Pharmacology Flashcards

1
Q

What is the general sayings about the Parasympathetic and Sympathetic Nervous Systems?

A

Parasympathetic = Rest and Digest Sympathetic = Fight and Flight

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

Describe the balance of ANS in the liver, explaining it

A

Sympathetic dominant - need glucose in case of a fight or flight situation and sympathetic increases glycogenolysis and gluconeogenesis

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

Describe the balance of ANS in the lungs, explaining why

A

Parasympathetic dominant - causes partial constriction so further constriction and dilation can occur

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

Describe the balance of ANS in the eyes, explaining why

A
  • causes partial constriction so further constriction and dilation can occur
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Describe baroreceptor action at rest

A
  • Baroreceptors are stimulatory to the parasympathetic nerve - They are inhibitory to the sympathetic nerve - At rest, the baroreceptor firing causes activation of the parasympathetic limb and inhibition of the sympathetic limb - PARASYMPATHETIC DOMINATES THE HEART AT REST
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

In terms of complexity, compare the PNS and the SNS

A

SNS = co-ordinated and divergent (1:20 pre vs post) - must be for fight or flight response PNS = discrete and localised (1:1 pre vs post)

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

State the pathway of the PNS

A

long pre-ganglionic fibre -> ACh -> short post-ganglionic fibre -> ACh -> effector organ

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

State the pathways of the SNS

A

short pre-ganglionic fibre -> ACh -> long post-ganglionic fibre -> NA -> effector organ short pre-ganglionic fibre -> ACh -> adrenal medulla -> A (80%) or NA (20%) -> effector organ short pre-ganglionic fibre -> ACh -> long post-ganglionic fibre -> ACh -> effector organ e.g. sweat gland

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

Name the small nervous system located in the gut and give a nickname due to it characteristic

A

Enteric Nervous System - ‘little brain’ - can act on its own accord

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

Describe Nicotinic receptors

A
  • found in all autonomic ganglia - are ion channel linked receptors (ionotropic type 1) therefore when ACh binds it opens and allows Na/Ca influx - very, very fast
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Describe Muscarinic receptors

A
  • found in any tissue innervated by post-ganglionic parasympathetic fibres and also sweat glands (sympathetic so odd) - are Type 2-G-protein coupled much slower than nicotinic
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

At rest, what would happen to the lungs if parasympathetic effects are lost?

A

Bronchodilation

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

At rest, what would happen to sweat glands if parasympathetic effects are lost?

A

Reduced sweat production - however at rest no sweat should be produced

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

At rest, what would happen to the bowel if parasympathetic effects are lost?

A

Constipation

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

At rest, what would happen to urine frequency if parasympathetic effects are lost?

A

Urine frequency would reduce

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

At rest, what would happen to your ability to focus your eyes if parasympathetic effects are lost?

A

You would become long-sighted and unable to focus on nearby objects

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

State the sub-types of muscarinic receptors.

A

M1 - neural (forebrain - learning and memory)

M2 - cardiac (brain - inhibitory autoreceptors)

M3 - exocrine and smooth muscle (hypothalamus - food intake)

M4 - periphery

M5 - striatal dopamine release

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

What adrenoreceptors do sympathetic nerves use to control vasculature

A

alpha 1 - constricts beta 2 - dilates

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

Blockade of which of the following receptor sub-types would induce both an increased heart rate and a reduction in sweat production during exercise?

a. Muscarinic receptors
b. α1 adrenoceptors
c. α2 adrenoceptors
d. β1 adrenoceptors
e. β2 adrenoceptors

A

a. Muscarinic receptors

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

Describe the actions that occur at a Muscarinic receptor

A
  1. Acetyl CoA + Choline -> ACh + CoA 2. An action potential causes Ca2+ influx which triggers the release of ACh 3. ACh diffuses across the synapse and binds to muscarinic receptor on post-synaptic ganglion triggering an action potential 4. Acetylcholin-esterase digests ACh into Choline and Acetate which is absorb back into pre-synaptic cleft
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Describe the actions that occur at a Adrenoreceptor

A
  1. Tryosine is converted to DOPA by Tryosine hydroxylase and the onto Dopamine by DOPA decarboxylase 2. Dopamine is packaged into vesicles and then converted to NA by Dopamine Beta Hydroxylase 3. NA is then released when the action potential comes along and triggers calcium influx 4. NA is NOT broken down in the synapse, it is either removed by: o Uptake 1 - back into the neuronal tissue o Uptake 2 - into extra-neuronal tissue 5. Once it has been taken up (by Uptake 1 or 2) it is broken down by: o Monoamine Oxidase (MAO) - mainly in neuronal tissue o Catechol-O-Methyl Transferase (COMT) - mainly in extraneuronal tissue
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

Blockade of which of the following targets would cause the most significant rise in synaptic noradrenaline concentrations?

a. Tyrosine hydroxylase
b. DOPA decarboxylase
c. Uptake 1 transport protein
d. Monoamine oxidase
e. Cathecol-O-methyl transferase

A

c. Uptake 1 Transport Protein - a and b are involved in synthesis of NA so would reduce NA in synapse - d and e will cause build up of NA in neuronal and extra-neuronal tissue so the gradient will reduce

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

Name the 4 drug target sites

A
  1. Receptors 2. Ion Channels 3. Transports systems 4. Enzymes NOTE: these are all proteins
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

Define Drug

A

A chemical substance that interacts with a biological system to produce a physiological effect

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

For what target site are the terms ‘agonist’ and ‘antagonist’ used

A

Receptors

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

Give 2 examples of drugs that target receptors

A

Acetylcholine - non-selective agonist of ACh receptors Atrophine - muscarinic cholinoceptor antagonist used as anaesthetic premediacation to dry up secretions

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

Give 2 examples of drugs that target ion channels

A

Local Anaesthetic - work by blocking voltage-gated NA+ channels in sensory axons so less action potential are propagated along the axons so perception of pain is reduced - usually ends in -caine (lidocaine) Calcium Channel Blockers - block voltage-sensitive Ca2+ channels - commonly used in cardiology for treatment of arrhythmias - usually end in -dipine (amlodipine)

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

Give 2 examples of drugs that target transport systems

A

Tricyclic antidepressants (TCAs) - prolongs the effect or noradrenaline which is below normal levels during clinical depression Cardiac Glycosides - these are cardiac stimulant drugs (e.g. digoxin) that interact with the Na+/K+ pump - if you give digoxin to a patient with heart failure, it will slow down their Na+/K+ pump and that has a knock on effect that increases the intracellular calcium concentration, therefore increasing the force of contraction

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

State the 3 types of drugs that target enzymes, naming examples for each one

A

Enzymes Inhibitors - e.g. anticholinesterases - neostigmine - increases the concentration of acetylcholine in the synapse by decreasing the rate of breakdown of acetylcholine - used to treat Myasthenia Gravis False Substrates - e.g. methyldopa (antihypertensive) - works by subverting the normal pathway that produces NA by taking the place of DOPA leading to the production of methyl noradrenaline is worse at causing vasoconstriction than NA so you get reduced TPR and hence reduced blood pressure this is a FALSE TRANSMITTER Prodrugs - e.g. chloral hydrate - must go to the liver to be metabolised into trichloroethanol before it is effective - used to treat insomnia

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

Describe an example of unwanted effects caused by drugs targeting enzymes

A

Paracetamol - if someone overdoses they saturate the liver’s microsomal enzymes so another set of enzymes (P450) starts breaking down the paracetamol and produces metabolites that cause damage to the liver and kidneys. Symptoms may not appear until 24-48 hours after the overdose, however at this point damage is irreversible

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

Explain the exceptions to the 4 target rule

A

Non-specific drugs produce responses due to their physicochemical properties - General Anaesthetics - dampen synaptic transmission - Antacids - reduces the acidity of the stomach contents - Osmotic Purgatives - stimulate the voiding of gut contents

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

Define Agonist

A

A molecule that binds to a receptor and stimulates it to generate a response (e.g. nicotine)

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

Define Antagonist

A

Substances that interact and bind to receptors but do not produce a response - they just get in the way of the agonist (e.g. atropine)

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

Define potency

A

How powerful the drug is, is dependent of affinity and efficacy

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

Define Affinity

A

How willingly a drug binds to its receptor

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

Define Efficacy

A

The ability of a drug to generate a response once it has bound to it receptor

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

Define Full Agonist

A

An agonist that generates the maximal response

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

Define Partial Agonist

A

An agonist that generates a less than maximal response

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

Define Selectivity

A

To have a preference for interacting with a particular receptor type (receptors are rarely specific - normally interact with a few other receptors)

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

Which of the following statements is most accurate?

a. a partial agonist will always have a higher efficacy than a full agonist
b. agonists have higher affinities than antagonists
c. full agonists that are selective for a given receptor will have the same efficacy
d. antagonists possess better efficacy than their respective agonists
e. competitive antagonists will preferentially occupy the relevant receptor in the presence of agonists

A

c. full agonists that are selective for a given receptor will have the same efficacy

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

A drug acting as an inhibitor at a particular drug target site prevents the removal of neurotransmitter from the synapse. Which type of drug target is this drug acting on? a. receptor b. voltage-sensitive ion channel c. receptor-linked ion channel d. transport protein e. non-proteinaceous target

A

d. transport protein

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

Name the 4 types of drug antagonisms.

A
  1. Receptor Blockade 2. Physiological Antagonism 3. Chemical Antagonism 4. Pharmacokinetic Antagonism
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

Explain how receptor blockade antagonists work

A

By simply binding to the receptor and therefore blocking the agonist from binding

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

What does Use Dependency refer to

A

Ion channel blockers - it means the more the tissue on which the drug is acting is being used (the more active they are) the more effective this type of blocker will be

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

Give an example which explain use dependency

A
  • normal neurones fire at a relatively low rate so if you put local anaesthetic on them, there’ll be relatively low blockage. - nociceptor neurones fire rapidly and because the action potentials are being generated rapidly so the ion channels are open more often - local anaesthetics work by binding to the inside of the ion channels after they open - if the channels are opening more often then there is more chance that they’ll be blocked by LAs - this gives local anaesthetics a selective action on nociceptor neurones
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

What does Physiological Antagonism refer to

A

Two drugs acting at different receptors causing opposite effects in the same tissue

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

Give an example of a physiological antagonism

A
  • NA causes vasoconstriction of vasculature by binding to adrenoceptors - co-administering histamine acts on different receptor to cause vasodilation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

Explain what is meant by Chemical Antagonism

A

Interactions between drugs in solution

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

Give examples and one relevant point about chemical antagonists

A
  • dimercaprol is a chelating agent which forms heavy metal complexes -> more rapidly excreted by the kidneys so, is useful for things like lead poisoning. Is very uncommonly used in practice
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

Explain what is meant by Pharmacokinetic Antagonism

A
  • when one drug reduces the concentration of the other drug at the site of its action (absorption, excretion etc)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

State a complication that must be looked for when using pharmacokinetic antagonisms

A
  • repeatedly administer barbiturates increases the production of microsomal enzymes so, if we administer another drug (e.g. warfarin) that is metabolised by the enzymes then it is going to be metabolised quicker and its effect will be reduced
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

Name the 5 main causes of drug tolerance.

