describe kininase II

A

a peptidyl dipeptidase that inactivates kinins by removing the two C-terminal amino acids, this enzyme is angiotensin converting enzyme (ACE)

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

What type of receptor are bradykinin receptors

A

bradykinin B1 and B2 receptors and are both Gq-coupled GPCRs

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

When/ where are bradykinin B1 and B2 receptors expressed and activated

A

B1 receptors are upregulated during inflammation through the action of cytokines, such as IL-1, and respond primarily to des-Arg-bradykinin (bradykinin itself is a very poor B1 agonist),

B2 receptors are constitutively expressed and potently activated by bradykinin and kallidin (des-Arg-bradykinin has virtually no efficacy)

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

What does the activation of bradykinin B1 and 2 receptors on vascular endothelium result in

A

causes an increase in [Ca2+], which activates cytosolic phospholipase A2 (cPLA2) resulting in prostacyclin (PGI2) production and endothelial nitric oxide synthase (eNOS) resulting in NO production

PGI2 and NO diffuse to the vascular smooth muscle cell layer and through increasing cAMP and cGMP levels respectively, mediate vasodilatation

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

Do ACE inhibitors only decrease vasoconstriction via ACE inhibition

A

no - not only reduce angiotensin II levels, thus reducing vasoconstriction, but they also cause an increase in bradykinin levels and thus enhance vasodilatation

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

How is bradykinin involved in pain (2)

A

Activation of bradykinin receptors causes activation of nociceptors and drives pain (due to increased [Ca2+]i).

Bradykinin also induces nociceptor sensitisation because activation of Gq GPCRs results in activation of PKC, which phosphorylates numerous ion channels involved in the sensation of pain.

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

What inhibits kallikrein

A

C1-esterase inhibitor (C1-INH).

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

What is HAE

what is the prevalence

A

hereditary angioedema

HAE, prevalence ~0.01%

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

What is the mutation in HAE

A

mutation in the gene encoding C1-INH causes excessive levels of bradykinin resulting in sufferers experiencing periods of severe and painful swelling

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

how does the cause of type I HAE differ from type II

A

Type I HAE results from mutations that compromise C1-INH synthesis/secretion,

whereas Type II HAE results from mutations that produce inactive C1-INH

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

How common is ACE inhibitor associated angioedema

what predisposes to it

A

0.1% but is higher (~0.5%) in African American

it is currently poorly understood what makes people susceptible to this, but there are indications that it may be linked to genetic variation in genes that regulate the immune system

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

How can you treat HAE

can anything else be treated in the same way

A

recombinant C1-INH, kallikrein inhibitors and the B2 antagonist icatibant can be used

anecdotal evidence for efficacy in treating ACE inhibitor associated angioedema.

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

Which drug is used to treat HAE

A

icatibant (B2 antagonist)

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

Can you exploit B1 receptors to treat pain

A

Plenty of research to develop B1 antagonists for inflammatory pain, but none clinically available

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

What are cytokines generally

A

a very broad group of proteins that are largely, but not exclusively, synthesised and released by cells of the immune system and generally help to coordinate the immune response

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

Why are mast cells unusual in terms of cytokines

why is this important pharmacologically

A

they contain granules containing pre-formed cytokines.

Thus, because most cells have to synthesise cytokines, drugs that inhibit cytokine transcription (e.g. corticosteroids, see later) have a major impact on immune responses

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

How many cytokines are there

are they all pro-inflammatory

A

over 100 cytokines, some of which are pro-inflammatory and some of which are anti-inflammatory.

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

What are the 4 main groups of cytokines

A

interleukins
chemokines
interferons
colony stimulating factors

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

What are the key pro inflammatory interleukins

what are they released from

A

are IL-1 (actually three cytokines: the agonists IL-1α and IL-1β and an endogenous receptor antagonist, IL-1ra) and TNFα

predominantly released from macrophages

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

What do IL-1 and TNFα do once released from macrophages

A

promote proliferation and maturation of other immune cells, as well as causing fever (IL-1).

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

are there any anti inflammatory interleukins

A

yes

which inhibit cytokine production and inhibit some T-cell responses (e.g. IL-10 and IL-1ra).

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

Why are ILs called interleukins

A

originally observed to communicate between leukocytes

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

Why is the name chemokines misleading

A

non-cytokines also act as chemoattractants and chemokines have other roles, e.g. CCL3 induces mast cell degranulation by acting at CCR1 receptors.

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

What does the nomenclature of chemokines relate to

A

the cysteine residues in the N-terminus of the peptide chain,

CXC chemokines have a single amino acid separating the two cysteines, CC chemokines have two adjacent chemokines, C chemokines only have one N-terminus cysteine and lastly, CX3C chemokines have three amino acids separating the two cysteines

these classes are also sometimes termed α, β, γ and δ.

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

What kind of receptor are chemokine receptors

A

GPCRs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
141
Q

What are the 3 classes of interferon and what is the role of each class

A

(IFN) α, β and γ, the former two have anti-viral activity and the latter has a role in induction of Th1 responses

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

What is the role of colony stimulating factors

A

stimulate the formation of maturing colonies of leukocytes and are primarily used to overcome deficits in a person’s white-blood cell count, e.g. following chemotherapy

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

What is NGF

A

Nerve growth factor

released from both macrophages and mast cells and is a potent sensitising agent, i.e. it does not excite nociceptors directly, but rather activates signalling pathways that result in a lesser stimulus causing pain

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

Give an example of the sensitising effect of NGF

A

following NGF injection, there is a >5°C lowering of the temperature required to generate pain in humans

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

What are the effects of NGF mediated through

A

mediated by the high affinity NGF receptor tropomyosin-related kinase A (TrkA).

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

How is NGF related to treatment of pain

A

in the treatment of inflammatory pain, blocking the actions of NGF is desirable and anti-NGF monoclonal antibodies such as tanezumab have been developed, but are still in clinical trials

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

name a monoclonal antibody against NGF

what is it hoped to treat

A

tanezumab

inflammatory pain

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

What is Annexin A1

A

a protein produced by many cells, which downregulates both inflammatory cell activation and mediator release, actions that are brought about by binding to the GPCR formylpeptide receptor 2 (FPR2)

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

What is FPR2 a receptor for

A

lipoxin

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

what are the 3 lipid mediators of inflammation

What is the main precursor for them? What is an exception to this?

A

leukotrienes,
platelet-activating factor
prostanoids

arachidonic acid (exception = PAF)

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

How are lipid mediators produced

A

on demand from membrane phospholipids by the action of phospholipases.

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

What is PAF

A

Platelet activating factor

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

Describe arachidonic acid

A

a 20-carbon unsaturated fatty acid containing four double bonds

AKA 5,8,11,14-eicosatetraenoic acid

eicosa refers to the 20 carbon atoms and tetraenoic to the 4 double bonds and thus lipid mediators are often referred to as eicosanoids

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

what is the rate limiting step for eicosanoid synthesis

A

liberation of arachidonic acid from membrane phospholipids and the usual one-step process involves PLA2.

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

What is the most important isoform of PLA2

A

cPLA2

which is activated by a combination of phosphorylation and Ca2+

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

cytosolic PLA2 (cPLA2) is activated by a combination of phosphorylation and Ca2+. Therefore, what else can stimulate it ?

A

can be stimulated by a plethora of substances, e.g.
bradykinin acting at B2 receptors raises [Ca2+]i

TNFα acts at TNFR1 to promote MAPK phosphorylation of cPLA2 at serine 505 and serine 727, as well raising [Ca2+]i via TNFR2.

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

What does the action of PLA2 result in

A

production of arachidonic acid (prostanoid and leukotriene precursor) and lysoglyceryl-phosphorylcholine (PAF precursor called lyso-PAF)

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

Which type of enzymes synthesise leukotrienes

A

synthesised by lipoxygenase enzymes from arachidonic acid

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

describe lipoxygenases

A

cytosolic enzymes, expressed primarily in the lungs and leukocytes

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

Describe how 2 lipoxygenases act of arachidonic acid

A

Arachidonic acid is converted by 12-lipoxygenase to produce 12-HETE (hydroxyeicosatetraenoic acid), which is a chemotaxin and 5-lipoxygenase to produce leukotriene A4 (LTA4).

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

Arachidonic acid is converted by 5-lipoxygenase to produce leukotriene A4 (LTA4). What further reactions occur to LTA4

A

converted in cells expressing the cytosolic enzyme LTA4 hydrolase to LTB4, whereas in cells expressing the membrane bound LTC4 synthase (also called glutathione S-transferase) LTA4 is converted into LTC4, which in turn is cleaved by peptidases to produce LTD4 and LTE4.

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

What are LTC4-LTE4 known as

A

the cysteinyl leukotrienes (CysLT)

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

What are the cysteinyl leukotrienes (CysLT)

A

LTC4-LTE4

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

What do different leukotrienes act on

what kind of receptor is each

A

LTB4 acts at BLT1 and BLT2 receptors,

the CysLTs act at CysLT1 and CysLT2 receptors;

BLT receptors can be either Gq- or Gi-coupled, whereas CysLT receptors are Gq-coupled.

all act on GPCRs

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

Different CysLTs have different efficacy at different receptors and thus CysLT metabolism has biological implications. What does this mean?

A

CysLT1: LTD4&raquo_space; LTC4 > LTE4

CysLT2: LTC4 = LTD4 > LTE4.

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

Give some of the roles of LTB4 (3)

A

potent chemoattractant and activator of neutrophils and macrophages,

it upregulates neutrophil adhesion molecule expression

promotes macrophage cytokine release

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

Name a leukotriene found in inflammatory exudate

what does this mean

A

LTB4

makes LTA4 hydrolase a therapeutic target

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

Why was a drug targeting LTA4 hydrolase withdrawn

A

caused dermatitis

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

What 3 things do all CysLTs do

A

cause bronchoconstriction,

increase vascular permeability

mucous secretion

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

What are CysLTs released from

why is this unsurprising?

A

from both mast cells and eosinophils

characteristic leukocytes in airways of asthmatics and all CysLTs cause bronchoconstriction

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

CysLTs are released from mast cells and eosinophils to cause bronchoconstriction. How has this knowledge been used pharmacologically?

A

montelukast (CysLT1 receptor antagonist) is used in maintenance treatment of asthma

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

What are the 2 main ways to synthesise lipoxins

A

either 12-lipoxygenase conversion of LTA4 to LXA4

or

15-lipoxygenase conversion of arachidonic acid to 15S-HETE, which is then converted by 5-lipoxgenase to LXA4

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

What does LXA4 bind to

what kind of receptor is this?

what is the effect?

A

binds to the FPR2,

Gi-coupled

reduces neutrophil chemotaxis and degranulation, as well as acting as an antagonist of CysLT1 receptors

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

How is PAF formed briefly

A

through the action of acetyltransferase on lyso-PAF

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

Why is PAF misleadingly named

A

it has effects upon a variety of cell types and acts through GPCRs that are variously coupled

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

What are the effects of PAF generally (4)

A

increases thromboxane (TXA2) production in platelets

can be spasmogenic;

chemotactic for neutrophils

activates PLA2

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

Where does the name ‘prostaglandin’ originate

A

experiments in the 1930s showed that semen contained a lipid that could evoke uterine smooth muscle, named prostaglandin (PG) because of originating from the prostate gland

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

How are prostaglandins formed

A

Arachidonic acid is metabolised to prostaglandins by cyclooxygenases (COX)

Following the production of PGH2, it is the cell specific enzymes that are responsible for the downstream production of a particular PG or thromboxane

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

How does COX convert arachidonic acid to PGH2

A

COX catalyses two reactions:

arachidonic acid is first cyclised and oxygenated forming the endoperoxide PGG2;

the hydroperoxyl in PGG2 is then reduced to form PGH2.

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

Where are COX 1 and 2 found

A

COX-1 is constitutively expressed in many cell types, whereas COX-2 expression is induced in inflammation, although some constitutive expression does occur.

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

What does prostanoid refer to

A

PGs and TX

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

What does the 2 in PGH2 refer to

A

the number of carbon-carbon double bonds in the final structure

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

What happens if an acid with a different amount of C=C double bonds is used to form PGs instead of eicosatetraenoic acid (four carbon-carbon double bonds) ?

A

if eicosatrienoic acid (three C=C double bonds) is used in place of eicosatetraenoic acid as a substrate then the resulting PGs have one fewer double bond and are named, for example, PGE1, and if eicosapentaenoic acid (five C=C double bonds) is used as a substrate then PGs have one more double bond and are named, for example, PGE3

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

What are the sources of prostanoids in inflammation?

A

varies

, PGE2 and PGI2 tend to be produced locally by tissues and blood vessels,

PGD2 is predominantly released by mast cells and in chronic inflammation macrophages release PGE2 and TXA2

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

True or false

Prostanoids act at RTK

A

false

Prostanoids act at GPCRs and some PGs can activate multiple receptors, e.g. PGD2 can activate TP and DP receptors

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

Describe the coupling of different prostanoid GPCRs

A
  • DP1, EP2, EP4 and IP receptors are Gs-coupled causing increased cAMP production
  • EP1, FP and TP receptors are Gq-coupled causing an increase in [Ca2+]i
  • DP2 and EP3 receptors are Gi-coupled causing decreased cAMP production
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
187
Q
What are the predominant effects of the following prostanoids in terms of inflammation:
PGD2
PGE2
PGI2
TXA2
PGF2α
A
  • PGD2 = vasodilatation, inhibition of platelet aggregation and relaxation of GI (GI)/uterine muscle via DP1 receptors; bronchoconstriction via TP receptors
    • PGE2 = bronchial/GI smooth muscle contraction (EP1), bronchodilation, vasodilatation and relaxation of GI smooth muscle (EP2), contraction of GI/uterine smooth muscle and fever (EP3), and sensitisation of nociceptors (EP4)
  • PGI2 = vasodilatation and inhibition of platelet aggregation
  • TXA2 = vasoconstriction, bronchoconstriction and platelet aggregation
  • PGF2α = uterine contraction in humans, bronchoconstriction in cats/dogs
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
188
Q

what is the balance in healthy blood vessels what is the balance that has to be struck with regards to platelet aggregation

how does this change if the endothelium is damaged

A

balance between the pro-aggregation effects of TXA2 (produced by platelets) and the anti-aggregation effects of PGI2 (produced by endothelial cells)

the balance shifts towards TXA2, platelets aggregate and the damage is sealed.

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

if the vascular endothelium is damaged then the balance shifts towards TXA2, platelets aggregate and the damage is sealed. How is this related to certain thromboembolic strokes

A

atherosclerotic plaque rupture damages the endothelium and the platelets adhere forming a clot;

part of the clot can break off and block a cerebral artery

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

Why do people with oily fish in their diet tend to have lower incidence of heart attacks

A

fish oil is high in eicosapentaenoic acids resulting in the production of PGI3 and TXA3, TXA2 is far more potent than TXA3, but PGI3 is more potent than PGI2

thus there is an overall tip in the balance towards anti-aggregation and less vasoconstriction

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

How do most NSAIDs work

Is it reversible?

A

inhibit COX by entering a hydrophobic channel on the enzyme and forming hydrogen bonds with an Arg120,

this prevents the entrance of fatty acids, like arachidonic acid, into the catalytic domain and thus PG production is stopped.

This inhibition is reversible

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

Why were selective COX inhibitors thought to be better than non-selective NSAIDs

A

COX-1 is constitutive and expressed in many tissues, whereas COX-2 is induced during inflammation.

It was thus hypothesised that drugs that only inhibited COX-2 would result in all of the benefits of COX inhibition (analgesia, decreased swelling and antipyresis etc.) without any of the side effects such as gastric bleeding

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

How can you make a COX inhibitor COX-2 selective

A

NSAIDs have to enter the hydrophobic tunnel of COX to reach Arg120 and whereas COX-1 has a narrow tunnel, COX-2 has a wider tunnel.

Consequently, drugs have been developed that have bulky sulphur-containing side groups and selectively act upon COX-2, but not COX-1 because they cannot enter the narrow, hydrophobic tunnel

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

Are non-selective NSAIDs common

A

yes
Most NSAIDs, such as ibuprofen, are non-selective because they are small and enter the hydrophobic tunnels of both COX-1 and COX-2

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

How can you recognise the name of a COX2 selective NSAID

A

generally name ends in -coxib

eg etoricoxib

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

Name a non selective and a COX2 selective NSAID

A

non-selective: ibuprofen

COX2 selective: etoricoxib

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

What is the most common side effect of non selective NSAIDs

Is this a trivial side effect

A

GI bleeding

no - accounts for approximately 5000 deaths in the UK each year

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

Name 3 groups NICE have identified to be at high risk of gastric bleeding

can you give these people NSAIDs

A

adults aged 65+, people with a history of gastric ulceration and those taking NSAIDs chronically

yes but must be co-administered with PPIs or with the PGE1 analogue misoprostol.

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

Why are COX2 selective NSAIDs not as safe as originally thoughout

A

although COX-2 is upregulated in inflammation, it is also constitutively expressed in some endothelial and vascular smooth muscle cells (e.g. in the kidney) and thus its inhibition results in decreased PGI2 production – PGI2 is vasodilatory and decreases platelet aggregation

also inhibiting COX-2 raises levels of endogenous inhibitors of eNOS, which would lead to decreased NO production. Consequently, some, but not all, COX-2 inhibitors have been associated with increased myocardial risk

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

Why did Merck withdraw rofecoxib

A

it was associated with increased myocardial risk (rofecoxib is a selective COX2 inhibitor)

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

Are COX2 inhibitors still in use?

A

Current guidance suggests that COX-2 inhibitors should be reserved for people suffering from chronic inflammation, e.g. osteoarthritis/rheumatoid arthritis, who are not thought to have a substantial cardiovascular risk and when conventional NSAIDs are thought to pose a high GI risk

NB GI problems can still occur with COX-2 inhibitors

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

Why might COX2 inhibitors still cause gastric ulcers

A

perhaps because they interfere with the healing of pre-existing ulcers

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

Do all NSAIDs pose a CV risk?

A

yes

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

True or false

naproxen carries a lower cardiovascular risk than other NSAIDs

A

false
although it was thought that the non-selective NSAID naproxen carried a lower risk, results from a recent large trial comparing naproxen, ibuprofen and a COX-2 inhibitor did not support this hypothesis (the COX-2 inhibitor did however show significantly lowered GI side effects)

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

How does aspirin differ from other NSAIDs

A

Rather than forming a hydrogen bond with Arg120, it acetylates Ser530 and irreversibly inactivates COX, an action that explains aspirin’s long-lasting actions

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

What type of molecule is aspirin

A

acetylsalicylate

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

Why is aspirin considered an acetylsalicylate

A

by donating an acetyl group to COX, salicylate is formed

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

What can salicylate do

A

reversibly inhibit COX (like most NSAIDs)

but the concentrations produced are unlikely to have a significant therapeutic effect

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

Explain how aspirin’s its irreversible action means that it is used at low doses for reducing the risk of platelet aggregation

A

platelets express COX-1 and TX synthase and are a major source of TXA2 (induces platelet aggregation and vasoconstriction), whereas endothelial cells predominantly produce PGI2 (inhibits platelet aggregation and vasodilatation);

acetylated COX-1 can be replaced by newly synthesised protein in endothelial cells, but TXA2 production by platelets is switched off for their lifetime (~10 days). Therefore, overall switch to beneficial, anti-thrombotic effects, with a lower risk of stomach ulcers (but PPIs are often co-prescribed to limit the risk.)

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

Is aspirin COX selective

A

Although weakly COX-1 selective, aspirin also acetylates COX-2

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

What does aspirins acetylation of COX2 result in

A

COX-2 no longer producing intermediates for PG and TXA2 synthesis, but instead 15R-HETE, which is the stereoisomer of 15S-HETE (product of 15-LO conversion of arachidonic acid, which is used to synthesise LXA4

5-LO then converts 15R-HETE to aspirin-triggered lipoxin – ATL, also known as 15R-epi-lipoxin A4, which has similar functions to LXA4.

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

What is ATL

A

aspirin triggered lipoxin

has similar functions to LXA2 so its production contributes to the anti-inflammatory action of aspirin

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

Give 4 predominant side effects of NSAIDs

Give the mechanism behind each side effect

A

GI bleeding (PGs inhibit gastric acid secretion and increase the release of protective mucin),

renal insufficiency and nephropathy (PGE2/PGI2 involved in maintenance of renal blood flow),

stroke/myocardial infarction (constitutive expression of COX-2 in endothelial/vascular smooth muscle cells, inhibition of which results in decreased PGI2 production, PGI2 is vasodilatory and decreases platelet aggregation),

bronchospasm (COX inhibition implicated, mechanism unclear).

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

What are the 2 specific side effects of aspirin

A

Reye’s Syndrome

Salicylism

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

Who gets Reye’s Syndrome

what is it characterised by

A

occurs almost exclusively in children (hence not recommended for use in children under the age of 16)

characterised by hepatic encephalopathy, often occurs when aspirin is taken for treating viral symptoms

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

Have health warnings about aspirin had any effect

A

yes
health warnings in the late 1980s have reduced the cases from 41/year in the 1980s to only 1 case of Reye’s Syndrome in 2002 and 3 further suspected cases up to 2009

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

When is salicylism seen

A

– essentially the result of an overdose of aspirin, often seen in children or in suicide attempts

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

What is the pathophysiology of salicylism

A

a progressive condition resulting from Kreb’s cycle inhibition and uncoupling of oxidative phosphorylation, primarily in skeletal muscle, which causes increased O2 consumption and thus increased CO2 production. CO2 stimulates peripheral and central chemoreceptors resulting in increased ventilatory rate causing respiratory alkalosis, which can be compensated for by increased renal bicarbonate excretion. However, larger doses depress the respiratory centres and CO2 accumulates, which is superimposed upon the reduced plasma bicarbonate to cause respiratory acidosis, which can combine with metabolic acidosis resulting from accumulated acidic metabolites due to disrupted carbohydrate metabolism. Fever and repeated vomiting occur, followed by dehydration, respiratory depression, coma and death

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

How do you treat salicylism (3)

A

fluids are administered, as is bicarbonate to enhance aspirin elimination,

activated charcoal adsorbs aspirin in the GI tract

in severe cases haemodialysis may be required.

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

What is paracetamol also known as

A

known as acetaminophen in the Canada, U.S., Iran and Japan

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

Where do the names paracetamol and acetaminophen come from

A

Both names are derived from a chemical name of the drug: N-acetyl-para-aminophenol and N-acetyl-para-aminophenol respectively

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

Why is paracetamol not really an NSAID

A

it is a v poor anti-inflammatory (but does have good anti-pyretic and analgesic effects)

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

Which enzymes does paracetamol inhibit

A

COX1 and 2 (but not through binding to arginine 120 or acetylation)

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

How does paracetamol inhibit COX enzymes

A

through reduction of the active site in COX enzymes required for the production of PGH2 from PGG2

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

How does paracetamol affect dogs

A

paracetamol inhibits the COX-1 splice variant COX-3 with greatest potency, and COX-3 might be preferentially expressed in the brain, however, COX-3 is likely non-functional in humans due to a shift in the reading frame producing a truncated COX protein.

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

How does paracetamol give analgesia

A

some paracetamol metabolites have also been demonstrated to have actions that contribute to analgesia

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

How is paracetamol eliminated usually

A

coagulation using hepatic enzymes

when these are saturated, oxidases act to metabolise paracetamol to NAPQI, which is usually conjugated to glutathione

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

What happens during paracetamol overdose

A

there is insufficient glutathione to eliminate paracetamol and NAPQI can oxidise the thiol groups of cellular proteins resulting in major hepato- and renal toxicity

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

how is NAPQI formed

A

when paracetamol dosage is high enough to saturate hepatic coagulation enzymes needed to eliminate the drug, oxidases (CYP2E1) metabolise paracetamol to NAPQI

NAPQI is also formed at therapeutic doses suggesting that other pathways for its formation exist

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

How does a paracetamol overdose present clinically

A

. Symptoms often do not occur until 24-48 hours post-ingestion and begin with nausea and vomiting and lead to liver failure induced death

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

can you reverse a paracetamol overdose

A

If the patient is seen soon after ingestion then prevention of liver damage can be attempted by administering substances that increase hepatic glutathione production, e.g. acetylcysteine

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

Name a drug given to treat paracetamol overdose

A

acetylcysteine

increases hepatic glutathione production

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

What is required to overdose on paracetamol

how has legislation tried to tackle this problem? has this been effective?

A

Toxicity occurs from as little as 150 mg/kg, for a 65 kg adult = 9.75 g, paracetamol often comes in packs of 16 x 500 mg = 8 g, e.g. not difficult to buy enough to overdose.

However, pre-1998, pack sizes were often 100 x 500 mg, legislation in England and Wales restricted over the counter packs in non-pharmacy premises to 16 x 500 mg,

studies show that this caused a significant reduction in paracetamol-related deaths

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
234
Q

What increases the risk of toxicity from paracetamol

A

Alcohol can increase risk of toxicity because it upregulates CYP2E1, which converts paracetamol to NAPQI, fasting is also a risk, perhaps because of decreased hepatic glutathione levels.

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

Which enzymes converts paracetamol to NAPQI

A

CYP2E1

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

Can you give aspirin and paracetamol to cats and dogs

A

aspirin is not suitable for use in cats because they lack the ability to conjugate salicylate with glycine.

Paracetamol should also be avoided in dogs and especially in cats because they lack enzymes that in humans enable efficient excretion

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
237
Q

What is ‘bute’

A

Phenylbutazone

NSAID used for treating horses but causes a plethora of side effects in humans (it is also abused in racehorses, which are regularly tested for bute use).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
238
Q

What is robenacoxib

A

an NSAID licensed for use in cats and dogs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
239
Q

What is firocoxib (species differences)

A

NSAID used for dogs and horses

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
240
Q

Where does the name ‘serotonin’ come from

A

because it was detected in serum following clotting of the blood and caused vasoconstriction

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

When are high 5-HT serum levels seen

A

after coagulation because platelets contain 5-HT, which is released during platelet activation and aggregation and induces further platelet aggregation, as well as causing vasoconstriction in damaged blood vessels

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

true or false

serotonin is a vasoconstrictor

A

true but can also dilate

5-HT can cause both vasodilatation and vasoconstriction depending upon receptor expression

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
243
Q

Which gut cells express high levels of 5-HT

A

intestinal enterochromaffin cells and in serotonergic neurones of the enteric (and central) nervous system.

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

How is 5-HT formed

A

from tryptophan through the actions of tryptophan hydroxylase (Tph) and L-aromatic acid decarboxylase (= dopa decarboxylase)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
245
Q

Where is 5-HT formed

A

from tryptophan in both serotonergic neurones and enterochromaffin cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
246
Q

What is the rate limiting step in 5-HT formation

A

Tph conversion of tryptophan to 5-hydroxytryptophan

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

What are the 2 Tph isoforms

A

Tph1 is primarily expressed in enterochromaffin cells and Tph2 is predominantly expressed in neurones

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
248
Q

True or false

Platelets produce serotonin

A

false

platelets expresses SERT which take up 5-HT from intestinal circulation

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

how do platelets take up 5-HT

A

Platelets express a serotonin transporter (SERT) enabling platelets to become loaded with 5-HT when they pass through the intestinal circulation (platelets are not thought to synthesis their own 5-HT), which has a high 5-HT concentration

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

Describe the degradation of serotonin

how is it excreted and why is this useful clinically

A

2 steps:
1) oxidative deamination via monoamine oxidase

2) through oxidation to produce 5-hydroxyindoleacetic acid (5-HIAA)

5-HIAA is excreted in the urine and levels of urine 5-HIAA are a measure of systemic 5-HT synthesis.