A
  1. Pharmacokinetic Factors
  2. Loss of Receptors
  3. Change in Receptors
  4. Exhaustion of Mediator Stores
  5. Physiological Adaptation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

Explain how pharmacokinetic factors can lead to drug tolerance, include to examples where this occurs

A

Metabolism of the drug increases when repeatable given - barbiturates and alcohol

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

Explain how the loss of receptors occurs and therefore how it leads to drug tolerance, giving a receptor that is susceptible to this

A

Cells repeatable stimulated by an agonist will endocytose some receptors leaving fewer on the cell surface - this is called down regulation - beta adrenoceptors

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

Give a time when cells up regulate receptors

A

Denervation supersensitivity

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

State how change in receptors can cause increased drug tolerance

A

A conformational change in the receptor occurs so that the drug can no longer bind

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

Using an example explain how the exhaustion of mediator stores leads to drug tolerance

A

Amphetamines - amphetamine is a CNS stimulant that causes the blood-brain barrier from the blood and acts on noradrenergic neurones in the brain where it binds to uptake 1 protein and is taken into the central NA synthesis system causing a big increase in NA production - if you take a second dose the response is far less severe because NA stores have been exhausted

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

How does physiological adaptation lead to drug tolerance

A
  • sort of like a homeostatic response -> the body is attempting the maintain a stable environment
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

What are the initial side effects of physiological adaptation causing drug tolerance

A
  • tiredness and nausea
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

Describe Ionotropic Receptors (Type 1)

A
  • are ion channels so are VERY, VERY FAST - 4 or 5 subunits including a transmembrane alpha helices section e.g. nicotinic acetylcholine receptor and GABA
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

Describe G-protein Coupled (Metabotropic) Receptors (Type 2)

A
  • must link to G protein so a MUCH SLOWER than Type 1 - 1 subunit and 7 transmembrane domains (alpha helices) e.g. beta-1 adrenoceptors in the heart
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

Describe (Tryosine) Kinase-linked Receptors (Type 3)

A
  • results in the phosphorylation of intracellular proteins so SLOWER than both Type 1 and 2 (usually minutes) - single protein with 1 transmembrane domain and 1 intracellular domain e.g. insulin receptor and growth factor receptors
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

Describe Intracellular Steroid Type Receptors (Type 4)

A
  • activated by steroid and thyroid hormones so is the SLOWEST (can take hours) of all 4 receptors - drug must pass through the cell membrane (these receptors regulate DNA transcription via zinc fingers
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

A 4 year old girl is bitten by a Tiger Snake whilst holidaying in Australia. An anti-venom (harvested antibodies) is administered. Which form of antagonism is utilised by anti-venom? a. competitive receptor blockade b. physiological antagonism c. chemical antagonism d. pharmacokinetic antagonism e. irreversible receptor blockade

A

c. chemical antagonism

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

Tolerance to the euphoric effects of drugs of abuse (heroin or cocaine etc) can occur after repeated use. Which form of tolerance would not involve any change in the cells that mediate the euphoric effect? a. receptor desensitisation b. receptor down-regulation c. exhaustion of mediator stores d. receptor up-regulation e. increased metabolic degradation

A

e. increased metabolic degradation

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

Describe the journey of a drug through the body.

A
  1. administration
  2. Absorption
  3. Distribution
  4. Metabolism
  5. Excretion
  6. voided

REMEMBER: ADME

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

Name 7 types of drug administration

A
  1. intravenous 2. intraperitoneal 3. intramuscular 4. dermal 5. subcutaneous 6. inhalation 7. ingestion
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
68
Q

State 2 approaches of drug administration, in terms of how much of the body is affected, that are used

A
  • systemic (entire organism exposed to the drug) - local (restricted to one area)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

What is the difference between enteral and parenteral administration routes?

A
  • enteral = via the GI tract (usually orally)
  • parenteral = avoids the GI tract (IV)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
70
Q

What are the two ways in which drug molecules move around the body?

A
  • bulk flow transfer -> in the bloodstream in bulk to tissue - diffusion transfer -> molecule by molecule over short distances
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

Which drug transport mechanism occurs with IV and oral administration

A
  • IV = bulk flow - oral = diffusion
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
72
Q

Name some methods that drug use to cross barriers

A
  • lipid soluble (non-polar solvents) - diffusing through aqueous pores in the lipid (if they are polar) - carrier molecules - pinocytosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
73
Q

What is the pH of most drugs

A

5-9 (weak acids and bases)

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

Explain the clinical relevance of drugs being either weak acids or weak bases

A
  • drugs exist in ionised and non-ionised form, forming a dynamic equilibrium of which the ratio depends on the pH of the environment and the pKa of the molecules - this alters the lipid solubility of the drug
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
75
Q

Use aspirin as an example to explain the pH partition hypothesis

A
  • aspirin a weak acid with a pKa of around 3.4 - when aspirin enters the stomach, which has a pH of less than the pKa of aspirin, it is more readily non-ionised -> aspirin exists mainly in the non-ionised form in the stomach meaning that it can readily diffuse across the lipid bilayer (as it is non-polar) - eventually the aspirin will make it into the small intestine, as the pH of the small intestine is greater than the pKa of aspirin, the aspirin becomes ionised -> now its more difficult to get through the membrane
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q

What is the clinical importance of the pH partition hypothesis when prescribing aspirin

A
  • for quick, short relief (e.g. headaches) give it as a soluble - for slow, long relief (e.g. arthritis) coat it in something that isn’t digested in the stomach
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
77
Q

What is ion trapping

A
  • once a drug goes through the liver and into the systemic circulation, the aspirin is in an aqueous environment so you find a proportion of aspirin in an ionised form - it is effectively TRAPPED
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
78
Q

State some factors which influence drug distribution

A
  • regional blood flow - extracellular binding (plasma protein binding) - capillary permeability - localisation in tissues
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
79
Q

Explain how regional blood flow affects drug distribution

A
  • tissues which are well perfused are likely exposed to a higher drug conc. - blood flow can change with activity such as skeletal muscle tissue
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
80
Q

State capillary wall architecture and how it effects distribution?

A
  • continuous -> normal vessels, not particularly permeable - fenestrated -> more permeable - discontinuous
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
81
Q

Explain how tissue properties can alter drug distribution

A
  • fat isn’t usually highly perfused - a very lipophilic environment -> drugs which are lipophilic tend to localise in fatty tissue (brain, testes etc)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
82
Q

Name the two major routes of drug excretion

A
  • liver - kidneys
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

Name other none major routes in which drugs can be excreted

A
  • exhalation/lungs - sweating/skin - GI secretions - saliva - milk - genital secretions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
84
Q

Explain drug excretion by the kidneys

A
  • do most of xenobiotic excretion - the majority of the drug that is excreted will get into the urine via active secretion rather than ultrafiltration -> you can’t filter large, protein-bound drug complexes - glomerulus - drug-protein complexes are NOT filtered - proximal Tubule - active secretion of acids and bases - proximal and Distal Tubules - lipid soluble drugs are reabsorbed
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
85
Q

Explain drug excretion by the liver

A
  • biliary excretion allows the concentration of large molecular weight molecules (most antibiotics) that are very lipophilic - active transport systems also secrete drugs into bile - active transport systems are geared for the transport of bile acids and glucuronides into the bile - drugs hitch a ride on this because they are non-polar and have a large molecular weight
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
86
Q

How can enterohepatic cycling cause problems

A
  • a drug or metabolite gets excreted into the gut but then it can get reabsorbed and returned to the liver via the enterohepatic circulation -> leading to drug persistence
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
87
Q

Why might treatment with IV sodium bicarbonate increase aspirin excretion?

A
  • IV sodium bicarbonate increases urine pH which ionises the aspirin making it less lipid soluble - less aspirin is reabsorbed in the proximal and distal tubules so there is an increase in the rate of excretion of aspirin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
88
Q

Define bioavailability

A
  • the proportion of the administered drug that is available within the body to exert its pharmacological effect
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
89
Q

Define apparent volume of distribution

A
  • the volume in which a drug appears to be distributed - an indicator of the pattern of distribution
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
90
Q

Define biological half-life

A
  • the time taken for the concentration of the drug (in the blood/plasma) to fall to half its original value
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
91
Q

Define clearance

A
  • the volume of plasma cleared of a drug per unit time
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
92
Q
A

a. ionised drug

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

Define first-order kinetics

A
  • the rate of elimination of a drug where the amount of drug decreases at a rate that is proportional to the concentration of drug remaining in the body
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
94
Q

Define zero-order kinetics

A
  • the rate of elimination of a drug where the amount is a constant per unit of time
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
95
Q

In terms of kinetics are most drugs first-or zeroth order, state an example of the minority?

A
  • first-order
  • alcohol/ethanol is zero-order because alcohol dehydrogenase becomes saturated
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
96
Q

Why is drug metabolsim required?

A
  • drugs tend to be lipophilic -> must be metabolised to become more water soluble so hey can be excreted easily
  • metabolism tends to eliminate or reduce the pharmacological and toxicological activity -> converts the drug into something more polar and, hence, soluble -> more easily be excreted by the kidneys
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
97
Q

Define hepatic first pass metabolism

A
  • metabolic conversion of the drug into something that is different before the drug enters the general circulation (the effect that occurs the very first time the drug passes through the liver)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
98
Q

Name some sites for drug metabolism, noting the major site

A
  • LIVER = MAJOR
  • gut
  • kidneys
  • brain
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
99
Q

What is meant by low bioavailability

A
  • when you administer the drug and it is absorbed and taken to the liver, you want it to be released in to the systemic circulation in a pharmacologically active form
  • f the metabolism is extensive you may only release a small amount of the active drug into the systemic circulation, this is low bioavailability
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
100
Q

Explain when low bioavailabitlity can be a problem and how it can be negotiated

A
  • it releases only a small amount of the active form of the drug meaning that if the problem is in the systemic circulation the conc. may not be higher enough to have an effect
  • this can be overcome by giving it intravenously
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
101
Q

Name some pre-hepatic sites for first pass metabolism

A
  • intestines
  • stomach
  • oesophagus and buccal cavity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
102
Q

What type of reactions occur in Phase I Reactions and what are the roles

A
  • oxidation -> create new functional groups
  • reduction -> create new functional groups
  • hydrolysis -> unmasks functional groups
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
103
Q

In what stage are prodrugs converted into their active form

A
  • Phase I Reactions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
104
Q

What happens to the polarity of the drug after phase I reactions

A
  • there is very little change in polarity, if it was lipophilic when it started it will still be pretty lipophilic etc
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
105
Q

Where do phase I reactions occur

A
  • primarily in the liver
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
106
Q

Exactly what metabolises most of the xenobiotics

A
  • the cytochrome P450 system in the liver
  • in humans, the system is made up of 57 enzymes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
107
Q

Specifically, where in cells of the liver is the cytochrome P450 enzyme system located?

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

State a problem that affects the CYP450 system which occurs with some drugs, explaining why it is an issue. Also name a drug which this occurs with.

A
  • some drugs can inhibit or induce the CYP450 system -> changing the ability of the system to handle certain drugs
  • the oral contraceptive pill is a common example -> can loss contraceptive properties with certain other drug combinations
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
109
Q

Give the equation for oxidation of a drug mediated by cytochrome P450

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

Describe the cyclic manner of work of cytochrom P450 oxidation

A
  1. the drug binds to the iron in the catalytic site and an electron is donated by NADPH
  2. this electron is picked up by the P450 complex and reductes Fe3+to Fe2+
  3. molecular oxygen binds to the catalytic site so P450 has Fe2+ and O2 bound
  4. Fe2+ loses an electron to become Fe3+ with oxygen picking up the extra electron and becoming unstable
  5. a second electron is donated by NADPH reducing Fe3+ to become Fe2+
  6. Fe2+ donates an electron to oxygen (becoming Fe3+) and making the oxygen very unstable, however the drug still hasn’t changed
  7. the drug is converted into its hydroxylated derivative and loses the reactive oxygen as water by picking up two protons
  8. drug is released and the P450 returns to the cycle with iron in its oxidised state (Fe3+) ready to undergo the next cycle
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
111
Q

Describe the structure of cytochrome P450

A
  • at the centre of P450’s catalytic site there is catalytic iron in its oxidised state (Fe3+)
  • all P450 enzymes have a porphyrin ring and iron (Fe3+) at its active site
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
112
Q

What kind of reaction do oxidations usually start with?

A
  • hydroxylation catalysed by CYP450
  • 9f ever unsure on an enzyme starting an oxidation guess CYP450
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
113
Q

What is pentabarbitone?

A
  • a narcotic, will put you to sleep
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
114
Q

How many isoforms are found after metabolsim by CYP450

A
  • depends on the drug, can be hundreds
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
115
Q

Give an example of a prodrug which is catalysed into active form by CYP450

A
  • acetanlide becomes paracetamol by oxidation of C4 in the benzene ring
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
116
Q

Why is acetanlide (paracetamol prdodrug) not for sale?

A
  • anlide is toxic to blood cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
117
Q

What happens if you oxidate a carbon in a carbon chain?

A
  • forms an alcohol group
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
118
Q

What happens if you oxidate a carbon in a benzene ring?

A
  • you form an alcohol group
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
119
Q

What happens if you oxidate a carbon attached to a nitrogen?

A
  • N-demethylation -> the carbon group that was attached to the nitrogen disassociates leaving a signle hydrogen behind if it was a tertiary amine, the remnants of the carbon group bind with an oxygen to forming a aldehyde, commonly formaldehyde (HCHO)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
120
Q

What happens if you oxidate a carbon attached to a oxygen?

A
  • O-demethylation -> an alcohol group forms in place of the carbon group when its a teritary structure and the remnants of the carbon group form an aldehyde or a ketone, formaldehyde (HCHO)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
121
Q

Why does codeine have such contrasting effects on people?