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

What type of receptors are each of the different 5-HT receptors (7)

A
  • 5-HT1A, B, D – F are Gi-coupled GPCRs (note that there is no 5-HT1C)
  • 5-HT2A – C are Gq-coupled GPCRs
  • 5-HT3 is an ionotropic receptor/ligand gated ion channel
  • 5-HT4 is a Gs-coupled GPCR
  • 5-HT5A is a Gi-coupled GPCR
  • 5-HT6 is a Gs-coupled GPCR
  • 5-HT7 is a Gs-coupled GPCR

(2A – C are Gq-coupled GPCRs; 3 is ionotropic; 1 and 5A are Gi GPCR; all the rest are Gs GPCR)

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

Give 2 reasons for emesis

A

chemicals in the intestine or blood or disturbance of the labyrinth in the ear

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

name 2 key components governing emesis

A

the vomiting centre and the chemoreceptor trigger zone (CTZ), both of which are located in the medulla

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

how do the vomiting centre and CTZ work together

A

circulating chemicals can activate the CTZ, which sends signals to the vomiting centre to produce emesis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
255
Q

Name a receptor important in vomiting

how is this exploited pharmacologically

A

Both visceral afferents that project to the CTZ and the CTZ itself express 5-HT3 receptors

inhibition of these receptors, eg by ondansetron is an effective preventative and treatment of vomiting

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
256
Q

What is ondansetron used to treat commonly

What is its main site of action

A

chemotherapy-induced nausea and vomiting (CINV), which commonly results from anti-cancer drugs

its main site of action appears to be the 5-HT3 receptors in the CTZ

257
Q

Now that ondansetron is off patent what can it be used to treat other than CINV

A

treat nausea and vomiting of most causes e.g. post-surgery, where anaesthetic drugs can lead to emesis.

258
Q

What are other drugs that can be used to stop emesis (not 5-HT3 receptor blockers)

A

H1 receptor antagonists (effective against motion sickness and other causes of emesis)

non-selective muscarinic receptor antagonists (anti-vomiting/nausea effects via M1 receptors)

Dopamine D2 receptor antagonists (act on CTZ)

259
Q

Why is metoclopramide less commonly used to treat emesis now?

A

can cause acute dystonia (due to effects upon dopaminergic signalling in the brain)

260
Q

How is domperidone designed to be an improvement on metoclopramide?

A

Domperidone was designed to be more peripherally selective and is observed to produce acute dystonia less frequently

(both are Dopamine D2 receptor antagonists )

261
Q

Can rodents vomit?

A

no

they appear to lack the brainstem component of vomiting and cannot vomit

262
Q

Should you fast rodents prior to surgery

A

no
Rodents appear to lack the brainstem component of vomiting and cannot vomit
in fact the hypoglycaemia that can result is highly dangerous

263
Q

Name 2 types of animal that cannot vomit

A

horses and rodents

264
Q

Why can horses not vomit

A

because of a muscle that acts as a one-way valve to allow food down, but not up, the oesophagus – humans have this muscle too, but it is not as strong and thus we can, and do, vomit

265
Q

What is a migraine

A

a severe headache that occurs in about 1 in 5 women and 1 in 15 men, usually beginning in young adulthood.

266
Q

What are the 2 forms of migraine

A

with or without aura

267
Q

What is aura

A

a progressive visual disturbance, which occurs in approximately 20% of migraineurs and is followed about 30 minutes later, by the migraine itself

268
Q

Describe the presentation of the migraine itself

A

throbbing headache, often starting unilaterally and accompanied by nausea and vomiting, photophobia and prostration; the length of the attack is usually several hours and people may often feel “hungover” following an attack, some suggest that hangovers are a form of migraine

269
Q

What is the etymology of ‘migraine’

A

likely derived from the Greek hemikrania, meaning pain on one side of the head

270
Q

What causes migraines

A

Although migraine can be triggered in some sufferers by specific stimuli (e.g. particular food such as red wine or chocolate), the underlying causes are poorly understood

271
Q

What are the 3 main hypotheses for the underlying cause of migraines

A

vascular hypothesis
brain hypothesis
inflammation hypothesis

(The brain and inflammation hypotheses are not necessarily mutually exclusive)

272
Q

Describe the vascular hypothesis for the underlying cause of migraines

A

intracerebral vasoconstriction produces an aura and a subsequent extracerebral vasodilatation causes headache

273
Q

Is there any evidence for the vascular migraine hypothesis

A

modern measurements have shown that although changes in cerebral blood flow occur during migraine, headache begins during vasoconstriction and not all stimuli that cause similar cerebral blood flow changes produce headache.

274
Q

What is the brain migraine hypothesis

A

– a wave of cortical spreading depression (CSD) spreads across the brain and is associated with the aura component of migraine. CSD is a slowly propagating 2-5mm/min wave of near-complete depolarisation, which results in silencing of neuronal electrical activity for several minutes (hence depression). CSD can be triggered by elevated extracellular [K+], such as that occurring following hyperactive neuronal firing and alongside neuronal depolarisation, ionic imbalance occurs and numerous mediators including H+, glutamate, NO, arachidonic acid, and 5-HT are released. CSD implies an important role for ion channels in migraine, and many preventative drugs work on ion channels.

275
Q

What is the inflammation hypothesis of the underlying cause of migraines

A

activation of trigeminal nociceptors that innervate the meninges and extracranial blood vessels (remember, the brain itself is not innervated by nociceptors = no pain) is the initial event in migraine resulting in pain and neurogenic inflammation through CGRP release.

276
Q

Do animals get migraines

A

some evidence
eg a Cocker Spaniel that would display signs of fearfulness and be very quiet for 1-2 hours before onset of vocalisation, low head carriage, refusal to eat and drink and occasional vomiting. Attacks occurred 1-2 times/month, persisted for up to 3-days and were followed by 1-2 days of quietness before a return to normal behaviour.

277
Q

In the case of the Cocker Spaniel who got migraines what drug was given to improve symptoms

A

Topiramate

opioids and NSAIDs were inefficacious

278
Q

What happens electrically during a migraine

A

as CSD occurs there is silencing of the electroencephalograph (EEG), coupled with an increase in [K+] and neuronal depolarisation (shown by a decreased direct-current, DC)

279
Q

Which mediators are released in migraines

A

NO, 5-HT and AA can cause activation and sensitisation of nociceptors innervating blood vessels and may even reach meningeal nociceptors due to the disruption of the BBB as a result of upregulation of matrix metalloprotease (MMP)

280
Q

How might the brain and inflammation hypotheses work together to cause migraines

A

During migraine, as CSD occurs there is silencing of the EEG, coupled with an increase in [K+] and neuronal depolarisation

Also, mediators released, eg NO, 5-HT and AA can cause activation and sensitisation of nociceptors innervating blood vessels and may even reach meningeal nociceptors due to the disruption of the BBB as a result of upregulation of MMP activity

Activation of nociceptors results in the release of CGRP, SP and neurokinin A, which further drive inflammatory pain and is enhanced by the activation of a parasympathetic reflex involving activation of the SSN and the sphenopalatine ganglion (SPG) leading to release of VIP, NO, and ACh.

281
Q

Why do we think the PNS is involved in migraines

A

nausea and vomiting is a common feature of migraine

282
Q

How do we know 5-HT is involved in migraines (3)

A

During migraine: the urine levels of 5-HIAA rise
and
blood levels of 5-HT drop,

many successful anti-migraine drugs target 5-HT function

283
Q

What is sumatriptan

what is it used to treat

MOA?

A

a 5-HT1B/D/F agonist

used to treat an acute migraine attack,

action at 5-HT1B causes vasoconstriction and 5-HT1D/F agonism inhibits nociceptor activity

284
Q

What is a side effect of sumatriptan?

A

vasoconstriction in peripheral vascular beds is an unwanted side-effect and is contraindicated in coronary disease

285
Q

What is the trouble with administering sumatriptan

A

poorly absorbed when taken orally, does not cross the BBB to any great extent and has short duration of action (t1/2 = 1.5 hours)

286
Q

How can sumtriptan be taken

A

orally
nasal spray
subcut injection

287
Q

sumatriptan is a 5-HT1B/D/F agonist but is contraindicated in coronary disease. What is being developed to improve on this

A

development of selective 5-HT1D agonists has proved therapeutically disappointing, which suggests that 5-HT1F agonists may be fruitful

288
Q

Name a triptan other than sumatriptan

How do their pharmacokinetics differ

A

naratriptan

naratriptan has a longer duration of action, can cross BBB and has fewer cardiac side effects

289
Q

Name a drug that became available in 2020 to treat migraines

A

Lasmiditan, a non-triptan 5-HT1F agonist

290
Q

What is a benefit of lasmiditan over sumatriptan (3)

A

lasmiditan is 440 times more potent for 5-HT1F receptors compared to 5-HT1B/D receptors and thus does not cause vasoconstriction,

it is also far more lipophilic

readily crosses the BBB where it inhibits nociceptor activity and CGRP release

291
Q

Can NSAIDs be used to treat migraine symptoms

A

can be used to help control acute symptoms

292
Q

How does the British Association for the Study of Headaches recommend you acutely treat a migraine

A

treatment ladder:

starts with an NSAID (e.g. ibuprofen), sometimes with an antiemetic, such as domperidone, before moving on to triptans.

Use of painkillers should be limited to no more than 2-3 days a week or 10 days a month to prevent overuse and to allow preventative agents the opportunity to work.

293
Q

Drugs can be used prophylactically for migraines. What is the principal here

A

to find a drug that suits the individual patient and effective drugs should generally be continued for at least 4-6 months before a trial of discontinuation

294
Q

Name some drugs that can be used prophylactically for migraines (5)

A
propranolol
amitriptyline
topiramate 
botulinum toxin A
erenumab
295
Q

How does propranolol prevent migraines

who shouldn’t take this as a prophylactic

A

β-adrenoceptor antagonists are thought to work by preventing the extracerebral vasodilation, or by acting on the central catecholaminergic system

asthmatics

296
Q

What is amitriptyline

A

an antidepressant used, at lower doses, as a migraine preventative

297
Q

Give the MOA for amitriptyline in migraine treatment

A

unclear, but is thought to involve inhibition of Nav channels and inhibition of noradrenaline and serotonin uptake

298
Q

What are side effects of taking amitriptyline prophylactically for migraines

A

can also be sedative and typically causes a dry mouth due to anticholinergic action

299
Q

What is topiramate?

MOA?

A

an anti-epileptic repurposed as a migraine preventative

acts on numerous ion channels including voltage-gated sodium channels (inhibition) and GABA-A receptors (facilitation)

300
Q

What is a common side effect of topiramate

A

tingling hands and feet

301
Q

Botulinum toxin A can be administered as a migraine prophylactic for which type of patient?

A

only advisable in patients suffering from chronic migraine (defined as headaches on 15 or more days/month) and in whom standard preventative agents have failed.

302
Q

How is Botulinum toxin A administered as a migraine preventative

A

by intramuscular injection to the head and neck

303
Q

How does botulinum toxin A provide migraine relief

A

thought unrelated to its muscular relaxant action, but may involve decreased release of neurotransmitters from nociceptors.

304
Q

Give evidence of CGRP having a role in migraine pathogenesis (2)

A

during an attack CGRP levels in the external jugular vein are elevated

CGRP infusion induces headaches and migraine-like attacks in migraineurs, but not in non-migraineurs.

305
Q

In recent years there have been a number of randomised clinical trials investigating the efficacy of using monoclonal antibodies against CGRP or the CGRP receptor. How do these work respectively?

What else similar has been developed to treat migraines?

A

the former sequestering CGRP, the latter functioning as receptor antagonists

small molecule CGRP receptor antagonists

306
Q

Give an example of an anti-CGRP receptor monoclonal antibody

what is it used for?

A

erenumab

for preventative treatment of migraine in adults and prophylaxis of migraines in adults who experience at least 4 migraines/month respectively.

307
Q

What is FHM

what does it involve

A

Familial hemiplegic migraine, involving migraine and half body paralysis (rare)

308
Q

True or false

Migraines can have sensory symptoms

A

true

it is common to for migraine with aura to cause one sided sensory symptoms

309
Q

How do you get FHM

A

inherited through mutations in different genes, one of which, CACNA1C, encodes the voltage-gated Ca2+ channel 1.2 α subunit (CaV1.2).

310
Q

How do mutations lead to FHM

A

Mutations produce variable effects in channel function and may lead to a lower threshold for CSD initiation.

311
Q

how can you treat FHM

A

Although randomised trials have not been conducted, there is some evidence that the CaV inhibitors are efficacious in both aborting attacks and as prophylactics.

312
Q

Give 2 migraine/ gene links

A

CACNA1C - encodes the voltage-gated Ca2+ channel 1.2 α subunit (CaV1.2) and is mutated in FHM

TRESK

313
Q

What is TRESK

what is it involved in

A

a 2-pore domain K+ channel

plays a role in regulating the resting membrane potential and some dominant negative mutations in TRESK are associated with migraine

314
Q

Why might TRESK lead to migraines

A

regulates resting potential and is expressed by nociceptors and loss of TRESK activity may predispose to migraine attacks

315
Q

Why is TRESK potentially useful therapeutically

A

TRESK agonists a potential therapeutic target in the treatment/prevention of migraine.

316
Q

What are the 2 phases of the adaptive immune response

A

the induction phase and the effector phase

317
Q

How does the induction phase of the adaptive immune response begin

A

by an antigen being presented to a naïve CD4+ or CD8+ T cell by an APC (macrophage or dendritic cell), which has ingested a pathogen, breaks it down by proteolysis and then presents peptide fragments

318
Q

What do CD4+ cells do once activated

A

synthesise and express IL-2 receptors and release IL-2, which acts in autocrine fashion (i.e. on itself) causing generation and proliferation of T-helper zero (Th0) cells.