A
  • it is metabolised into morphine by the CYP450 complex causing all the relative effects, specifically as a painkiller
  • however, some people are a lot better at doing this metabolism compared to others hence to disparity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
122
Q

Name two alternative oxidation of nitrogen than N-demethylation, stating the enzymes that carries it out in humans.

A
  • N-oxidation -> requires the formation of a dative bond, flavin containing monoxygenase
  • alcohol oxidation by alcohol dehydrogenase
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
123
Q

What is the clinical relevance of flavin containing monoxygenase?

A
  • flavin containing monoxygenase defiiciency (aka fish odour syndrome)
  • humans generate trimethylamine in their GI tract (product of protein metabolsim -> rimethylamine smells terrible
  • in the liver, flavin containing monooxygenase converts trimethylamine into trimethylamine N-oxide which is odourless and polar so it can be readily excreted in the urine
  • a small subset of the population has defective flavin containing monooxygenase so they can’t metabolise trimethylamine and must sweat and breathe it out
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
124
Q

What are the consequences of fish odour syndrome?

A
  • strange behavouirs
  • smoke or wear lots of perfume to mask the smell
  • despression -> very high suicide rate
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
125
Q

Where are most reductases found?

A
  • in the gut as they are bacterial enzymes colonising the gut
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
126
Q

In phase I reactions are oxidations or reductions more common?

A
  • oxidations are far more common, hydrolysis is even less less common than reduction
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
127
Q

In phase I reactions, when does hydrolysis occur?

A
  • when there is an ester or an amide
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
128
Q

What enzymes carry out hydrolysis

A
  • esterases = hydrolysis of an ester
  • amidase = hydrolysis of an amide
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
129
Q

True or false, metabolism of prodrugs activates their pharmacological activity.

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

True or false, xenobiotic metabolism only occurs in the liver

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

True or false, hydrolysis is a Phase 1 reaction

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

Does cytochrome P450 uses NADH+ as cofactor ?

A
  • can do but more readily uses NADPH
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
133
Q

True or false, cytochrome P450 contains Cu2+ at its active site

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

Name the types of reactions that occur in phase II metabolsim

A
  • glucuronidation (most common)
  • methylation
  • sulphation
  • acetylation
  • aminoacid conjugation
  • gluathione
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
135
Q

State the rule about enzymes carrying out phase II reaction.

A
  • the name of the reaction and add transferase to the end of it to find the enzyme name
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
136
Q

What are the results of phase II drug metabolsim

A
  • the conjugate that is formed is almost always inactive (morphine is an exception)
  • less lipid soluble
  • more polar
  • easier to excrete
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
137
Q

What is the conjugating agent for glucuronidation?

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

What occurs in a glucuronidation reaction?

A
  • xenobiotic is catalysed by glucuronyl transferase to form the glucuronide derivative of the xenobiotic
  • this derivate is polar so can be removed, usuallt though bile due to large molecular weight
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
139
Q

Give an example of a drug that goes through glucuronidation.

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

What kind of drugs go through acetulation?

A
  • drugs with a amino group
  • acetylation occurs to an electron rich atom
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
141
Q

Explain acetylation reaction as part of phase II drug metabolsim

A
  • produce acetylated derivative of the drug and CoA which then goes into intermediary metabolism
  • use acetyl CoA as the high energy intermediate
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
142
Q

What happens in methylation as part of phase II drug metabolsim?

A
  • a methyl group is donated to an atom rich atom (N, S or O etc) via the high energy intermediate S-adenosyl methionine using the enzyme methyl transferase
  • methylation DECREASES polarity
  • in ZH, Z can be N, O or S
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
143
Q

As methylation makes molecules less polar and therefore hard to excrete why does it occur?

A
  • for endogenous molecules such as noradrenaline -> allows NA to convert into adrenaline
  • only occurs to drugs such as amphetamines which have a similar structure to NA
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
144
Q

Which phase II drug metabolism reaction occurs with paracetamol?

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

What happens in the phase II drug metabolsim reaction sulphation?

A
  • xenobiotic is taken with PAPS which is the sulfate donor and gets sulfated to produce the sulfuric acid derivative of the molecule - the derivative is very polar and water soluble

the reaction is catalysed by sulfotransferases

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

What is glutathione?

A
  • a tripeptide molecule kept in large quantities in the liver and kidneys that protects the bodies from toxic metabolites such as electrophiles
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
147
Q

Which part of a molecule of glutathione is important?

A
  • cysteine because it has the thiol part
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
148
Q

True or false, metabolism of lipophilic chemicals facilitates their excretion?

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

True or false, metabolism of drugs prior to entering the systemic circulation is known as “first pass metabolism”?

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

True or false, phase 2 metabolism includes reduction and acetylation.

A
  • false, reduction is phase I
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
151
Q

True or false, phase II metabolism generally increases the polarity of drugs.

A
  • true, to aid excretion
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
152
Q

True or false, conjugation of drugs with glutathione is the most common Phase II route of metabolism.

A
  • false, glucuronidation is
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
153
Q

What is the importance of drug metabolsim?

A
  • biological half-life of a drug is reduced
  • duration of exposure is reduced
  • accumulation of the drug in the body is avoided
  • potency/duration of the biological activity of the chemical can be altered
  • pharmacology/toxicology of the drug is governed by its metabolism
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
154
Q

What are cholinomimetic drugs?

A
  • drugs that mimic the action of acteylcholine in the body
  • therefore they are parasympathetic drugs
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
155
Q

What are the cholinergic target sites/systems?

A
  • eyes
  • salivary glands
  • sweat glands
  • lungs
  • heart
  • gut
  • bladder
  • vasculature
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
156
Q

What are the 3 main muscarinic effects on the eye?

A
  • contraction of the ciliary muscle - accommodates for near vision
  • contraction of sphincter pupillae (circular muscle of the iris) - this constricts the pupil (miosis) and increases drainage of intraocular fluid
  • lacrimation (tears)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
157
Q

What is glaucoma?

A
  • an increase in intraocular pressure -> can cause damage to the optic nerves and retina and can ultimately lead to blindness
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
158
Q

Explain the mechanism of angle-closure glaucoma

A
  • aqueous humour is generated by the capillaries of the ciliary body, it flows into the anterior chamber of the eye
  • its role is to supply oxygen and nutrients to the lens and cornea because they don’t have a blood supply
  • the aqueous humour diffuses forwards across the lens, then across the cornea and it drains through the canals of Schlemm back into the venous system
  • in angle-closure glaucoma, the angle between the cornea and the iris becomes narrowed => reduces the drainage of intraocular fluid via the canals of Schlemm
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
159
Q

Describe the treatment for angle-closure glaucoma

A
  • muscarinic agonist -> causes contraction of the iris opening up the angle and increasing the drainage of intraocular fluid through the canals of Schlemm (has no effect on fluid production)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
160
Q

Explain the muscarinic effects on the heart.

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

What are the muscarinic effects on the vasculature?

A
  • most blood vessels do NOT have parasympathetic innervation
  • acetylcholine acts on vascular endothelial cells to stimulate NO release via M3 receptors -> NO induces vascular smooth muscle relaxation -> decrease in TPR
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
162
Q

What are the overall muscarinic effects on the CVS?

A
  • decrease heart rate
  • decreased cardiac output (due to decreased atrial contraction)
  • vasodilation (via stimulation of NO release)
  • all of these can lead to a sharp DROP BLOOD PRESSURE
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
163
Q

State the muscarinic effects on non-vascular smooth muscle

A
  • non-vascular smooth muscle that has parasympathetic innervation responds in the opposite way to vascular smooth muscle - it contracts

· Lungs - bronchoconstriction

· Gut - increased peristalsis/motility

· Bladder - increased bladder emptying

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

What are the muscarinic effects on exocrine glands?

A

· Salivation

· Increased bronchial secretions

· Increased GI secretions (including gastric HCl production)

· Increased sweating (SNS-mediated)

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

Summarise the muscarinic effect on the body

A

· Decreased heart rate

· Decreased blood pressure

· Increased sweating

· Difficulty breathing

· Bladder emptying

· GI pain

· Increased salivation and tears

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

Name the two types of cholinomimetic drugs

A
  1. Directly Acting
  2. Indirectly Acting
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
167
Q

State what directly acting cholinomimetics are

A
  • muscarinic receptor agonists
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
168
Q

Name the two types of muscarninic receptor agonists, with an example for each

A

o Choline esters (bethanechol)

o Alkaloids (pilocarpine)

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

Describe the structures of bethanechol and pilocarpine

A
  • both very similar to acetylcholine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
170
Q

State some properties of pilocarpine

A
  • non selective -> stimulates all muscarinic receptors
  • good lipid solubility
  • half-life of 3-4 hours
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
171
Q

Give a disease which is treated with pilocarpine

A
  • glaucomao -> constricts sphincter pupillae and opens up the canals of Schlemm to increase the drainage of intraocular fluid
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
172
Q

State the side effects of pilocarpine

A
  • general effects of parasympathetic discharge:

o Blurred vision

o Sweating

o GI disturbance and pain

o Nausea

o Bradycardia

o Hypotension

o Respiratory distress

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

How are the side effects of pilocarpine avoided?

A
  • can be given successfully as an eye drop -> therefore a very low conc. reaches the systemic circulation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
174
Q

State some properties of bethanechol

A

· M3 receptor selective agonist, still has an effect on all muscarinic receptors

· It is resistant to degradation by acetylcholinesterase

· It is orally active and has limited access to the brain

· Half-life = 3-4 hours

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

Why is bethanechol administrated?

A
  • mainly used to assist bladder emptying and enhance gastric motility
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
176
Q

What are the symptoms of bethanechol?

A

o Sweating

o GI disturbance and pain

o Respiratory distress

o Impaired vision

o Nausea

o Bradycardia

o Hypotension

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

How do indirectly acting cholinomimetic drugs work?

A
  • inhibit acetylcholinesterase -> decrease acetylcholine breakdown -> increase the amount of acetylcholine in the synapse -> increase the effect of normal parasympathetic nerve stimulation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
178
Q

State the categories of indirecty acting cholinomimetic drugs, with examples

A
  • Reversible Anticholinesterases (PHYSOSTIGMINE, Neostigmine, Donepezil)
  • Irreversible Anticholinesterases (ECOTHIOPATE, Dyflos, Sarin)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
179
Q

Name the two types of cholinesterass

A

o Acetylcholinesterase (true or specific cholinesterase)

o Butyrylcholinesterase (pseudocholinesterase)

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

Describe acetylcholinesterase

A

o Found in ALL cholinergic synapses (peripheral and central)

o Very rapid action (> 10 000 reactions per second)

o It is highly selective for acetylcholine

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

Describe butyrylcholinesterase

A

o Found in plasma and in most tissues but NOT in cholinergic synapses

o Broad substrate specificity - hydrolyses other esters e.g. suxamethonium

o It is the principal reason for low plasma acetylcholine

o Shows genetic variation so different people show different reactions to suxamethonium

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

What are the effects of cholinesterases at a low dose

A

o Enhanced muscarinic activity

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

What are the effects of cholinesterases at a moderate dose?

A

o Further enhancement of muscarinic activity

o Increased transmission at ALL autonomic ganglia (nAChRs)

· This is because the anticholinesterase increases the acetylcholine concentration at ALL cholinergic synapses, muscarinic and nicotinic

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

What are the effects of cholinesterases at a high dose?

A

o Becomes toxic

o Depolarising block at autonomic ganglia and neuromuscular junction

o The nicotinic receptors get overstimulated so they shut down

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

Describe the general action of Reversible Anticholinesterase Drugs

A

· They compete with acetylcholine for the active site on acetylcholinesterase

· These drugs donate a carbamyl group to the enzyme, blocking the active site and preventing acetylcholine from binding

· Carbamyl groups are removed by slow hydrolysis (this takes minutes rather than miliseconds)

· This increases the duration of acetylcholine activity in the synapse

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

Where does physostigmine act?

A
  • postganglionic parasympathetic synapses
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
187
Q

What is the half-life of physostigmine

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

What is physostigmine used to treat?

A

o Glaucoma - increases drainage of intraocular fluid

o Atropine poisoning (common in children who eat atropa belladonna berries)

· Atropine is a competitive muscarinic antagonist

· Physostigmine increases the concentration of acetylcholine at the synapse so that the acetylcholine can outcompete the atropine until the atropine is cleared

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

Describe the general mechanism of irreversible anticholinemimetic drugs

A

· Irreversible anticholinesterase drugs are ORGANOPHOSPHATE compounds

· Rapidly react with the enzyme active site, leaving a large blocking group

· The blocking group is stable and resistant to hydrolysis so recovery requires the production of new enzymes (this takes days/weeks)

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

What are the break down of uses of Irreversible Anticholinesterase Drugs?

A

· Ecothiopate is the only one in clinical use but the others are used as insecticides and nerve gas

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

What are the side effects of physostigmine

A

o Side-effects are that of parasympathetic discharge:

· Sweating

· Blurred vision

· GI disturbance and pain

· Bradycardia

· Hypotension

· Respiratory difficulty

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

What are the side effects of ecothiopate?

A

o Side-effects are that of parasympathetic discharge:

· Sweating

· Blurred vision

· GI disturbance and pain

· Bradycardia

· Hypotension

· Respiratory difficulty

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

What is the mechanism of ecothiopate?

A

o Potent inhibitor of acetylcholinesterase

o Slow reactivation of the enzyme by hydrolysis takes several days

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

Name a disease which ecotiopate is used to treat?

A

o eye drops to treatment glaucoma -> increases the drainage of intraocular fluid and has a prolonged duration of action

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

What are the effects of Anticholinesterase Drugs on the CNS?

A

· Non-polar anticholinesterases (physostigmine, nerve agents) can cross the BBB

· LOW doses -> CNS excitation with the possibility of convulsions

· HIGH doses -> Unconsciousness, respiratory depression and death

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

What happens as consequences of organophosphate poisoning?

A
  • severely toxic
  • increase in muscarinic activity –> CNS excitation –> depolarising NM block
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
197
Q

What is the treatment of organophosphate poisoning

A

o INTRAVENOUS ATROPINE

o Patient is put on a respirator because of the respiratory depression

o If found in the first few hours the patient can be given Pralidoxime (IV) - this unblocks the enzymes

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

B

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

B

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

Which of the following drugs has efficacy for the muscarinic acetylcholine receptor?

a. Acetylcholine
b. Atropine
c. Acetyl-cholinesterase
d. Adrenaline
e. Acetate

A

a. Acetylcholine

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

What is the other name for nicotinic receptor antagonists?

A
  • ganglion blocking drugs
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
202
Q

State the two ways in which nicotinic receptor antagonists function

A
  • blocking the receptor
  • blocking the ion channel itself
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
203
Q

Do nicotinic receptor antagonists affect the parasympathetic or the sympathetic nervous systems?

A
  • effect the systme which is more dominant within the tissue at that time due to use dependency
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
204
Q

Which of the following effects would be observed, at rest, after treatment with a ganglion blocking drug?

a. Increased heart rate
b. Pupil constriction
c. Bronchodilation
d. Detrusor contraction
e. Increased gut motility

A

a and b, both sympathetic effects as the parasymapthetic is blocked

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

Why do nicotinic receptor antagonists cause hypotension?

A
  • both vasocnstriction and renin production are inhibited leading to less TPR and water reabsorption
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
206
Q

Apart from cauing hypotension, name some other side effects of nicotinic receptor antagonists

A
  • smooth muscle: pupil dilation, decreased GI tone, bladder dysfunction and bronchodilation
  • exocrine secretions: decreased secretions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
207
Q

What was the clinical use of hexamethonium?

A
  • 1st anti-hypertensive
  • was to general so had horrible side effects so has be superseded by more selective agents
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
208
Q

When is trimetaphen used?

A
  • only in surgery when controlled hypotension is required -> because it is very potent but the effcts are very short lived so patient will find after the effects of the aneasthetic have worn off
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
209
Q

What other agents are often receptor blockade antagonists?

A
  • toxins and venoms -> often bind irreversible unlike the drugs and prevent channel opening which causes total loss of autonomic function
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
210
Q

What is the effect of atropine on the CNS?

A

Normal dose - little effect

Toxic dose - mild restlessness through to severe agitation

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

What is the effect of atropine on the CNS?

A

Normal dose - sedation, amnesia

Toxic dose - CNS depression or paradoxical CNS excitation (associated with pain)

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

What is the action of tropicamide?

A
  • is a muscarinic receptor antagonist which acts on receptors within the iris of the eye to cause pupil dilation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
213
Q

Name a use for tropicamide

A
  • used in eye exams in order to examine the retina
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
214
Q

Explain why muscarinic receptor antagonists are used a anaesthetic pre-medications

A
  • causes airways dilation
  • dry the throat by decreasing saliva produciton (reduces the risk of aspiration)
  • slightly increases the heart rate
  • if correctly chosen (hyoscine) has a sedative effect
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
215
Q

What is motion sickness?

A
  • sensory information from the ear (labyrinth) and the eye are both coming in and meeting at the memory centre, the hippocampus
  • if there is a mismatch in the two bit of information the hippocampus triggers the vomitting centre (Area postrema - CVO) and nausea begins
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
216
Q

How do muscarinic receptor antagonists, such as hyoscine, work a treatment for motion sickness?

A
  • reduce the flow of information from the labyrinth to the brain -> although there is still mismatch the response doesn’t occur so neither does nausea
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
217
Q

What is Parkinson’s Disease?

A
  • dopaminergic neurones from the substantia nigra releases dopamine into the striatum which binds to D1 receptors, activating these allows the fine control of movement
  • M4 receptor has an inhibitory effect on this process
  • in Parkinson’s you lose dopaminergic neurones so there is less stimualtion and therefore a loss of fine control
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
218
Q

How can muscarinic receptors antagonists be used in treating Parkinson’s Disease?

A
  • as 60-70% of dopaminergic neurones have been lost the inhibitory effect of M4 receptors are no longer wanted so the antagonists can be used to take out the M4 receptors and the inhibitory effect
  • by no means a first line treatment
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
219
Q

How does ipratropium bromide work to treat asthma and COPD?

A
  • is a muscarinic recepotr which means it blocks the parasympathetic induced bronchoconstriction
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
220
Q

Why is ipratropim bromide used to treat asthma and COPD instead of atropine?

A
  • ipratropim bromide has a large quaternary amine structure which means when given as an aerosol it doesnt cross the lipid membrane of the lungs to exit because of its positive charge -> remains localised
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
221
Q

How can muscarinic receptor antagonists be used to treat irritable bowel syndrome

A
  • knocking out parasympathetic effects within the gut reduces smooth muscle contraction, gut motility and secretions to relieve some of the symptoms
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
222
Q

Name the unwanted side effects of muscarinic receptor antagonists

A
  • hot as hell -> decreased sweating interferes with thermoregulation
  • dry as a bone -> reduced secretions
  • blind as a bat -> cyclopegia
  • mad as a hatter -> CNS disturbance
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
223
Q

Which of the following drugs would you administer to treat an atropine overdose?

a. bethanechol
b. ecothiopate
c. hyoscine
d. physostigmine
e. pralidoxime

A

d- physostigmine - reversibly blocks anti-cholinesterase so acetyl choline can outcompete the atropine until it is metabolised out of the system

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

State the role of different adrenoceptors.

A
  • alpha 1 - vasoconstriction, relaxation of GIT
  • alpha 2 - inhibition of transmitter release, contraction of vascular smooth muscle, CNS actions
  • beta 1 - increased cardiac rate and force, relaxation of GIT, renin release from kidney
  • beta 2 - bronchodilation, vasodilation, relaxation of visceral smooth muscle, hepatic glycogenolysis
  • beta 3 - lipolysis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
225
Q

What are the affects of stimulating A1 adrenoceptors?

A

PLC —> IP3 and DAG

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

What are the affects of stimulating A2 adrenoceptors?

A

decreased cAMP

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

What are the affects of stimulating B2 adrenoceptors?

A

increased cAMP

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

What do cholinoceptor antagonists do?

A
  • cause the loss of sympathetic functions -> enhance parasymathetic function
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
229
Q

What do SNS agonist cause?

A
  • cause the loss of parasympathetic functions -> enhance symathetic function
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
230
Q

Describe the selectivity of SNS agonists

A
  • ALL adrenoceptors can be activated by NA and A
  • NORADRENALINE is more selective for ALPHA receptors
  • ADRENALINE is more selective for BETA receptors
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
231
Q

Name 5 directly acting SNS agonists

A
  • adrenaline
  • phenylephrine
  • clonidine
  • dobutamine
  • salbutamol
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
232
Q

Which is adrenoceptor is adreanline selective too?

A
  • non-selective
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
233
Q

Which receptor is phenylephine selective too?

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

Which receptor is cloninide selective too?

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

Which receptor is dobutamine selective too?

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

Which receptor is salbutamol selective too?

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

What does selectivity depend on?

A

o CONCENTRATION - at low concentrations these drugs will be relatively selective but if you increase its concentration, the chance of binding to other receptors increases

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

Why is adrenaline used in the treatment of anaphylaxis instead of noradrenaline?

A

o the key thing to consider here is BREATHING -> the patient breathing must be breathing, even if you have a functioning cardiovascular system, you will have nothing to pump around

o adrenaline acts MORE ON BETA RECEPTORS than noradrenaline and this stimulates bronchodilation and relaxation of the throat muscles

o will also stimulate the heart so you get tachycardia, this will support the blood pressure

o acts on alpha receptors and causes vasoconstriction

o via the beta 2 receptors, slows down the release of histamine from the mast cells

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

What are the symptoms of hypersensitivity?

A

o endothelial cells within the membranes of the blood vessels move apart so you get a lot of fluid moving into the tissues -> leads to a fall in circulating fluid volume -> fall in bp -> ANAPHYLACTIC SHOCK and collapse of the circulatory system that leads to unconsciousness

o contraction of bronchial smooth muscle and constriction of the muscles around the throat -> causing respiratory distress

o constriction of smooth muscle in the GI tract causing vomiting and diarrhoea

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

In what instances would you use adrenaline for treatment of pulmonary disorders?

A
  • obstructive conditions -> asthma emergency or acute bronchospasm
  • selective beta-2 agonists are preferable
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
241
Q

What is adrenaline used to treat?

A
  • allergic reactions and anaphylactic shock
  • pulmonary obstructive conditions -> astham and acute bronchospasm
  • glaucoma

cardiogenic shock

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

In what pulmonary event would adrenaline be given as a treatment?

A
  • cardiogenic shock - the sudden inability of the heart to pump sufficient oxygen-rich blood (occurs in MI or cardiac arrest)

o beta 1 receptor is the main target -> positive inotropic action

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

When is adrenaline given in which the main target site is alpha 1?

A
  • spinal and local anaestheisa
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
244
Q

Why is adreanline given with spinal anaesthsia

A

o anaesthetising through the spine takes away the sympathetic output to the peripheral resistance vessels

o meaning you get relaxation of peripheral resistance vessels so the patient won’t be able to maintain their blood pressure

  • giving a little bit of adrenaline at the same time you can constrict the blood vessels so you can maintain the blood pressure
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
245
Q

Why is adrenaline given with local anaesthesia?

A

o adrenaline causes constriction of the blood vessels in the local area thus preventing the clearance of the anaesthetic from the area

o if the anaesthetic was given without adrenaline then it will wear off faster

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

What are the side effects of adrenaline?

A
  • Secretions - reduced and thick
  • CNS - minimal
  • CVS Effects:

o Tachycardia, palpitations, arrhythmias

o Cold extremities, hypertension

o OVERDOSE - cerebral haemorrhage, pulmonary embolism

  • GIT - minimal
  • Skeletal muscle - tremor
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
247
Q

Out of the unwanted effects of adrenaline a important to monitor?

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

Describe the selectivity of phenylephrine

A
  • very selective to alpha 1
  • minorly selective to alpha 2
  • no really selectivity to beta 1 or 2
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
249
Q

Phenylephrine is structually similar to adrenaline, so why is it ever used?

A
  • more resistant to COMT degradation so stays in the system for a bit longer
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
250
Q

What are the clinical uses of phenylephrine?

A

o Vasoconstriction

o Mydriatic (dilates pupil)

o Nasal Decongestant -> vasoconstriction in nasal sinus -> less fluid leakage -> less blockage

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

What is aside effect of phenylephrine which must be watched out for?