Autocrine action of IL-4 causes the production of Th2 cells, which in turn activate B cells to proliferate and give rise to plasma cells that secrete antibodies and memory B cells

319
Q

Name 4 elements involved in antibody mediated immunity

A

activation of complement,

labelling of bacteria for phagocytosis,

linkage with natural killer cells to kill antibody coated cells

activation of mast cells/basophils

320
Q

What are follicular T cells involved in

A

B cell proliferation and differentiation.

321
Q

what is iTreg and what does it act with

A

inducible Treg

acts in concert with nTreg cells (naturally occurring Treg) and is responsible for restraining the immune response to prevent excessive immune responses

322
Q

Compare, briefly, the CD8+ and CD4+ response

A

Like CD4+ cells, CD8+ cells also synthesise and express IL-2 receptors and release IL-2, which acts in an autocrine manner to generate cytotoxic T cells (TC), which kill virally infected cells.

323
Q

What type of immune response dominants type 1 diabetes

what about asthma

A

Th1 responses

Th2 response

324
Q

Give 4 autoimmune reactions which involve the Th1 response

A

Type 1 (insulin-dependent) diabetes mellitus, multiple sclerosis, rheumatoid arthritis and graft rejection

325
Q

Distinguish corticosteroids from glucocorticoids

how are mineralocorticoids

A

glucocorticoids is reserved for endogenous steroids that regulate glucose metabolism (e.g. cortisol), whereas corticosteroids includes both endogenous glucocorticoids and artificial steroids, such as prednisolone, that are used in therapy;

mineralocorticoids are also a subclass of corticosteroids, but do not concern us here

326
Q

Where are glucocorticoids synthesised

A

in the adrenal cortex in response to circulating levels of adrenocorticotrophic hormone (ACTH, released from the pituitary gland)

327
Q

What is ACTH release dependent upon

A

under the control of corticotrophin-releasing factor (CRF, release from the hypothalamus) and one function of glucocorticoids is to inhibit CRF production and thus their own systemic levels as well

328
Q

Give 5 metabolic actions of corticosteroids

A
decreased uptake of glucose by muscle/fat (consider the impact for diabetic humans), 
increase gluconeogenesis, 
increased protein catabolism, 
decreased protein anabolism 
redistribution of fat
329
Q

Give 6 anti-inflammatory actions of corticosteroids

A

decrease:

  • influx/activity of leukocytes
  • activity of monocytes
  • clonal expansion of T and B cells
  • pro-inflammatory cytokine production/action (e.g. IL-1, IL-2, IL-5, TNF-α, etc.)
  • eicosanoid production

increase
- release of anti-inflammatory factors, e.g. IL-10, IL-1ra, lipocortin 1, annexin-A1 and IκB

330
Q

What is the overall effect of corticosteroids on the immune system

A

to reduced activity of both the innate and adaptive immune response

331
Q

what do corticosteroids bind to

A

the glucocorticoid receptor (GRα), which is a nuclear receptor.

332
Q

How does the glucocorticoid receptor exist normally and what happens in the presence of corticosteroids

A

GRα is present as homomers in the cytoplasm and are bound to heat shock protein 90 (Hsp90) and other proteins.

Binding of ligand to the receptor causes dissociation of Hsp90 and enables ligand-bound GRα to form homodimers, which translocate to the nucleus and can either transactivate or transrepress a wide range of genes

333
Q

How much of the genome can corticosteroids regulate

A

GRα can regulate 1% of the total genome

334
Q

What are the 4 ways ligand-bound GRα can regulate gene expression

A

A, GRα binds to a positive glucocorticoid response element (GRE) within a promoter region for a gene with low transcriptional activity and activates the transcriptional machinery (TM)

B, the TM is constitutively driven by transcription factors (TF) and GRα binds to negative GREs (nGRE) causing TF displacement and repression of the TM;

C, the TM is driven at a high level by Fos/Jun TFs binding to their AP-1 regulatory site, the effect of which is reduced by GRα binding

D, the TFs P65 and P50 bind to the NFκB site promoting the TM, an effect that is prevented by prior binding of GRα.

335
Q

Give 3 examples of corticosteroids having non-genomic effects

A

hydrocortisone rapidly inhibits neutrophil degranulation and neither GRα antagonists nor inhibitors of translation prevent this phenomenon.

IgE-mediated mast cell degranulation is rapidly inhibited by corticosteroids in a non-genomic manner, such that membrane impermeable versions of corticosteroids have identical effects to membrane permeable versions, i.e. binding to intracellular GRα is not required.

in healthy human volunteers prone to allergic rhinitis, following induction of nasal irritation with a pollen suspension, betamethasone administration markedly reduced nasal itching within 10 minutes, the speed of this response cannot be explained by genomic effects.

336
Q

Give 9 side effects of corticosteroids

A
  • opportunistic infection
  • thinning of skin and impaired wound healing
  • candidiasis
  • osteoporosis
  • hyperglycaemia – because of glucose metabolism effects (particularly problematic in diabetic humans and animals)
  • muscle wasting
  • stomach ulcer
  • avascular necrosis of the femoral head (rare and idiosyncratic leading to a need for hip replacement)
  • adverse psychiatric effects, e.g. disturbed sleep and even psychosis
337
Q

Why is osteoporosis a side effect of corticosteroids

A

several reasons, including the reduced osteoblast/increased osteoclast activity

338
Q

Why is oral thrush a symptom of corticosteroids

A

suppression of local anti-infective mechanisms when taken orally

339
Q

name 2 drugs prescribed alongside corticosteroids

which side effects do they protect against

A

a bisphosphonate (bone protection)

a proton pump inhibitor (ulcer).

340
Q

What can happen if corticosteroid treatment in continued long term

A

Cushing’s syndrome can develop

341
Q

How can Cushing’s develop in the wild

A

from an ACTH secreting tumour

342
Q

How can Cushing’s develop in dogs

Name some symptoms if this happens

A

in dogs receiving excessive corticosteroid treatment, or due to ACTH secreting tumours

symptoms include: pot-bellied appearance, hair loss (primarily body, not face/legs), polydipsia and polyuria.

343
Q

Why should you not withdraw corticosteroids abruptly after lengthy (>1-2 weeks) therapy

A

can result in acute adrenal insufficiency (Addisonian crisis as the patient fails to synthesise enough steroids (and hence patients carry a “steroid card”)

withdrawal should thus be tapered to enable full HPA recovery and schedules are often patient specific.

344
Q

Describe the potency and length of action of 2 corticosteroids

A

hydrocortisone (name given to cortisol when used as a medication) is short acting <24hrs

dexamethasone (for treating swelling and inflammation in metastatic cancer, ) is long acting with a biological half-life of >48hrs.

345
Q

What is dexamethasone used to treat

A

for treating swelling and inflammation in metastatic cancer, as well as to Covid-19 patients requiring supplemental oxygen and/or mechanical ventilation

346
Q

Give an eg of an inflammatory skin condition that can be treated using corticosteroids

A

eczema can be treated with a 1% hydrocortisone topical cream

347
Q

name 2 brain conditions that can be treated with corticosteroids

A

reduce cerebral oedema in patients with brain tumours and those suffering from high altitude cerebral oedema

348
Q

Would you give corticosteriods to treat Addison’s or Cushing’s disease?

A

used to replace physiological levels of endogenous steroids in Addison’s disease (where the adrenal glands fail to produce sufficient steroid hormone)

349
Q

Give 2 causes of Addison’s disease

A

autoimmune destruction of the adrenal cortex and infection of the adrenal glands by tuberculosis

350
Q

How many people suffer from asthma

A

In the UK, approximately 5.5 million people suffer from asthma, about 1 in 12 people

351
Q

What disease does the underlying pathology of asthma resemble

A

COPD

352
Q

What are 3 key characteristics of asthma

A

1) inflammation of the airways
2) bronchial hyper-reactivity (hypersensitivity to irritant chemicals, cold etc. and in allergic asthma, specific allergens)
3) reversible bronchoconstriction

353
Q

Numerous cell types are involved in asthma pathogenesis, but what is the key leukocyte involved

A

Th2

354
Q

give the pathophysiology of asthma

A

a dendritic cell in the airway submucosa takes up an allergen and presents it to a CD4+ T cell, resulting in the development of a Th0 cell, which gives rise to Th2

Th2 cells:
release IL-5 (eosinophil priming) and IL-4 and IL-13 (switch B cells/plasma cells into producing IgE)
induce IgE receptor (FcεRI) expression in mast cells and eosinophils

high circulating IgE levels also increase mast cell FcεRI expression

355
Q

Why is the allergen-induced FcεRI cross-linking on mast cells in asthma bad

A

causes degranulation and release of histamine and CysLTs, which are both powerful bronchoconstrictors and increase vascular permeability – process is characteristic of the immediate/acute phase of an asthma attack

356
Q

What do mast cells release which is involved in the late phase of asthma (5)

A

release IL-4, IL-5, IL-13 and TNFα, as well as a variety of chemotaxins that recruit both eosinophils and macrophages

357
Q

What happens in the late phase of asthma

A

smooth muscle hypertrophy occurs and the eosinophils recruited to the mucosal surface release CysLTs and granule proteins such as eosinophil cationic protein and eosinophil major basic protein which both damage the epithelium resulting in airway hypersensitivity

358
Q

Epithelial cells are damaged/ lost in asthma. Why is this important?

A

epithelial cell loss means that nociceptive C-fibres are more accessible to irritant stimuli, which is an important hypersensitivity mechanism

359
Q

What are the 2 distinct ways to treat asthma

A

bronchodilators (predominantly for use in acute attacks)

anti-inflammatories (to limit the inflammatory components of both acute and late phases), which are usually taken as prophylactics

360
Q

Which asthma patients take preventative treatment

A

not everyone but recommended recommended if a person experiences asthmatic symptoms/uses a bronchodilator more than twice a week, or wakes up once a week with asthmatic symptoms

361
Q

What is the step wise approach to asthma treatment (4)

A

starts with a short-acting inhaled β2-adrenoceptor agonist used as required (e.g. salbutamol),

moving up to a regular inhaled corticosteroid (e.g. beclomethasone) to which a long-acting inhaled β2 agonist (LABA, e.g. formoterol) can be added,

then the addition of either a CysLT1 receptor antagonist (e.g. montelukast), theophylline or oral LABA (each can be tried in turn)

finally daily oral corticosteroids at the lowest dose that controls the asthma. Omalizumab can also be considered

362
Q

How do β2-adrenoceptor agonists help in asthma

A

act upon Gs-coupled β2-adrenoceptors expressed by bronchial smooth muscle to induce relaxation and bronchodilation and β2-adrenoceptors expressed by mast cells to inhibit degranulation

363
Q

How do beta2 AR agonists cause bronchodilation

A

increase adenylate cyclase activity = increase cAMP/PKA = myosin light chain kinase phosphorylation and inactivation

364
Q

Name a short and long term β2-adrenoceptor agonist used to treat asthma

how long does each last and when is each used

A

salbutamol is a short-acting drug taken as needed to control symptoms, the maximum effect occurs within 30 minutes and the duration of action is 3-5 hours;

formoterol is a LABA used prophylactically and has a duration of action of 8-12 hours.

365
Q

Why does formoterol have a longer action than salbutamol

A

because its long, lipophilic tail enables it to be incorporated into the plasma membrane, which acts as a drug reservoir, and in addition, the lipophilic tail binds to an extra binding site on the β2-adrenoceptor itself

366
Q

Give 2 side effects of β2-adrenoceptor agonists for asthma treatment

A

tremor (excitation of β2-adrenoceptors in skeletal muscle) and tachycardia (excitation of facilitatory β2-adrenoceptors in cardiac noradrenergic nerve terminals).

367
Q

Are LABAs used to treat COPD?

A

yes

including the “ultra” long-acting β2-adrenoceptor agonist indacaterol

368
Q

What is beclomethasone

A

an inhaled corticosteroid, commonly used in the prophylactic treatment of asthma

369
Q

Why does beclomethasone take some time to work

What does this mean they are not useful for

A

action is genomic

so are not useful reversing the acute bronchoconstriction during an asthma attack

370
Q

How can you reduce the systemic dose of beclomethasone

what does this do

A

when treating asthma, can be inhaled directly

reduces side effect profile but oral candidiasis (thrush) infections can also occur

371
Q

How can you aid patient compliance for asthmatics taking beclomethasone

A

Beclomethasone can be combined with formoterol to lessen the number of inhalers used by patients

372
Q

What is the MOA of theophylline

A

inhibits PDE to increase [cAMP] and therefore induces bronchodilation/mast cell stabilisation by the same mechanism as β2-adrenoceptor agonists

373
Q

What is theophylline used for

A

it is used prophylactically against asthma and not advised in treating acute attacks

374
Q

Give a side effect of theophylline

A

commonly cardiovascular, e.g. tachycardia

375
Q

Why should theophylline be used with caution in polypharmacy

A

is metabolised by CYP450, which can be induced/inhibited by many drugs

376
Q

What is Montelukast

how does it compare to other drugs that treat the same disease

A

a CysLT1 receptor antagonist and can induce bronchodilation to treat asthma,

less effective than salbutamol and is used prophylactically.

377
Q

Name a muscarinic agonist used to treat asthma and COPD

A

ipratropium (used in management of acute attacks)

378
Q

What is ipratropium

give brief MOA description

A

a short-acting muscarinic antagonist

produces bronchodilation by inhibiting M3 receptors on the smooth muscle

379
Q

true or false

You cannot use sodium cromoglycate to treat asthma

A

false
Sodium cromoglycate is a mast cell stabiliser and can be used prophylactically in the treatment of allergic asthma where mast cells are key players in the pathology, such as in research scientists who develop allergies to laboratory animals.

380
Q

What is omalizumab?

What is it used against?