A
  • hypertension due to vasoconstriction of vessels
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
252
Q

Describe the selectivity of clonidine

A
  • very selective to alpha 2
  • moderately selective to alpha 1
  • no really selectivity to beta 1 or 2
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
253
Q

Describe the mechanism of action of clonidine

A
  • clonidine will stimulate the pre-synaptic alpha 2 receptors -> has a negative effect on the synthesis and release of noradrenaline
  • less noradrenaline -> less stimulation at the effector organ -> less vasoconstriction -> fall in TPR and bp
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
254
Q

What are the clinical uses of clonidine

A
  • hypertension
  • migraines
  • glaucoma
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
255
Q

Excluding blood vessels, where else are alpha 2 receptors found and explain whether clonidine acts on them

A
  • the brain
  • clonidine has a central action on the brainstem

o within the brainstem it works on the baroreceptors in this pathway and reduces the sympathetic drive coming out of the brain

o the reduction in sympathetic activity -> reduced TPR and noradrenaline release at the nerve terminal thus reducing TPR further

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

Describe the selectivity of isoprenaline

A
  • selective for beta 1 and beta 2
  • no selectivity for alpha 1 or alpha 2
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
257
Q

Why is isoprenaline clinically relevant seeing that it is structually similar to adrenaline?

A
  • less susceptible to uptake 1 and MAO breakdown -> much longer plasma half-life (2 hours)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
258
Q

What are the clinical uses of isoprenaline?

A
  • Cardiogenic Shock
  • Acute Heart Failure
  • Myocardial Infarction
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
259
Q

What is the issue with using isoprenaline to treat cardiac problems?

A
  • you are trying to restore cardiac output
  • beta-2 receptors in vascular smooth muscle which causes dilation of the blood vessels in the muscles -> pooling of blood within the muscles -> decreased venous return -> via baroreceptors, you get REFLEX TACHYCARDIA
  • the caused effect is opposite to what you are trying to achieve
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
260
Q

Describe the selectivity of dobutamine

A
  • selective to beta 1
  • some selectivity to beta 2
  • very light selectvitiy to alpha 1 or alpha 2
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
261
Q

What are the clinical uses of dobutamine

A

o Cardiogenic Shock

o Acute Heart Failure

o Myocardial Infarction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
262
Q

Why is dobutamine used for cardiac disorders instead of isprenaline

A
  • lacks reflex tachycardia because it is selective for beta 1
  • short plasma half-life (2 minutes) so gives a lot of control
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
263
Q

How is botutamine administrated?

A
  • IV infusion
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
264
Q

Describe the selectivity of salbutamol

A
  • beta 2 selectivity
  • some selectivity to beta 1
  • not selective for alpha 1 or alpha 2
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
265
Q

What are the clinical uses of salbutamol?

A
  • asthma
  • threatened premature labour
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
266
Q

How does salbutamol work as a treatment for asthma?

A

· Beta-2 mediated relaxation of bronchial smooth muscle

· Inhibition of release of bronchoconstrictor substances from mast cells

· The inhaler gives relatively localised effects on the lungs

· You can also give salbutamol as tablets to control asthma - for chronic asthma conditions in hospital

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

How does salbutamol work as a treatment for threatening premature labour

A
  • beta-2 mediated relaxation of uterine smooth muscle -> prevents abortion of a foetus
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
268
Q

What are the side effects of salbutamol?

A

o Reflex tachycardia

o Tremor

o Blood sugar dysregulation

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

Name two indirectly acting SNS agonists?

A
  • cocaine
  • tyramine -> the cheese reaction
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
270
Q

WHat is the mechanism of cocaine?

A
  • Uptake 1 blocker for dopamine and noradrenaline
  • gives a high by causing an increase of dopamine in the synapse
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
271
Q

What are the actions/side effects of cocaine on the CNS?

A
  • low doses: euphoria, excitement, increased motor activity
  • high doses: activation of chemotactic trigger zones -> vomiting, CNS depression, respiratory failure, convulsions and death
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
272
Q

What are the actions/side effects of cocaine on the CVS?

A
  • low doses: tachycardia, vasoconstriction and raised blood pressure
  • high doses: ventricular fibrillation and cardiac arrest
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
273
Q

What is the mechanism of tyramine?

A
  • a dietary amino acid - commonly found in cheese (hence being dubbed the cheese reaction) , red wine and soy sauce
  • it acts as a false neurotransmitter -> isn’t a problem in normal people (except from stimulating dreams)
  • tyramine acts as a weak agonist at the effector organ at which the noradrenaline will be stimulating the receptors while aslo piggy backing on the uptake systems and competing with NA for the uptake 1 site
  • this means that you get more NA hanging around in the synapse before displacing NA from the vesicles
  • normally, this displaced NA will be broken down by MAO but the tyramine competes with the NA for the binding site on the MAO
  • this means that there is less break down of the NA that has been displaced from the vesicles -> builds up and leaks out into the synaptic cleft
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
274
Q

What kind of people have to be careful of tyramine?

A
  • people on MAO inhibitors (e.g. to control depression)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
275
Q

What is the problem with the combination of tyramine and MAO inhibitors?

A
  • patients on MAO inhibitors have their MAO capabilities completely blocked
  • this inhibits the breakdown of NA -> accumulation of NA in the synapse that leads to a hypertensive crisis
  • patients on MAO inhibitors are told to avoid cheese, red wine and soy sauce and anything else that has high concentrations of tyramine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
276
Q

What do alpha 1 adrenoceptors cause?

A
  • vasoconstriction, relaxation of the GIT
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
277
Q

What do alpha 2 adrenoceptors cause?

A
  • inhibit transmitter release
  • contraction of vascular smooth muscle
  • CNS actions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
278
Q

What do beta 1 adrenoceptors cause?

A
  • increase cardiac force and rate
  • relaxation of GIT
  • renin release from the kidneys
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
279
Q

What do beta 2 adrenoceptors cause?

A
  • bronchodilation
  • vasodilation
  • relaxation of visceral smooth muscle
  • hepatic glycogenolysis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
280
Q

What do beta 3 adrenoceptors cause?

A
  • lipolysis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
281
Q

What is different about alpha 2 receptors compared to other adrenoceptors?

A
  • appear on neurones and pre-synapses
  • act as a negative feedback regulator to prevent prolonged action
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
282
Q

What receptors are targeted by carvedilol?

A
  • alpha 1 and beta 1
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
283
Q

What receptors are targeted by labetalol?

A
  • alpha 1 and beta 1
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
284
Q

What receptors are targeted by prazosin?

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

What receptors are targeted by propanolol?

A
  • beta 1 and 2
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
286
Q

What receptors are targeted by atenolol?

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

What diseases provide the main clinical uses of SNS antagonists and false transmitters?

A

· Hypertension

· Cardiac Arrhythmias

· Angina

· Glaucoma

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

What is the major controller of blood pressure?

A
  • renin release -> angiotensin II and aldosterone
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
289
Q

Name the targets for anti-hypertensive treatments.

A

· Sympathetic nerves that release the vasoconstrictor noradrenaline

· Kidneys - regulates blood volume and vasoconstriction

· Heart

· Arterioles determine peripheral resistance (alpha 1 mediated vasoconstriction)

· CNS - determines blood pressure set point and regulates some systems involved in blood pressure control and the autonomic nervous system

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

Why were cardioselective beta antagonists made?

A
  • most of the POSITIVE effects are from Beta 1 blockade and most of the NEGATIVE effects are from Beta 2 blockade
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
291
Q

What are the target sites for beta blcokers?

A

o act as anti-hypertensives mainly because of their antagonism of beta 1 adrenoceptors

  • in the heart to reduce heart rate and cardiac output but this effect disappears with chronic treatment - the heart resets itself
  • KIDNEYS to REDUCE RENIN PRODUCTION - PRIMARY EFFECT

o beta 2 antagonism may be important, but it is not clear to what extent due to decreased noradrenaline

o act in the CNS, if lipid soluble enough, to reduce sympathetic tone (beta 1 and 2)

Back

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

Name some side effects of beta blockers?

A
  • bronchoconstriction
  • cardiac failure -> caused by slowing the heart too much
  • hypoglycemia
  • fatigue -> due to reduced cardiac output and reduced muscle perfusion
  • cold extremities -> loss of beta receptor mediated vasodilation of cutaneous vessels
  • bad dreams
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
293
Q

Name 4 disease that if a patient is suffering from should not be given no-selective beta blockers?

A

o ASTHMA

o COPD

o CARDIAC FAILURE

o DIABETES -> masks symptoms of hypoglycemia - don’t even know they are haing an attack

  • all due to beta 2 blockade
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
294
Q

What is the mechanism of action of Nebivolol?

A
  • binds to beta 1 receptors
  • causes NO release
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
295
Q

How does Sotalol function?

A
  • binds to beta 1 and 2
  • inhibits K+ channels
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
296
Q

What is the selectivity of Propanolol?

A
  • non-selective beta antagonist
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
297
Q

What are the effects of Propanolol?

A
  • at rest, propranolol causes very little change in heart rate, cardiac output or arterial pressure
  • in exercise that you see the effects of propranolol on these variables
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
298
Q

Does Propanolol have any negative effects?

A
  • all the typical adverse effects, caused by beta 2 antagonism
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
299
Q

What is the selectivity of Atenolol?

A
  • historically called cardioselective beta blockers -> are selective for beta-1
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
300
Q

How does Antenolol bring about its effects?

A
  • mainly antagonises the effects of noradrenaline in the heart
  • also affect any tissue with beta 1 receptor e.g. kidneys
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
301
Q

What does Atendolol’s selectivity depend on?

A
  • its concentration -> therefore it isnt safe to give to asthmatic patients despite having less of an effect on the airways
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
302
Q

When might Sotalol be the best anti-hypertensive?

A
  • when the hypertension is associated with an arrhythmia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
303
Q

What are the advantages of Atanolol over Propanolol?

A
  • Atanolol is beta 1 specific so doesn’t cause the side effects
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
304
Q

What are the targets for Carvedilol?

A
  • mixed beta and alpha -> beta 1, beta 2 and alpha 1
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
305
Q

What is the advantage of Carvedilol?

A
  • alpha 1 blockade -> additional vasoconstrictor properties
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
306
Q

What advantage does carvedilol have over atenolol and propranolol?

A
  • reduces the vasoconstriction due to targeting of alpha 1
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
307
Q

What are the affects of alpha receptors?

A
  • a1-receptors -> postsynaptic on vascular smooth muscle -> vasoconstriction
  • a2-receptors -> presynaptic autoreceptors inhibiting NE release
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
308
Q

What is the selectivity of Phentolamine?

A
  • non-selective alpha antagonist
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
309
Q

How does Phentolamine act as a anti-hypertensive agent?

A
  • causes vasodilation due to alpha 1 blockade
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
310
Q

What is the problem with Phentolamine?

A
  • blockade of presynaptic alpha-2 receptors removes the inhibitory effect of the alpha-2 receptors on noradrenaline release -> increase in noradrenaline release -> enhances the reflex tachycardia that appears with any blood pressure lowering agent
  • increased GIT motility
  • diarrhoea

o NO LONGER IN CLINICAL USE

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

What is the selectivity of Prazosin?

A
  • highly alpha 1 selective antagonists
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
312
Q

What is the effect of Prazosin?

A
  • vasodilation
  • less tachycardia than non-selective antagonists since they do NOT increase noradrenaline release from nerve terminals
  • cardiac output decreases, due to a fall in venous pressure due to dilation of capacitance vessels
  • hypotensive effect is dramatic -> the postural hypotension is troublesome
  • unlike other anti-hypertensives, alpha-1 antagonists cause a decrease in LDL and an increase in HDL cholesterol -> becoming popular again as an anti-hypertensive agent
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
313
Q

Name 6 beta blockers.

A
  • Propanolol
  • Pindolol
  • Atenolol
  • Carvedilol
  • Nebivolol
  • Sotalol
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
314
Q

Name 2 alpha blockers.

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

What is the current role of beta and alpha blockers in the treatment of heart failure?

A
  • not front-line treatment
  • are add ons if 2 or 3 methods don’t work
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
316
Q

Why do alpha 2 receptors and baroreceptors reduce the effectiveness of phentolamine?

A
  • removes inhibitory effect of alpha 2 receptors on noradrenaline release -> increase NA release -> enhanced reflex tachycardia to low bp
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
317
Q

Name a false transmitter used to treat hypertension.

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

What is the mechanism of action os Methyldopa?

A
  • Methyldopa is taken up by the noradrenergic neurons and is decarboxylate and hydroxylated to form the false transmitter - alpha-methyl noradrenaline
  • it is NOT deaminated within the neuron by MAO and tends to accumulate in larger quantities than noradrenaline therefore it displaces noradrenaline from the synaptic vesicles
  • is less active than noradrenaline on alpha-1 receptors so is LESS EFFECTIVE AT CAUSING VASOCONSTRICTION
  • more active on presynaptic alpha-2 receptors, this means that the auto-inhibitory feedback mechanism operates more strongly and reduces noradrenaline release below normal levels
  • has some CNS effects, it stimulates the vasopressor centre in the brainstem to inhibit sympathetic outflow
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
319
Q

What are the other effects of Methyldopa?