A

humanised monoclonal Ab against IgE (5% mouse, 95% human sequence)

recommended cases of severe, persistent allergic asthma that are refractory to treatment with corticosteroids and long-acting β2-adrenoceptor agonists.

381
Q

What is the MOA of omalizumab

A

humanised monoclonal Ab against IgE

By binding to the Cε3 region of IgE (part of the heavy chain that binds to FcεRI), omalizumab prevents IgE binding to FcεRI, which reduces FcεRI expression and inhibits allergen induced mast cell degranulation

382
Q

Why does omalizumab not activate FcεRI itself

A

Omalizumab is built on an IgG framework

383
Q

How is Omalizumab administered

A

given as an injection every 2-4 weeks

384
Q

Who is omalizumab recommended for

A

only recommended for people who frequently suffer from severe allergic asthma attacks necessitating frequent hospital attendances, where other treatments have failed

385
Q

can cats get asthma

A

Cats can also develop pulmonary conditions showing similarities to asthma and they, like humans, can be treated with β2-adrenoceptor agonist bronchodilators like salbutamol and corticosteroids, such as fluticasone – harder to administer to a cat than a human, but inventions like the AeroKat perhaps make it (slightly) easier.

386
Q

what is a pulmonary condition that horses get that is similar to asthma

how is it treated

A

Recurrent Airway Obstruction (RAO, or heaves), which can be treated with both bronchodilators (e.g. clenbutarol administered orally with feed) and corticosteroids (e.g. fluticasone). In contrast to treating humans, sodium cromoglycate has little efficacy, which likely reflects a more important role for neutrophils than mast cells in the pathophysiology of RAO.

387
Q

What is COPD characterised by

A

chronic inflammation of the airways and lung tissue leading to narrowing of the airways and shortness of breath.

388
Q

What accounts for the majority of cases of COPD in the a) developed world and b) the developing world

A

a) smoking

b) significant contribution from pollutants (e.g. smoke from cooking with wood with poorly ventilation).

389
Q

what are bronchial biopsies from asthmatics characterised by

how does this differ from those from COPD patients

A

asthma: characterised by activated mast cells and eosinophils

COPD: enriched in neutrophils and macrophages + fibroblast proliferation (leading to small airway fibrosis)

390
Q

What contributes the irreversible nature of airway narrowing in COPD

A

fibroblast proliferation leading to small airway fibrosis (common in COPD)

this is a stark contrast to the reversible inhibition seen in asthma

391
Q

What differentiates COPD from asthma (4)

A

Whereas bronchial biopsies from asthmatics are characterised by activated mast cells and eosinophils, biopsies from those with COPD are enriched in neutrophils and macrophages

airway narrowing is irreversible in COPD but reversible inhibition is seen in asthma

Alveolar tissue damage is important in COPD but not asthma

whereas small airway epithelial loss is common in asthma (leading to exposure of nociceptors), epithelial cells often proliferate in COPD

392
Q

Can you use SABA bronchodilators to treat COPD

A

not particularly effective because of the limited reversible nature of the bronchoconstriction occurring in COPD

They are not recommended for treatment on a regular basis, but can be used in acute exacerbations

393
Q

Why are short acting bronchodilators not particularly effective in COPD?

A

the limited reversible nature of the bronchoconstriction occurring in COPD (remember, unlike in asthma that fibrosis also occurs and affects tissue elasticity).

394
Q

Can LABAs be used in COPD

A

yes: formoterol and indacaterol are used, often in combination with long-acting muscarinic antagonists (LAMAs, e.g. tiotropium, rather than short-acting ipratropium, which is used for treating milder COPD as determined by lung function tests)

395
Q

How are LABAs usually administered to COPD patients

A

in LABA-LAMA combination inhalers

396
Q

Why is the LABA-LAMA combination effective in treating COPD

A

different mechanisms of action leads to additive effects: LABA = β2-adrenoceptor activation = increase AC activity = increase cAMP/PKA = myosin light chain kinase phosphorylation and inactivation
+
LAMA = M3 inhibition = prevention of PLC activation and IP3 generation

397
Q

Are orally given corticosteroids used to treat COPD

A

they are largely ineffective, although are used in the treatment of acute exacerbations of COPD, for example when there is a coexistent infection, or where small airway inflammation is predominant

398
Q

Explain the lack of efficacy of corticosteroids in treating COPD

A

in COPD, there is a reduction in expression and activity of histone deacetylase 2 (HDAC2): superoxide (O2-) and nitric oxide (NO) produced from cigarette smoke and activated immune cells results in the formation of peroxynitrite (ONOO-), which nitrates HDAC2 at the active site leading to its inactivation, ubiquitination and degradation.

With lowered levels of HDAC2, there is increased acetylation of GRα, which prevents it from inhibiting NFκB-driven inflammation.

399
Q

What is a promising therapy for COPD

A

Reversing corticosteroid resistance

400
Q

Can you use theophylline to treat COPD

A

yes: used as a sustained release preparation to treat COPD

401
Q

How does theophylline work in treating COPD

A

bronchodilator effects (due to increased cAMP levels)

raises cAMP levels in neutrophils and other immune cells causing a decrease in chemotaxis and activation

402
Q

What is the main PDE expressed in neutrophils

why is this important for COPD treatment

A

PDEIV (also main PDE expressed in neutrophils, T cells, ad macrophages)

neutrophils highly expressed in COPD
specific PDEIV inhibitor roflumilast is approved for treating COPD; it is used as an add on therapy to LABAs.

403
Q

When is O2 therapy used in COPD patients

A

for those with low blood O2 levels, long-term O2 therapy is used

sometimes only necessary during exercise

NOT for smokers

404
Q

Which COPD patients cannot receive O2 therapy

A

O2 and cigarettes do not mix: home O2 therapy should not be prescribed to patients who continue to smoke

405
Q

What is ambulatory O2 therapy

A

when O2 therapy is only necessary during exercise

406
Q

Give 3 potential COPD therapies

A

modulation of neutrophil chemotaxis

anti-fibrotic therapy,

inhibition of elastase activity

407
Q

How are trials for drugs which modulate neutrophil chemotaxis to treat COPD going?

A

initial trials with CXCR2 antagonists were promising, but were discontinued when larger trials produced negative results

408
Q

Why might drugs which inhibit elastase work to treat COPD

A

elastase elevated in COPD and may contribute to the development of emphysema

409
Q

Which sex displays higher prevalence of autoimmune diseases

A

female

410
Q

Give 2 examples of autoimmune diseases that involve the thyroid

A

Graves’ disease (autoantibodies activate the thyrotropin receptor causing increased thyroxine release)

Hashimoto’s disease (autoantibodies against proteins involved in thyroxine synthesis, e.g. thyroglobulin; this typically leads first to hyperthyroidism due to cell death and release of thyroxine, followed by hypothyroidism as thyroxine supplies are exhausted and not replaced)

411
Q

What happens in myasthenia gravis

A

autoantibodies against the nicotinic receptor that prevent the effects of acetylcholine

412
Q

What happens in the following:

  • primary biliary cirrhosis
  • rheumatoid arthritis
A
  • primary biliary cirrhosis -> immune attack against bile ducts of the liver)
  • rheumatoid arthritis -> immune attack on the synovium surrounding joints
413
Q

What is the prevalence of RA

How common is severe disability following development of RA

A

1%

one third of whom were likely to become severely disabled due to joint damage, prior to the development of disease modifying therapies

414
Q

What causes joint damage in RA

A

a combination of factors that are driven by activated Th1 cells:

macrophages are stimulated to release IL-1 and TNFα causing release of metalloproteinases from osteoclasts and fibroblasts that cause cartilage and bone destruction, an effect exacerbated by infiltrated inflammatory cells, e.g. neutrophils release proteases and ROS that both cause damage.

Hyperplasia of the synovium also occurs resulting in pannus formation as fibroblast-like synoviocytes proliferate and invade the joint and release proinflammatory cytokines and proteases.

415
Q

True or false

RA treatment doesn’t treat underlying disease progression

A

some treat only the symptoms, whereas others reduce disease progression, so-called disease-modifying antirheumatic drugs (DMARDs).

416
Q

How do NSAIDs affect RA

A

used to relieve pain and lessen inflammation but do not alter the underlying disease process

417
Q

Which NSAIDs are used for RA treatment (name 4)

A

Diclofenac (or the misoprostol combination drug Arthrotec) has largely been superseded by naproxen

etorixcoxib

418
Q

why has naproxen superseded Arthrotec for RA treatment

why is etorixcoxib also preferable

A

lower cardiovascular risk

COX2 selective so significant reduction in risk of GI bleeding

419
Q

name an oral corticosteroid used to treat RA

when are they given as treatment

A

prednisolone (provides symptomatic relief and suppresses disease activity)

Due to the side-effects associated with systemic/chronic corticosteroid use, they are usually only used to treat disease flare-ups, or as a bridging therapy when waiting for a DMARD to take effect

420
Q

What are the major mechanisms of corticosteroids for the treatment of RA

A

decreased transcription of IL-2, which is important for driving Th cell proliferation, and the decreased transcription of IL-1/TNFα

421
Q

How can corticosteroids be administered in RA

A

orally

can also be delivered locally, i.e. intra-articular injections

422
Q

Which drug is the mainstay of management of RA

is it used alone?

A

methotrexate

can be used alone or in combination with other DMARDs

423
Q

How does methotrexate work in RA treatment

A

folic acid inhibition lowering the production of tetrahydrofolate that is required for purine nucleotide and thymidylate synthesis and thus is anti-proliferative, reducing the immune response

424
Q

How does methotrexate provide folic acid inhibition

A

inhibition of dihydrofolate reductase and competition with folic acid transport into cells

425
Q

How long does it take for the effects of methotrexate in RA treatment

How is it usually administered

A

3–12 weeks

orally to be taken once a week, but can also be administered by weekly subcutaneous injection; weekly administration is important (NOT daily)

426
Q

give 2 side effects of methotrexate

how can these be limited

A

bone marrow suppression and GI disturbance

weekly folic acid supplement

427
Q

What is a common but deadly prescribing error seen with methotraxate

A

incorrect daily administration of methotrexate is a recognised prescribing error that can prove fatal due to the profound effects on bone marrow suppression and impairment of immune system function

428
Q

What is a popular first choice DMARD for IBS (not methotrexate)

A

Sulfasalazine

429
Q

How is Sulfasalazine administered

how is it metabolised

what is a likely MOA

A

taken orally daily

broken down by colonic bacteria to produce sulfapyridine (a sulphonamide) and 5-aminosalicylic acid (a salicylate) and one likely MOA is that the 5-aminosalicylic acid scavenges ROS released from neutrophils

430
Q

When do the therapeutic benefits of sulfasalazine seen

A

not until after 3 months

431
Q

What are unwanted effects of sulfasalazine

A
GI disturbances, skin rashes, 
bone marrow suppression 
hepatotoxicity 
tears and contact lenses can be stained yellow 
urine may appear orange
432
Q

What was hydroxychloroquine developed to treat

A

malaria (also now used as a DMARD)

433
Q

MOA of hydroxychloroquine

A

As a lipophilic weak base, hydroxychloroquine accumulates in cytoplasmic acidic vesicles, which can reduce antigen presentation by macrophages and reactive oxide species generation in neutrophils

434
Q

What are some side effects of hydroxychloroquine (6)

A
ocular toxicity,
GI upset, 
skin reactions, 
seizures,
myopathy, 
psychiatric disturbance.
435
Q

What does leflunomide do

A

inhibits dihydroorotate dehydrogenase, a key enzyme in pyrimidine synthesis, and thus it inhibits synthesis of DNA and RNA, with its main effect being on rapidly dividing cells such as B and T cells.

436
Q

What can leflunomide be used to treat

side effects?

A

RA

Adverse effects include GI disturbance, bone marrow suppression and hepatotoxicity.

437
Q

Which drugs are reserved for RA patients who have failed conventional DMARDS (5)

what are these often more effective with

A

biological DMARDS (Anti TNFα therapies, Anti-IL-6 therapy, Anti-B cell therapy, Anti-T cell therapy, Anti-IL-1 therapy)

as combination treatments with methotrexate

438
Q

Name 3 drugs used in anti TNFα therapies

A

Etanercept
Infliximab
adalimumab

439
Q

What is etanercept

A

a fusion protein of the soluble TNFα receptor and the Fc portion of IgG1, which acts to mop up the high levels of TNFα that occur in RA

440
Q

What are Infliximab and adalimumab

A

monoclonal anti-TNFα antibodies (mAb, chimeric and human respectively) that, like etanercept, sequester TNFα.

441
Q

What are adverse effects of anti TNFα therapies (4)

A

allergic reactions, bone marrow suppression,
increased susceptibility to infections (particularly tuberculosis and hepatitis B reactivation)
MS

442
Q

Give an unexpected side effect of anti TNFα therapies

A

several cases of multiple sclerosis have been reported in patients following anti-TNFα treatment for inflammatory arthritis

443
Q

Name a drug used in anti IL-6 therapy

what does it target

A

Tocilizumab (a mAb)

targets the IL-6 receptor, inhibiting the action of IL-6.

444
Q

Why can tocilizumab be used to treat RA

A

IL-6 is a pro-inflammatory cytokine and, like TNFα and IL-1 is upregulated in RA. IL-6 also drives the production of c-reactive protein (CRP), which is commonly measured in blood tests, and so tocilizumab tends to be targeted towards patients with significantly elevated levels of CRP.

445
Q

Name a drug used in anti-B cell therapy

A

rituximab (a mAb)

446
Q

What does rituximab target

Name a disease that it can be used to treat

A

CD20 that is primarily expressed by B cells,

can be used in the treatment of RA, its therapeutic effect being through B cell destruction.

447
Q

What risks are associated with rituximab

A

increased risk of infection (like all DMARDs)
especially, an increased risk of a potentially fatal brain infection called progressive multifocal leukoencephalopathy, which is caused by reactivation of the John Cunningham (JC) virus.

448
Q

Name a drug used in anti-T cell therapy

describe it

A

abatacept

a synthetic fusion protein of the costimulatory cytotoxic T-lymphocyte protein 4 (CTLA-4) and the Fc fragment of human IgG1

449
Q

Give MOA of abatacept

A

binds to CD80 and CD86 on antigen presenting cells, so interferes with the ability of antigen presenting cells to activate T cells

450
Q

Name a drug used in anti-IL1 therapy

describe the drug

A

anakinra

a recombinant version of the IL-1 receptor antagonist, which binds to the IL-1R with a higher affinity than IL-1 itself, but does not initiate receptor signalling

451
Q

When do you use anakinra to treat RA

A

On the balance of clinical benefits and cost-effectiveness, it is not recommended for treatment of RA in the UK, but is used in other countries

452
Q

Is methotrexate used in animals

A

although usually reserved for treating certain forms of cancer methotrexate can also be used to treat dogs with arthritis.

453
Q

How do you treat RA in horses and dogs

A

NSAIDs and corticosteroids

454
Q

How do the different classes of biological DMARD compare (4)

A

All biological DMARDs are given by injection, either intravenous or subcutaneous,

all are expensive.

They are all associated with an increased risk of infections.

Each class has similar efficacy, but failure to respond to one class does not determine response to another class, so they tend to be tried serially

455
Q

What is a major drawback of the therapeutic avenue of immunosuppression to prevent eg graft rejection

A

the immune system is compromised and thus less able to respond to infection

456
Q

Name 3 steroid sparing agents

A

azathioprine
methotrexate
mycophenolic acid.

457
Q

How do corticosteroids exert their immunosuppressive effects

A

via decreasing the transcription of a variety of cytokines, including those that drive Th cell proliferation and those that drive inflammatory responses, (e.g. IL-1 and TNFα), while at the same time upregulating anti-inflammatory factors such as IL-1ra

458
Q

Which cytokine that drives Th0 proliferation is inhibited by corticosteroids

what does it cause

A

IL-2 which causes generation and proliferation of Th0 cells and proliferation of Th0 cells into Th1 cells

459
Q

Name an anti inflammatory factor that is upregulated by corticosteroids

A

IL-1ra

460
Q

How does azathioprine work

A

is a prodrug for 6-mercaptopurine (6-MP), which is converted to thioinosic monophosphate (TIMP), which is converted by two different enzymes to produce two products that inhibit DNA function:

Thiopurine methyltransferase (TPMT) converts TIMP to 6-methyl-TIMP (MeTIMP), which inhibits de novo purine synthesis

and

inosine monophosphate dehydrogenase (IMPDH) converts TIMP to 6-thioguanosine nucleotides (6-TGN), which are incorporated into cellular nucleic acids resulting in inhibition of nucleotide and protein synthesis

461
Q

How is 6-MP converted to TIMP

A

by hypoxanthine phosphoribosyl transferase (HPRT)

462
Q

What is the overall effect of azathioprine

why is this useful

A

inhibits cellular proliferation

has a greater effect upon rapidly proliferating cells, such as T cells and B cells when activated, and thus azathioprine inhibits T cell and B cell proliferation making it a useful drug in preventing transplant rejection and autoimmunity

463
Q

Name 3 conditions you can use azathioprine to treat

A

IBS
RA
graft rejection

464
Q

What are the side effects of azathioprine

A

effects include GI upset, hepatotoxicity and bone marrow depression
(you can also get accumulation of cytotoxic 6-thioguanine nucleotide analogues leading to toxicity)

465
Q

How common is accumulation of cytotoxic 6-thioguanine nucleotide analogues as a side effect of azathioprine

why is this

how can this be prevented

A

10%

TPMT levels vary, 10% of people having very low activity and thus accumulation of cytotoxic 6-thioguanine nucleotide analogues leading to toxicity

by checking TPMT levels, or close monitoring of blood counts/liver function on starting treatment

466
Q

Can azathioprine be used in dogs

A

yes as an immunosuppressant

467
Q

True or false

methotrexate is a folic acid analogue

A

true
targets rapidly proliferating cells, such as those of the immune system. It is used in the treatment of many autoimmune conditions such as RA

468
Q

What is mycophenolic acid

A

derived from the fungus Penicillium stoloniferum, which inhibits IMPDH, an enzyme crucial for de novo guanosine synthesis (so has greatest effect on rapidly dividing B and T cells)

469
Q

What is mycophenolic acid used for

A

used to diminish transplant rejection but also used in autoimmune diseases such as myasthenia gravis.

470
Q

How is cyclophosphamide activated

A

a prodrug, being converted by hepatic P450 enzymes first to aldophosphamide and then the active phosphoramide mustard

(alkylating agent)

471
Q

MOA of active phosphoramide mustard

side effects?

A

has two alkylating groups and cross links guanine in DNA via N7 groups

bischloroethylamine undergoes cyclisation, releasing Cl- and forms an unstable immonium cation, the strained ring opens to form a reactive cabonium ion, which reacts with guanine’s N7 to produce 7-alkylguanine.
DNA replication is thus defective because 7-alkylguanine pairs with thymine producing substitution of A-T for G-C, it can also cause guanine excision and chain breakage.

immunosuppressive effect is due to killing of dividing T and B cells, but this occurs alongside side effects: bone marrow depression, potential infertility, bladder irritation and cancer

472
Q

Why can cyclophosphamide lead to cancer

is this seen anywhere else

A

its metabolite acrolein activates TRPA1 expressed by bladder innervating nociceptors,

acrolein is also present in cigarette smoke and some forms of tear gas

473
Q

What is cyclophosphamide used to treat

A

used to treat refractory autoimmune conditions and some cancers

474
Q

What is CsA

A

Ciclosporin A

an immunosuppressive isolated from the fungus Tolypocladium inflatum

475
Q

Describe the the structure of CsA

What is its main action

A

a cyclic, 11 amino acid peptide

predominant action is to prevent T-cell proliferation via suppression of IL-2 synthesis.

476
Q

What normally happens within the T cell when an antigen interacts with the TCR?

How does this differ when CsA is present

A

increase in [Ca2+]i that stimulates the activity of a serine-threonine phosphatase called calcineurin (CaN), which then dephosphorylates the nuclear factor of activated T-cells (NF-AT) enabling its translocation to the nucleus and consequent upregulation of IL-2 transcription

CsA binds to a cytoplasmic protein called cyclophilin (CpN), which is member of the immunophilin group of proteins. The CsA-CpN complex binds to and inhibits CaN, which thus prevents NF-AT dephosphorylation and it is retained in the cytoplasm preventing IL-2 upregulation and is thus used in suppressing the rejection of transplanted organs

477
Q

What is CsA used to treat in dogs

A

atopic dermatitis

478
Q

What is another name for Tacrolimus

What is this drug

A

FK506

macrolide antibiotic produced by Streptomyces tsukubaensis, which binds to an immunophilin called FKBP

479
Q

What is the importance of FK506 binding to FKBP

A

the complex inhibits CaN (similar to the action of the CsA-CpN complex)

thus thus NF-AT is retained in the cytoplasm and IL-2 synthesis is inhibited

480
Q

What are tacrolimus and CsA used to treat

A

transplant rejection

481
Q

What is basiliximab t

A

a monoclonal antibody against the CD25 alpha subunit of the IL-2 receptor and thus functions as an IL-2 receptor antagonist

482
Q

Why is basiliximab effective in preventing transplant rejection

A

it is an IL-2 receptor antagonist. IL-2 receptors are upregulated on activated T-cells and B-cells so basiliximab decreases the incidence and severity of acute rejection following transplantation.

483
Q

Name a immunosuppressive drug that works in a similar way to abatacept

A

belatacept

484
Q

Describe the structure of belatacept

A

fusion protein of the extracellular domain of cytotoxic T-lymphocyte protein 4 (CTLA-4) and the Fc fragment of human IgG1

485
Q

T cells require co-stimulation for activation. What does this involve?

Which drug exploits this

A

2 signals: 1) interaction between the major histocompatibility complex (MHC) of the APC with the T cell receptor

and

2) interaction between CD80/86 ligands of the APC with CD28 of the T cell

Belatacept - acts to bind CD80/86 and prevent T cell costimulation

486
Q

What does the CTLA-4 of a T Cell do

Why is the CTLA-4 domain of belatacept special

A

its activation by CD80/86 sends an inhibitory signal to the T cell

has 2 amino acid variations that confer greater affinity for, and slower dissociation from, CD80/86

487
Q

When is belatacept used

A

used in combination with other drugs to prevent graft transplant rejection

488
Q

What can happen if you stimulate CD28 on T cells alone

How was this information used pharmacologically

A

can cause activation and proliferation of regulatory T cells, which are involved in preventing autoimmune reactions

an anti-CD28 agonist monoclonal antibody TGN1412 was trialled following promising results from non-human animal tests, but was abruptly ended after causing a cytokine storm in humans due to its activating effects being non-selective, i.e. in humans at the dose used pro-inflammatory memory T cells were activated alongside regulatory T cells

489
Q

Does TGN1412 have a future?

A

maybe but must be administered at lower doses than in original trial to enable selective activation of Treg cells

490
Q

Give other name of rapamycin

what is this drug

A

sirolimus

a macrolide antibiotic produced by Streptomyces hygroscopicus, which like tacrolimus binds to FKBP and inhibits mTOR

491
Q

How does sirolimus differ from tacrolimus

A

both bind to FKBP but sirolimus-FKBP inhibits mTOR whereas tacrolimus-FKBP inhibits CaN

492
Q

What is mTOR

what is the main target of sirolimus

A

mammalian target of rapamycin (mTOR), which is a serine/threonine kinase involved in cell cycle progression and protein synthesis

mTORC1 (mTOR complex 1)

493
Q

What is the overall MOA of sirolimus

A

binds FKBP, forming a complex which inhibits mTOR, inhibiting mTORC1 especially.

this leads to decreased T cell activation and proliferation and a decreased immune response

494
Q

What are the uses of sirolimus (2)

A

prevent transplant rejection

used to coat coronary stents to prevent restenosis (thanks to its antiproliferative effects)

495
Q

How do you create a polyclonal antibody

A

host organism can be injected with an antigen (e.g. inactivated bacterial toxin), serum extracted from the host would contain antibodies against the antigen, but they would be a mixture of antibodies because numerous plasma cells react to a particular antigen and produce different antibodies

496
Q

What are 2 drawbacks of polyclonal antobodies

A

Each serum generated in this manner will be of variable potency and there is only so much serum that can be collected

497
Q

What is the limit of collecting serum for polyclonal antibodies

A

for generating antibodies for the lab, one standard protocol involves injecting two rabbits with antigen and collecting a total of 75 ml/rabbit over 70 days

498
Q

How did Milstein and Köhler describe making a mAb

A

fusing single mouse B-cells with immortalised tumour cells to produce a hybridoma that could grow indefinitely to produce a single antibody

499
Q

2 advantages of mAbs over polyclonal Abs

A

mAbs have known potency and there is no limitation to the amount that can be produced.

500
Q

Give the full method for creating a mAb (3)

A

Mice are firstly injected with an antigen and antibody producing B cells are isolated from the spleen, these are then mixed with myeloma cells (HPRT-ve) in polyethylene glycol to produce hybridomas;

hybridomas are selected by growing in a medium containing a dihydrofolate reductase inhibitor, which kills off unfused myeloma cells, whereas hybridomas can create new nucleotides from supplements in the medium because they express B cell HPRT, unfused B cells have a short life and die off.

B cells are then screened for specificity (e.g. using an ELISA) and then using limiting dilution individual cells/clones are isolated, which can be propagated and then mAb collected

501
Q

Describe the 2 key regions of an antibody

A

2 key domains:
Fc region - reacts with cell surface Fc receptors that are expressed by neutrophils (FcγRI, respiratory burst and phagocytosis), macrophages (FcγRII, phagocytosis) and natural killer cells (FcγRIII, antibody-dependent cell-mediated cytoxicity, ADCC)

Fab- contains the variable and hypervariable regions (also known as the complementarity determining regions, CDR), which bind the antigen in question

502
Q

Why were 1st gen mAbs problematic (3)

A

a mouse injected with, for example, human TNFα will make mouse antibodies (which can be isolated as mAbs using hybridoma technology) and thus when injected into humans they were observed to produce an immune response in over half of recipients because of the generation of human anti-mouse antibodies

relatively low circulatory half-life

unable to activate human complement

503
Q

What is Muromonab-CD3

A

murine mAb that targets CD3

was the first mAb to be approved for clinical use in humans to reduce acute transplant rejection

504
Q

Go through the stages of mAb development and give an example of each

A

Using recombinant DNA technology, the first strategy was to create chimeric mAbs that contained the human Fc region, but mouse variable region (e.g. infliximab).

Next, humanised mAbs were developed where only the CDR was of mouse origin and these produce very little immune response (e.g. omalizumab, alemtuzumab and rituximab)

505
Q

What is the benefit of chimeric and humanised mAbs over murine ones

A

In both chimeric and humanised mAbs, the presence of the human Fc region lengthens the plasma half-life because they can bind to neonatal Fc receptor (FcRn) on endothelial cells and are recycled.

506
Q

How can human mAbs be made (2)

name one that is in clinical use

A

either by using transgenic mice in which the mouse immunoglobulin genes are swapped with the human equivalents, or through phage display

adalimumab

507
Q

How are mAbs administered

A

By injection every 2 weeks (although this differs greatly e.g., infliximab is administered every 2 weeks and others once per month)

508
Q

Are there still risks with taking mAb medication

A

Due to their high specificity, there are few off-target effects, but mAbs that target functioning of the immune system can precipitate latent disease or encourage opportunistic infections.