A

o Renal and CNS blood flow is well maintained so it is widely used in patients renal insufficiency or cerebrovascular disease

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

For what group of people is Methyldopa recommended?

A

o pregnant women because it has no adverse effects on the foetus despite crossing the blood-placenta barrier

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

What are the adverse effects of Methyldopa?

A
  • dry mouth
  • sedation
  • orthostatic hypotension
  • male sexual dysfunction

o It is RARELY USED -> causes postural hypotension

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

What is the main cause of myocardial ischaemia?

A
  • arrhythmias
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
323
Q

Which drug is used in the majority of arrhythmias?

A
  • Propanolol
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
324
Q

What class of drugs are used to treat arrhythmias?

A

o beta antagonists (beta blockers)

  • an increase in sympathetic drive to the heart via beta-1 can precipitate or aggravate arrhythmias due to its control on the pacemaker current and AV conductance
  • refractory period of the AV node is increased by beta antagonists -> interferes with AV conduction in atrial tachycardias and slows down ventricular rate -> even if with strange re-entry type electrical activity in the damaged tissue, it wont stimulate another heart beat because it’s still in the refractory period
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
325
Q

Define Angina.

A
  • pain that occurs when the oxygen supply to the myocardium is insufficient for its needs
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
326
Q

Where are the most common places to feel pain with angina?

A
  • chest
  • arms
  • neck
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
327
Q

What tends to bring angina on?

A
  • exertion
  • excitement
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
328
Q

Describe Stable Angina.

A

o due to a fixed narrowing of the coronary vessels e.g. atheroma

o pain on exertion, but is enough flow for normal activity

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
329
Q

Explain what is meant by Unstable Angina?

A

o pain with less and less exertion, culminating with pain at rest due to atheromatous plaque beginning to rupture

o you get a platelet-fibrin thrombus associated with the ruptured atheromatous plaque but without complete occlusion of the vessel

o high risk of infarction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
330
Q

What causes Variable Angina?

A

o coronary artery spasm -> associated with atheromatous disease

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
331
Q

What drugs can be used to treat Angina?

A

o decrease heart rate and cardiac contractility -> decreased CO -> decreased systolic blood pressure

o at low doses, beta-1 selective antagonists, METOPROLOL, reduce heart rate and myocardial contractile activity without affecting bronchial smooth muscle -> reduce the oxygen demand whilst maintaining the same degree of effort

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
332
Q

What is Glaucoma?

A

o an increase in intraocular pressure due to aqueous humour produced by the blood vessels in the ciliary body via the actions of carbonic anhydrase

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
333
Q

What is directly related to the amount of aqueous humour that is produced?

A

o blood flow in the ciliary body

o blood pressure

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
334
Q

State the path of aqueous humour?

A

o posterior chamber -> through the pupil -> anterior chamber -> trabecular network in the canal of Schlemm

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
335
Q

What type of drugs are used to reduce the rate of aqueous humour formation?

A

o beta-blockers

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

How do beta blockers help with migraines?

A
  • maintains good blood supply to the CNS so reduces the risks of a migraine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
337
Q

In what nerves do you find neuromuscular transmission?

A
  • somatic
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
338
Q

Describe neuromuscular transmission?

A

o motor nerves are Cholinergic

  • action potential –> depolarisation of the membrane –> opening of voltage sensitive calcium channels –> calcium influx –> vesicles exocytosis
  • acetylcholine is synthesised from Acetyl CoA and Choline by choline acetyltransferase (CAT) (only found in cholinergic nerve terminals)
  • the targets for the acetylcholine at neuromuscular junctions are the nicotinic acetylcholine receptors on the END PLATE
  • nicotinic receptors are ion channel linked, when you stimulate the receptor you get a change in conformation and an influx of sodium ions
  • this results in a depolarisation of the membrane called the end plate potential
  • this is a GRADED potential - depends on how much acetylcholine is released and how many receptors are stimulated
  • once the end plate potential reaches a threshold then it generates an action potential that propagates in both directions from the end plate
  • the acetylcholinesterase, which breaks down the acetylcholine once it’s had its effect, is bound to the basement membrane in the synaptic cleft
  • acetylcholinesterase breaks down acetylcholine to acetate and choline
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
339
Q

What are the two main subsets of nicotinic acetylcholine receptors?

A

o ganglionic (aka neuronal)

o muscle

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

Describe the distribution of nicotinic receptors.

A

o span the membrane and have both intra- and extra-cellular components

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
341
Q

How many subunits make up a nicotinic acetylcholine receptor?

A

o 5

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

What is the significance of there being two alpha subunits when it comes to receptor activation?

A

o both must bind to acetylcholine before it becomes activated

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
343
Q

Name a drug which targets the central processes with the CNS?

A
  • baclofen/diazepam -> GABA receptor agonist
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
344
Q

When is diazepam useful?

A

o is a benzodeizepam -> used to treat spasticity following strokes or due to cerebral palsy

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

How do local anaesthetic work?

A
  • prevent conduction of action potentials down motor neurones
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
346
Q

How can local anaesthetics cause muscle weakness?

A
  • if it injected too close/directly into a motor neurone
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
347
Q

Name both two therapeutic drugs and one toxin that can interfere with acetylcholine release?

A
  • therapeutic = hemicholinium and calcium entry blockers
  • toxin = botulinium toxin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
348
Q

What are the two types of neuromuscular blocking drugs?

A
  • depolarising
  • non-depolarising
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
349
Q

Name a depolarising and a non-depolarising drug which acts at the motor end plate?

A
  • depolarising - suxamethonium
  • non-depolarising - tubocurarine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
350
Q

Where is Dantrolenes sit of action?

A
  • inhibits calcium release within the muscle fibres themselves -> prevents propagation – acts as a spasmolytic
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
351
Q

Where is the site of action of neuromuscular blocking drugs?

A
  • post-synaptic -> act on nicotinic receptors on the motor end plate
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
352
Q

What are non-depolarising NM blocking drugs?

A
  • competitive nicotinic receptor antagonists
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
353
Q

What are depolarising NM blocking drugs?

A
  • nicotinic receptor agonists
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
354
Q

Do NM blocking drugs affect consciousness?

A
  • no
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
355
Q

Do NM blocking drugs affect pain sensation?

A
  • no
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
356
Q

What must be done to help patients when on NM blocking drugs?

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

Describe the structure of a non-polarising NM blocking drug?

A
  • big molecules with relatively restricted movement around the bonds
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
358
Q

Describe the structure of a polarising NM blocking drug?

A
  • small molecules with rotationally movement around the bonds
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
359
Q

What is the structure of suxamethonium?

A
  • two bonded acetylcholine molecules
  • due to the flexibility one molecule can bind to two alpha subunits -> therefore stimulating the receptor
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
360
Q

Describe the mechanism of action of suxamethonium.

A
  • causes an extended end plate depolarisation -> isn’t metabolised as quickly as acetylcholine so remains bound for longer
  • this leads to a depolarisation block of the NMJ (aka phase 1 block) due to overstimulation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
361
Q

How are NM blockers administrated?

A
  • IV shot – slowly seeps into muscle fibres
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
362
Q

What does suxamethonium cause?

A
  • fasciculations (individual fibre twitches) -> leads to flaccid paralysis (up to 5 minutes)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
363
Q

What metabolises suxamethionium?

A
  • pseudocholinesterase in the liver and plasma
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
364
Q

What are the uses of suxamethinium?

A
  • endotracheal intubation -> relaxes skeletal muscle of the airways
  • muscle relaxant for electroconvulsive therapy -> treatment for severe clinical depression if talking therapy and anti-depressants haven’t worked
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
365
Q

Name 4 unwanted effects of suxamethonium.

A
  • post-operative muscle pain
  • bradycardia
  • hyperkalaemia
  • raised intraocular pressure
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
366
Q

What causes the post-operative muscle pain after suxamethonium use?

A
  • initial fasciculation ripping the muscle slightly
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
367
Q

How does suxamethium cause bradycardia?

A
  • direct muscarinic action in the heart
  • generally avoid as suxamethonium is given after general anaesthetic which has atropine (competitive muscarinic antagonist) with it
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
368
Q

Name 3 types of people who should not be given suxamethonium.

A
  • burn patients
  • soft tissue injury
  • eye injuries or glaucoma
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
369
Q

What is the mechanism of action of tubocurarine?

A
  • competitive acetylcholine receptor antagonist
  • 70-80% is required to achieve full relaxation of muscles as it prevents the potential from reaching the threshold
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
370
Q

What are the effects of tubocurarine?

A
  • fasciculations (individual fibre twitches) -> leads to flaccid paralysis (40-100 minutes)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
371
Q

In which order does skeletal muscle relax in when administrated with tubocarinine?

A

o extrinsic eye muscles (relaxes first, gets back to normal last)

o small muscles of the face, limbs, pharynx

o respiratory muscles (relaxes last, gets back to normal first)

  • return in the opposite order
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
372
Q

What are the uses of tubocarinine?

A
  • relaxation of skeletal muscle during surgery -> means less anaesthetic is needed
  • permit artificial ventilation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
373
Q

What reverses the actions of non-polarising NM blockers

A
  • anti-cholinesterases (neostigmine) -> increase acetylcholine conc.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
374
Q

Describe the pharmacokinetics of tubocurarine.

A
  • does NOT cross the BBB or placenta
  • NOT metabolised at all -> excreted in the urine (70%) and the bile (30%)
  • if there is any impairment in hepatic or renal function then it increases the duration of action of tubocurarine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
375
Q

What is atracurium?

A
  • a non-depolarising NM blocker used instead of tubocurarine (same mechanism) in patients with hepatic or renal impairment
  • a chemically unstable molecule - due to the pH of the plasma it gets hydrolysed into two inactive fragments -> duration of action isn’t affected by functionality
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
376
Q

What are the unwanted effects of tubocurarine?

A

o ganglion block - it could block some of the nicotinic receptors in the ganglia

o histamine release from mast cells

  • hypotension -> due to ganglion blockade and histamines acting on the H1 receptors on the vasculature and cause vasodilation
  • tachycardia -> may lead to arrhythmias -> is a reflex in response to the hypotension or blockade of vagal ganglia
  • bronchospasm -> caused by the histamine release
  • excessive secretions (bronchial and salivary) -> caused by the histamine release
  • apnoea -> why you always assist respiration in someone who’s taking tubocurarine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
377
Q

Which of the following effects would be observed with a non-depolarising neuromuscular block?

A: Initial muscle fasciculations

B: Irreversible nAChR blockade

C: The block would be enhanced by anti-cholinesterase drugs

D: A flaccid paralysis

E: Increased arterial pressure

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

The clinical use of neuromuscular blocking drugs will most likely involve interference with which of following physiological processes?

A: Kidney function

B: Consciousness

C: Body temperature regulation

D: Pain sensation

E: Respiration

A
  • E
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
379
Q

Describe action potentials of the heart.

A
  • several channels are important in regulating the sinoatrial action potential
  • If Channel is responsible for allowing the action potentia to propagate -> is a sodium channel that opens at the most negative potential -> initially sodium influx occurs until a certain amount of depolarisation and then the calcium channels open
  • calcium channels come in TWO forms: -> T type = transient and L type = long lasting
  • opening of the calcium channels increases the depolarisation
  • potassium channels opening is responsible for repolarisation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
380
Q

How does the sympathetic nervous system bring about an increase in herat rate?

A
  • beta adrenoceptors are coupled with adenylate cyclase and cause an increase in cAMP -> is important in opening the If channel
  • sympathetic nervous system is responsible for causing this increase in cAMP and it also has a positive effect on calcium entry
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
381
Q

How does the parasympathetic system slow heart rate?

A
  • parasympathetic nervous system is negatively coupled with adenylate cyclase via muscarinic receptors -> promotes the opening of potassium channels and prolongs repolarisation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
382
Q

What 4 factors influence myocardial oxygen demand?

A
  • heart rate
  • afterload
  • contractility
  • preload
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
383
Q

Name three drugs which can slow heart rate.

A
  • beta blockers
  • calcium channel antagonists
  • ivabradine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
384
Q

How do beta blockers slow heart rate?

A
  • block beta receptors -> less cAMP is made so less downstream signalling -> longer initial depolarisation phase before calcium channels open
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
385
Q

How do calcium channel antagonists reduce heart rate?