509
Q

What does alemtuzumab do

what is it used to treat

A

binds to CD52, an antigen on mature lymphocytes, but not the stem cells from which they derive, and targets them for destruction

used in treating chronic lymphocytic leukaemia, cutaneous T cell lymphoma, T cell lymphoma and multiple sclerosis

510
Q

What do each of the mAb suffixes mean

A
  • ximab = chimeric
    -axomab = rat/mouse hybrid
    -zumab = humanised
    –umab = human
511
Q

True or false

mAbs are only used in human medicine

A

false (but used to be true)
Nexvet used a process called PETization to make canine, feline and equine mAbs and was bought out by Zoetis, who have marketed an anti-IL-31 mAb for treating dermatitis in dogs. Publications suggest that anti-NGF mAbs are in late-stage development for treating chronic pain such as that experienced in feline/canine osteoarthritis.

512
Q

What do bi-specific Abs do

A

involve stitching together two halves of different antibodies, the idea is that one antibody binds the target cell and the other binds a cytotoxic cell, i.e. the killing machinery is localised to the target

513
Q

Give an example of a bi-specific mAb

describe

A

catumaxomab

a rat/mouse hybrid mAb that binds epithelial cell adhesion molecule and CD3 on tumour cells and T cells respectively leading to the destruction of the cancer cell

514
Q

What is catumaxomab licensed to treat

A

ascite in cancer patients

515
Q

Why has catumaxomab been termed trifunctional

A

the Fc portion can bind the Fc receptors on macrophages etc

516
Q

Give a possivle option for development of a bi-specific mAb to treat inflammation

A

one that can sequester two molecules simultaneously, e.g. TNFα and IL-1.

517
Q

How can you engineer improvement of Ab-NK cell coupling

A

most antibodies and serum IgG are fucosylated, but antibodies engineered to be afucosylated have a higher affinity for the FcγRIII on NK cells and overcome competition with serum IgG thus increasing the window of time for ADCC to occur.

518
Q

How does fucosylation of Fc affect its receptor

A

Fucosylated Fc causes displacement of the oligosaccharide tree connected to Asn162 of FcγRIII, which leads to an increased distance of the carbohydrate-carbohydrate contact between Fc and FcγRIII and thus reduced bond strength that explains the reduced affinity of fucosylated Abs

519
Q

True or false

fucosylated Fc Abs can outcompete afucosylated, serum IgG

A

false
afucosylated Fc Abs can outcompete fucosylated, serum IgG

Fucosylated Fc causes displacement of the oligosaccharide tree connected to Asn162 of FcγRIII, which leads to an increased distance of the carbohydrate-carbohydrate contact between Fc and FcγRIII and thus reduced bond strength

520
Q

What is the other name for Herceptin

what is Herceptin used to treat

Is this an effective treatment

A

trastuzumab

used to treat HER2 positive breast cancers

yes but resistance often arises

521
Q

How can Herceptin treatment of HER2 positive breast cancer be improved (give details of MOA)

A

Trastuzumab emtansine (T-DM1) is a combination of trastuzumab (T) with a derivative of the antimicrotuble compound maytansine (DM1).

Binding of T-DM1 to HER2 causes internalisation, lysosomal degradation and release of DM1, which can bind to tubulin and prevent polymerisation causing cytotoxicity;

T-DM1 also retains the basic actions of trastuzumab: disruption of HER2 signalling and targeting tumour cells for ADCC

522
Q

Why would pharmacologists mutate the Fc region of some mAb
(think pH)

give an example

A

improved pharmacokinetics by mutation of the Fc region to improve binding to FcRn at low pH so that more is recycled out of cells and not broken down by intracellular degradation machinery.

Fc portion of IgG binds FcRn with high affinity at acidic pH (e.g. in endosome), but lower affinity at physiological pH, increasing this affinity further would increase the circulation time.

523
Q

What is the problem with mAbs being large proteins

A

diffusion into tissues is slow and access to intracellular targets is problematic

524
Q

mAbs are large proteins and thus diffusion into tissues is slow and access to intracellular targets is problematic. How can we overcome this? (3)

A

shrink the peptide scaffold that displays the antigen binding loops

create bicyclic peptides, which due to their rigid conformation can bind with high affinity and specificity to protein targets, but are more resistant to serum proteases than linear peptides

creation of nanobodies

525
Q

What are nanobodies

A

the isolated variable heavy chains, containing the CDRs, from Abs produced by camelidae and certain cartilaginous fish - they are single domain abs with low Mr

526
Q

What are 2 advantages of using nanobodies

disadvantage?

A

+larger loops in camelidae CDRs enables greater antigen-interacting surface than heavy chain CDRs from standard IgG Abs.
+a low molecular weight (~12-15 kDa vs. ~150 kDa of conventional mAbs) means they are likely to have better tissue permeability and tissue penetration

  • also means they have a lower plasma half-life
527
Q

What is osteoarthritis (OA)

A

a degenerative joint disease

528
Q

How is RA connected to OA

A

Unlike RA, osteoarthritis (OA) is not an autoimmune condition, but rather a degenerative joint disease that sometimes occurs as a consequence of rheumatoid arthritis

529
Q

What is another name for OA

A

degenerative joint disease (DJD).

530
Q

What characterises OA

A

disruption of the equilibrium between cartilage breakdown and repair, which results in a net loss of articular chondrocytes

531
Q

True or false

cartilage damage is an inevitable result of ageing

A

true

however, this does not mean everyone will develop OA

532
Q

What are risk factors for developing OA (3)

A

age – onset is most common from late 40s onwards,
gender – OA is more common in women than men,
obesity – increased weight bearing is a key factor in OA of the knee, and joint injury/abnormality

533
Q

What is OA usually a consequence of in dogs and cats

A

usually a consequence of a developmental orthopaedic disease, such as hip dysplasia (very much breed specific, and if I had a choice I would rather be born a whippet, 1.0% dysplastic, than a pug, 71.7% dysplastic)

although the aetiology is less well understood in cats the degenerative process itself is similar to that observed in other species and affects similar joints to as in dogs, i.e. hips, stifle and elbow.

534
Q

What is considered a key hallmark of OA (ie differentiates it from cartilage damage in non-OA patients)

A

bone remodelling

535
Q

How is the bone remodeled in OA

early and late

A

early-stage OA is associated with thinning of the subchondral plate,

but in late-stage disease there is a decrease in bone resorption, but no decrease in bone formation leading to the development of subchondral sclerosis (increased bone density) and bony outgrowths at joint margins called osteophytes

536
Q

What happens to the synovium in OA

A

often inflamed , sometimes being observed in early-stage, late-stage or both stages of OA

537
Q

Is OA considered to be an inflammatory disease?

A

no
despite inflammation of the synovium it is considered non-inflammatory because the synovial fluid typically contains a lower leukocyte count than that which defines an inflammatory condition

538
Q

What are 2 key symptoms associated with OA

A

decreased range in joint movement and pain

539
Q

True or false

OA is the most common cause of joint pain

A

true

globally ~20% of chronic pain is related to OA.

540
Q

How can you treat OA

A

NO CURE
but
NSAIDs used for pain control and corticosteroids can be used
further analgesia may be necessary depending upon the level of pain

541
Q

Which NSAIDs are used to treat OA in:

a) humans (2)
b) animals

A

a) e.g. diclofenac and etoricoxib

b) e.g. robenacoxib

542
Q

describe a further analgesia that can be applied topically onto joints in OA patients (not NSAID or corticosteroid)

A

capsaicin

activates TRPV1 expressed by nociceptors and constant presence of the agonist (after an initial warming sensation) causes depolarising block of nociceptors, as well as nociceptor degeneration (depending on the concentration used), leading to pain relief

543
Q

is capsaicin administered to OA patients

what is a treatment with a similar MOA used in dogs

A

Capsaicin creams can be applied to the affected area 3-4 times a day in humans

intra-articular injection of TRPV1 agonists has been successfully trialled in dogs

544
Q

Name some cytokines that are raised in OA

has this been used in treatment strategies

A

osteoarthritic synovitis is associated with raised levels of IL-1, IL-6 and TNFα

unfortunately related treatments used to treat RA are largely ineffective

545
Q

What is IRAP

A

IL-1 receptor antagonist protein - used to treat OA in horses

546
Q

Describe IRAP collection and use

A

: blood is collected from a horse into a syringe with glass beads coated in a way that induces IRAP production, 24-hours later serum containing IRAP is collected and can be stored at -80°C and injected into affected joints (of the same horse!) when needed.

547
Q

What growth factor is raised in OA synovial fluid

what does this mean for possible OA treatment

A

NGF, which causes pain

tanezumab - an anti-NGF mAb

548
Q

What are the prospects of tanezumab for treating OA

A

looks efficacious compared to other treatments, but concerns remain with regard to increased observation of rapidly progressing OA in patients treated with tanezumab compared to controls.

549
Q

Name some non-pharmacological treatments for OA (4)

A
  • autologous chondrocyte implantation/ transplantation
  • injection of mesenchymal/bone marrow-derived stem cells
  • arthroplasty (joint replacement) in severe cases
  • possibly glucosamines and chondroitin supplements
550
Q

What does autologous chondrocyte implantation/ transplantation involve

A

collecting healthy cartilage tissue, isolating chondrocytes and implanting them into the damaged area

551
Q

What are the results of chondrocyte trans/implantation and injection of bone marrow derived stem cells in treating OA

A

both procedures reduce pain and increase function although the latter is very much in the trial phase

552
Q

Why might glucosamine and chondroitin supplements help treat OA

what does trial data suggest about this treatment

A

they are constituents of joint cartilage

trial data indicates that these supplements are safe to take, but little convincing evidence that they reduce pain, or impact the joint infrastructure compared to placebo

553
Q

What is lupus

A

an autoimmune condition that occurs in two main forms, discoid lupus, which only affects the skin, and systemic lupus erythematosus (SLE), which affects skin and joints and frequently involves internal organs (e.g. heart and kidneys)

554
Q

Why is SLE hard to diagnose

A

wide range of symptoms

555
Q

What is the leading cause of death from SLE

A

cardiovascular problems eg patients often suffer from pericarditis (inflammation of the pericardium) and pleurisy with accelerated atherosclerosis also common in SLE

556
Q

Give 5 common symptoms of SLE

A

pains,
rashes (especially over parts of the body exposed to the sun)
extreme fatigue fever
swelling of the lymph glands

557
Q

What is the ratio of female SLE patients to male ones

which type of woman does it most commonly affect

A

9:1

those of child bearing age

558
Q

Is SLE genetic?

A

although SLE runs in families, no single gene has been identified as causal, but susceptibility genes are known. Similarly, numerous environmental triggers are associated with SLE, e.g. UV radiation and viral infectio

559
Q

Which abnormalities in the immune system are associated with SLE (5)

A

defective clearance of apoptotic cells,
increased response to antigens containing nucleic acids

increased levels of several cytokines including: TNFα, IL-6, interferons and B lymphocyte stimulator (BLyS)

decreased NET degradation

a decreased threshold for activation of autoantibody producing B cells

560
Q

What is a leading theory for the pathophysiology of SLE

why would decreased NET degradation contribute to this

A

defective clearing of apoptotic cells by macrophages results in APCs presenting apoptotic material as autoantigens, which results in the development of anti-nuclear antibodies (ANA)

provide a further source of common SLE autoantigens such as histones

561
Q

What are the main diagnostic tests for SLE (3)

A

physical examination,

blood tests against ANA and anti-dsDNA.

562
Q

What is BLyS

A

B lymphocyte stimulator

important for B cell survival, differentiation and antibody production

563
Q

Is there are cure for SLE

A

NO but treatments available have markedly changed life expectancy from ~5 years after diagnosis in the 1950s to today where >80% can expect to live a normal lifespan

564
Q

Give 6 drugs that can be prescribed for SLE

A
  • NSAIDs such as diclofenac to reduce joint pains and fever
  • hydroxychloroquine reduces inflammation, likely via reducing interferon production
  • corticosteroids to reduce inflammation during a flare-up
  • immunosuppressants like methotrexate and azathioprine
  • rituximab, anti-CD20 on B cells thus causing B cell destruction (limited trial evidence, likely resulting from there being multiple B cell subsets, perhaps CD20+ve are not that important in SLE)
  • belimumab – anti-BLyS, sequestration of BLyS reduces B cell activity
565
Q

Which drugs can cause drug induced lupus

how is this treated

A

e.g. etanercept and infliximab,

discontinuation of use usually reverses the symptoms.

566
Q

Which dogs are most likely to get lupus

describe clinical symptoms

A

genetic predisposition such that German shepherds and Shetland sheepdogs are most affected

similar to in humans, with skin, joints and internal organs being affected and diagnostic tests also involve an anti-nuclear antibody test.

567
Q

Has lupus been seen in cats

A

yes but rarely

568
Q

How do you treat lupus in non-human animals

A

As in humans, corticosteroids can be used, as can immunosuppressive therapies such as azathioprine.

569
Q

Which organ is chiefly responsible for glucose regulation

A

pancreas (islets of Langerhans)

570
Q

What happens in the islets of Langerhans a) when blood glucose falls
b) after a meal

A

a) glucagon is released from α-cells to induce hepatic glycogenolysis (breakdown of stored liver glycogen to glucose) and gluconeogenesis, and lipolysis in fat
b) a rise in blood glucose causes insulin release from β-cells

571
Q

Describe the cascade that occurs when insulin binds to its receptor

A

Insulin binds to the insulin receptor (a RTK) causing tyrosine autophosphorylation and binding via SH2 domains to insulin receptor substrate-1 (IRS-1), IRS-1 is phosphorylated and interacts with SH2-domain containing proteins such as PI3K

572
Q

What is the effect (increase or decrease) of insulin on the following processes:

  • glucose uptake by muscle/fat,
  • glycogenesis (glucose to glycogen) in muscle/liver,
  • lipogenesis (glucose to lipids)
  • hepatic gluconeogenesis and glycogenolysis
A

all increase apart from hepatic gluconeogenesis and glycogenolysis

573
Q

How does insulin release occur

A

Glucose enters β-cells via the glucose transporter GLUT2 and is metabolised to ATP.