A
  • block calcium channels (L-type) in the plasma membrane -> reduced calcium influx of external calcium -> reduced influx of calcium from the sacroplamic reticulum
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
386
Q

What part of cardiac myocytes holds the most calcium?

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

How does ivabradine reduce heart rate?

A
  • blocks sodium channels (If) -> delays depolarisation to threshold levels
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
388
Q

Name 2 drugs that can reduce cardiac contractility.

A
  • beta blockers
  • calcium channel blockers
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
389
Q

Name the 2 classes of calcium channel antagonists, also state the major difference between the two.

A
  • rate slowing -> act on the CARDIAC and smooth muscle
  • non-rate slowing -> act on the smooth muscle
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
390
Q

What is the major consequence of non-rate slowing calcium channel antagonists?

A
  • reflex tachycardia -> profound vasodilation triggers baroreceptors which signal to the heart
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
391
Q

What is the mechanism of action of organic nitrate?

A
  • organic nitrates are substrates for nitric oxide production
  • enters endothelial cells and promote NO production -> NO then diffuses into the vascular smooth muscle and causes smooth muscle relaxation by activating guanylate cyclase
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
392
Q

Name a condition in which organic nitrates are commonly prescribed.

A
  • angina when there is profound atherosclerosis reducing the blood flow to the heart
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
393
Q

What is the mechanism of potassium channel openers?

A
  • promote potassium efflux so smooth muscle becomes hyperpolarised -> has a reduced ability to contract -> overall it promotes smooth muscle contraction/vasodilation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
394
Q

How do organic nitrates and potassium channel openers influence the myocardial oxygen supply/demand relationship?

A
  • increase coronary blood flow
  • vasodilation reduces TPR hence reduces afterload -> heart has less work to do against the resistance
  • venodilation, which reduces venous return to the heart and hence reduces preload and contractility
  • reduction in afterload and preload causes a decrease in myocardial oxygen demand
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
395
Q

What are the effects of beta blockers?

No action

Mouse Gestures

Back

Forward

Scroll up

Scroll down

Switch to previous tab

Switch to next tab

Close all tabs except current

Close current tab

Open new tab

Close all tabs

Refresh current tab

Stop loading

Scroll to bottom

Scroll to top

Reopen closed tab

Go to home page

A
  • decreased heart rate and contractility
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
396
Q

What are the effects of calcium channel antagonists?

A
  • decreased heart rate and contractility
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
397
Q

What are the effects of organic nitrates?

A
  • increased coronary blood flow, decreased preload and afterload
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
398
Q

What are the effects of potassium channel openers?

A
  • increased coronary blood flow, decreased preload and afterload
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
399
Q

What is the effects ivabradine?

A
  • decreased heart rate
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
400
Q

Name treatments for angina.

A
  • beta blocker or calcium antagonist as background anti-angina treatment
  • ivabradine is a newer treatment
  • nitrate as symptomatic treatment (short acting)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
401
Q

What are the side effects of beta blockers?

A
  • worsening of cardiac failure (CO reduction) -> in angina with HF beta-blockers will reduce the work the heart has to do but, because of the HF it is dangerous -> CO goes down -> HF is worsened
  • bradycardia (heart block) -> due to less conduction through AV node
  • bronchoconstriction (blockade of β2 in airways)
  • hypoglycaemia (in diabetics on insulin) due to decreased glycogenolysis/gluconeogenesis
  • cold extremities and worsening of peripheral arterial disease (β2 blockade in skeletal muscle vessels) -> beta-2 receptors cause vasodilation, especially in peripheral tissue -> e.g. blood flow to the hands is dependent on beta-2 mediated vasodilation
  • fatigue
  • impotence (sexual dysfunction)
  • depression
  • CNS effects (lipophilic agents) e.g. nightmares
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
402
Q

If a beta blocker is being used as a treatment for heart failure what properties should you look for?

A
  • use a beta blocker whihc has vasodilator properties -> should have equal affinity for beta 1 and 2 receptors or also antagonise a 1 receptors -> e.g. pindolol or carvedilol
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
403
Q

Name two groups of patients that shouldn’t be given beta blockers.

A
  • asthmatics -> due to bronchoconstriction
  • diabetics -> due to hypoglycaemic properties
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
404
Q

What are the side effects of rate limiting calcium channel blockers?

A
  • bradycardia and AV block (Ca2+ channel block)
  • constipation -> less gut contractions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
405
Q

Name a rate limiting/slowing calcium channel blocker.

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

What are the side effects of non-rate slowing calcium channel blockers?

A
  • ankle oedema -> vasodilation means more pressure on capillary vessels
  • headache/flushing
  • palpitations -> baroreceptor reflex response
  • vasodilation/reflex adrenergic activation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
407
Q

Name a non-rate slowing calcium channel blocker.

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

Describe the simple classification of arrhythmias based on the site of origin.

A
  • supraventricular arrhythmias (e.g. amiodarone, verapamil) – can be atria/nodal tissue
  • ventricular arrhythmias (e.g. flecainide, lidocaine)
  • complex (supraventricular + ventricular arrhythmias) (e.g. disopyramide)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
409
Q

What is Vaughan-Williams classification.

A
  • classification of anti-arrhythia drugs

o CLASS I: Sodium channel blockade

o CLASS II: Beta-adrenergic blockade

o CLASS III: prolongation of repolarisation (membrane stabilisation, mainly due to potassium channel blockade)

o CLASS IV: Calcium channel blockade

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
410
Q

What is the problem with the Vaughan-Williams classification system?

A
  • despite still being used by most cardiologist lots of of modern dugs don’t fit it -> many drugs cross classes, particularly Na and K channel blockers -> couldn’t be classified -> e.g. amiodarone
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
411
Q

What is the mechanism of adenosine?

A
  • nodal tissue via the A1 receptor -> negative effects on cAMP -> decreased Ca channel opening and prolonged K channel opening -> prolonged depolarisation and slower repolarization -> helps to normalise tachyarrhythmia to normal sinus rhythm
  • vascular effects -> stimulation of adenylate cyclase à increased cAMP -> associated with relaxatiob of smooth muscle
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
412
Q

What is the use of adenosine?

A
  • supraventricular tachyarrhythmias
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
413
Q

What is the mechanism of action of verapamil?

A
  • depresses SAN automatically and subsequently AV node conduction
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
414
Q

What is the use of verapamil?

A
  • reduction of ventricular responsiveness to atrial arrhythmias
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
415
Q

What is the mechanism of amiodarone?

A
  • complex action involving beta-blocking, calcium and potssium channel effects
  • major effects are attributable to potassium channel blockade
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
416
Q

What are the uses of amiodarone?

A
  • supraventricular and ventricular tachyarrhythmias -> often due to re-entry problems
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
417
Q

What are the adverse effects of amiodarone?

A
  • accumulates in the body -> t½ 10 - 100 days causing:
  • photosensitive skin rashes
  • hypo- or hyper-thyroidism
  • pulmonary fibrosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
418
Q

What is re-entry rhythms?

A
  • when APs meet they tend to cancel each other out
  • in healthy tissue APs would pass down on either side, and if they crossed paths, they would MEET and CANCEL EACH OTHER OUT. This ensures that the AP proceeds in one direction.
  • tissue blocks leads to re-entry rhythm -> if the tissue with a block has undergone its repolarisation phase, APs can reactivate the tissue -> tachyarrhythmia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
419
Q

How are re-entry rhythms overcome?

A
  • work against it to prolong repolarisation -> the re-entry AP comes into the tissue and reaches the AP point, the tissue hasn’t repolarised yet -> the AP just dies and can’t be propagated -> reduce some of these arrhythmias
  • done with a potassium channel blocker like AMIODARONE
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
420
Q

What is the mechanism of cardiac glycosides?

A
  • inhibition of Na-K-ATPase -> increased intracellular Ca2+ via effects on Na+/Ca2+ exchange -> positive inotropic effect
  • central vagal stimulation by digoxin causes increased refractory period and reduced rate of conduction through the AV node (parasympathetic NS slows the heart)

o overall they slow the heart rate and improve ventricular contraction -> improves CO and restores normal rhythm

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
421
Q

What are the uses of cardiac glycosides?

A
  • AF and flutter lead to a rapid ventricular rate that can impair ventricular filling (due to decreased filling time) and reduce CO
  • digoxin reduces the conduction of electrical impulses within the AV node via the vagal stimulation -> fewer impulses reach the ventricles and ventricular rate falls
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
422
Q

Why does hypokalaemia lower the threshold for digoxin toxicity?

A
  • digoxin competes with potassium -> if you have little potassium in your blood, then digoxin over-inhibits
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
423
Q

What is the treatment pathway for hypertension?

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
424
Q

Name the 3 major stimuli for renin production.

A
  • decreased renal Na absorption
  • decreased renal perfusion pressure
  • increased sympathetic activity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
425
Q

What are the actions of angiotensin II?

A

o very powerful vasoconstrictor à increased TPR

o direct and indirect effects on the kidney

  • directly affects kidney to promote salt and water retention à increased blood volume
  • indirectly affects kidney by stimulating aldosterone production -> aldosterone acts on the kidney

o sympathetic activation

o thirst activation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
426
Q

Name an ACE inhibitor.

A
  • enalapril -> any drug ending with pril
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
427
Q

What are ACE inhibitors used to treat?

A
  • hypertension
  • heart failure
  • post-myocardial infarction
  • diabetic nephropathy
  • progressive renal insufficiency
  • procaution for patients at high risk of cardiovascular disease
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
428
Q

How do ACE inhibitors treat hypertension?

A
  • an effect on TPR -> angiotensin II is a vasoconstrictor -> blocking it will reduce TPR -> decrease BP
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
429
Q

How do ACE inhibitors treat heart failure?

A
  • angiotensin II increases salt and water retention -> drives up blood volume -> increase in venous return
  • ACE inhibitors decrease venous return in HF patients -> reduce work of the heart and decreased congestion in the system -> stress on heart alleviated
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
430
Q

What can angiotensin II receptor blockers be used to treat?

A
  • hypertension
  • heart failure
  • very similar to ACE inhibitors -> have the same effects -> decreased TPr and decreased venous return
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
431
Q

What are the side effects of ACEi and ARBs?

A
  • GENERALLY WELL TOLERATED. SIDE EFFECTS:
  • cough -> ACEi only -> prevents the breakdown of bradykinin -> builds up and stimulates coughing
  • hypotension
  • urticaria/angioedema (rare)
  • hyperkalaemia
  • foetal injury
  • renal failure in patients with renal artery stenosis -> angiotension can’t bind to efferent arteriole so glomerulus pressure isn’t maintained
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
432
Q

What is the mechanism of smooth muscle contraction?

A
  1. membrane depolarisation opens voltage-gated Ca2+ channels (VGCCs)
  2. Ca2+ enters & binds to calmodulin (CaM)
  3. Ca2+-CaM complex binds to & activates myosin light chain kinase (MLCK)
  4. MLCK mediated phosphorylation -> smooth muscle contraction
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
433
Q

Why are CCBs or thiazide diuretics used instead of ACEi or ARBs in the elderly adn Afro-Caribbean population?

A

elderly = uncoupling of BP from the RAS system as you get older -> more due atherosclerosis etc

  • afro-Caribbean = to low plasma renin activity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
434
Q

Why would you give an alpha blocker in resistant hypertension?

A
  • alpha 1 adrenoreceptor antagonists are used as a last resort as antihypertensive treatment
  • alpha 1 receptor is the predominant vasoconstricting receptor in the vasculature -> blocking alpha 1 receptors reduces vasoconstriction -> reduce TPR -> decrease BP
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
435
Q

Why do people abuse drugs?

A

primarily centred on the reward pathway (mesolimbic dopamine system) in the brain

  • reward pathway is a collection of dopaminergic neurones that originate in the ventral tegmental area and project down to the ventral striatum -> in particular the nucleus accumbens
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
436
Q

Define euphoria.

A
  • excessive dopamine release within the nucleus accumbens
437
Q

Describe the intranasal route of drug administration.

A
  • snorting
  • drug enters nasal sinus -> venous drainage -> lung -> heart -> brain

o mucous membranes of the nasal sinuses slow absorption

438
Q

Describe the oral route of drug administration.

A
  • eat or drink
  • stomach -> small intestine -> portal system -> liver -> heart -> brain

o very slow absorption due to the GI tract

439
Q

Describe the inhalational route of drug administration.