β-cells express ATP-sensitive K+-channels (KATP, i.e. ATP binding = inhibition), which are usually open and contribute to the resting membrane potential: K+ moves out of cells down its concentration gradient.

KATP closure increases intracellular [K+], depolarisation activates CaV

Ca2+ influx induces fusion of insulin containing vesicles: insulin is released

574
Q

Describe the structure of K(ATP) channels in pancreatic β-cells

where does the ligand bind and what is the effect

A

octameric

containing a core of 4 inwardly rectifying K+ channel subunits, Kir6.2, which form the pore of the ion channel,
and
surrounding this are 4 sulphonylurea receptor 1 (SUR1) subunits.

ATP binds to the Kir6.2 subunits to induce channel closure and β-cell depolarisation.

575
Q

What % of diabetes cases are T1

When does it usually present

describe its onset

A

10%

presents in childhood (peak incidence at age of 9-14);

onset can be rapid and potentially life threatening if left untreated.

576
Q

When is the usual onset of T2 diabetes

A

typically aged 40+, although this varies depending upon ethnicity, and unlike Type 1 DM the condition generally develops slowly

577
Q

explain why glycosuria, polydipsia, and polyuria are symptoms of diabetes

A

Excess glucose in the blood results in urinary excretion of glucose (glycosuria) producing osmotic diuresis and excessive urination (polyuria), dehydration, thirst and increased drinking (polydipsia)

578
Q

Why do untreated diabetics lose weight

A

Reduced insulin function also increases breakdown of protein and fat, which leads to weight loss and the increased production of ketoacids from lipid can result in diabetic ketoacidosis

579
Q

Give some of the long term complications of diabetes mellitus (7)

A

neuropathy and damage to blood vessels, which can lead to blindness (diabetic retinopathy), nephropathy and an increased risk of cardiac/cerebral infarct, gangrene and limb amputations

580
Q

What about the incidence of diabetes in dogs suggests a genetic component

A

the prevalence ranges from 0.03 – 1.3% and the fact that it occurs in certain breeds (e.g. pugs, Samoyeds and Swedish elkhounds) more than others (e.g. boxers and collies) suggests a genetic component

581
Q

Do dogs get type 1 and 2 DM?

A

. Most dogs have a form of diabetes similar to Type 1 DM in humans, i.e. autoimmune β-cell destruction, but onset tends to be in older dogs as opposed to young humans. Dogs also present with DM that is more similar to human Type 2 DM, but obesity does not appear to be a clear-cut risk factor, although larger epidemiological studies are likely needed

582
Q

Do cats get DM

A

nearly all cases observed are Type 2 DM with males being more likely to be diabetic than females, certain breeds are more susceptible (e.g. Burmese vs. Persian) and as in humans obesity is a clear risk factor

583
Q

What is the only real treatment for T1 DM

A

insulin

584
Q

Describe the structure of insulin

A

51-amino acid protein consisting of 2 polypeptide chains (A and B) linked by disulphide bonds

585
Q

How is insulin administered to a T1 diabetic

A

injection - usually subcutaneously, but intravenous administration can be used in hospital

586
Q

Why is insulin not given to T1 DM patients orally

A

Oral administration would result in too low bioavailability due to destruction in the GI tract

587
Q

How was insulin usually obtained for DM patients

what was the problem with this

A

bovine/porcine insulin were once used

they had potential to induce an immune response

588
Q

What is now used instead of bovine insulin for T1 DM patients

give 2 benefits of it over bovine

A

Recombinant human insulin

greater quality consistency
no immune response

589
Q

What happens to insulin after being injected under the skin

A

forms hexamers, which then dissociate to be absorbed into the blood stream

590
Q

What are the different types of insulin available

A
  • fast-acting, e.g. insulin lispro
  • short-acting, e.g. insulin actrapid
  • intermediate-acting, e.g. Neutral Protamine Hagedorn (NPH) insulin
  • long-acting, e.g. glargine
591
Q

Give a fast acting insulin

describe its structure

what is the time of onset and duration

A

insulin lispro

analogue swapping of a lysine and proline residue towards the end of the C-terminus of the B chain
this reduces dimer and hexamer formation, i.e. larger amounts of the active monomeric insulin are available after injection;

onset after s.c. injection within 5-15 minutes, duration 4-6 hours

592
Q

Give a short acting insulin

what is the time for onset and duration

A

insulin actrapid

human sequence with onset after sc injection within 30-60 minutes, duration 8-10 hours

593
Q

What is NPH insulin

A

intermediate acting insulin

a suspension of protamine polypeptide and insulin, which forms relatively insoluble crystals thus slowing absorption

onset within 2-4 hours, duration up to 12-18

594
Q

Give a long acting insulin

how is this analogue different

what is the benefit of this structure

A

glargine

amino acid changes to the A and B chains change the molecule’s isoelectric point to a more neutral pH.

When injected subcutaneously glargine forms a microprecipitate of stable hexamers and higher aggregates, which retard and prolong absorption

595
Q

What is the time of onset and duration for glargine

A

onset within 2-4 hours, duration 20-24 hours

596
Q

How are the different insulins used to treat DM

A

can be combined into various dose regimens, e.g. once daily injection of a long-acting insulin at night and multiple injections of fast-acting insulin before eating or injection of a fast- and intermediate-acting insulin twice daily (available as pre-mixed preparations in fixed ratios) before breakfast and the evening meal

597
Q

How do insulin pumps work

A

infuse fast-acting insulin continuously into subcutaneous tissues

598
Q

Describe administration of insulin to diabetic cats and dogs

A

In dogs and cats insulin must of course be administered by the owner, this is usually twice daily in cats and once/twice daily in dogs with monitoring of blood glucose as in humans

599
Q

What are the risks of hypoglycemia

A

unpleasant and can be dangerous and may be initially signalled by trembling and sweating followed by confusion and irritability (in humans or animals alike!) and potentially resulting in drowsiness and coma.

600
Q

How is hypoglycaemia from insulin administration countered

A

by eating/drinking something sugary or intravenous glucose or glucagon by injection if unconscious

601
Q

Give a side effect of insulin injection (not hypoglycaemia) (3)

A

lipodystrophy at the site of injection, which can take the form of lipohypertrophy (fat build up due to local anabolic action of insulin)

lipoatrophy (fat loss, likely resulting from an immune response and is far less common with recombinant forms of insulin),

allergic reactions to insulin/additives can also develop.

602
Q

Can a T2 DM patient produce insulin

A

many still have the potential to and treatments are aimed at enhancing insulin release or increasing insulin sensitivity

603
Q

Is insulin given to T2 DM patients

A

it can be if body’s insulin production / sensitivity are inadequate
but is rarely first line treatment

604
Q

Is pharmacological treatment always necessary for T2 DM patients

A

no
. In some cases changes to diet and level of exercise alone can be sufficient to prevent any need for pharmacological treatment, many studies show that exercise increases insulin sensitivity through a number of mechanisms

605
Q

Give an example of a mechanism where exercise increases insulin sensitivity

A

insulin-stimulated PI3K activation is increased in human skeletal muscle, which results in increased GLUT4 expression (GLUT4 transports glucose into skeletal muscle) thus facilitating insulin-mediated glucose uptake.

606
Q

What is the most common first-line Type 2 DM medication

A

metformin

607
Q

What are some of the beneficial effects of metformin (5)

A

increase glucose uptake in skeletal muscle,
decrease hepatic gluconeogenesis,
decrease intestinal carbohydrate absorption,
decrease circulating levels of VLDL and LDL

considering the fact that many Type 2 diabetics are overweight, an added bonus is decreased appetite

608
Q

Which protein do we think is important to the MOA of metformin
(MOA is poorly understood)

A

AMPK

609
Q

How does AMPK affect certain genes (important for DM treatment)

A

. Activation of AMPK decreases expression of genes involved in hepatic gluconeogenesis

610
Q

What re the most common side effects of metformin (3 points)

A

GI disturbances

lactic acidosis -metformin inhibits hepatic lactate uptake and thus people susceptible to lactate build up

Importantly, metformin does not cause hypoglycaemia

611
Q

Who cannot take metformin

A

those experiencing renal dysfunction or alcoholics as it makes patients susceptible to lactate build up

612
Q

Name 2 sulphonylurea compounds

A

glibenclamide

glipizide

613
Q

What is the MOA of SU compounds

A

bind to the SUR1 subunit of KATP channels in pancreatic β-cells causing closure of the channel, triggering depolarisation, Ca2+ influx and insulin release

(thus only useful to someone who has β-cells )

614
Q

Give a brief overview of the PK of SU compounds eg glipizide

A

well absorbed, peak plasma concentrations are reached within a few hours and the duration of action is up to 24 hours.

615
Q

Compare the metabolism of 2 sulphonylurea drugs

A

glipizide is mainly metabolised in the liver to inactive products and excreted in the urine

there is significant conversion of glibenclamide to active products in the liver before urinary excretion, therefore in people with renal inefficiency (i.e. the elderly) the action of glibenclamide is potentiated

616
Q

Who shouldn’t take SU compounds

A

pregnant women (SUs cross the placenta and enter breast milk)

people who do not have beta cells

617
Q

Give some side effects of SU compounds (4)

A

can evoke hypoglycaemia and also tend to increase appetite and thus cause weight gain. Blockade of cardiac KATP could be a potential issue

618
Q

Why are cardiac issues only a POTENTIAL problem with SU compounds ?

A

Blockade of cardiac KATP could be a potential issue, but the SUR2A subunit, not SUR1, is expressed in the heart and SUs show slightly higher affinity and efficacy at SUR1 containing KATP channels than SUR2A containing KATP channels

619
Q

Name a Meglitinide

what is the MOA

A

repaglinide,

also inhibit KATP by binding to SUR1 like SU drugs

620
Q

Compare meglitinide and SU drugs

A

both inhibit KATP by binding to SUR1

meglitinides have faster onset and offset kinetics than SUs meaning that they are often taken just before a meal and are less likely to cause hypoglycaemia

621
Q

Name 2 incretins

where are they secreted from

A

Glucagon-like insulinotropic peptide (GIP) - from K cells

glucagon-like peptide-1 (GLP-1) - from L cells

622
Q

What are the effects of incretins

A

stimulate insulin secretion and inhibit glucagon secretion, as well as reducing gastric emptying thus slowing the rate of food absorption. GLP-1 also acts in brain to reduce appetite and thus reduces body weight

623
Q

How are incretins broken down

A

Both GLP-1 and GIP are broken down by dipeptidyl peptidase-4 (DPP-4)

624
Q

What is exenatide

A

a synthetic version of exendin-4, a peptide found in the saliva of the Gila monster (Heloderma suspectum) and mimics the effect of GLP-1 but is longer acting

625
Q

how is exenatide administered

A

Exenatide is injected subcutaneously and being more stable than GLP-1 can be administered twice daily, but a long release formulation enabling a weekly injection is also available and oral preparations are in phase 3 clinical trials.

626
Q

which drugs inhibit DPP-4

A

a class of drugs called gliptins, e.g. sitagliptin

627
Q

How is sitagliptin administered

A

taken orally in combination with other drugs

628
Q

What are SGLT1 and 2 responsible for

A

SGLT1 allows absorption of glucose in the small bowel and SGLT2 is responsible for reabsorption of glucose filtered in the kidneys at the PCT

629
Q

What are gliflozins

name one

A

inhibit either SGLT2 (e.g. dapagliflozin) or combined SGLT1/2 inhibition

630
Q

What is the effect of gliflozins

A

predominantly increase urinary glucose loss (and thus loss of calories so also allow weight loss), but also reducing GI glucose absorption.

631
Q

When are the effects of gliflozins decreased

what are side effects of these drugs (3)

A

efficacy is reduced where renal function is reduced due to less glucose in PCT to be excreted.

Side effects are increased urinary and genital infections, increased urinary frequency and an as yet unexplained increase in ketone production in the liver

632
Q

Name a thiazolidinedione

what do these drugs do

A

pioglitazone

activate the peroxisome proliferator-activated receptor γ (PPARγ), a transcription factor highly expressed in adipose tissue, but also present in liver and muscle. PPARγ activation increases lipogenesis, glucose/fatty acid uptake and increases transcription of numerous genes including GLUT4.

633
Q

When do you usually see the effects of thiazolidinedione drugs

A

effects are not observed immediately, and pioglitazone can be given as an add-on therapy

634
Q

What are the side effects of thiazolidinedione drugs

A

weight gain

fluid retention due to enhancement of amiloride-sensitive Na+ reabsorption.

635
Q

How can you reduce the amount of glucose uptake in the GI tract

A

α-glucosidase inhibitors, e.g. acarbose, slow the breakdown of starch/disaccharides into glucose in the GI tract and reduce the amount of glucose entering the bloodstream, as such they also aid weight loss

636
Q

α-glucosidase inhibitors, e.g. acarbose are rarely used. when are they given?

what are side effects

A

Given in combination with other treatments,

side effects include flatulence and diarrhoea

637
Q

What is used clinically as a surrogate measure of blood glucose

A

the concentration of glycated haemoglobin (HbA1c), i.e. the
level of covalent bonding of glucose with haemoglobin, the poorer the control of blood glucose and thus the more persistent hyperglycaemia is results in higher HbA1c values

638
Q

Describe the stages for T2 DM treatment according to NICE’s NG28 algorithm

A

(this is used when diet and exercise is insufficient)

Stage 1 involves metformin alone,

Stage 2 involves combining metformin with a second compound

Stage 3 means adding a third compound or moving to insulin based treatment.

A separate algorithm is used for those for whom metformin is contraindicated.

639
Q

How are animals treated for T2 DM?

A

In animals, insulin is always the treatment of choice, regardless of Type 1 or Type 2 DM.