A
  • smoke
  • > lungs -> heart -> brain
  • rapid absorption (seconds) -> because the lungs are next to the heart -> is the fastest administration route (slightly faster than IV)
440
Q

Describe the intravenous route of drug administration.

A
  • injecting
  • vein -> heart -> brain

o rapid absorption (seconds)

441
Q

What are the 4 classes of drugs of abuse?

A
  • marcotics/painkillers
  • depressants
  • stimulants
  • miscellaneous
442
Q

Name some narcotics.

A
  • opiate like drugs -> heroin, morphine
443
Q

Name some depressants.

A
  • alcohol
  • benzodiaepines (valium)
  • barbiturates
444
Q

Name some stimulants.

A
  • cocaine
  • amphetamine (speed)
  • caffeine
  • metamphetamine (crystal meth)
445
Q

Name some miscellaneous drugs of abuse.

A
  • cannabis
  • ecstasy
446
Q

What are cannabinoids?

A
  • the compounds that prodcue the high effect when taking cannabis
  • 60 out of the 400 compounds in cannabis sativa are cannabinoids
447
Q

What is the most potent cannabinoid?

A
  • tetrahyrdocannabinoid (THC)
448
Q

What is cannabidiol?

A
  • a compound believed to have a protective effect from the negative effects of THC
449
Q

Describe the bioavailability of the cannabis from its two main royte of administration.

A
  • oral = 5-15% of dose enters bloodstream
  • inhalation = 25-35% of dose enters bloodstream -> anything inhaled suffers from about 50% loss automatically as only 50% gets far enough down into the lungs to diffuse into the bloodstream
450
Q

Describe the solubility of cannabis/cannabinoids.

A
  • cannabis is very lipid soluble -> accumulates in poorly perfused fatty tissues
  • in chronic cannabis users it will leak back out and can even be stored to a 4 times high concentration than in the bloodstream
451
Q

What is 11-hydroxy-THC?

A
  • the major metabolite of THC
  • is even more potent that THC
452
Q

Describe cannabis secretion.

A
  • a large amount of cannabis is secreted by the bile (enterohepatic recycling) -> therefore it goes down the bile duct -> small intestine -> potential reabsorption
  • 65% of 11-hydroxy-THC is secreted into the gut, which is then heavily reabsorbed back into the system
  • eventually the cannabis will be cleared -> urine loss accounts for 25%
453
Q

What is the correlation betwen plasma cannabinoid concentration and the degree of intoxication?

A
  • the correlation is very poor due to the storage/accumulation in fatty tisses (the brain is key in this due to its high fat content) -> significantly worse in chronic user
454
Q

How long after smoking a cannabis cigarette will the effects persist in the body?

a. 5 hours
b. 12 hours
c. 7 days
d. 30 days
e. 10 years

A

d. 30 days -> due to accumulation and then seaping out of adipose tissue

455
Q

What is anandamide?

A
  • an endogenous substance produced by the endogenous cannabinoid system -> behaves similarly to cannabis
456
Q

What are the receptors for endogenous anandamide?

A
  • CB1 RECEPTORS ->predominantly brain (hippocampus, cerebellum, cerebral cortex, basal ganglia)
  • CB2 RECEPTORS -> predominantly on immune cells in the periphery
457
Q

Explain the mechanism of anandamide.

A

Anandamide binds to receptors -> G-protein is negatively coupled to adenylate cyclase -> depress the inhibition to the dopaminergic neurones by GABA

  • CB1 receptor is the most prevalent G-protein coupled receptor in the brain
458
Q

What kind of drug is cannabis?

A
  • a depressant
459
Q

What is the mechanism of cannabis that brings about euphoria?

A
  • activation of CB1 and CB2 receptors induce euphoria -> stimulation of the CB1 receptor will switch off and blocks GABA transmission -> GABA suppresses the reward pathway -> when the reward pathway needs to be activated, GABA is inhibited
  • cannabis hijacks this system -> cannabis binds to CB1 receptors on GABA neurones -> reduces firing rate of GABAergic neurones (depressant) -> dopaminergic neurone firing increases -> euphoria
460
Q

What is the function of the anterior cingulate cortex?

A
  • the part of the brain important in error detection and monitoring of behaviour
  • is heavily involved with performance monitoring and behavioural adjustments to avoid losses (ensure good things occur)
461
Q

What is seen in the anterior cingulate cortex in cannabis users?

A
  • hypoactivity in the ACC -> cannabis users have problems adjusting their behaviour appropriately -> an element of psychosis and schizophrenia
  • isn’t clear whether cannabis causes psychosis and schizophrenia or whether suffers use cannabis to help symptoms
462
Q

How does cannabis increase appetite?

A

o cannabis has a positive effect on orexigenic neurones in lateral hypothalamus

  • cannabis switches off GABA -> increases MCH neuronal activity
  • increased orexin production

o also directly effects the AgRP neurones of the arcuate nucleus -> stimulates appetite

463
Q

What is the effects of cannabis on the immune system?

A
  • CB2 receptors are found in the periphery on immune cells -> down regulation of immune response
  • affects the activity of NK cells, macrophages, mast cells, T lymphocytes and B lymphocytes
  • chronic cannabis users have an impaired immune system
464
Q

What are the effects of cannabis?

A
  • psychosis and possible schizophrenia
  • increased appetite via the hypothalamus
  • memory loss -> Limbic regions (amnestic effects/↓BDNF) -> similar to alcohol
  • psychomotor performance -> cerebral cortex -> similar to alcohol
  • immunosuppressant
  • tachycardia and vasodilation -> conjunctivae (blood shot eyes)
465
Q

Explain why it is very difficult to die from cannabis.

A
  • there is very low CB1 receptor expression in the medulla -> important because the medulla contains the cardio-respiratory centre
  • substances such as alcohol and heroin have profound effects in the medulla, so can cause death cannabis doesn’t
466
Q

What disease cause an upregulation of CB receptors?

A
  • MS, chronic pain, stroke -> CB receptor are trying to compensate for the disease process
  • infertility, obesity -> CB receptor are part of the pathology of the disease
467
Q

In what diseases would you want to you a CB receptor agonist?

A
  • diseases with chronic pain or upregualtion of CB receptors as a consequence of the disease -> stroke or MS
  • examples of agonists are dronabinol, nabilone (both used in chemo patients to prevent nausea or to stimulate appetite in AIDS) and sativex (analgesic)
468
Q

In what diseases would you want to you a CB receptor antagonist?

A
  • obesity -> rimonabant was one of the first anti-obesity agents to target the cannabinoid receptor -> it was affective, but the problem was its propensity to cause depression, and suicide
469
Q

What enzyme break down endogenous cannabinoids?

A
  • fatty acid amide hydrolase
470
Q

What kind of drug is cocaine?

A
  • stimulant
471
Q

What kind of drug is nicotine?

A
  • stimulant
472
Q

Why is cocaine linked with nasal issues?

A
  • it is a powerful vasoconstrictor -> constantly snorting cocaine leads to vasoconstriction of the blood vessels in the nose -> necrosis then kicks in
473
Q

How is cocaine paste made?

A
  • crushing up the leaves with an organic solvent to form a paste -> extract is around 80% cocaine
474
Q

How is cocaine HCl made?

A
  • cocaine (erythroxylum) leaves are dissolved in acidic solution
475
Q

What form of cocaine was an early anaesthetic?

A
  • cocaine HCl -> however it was also abused
476
Q

How can cocaine paste and cocaine HCl be administrated?

A
  • IV
  • oral
  • intranasal
477
Q

How is crack cocaine made, and how can it be purified?

A
  • precipitate cocaine HCl mixed with an alkaline solution (e.g. baking soda) -> once this solution dries and hardens, you end up with little cocaine rocks
  • crack can be purified by dissolving it in a non-polar solvent (e.g. ammonia or ether) -> this is called freebase
478
Q

How can crack cocaine and freebase be administrated?

A
  • IV, orally and intranasal like other forms but also inhalation -> faster onset
  • crack cocaine is probably the most prelevant drug on the planet
479
Q

Which administration methods of cocaine have the longest time of action?

A
  • intranasal and oral -> slower onset but also a slower clearance/metabolsim
480
Q

What is the half-life of cocaine?

A
  • 20-90 minutes
481
Q

Why is the half-life of cocaine relatively short?

A
  • metabolsied rapidly by liver AND plasma cholinesterases -> is metabolised in th bloodstream
  • 75-90% of cocaine is broken down into inactive inert metabolites
482
Q

Why is cocaine so addictive?

A
  • speed of onset -> addiction is very closely correlated with how fast the effect comes on
  • speed of breakdown -> is a powerful euphoric high very quickly, and it is then lost very quickly too -> is a reason for continuously taking cocaine -> reinforce the effect
483
Q

What are the pharmacodynamics of a high dose of cocaine?

A
  • a local anaesthetic -> blocks sodium channelo within nerves -> reduced propagation of APs -> suppression of pain sensation
484
Q

What are the pharmacodynamics of a low dose of cocaine?

A
  • dopamine transporter takes molecules of dopamine from the synapse, and flips it back into the pre-synaptic cell -> cocaine blocks these dopamine transporters -> increase dopamine synaptic concentraion and therefore NA and serotonin are also affected
485
Q

Does cocaine influence dopamine affinity/efficacy for the dopamine receptor?

A
  • no -> cocaine blocks the reuptake protein and affinity and efficacy are related to interactions at the receptor
486
Q

Why must cocaine be a neutral/uncharged molecule to have it effects?

A
  • must diffue across the plasma membrane -> can only access sodium channels from the membrane/cytoplasmic side
487
Q

How does cocaine bring about euphoria?

A
  • cocaine goes straight to the nucleus accumbens and blocks the dopamine transporter -> results in many more dopamine molecules in the synapse, binding to dopamine receptors and stimulating euphoria
488
Q

Name some mild-moderate effects of cocaine on the brain/behaviour.

A

o mild-moderate -> euphoria and dysphora, heightened energy, motor excitement, restlessness, insomnia, increased libido, mild to moderate anorexia, anger and inflated self-esteem

489
Q

Name some severe effects of cocaine on the brain/behaviour.

A
  • irritability, hostility, anxiety, fear, withdrawl
  • extreme energy or exhaustion
  • total insomnia
  • compulsive motor sterotypies
  • incoherent speech
  • decreased libido
  • possible extreme violence
  • total anorexia
  • delusion of grandiosity
490
Q

Why does cocaine binging lead to a change from positive/reinforcing euphoria to negative/stereotypic effects?

A
  • reuptake is being blocked -> dopamine is not being reabsorbed back into the presynaptic neurone/is not being recycled back into the neurone frequently enough
  • binging on cocaine leads to reuptake being prevented -> dopamine will eventually be metabolised to a degree -> will not get any dopamine back into the neurone -> dopamine vesicles become fewer in number -> loss in the positive/reinforcing euphoria, and more negative/stereotypic effects
491
Q

What are the effects of cocaine of the cardiovascular system?

A

o effects on the sympathetic nervous system - due to increased catecholamines (predominantly NA)

  • increased NA -> increased work of the heart -> increased BP -> increased oxygen demand
  • increased NA (also inhibits NO) -> vasoconstriction -> decreased blood flow to the heart
  • sympathetic nervous system activates platelets -> platelet adherence
  • overall it leads to decreased oxygen supply despite increased demand -> ISCHAEMIA AND INFARCTION

o decreased sodium transport creating a local anaesthetic effect -> interferes with rhythm and left ventricular function

492
Q

How can cocaine overdose lead to hyperthermia?

A
  • causes increased agitation, locomotor activity and involuntary muscle contraction -> all lead to an increase in body temperature -> if in a hot environment htis can lead to hyperthermia
  • cocaine also increases the threshold of sweating and cutaneous vasoldilation three fold (must rise be 3 times as much) -> effects the bodies mechanisms of cooling down
493
Q

What methods of administration exist for nicotine?

A
  • cigarettes -> tar droplets are the transporter for the nicotine
  • nicotine spray
  • nicotine gum
  • nicotine patch

o last three are replacement therapies to wean off smoking

494
Q

State the bioavailability of nicotine in its different administartion routes.

A
  • nicotine spray 1mg (20-50%)
  • nicotine gum 2-4mg (50-70%)
  • cigarettes 9-17mg (20%)
  • nicotine patch 15-22mg/day (70%)
495
Q

Explain why nicotine has such a poor bioavailability in cigarettes.

A
  • cigarette smoke is quite acidic and nicotine has a high pKA of 7.9
  • means that nicotine is very poorly absorbed in the buccal cavity as within the smoke it is ionised and in the alveoli it depends on pH
496
Q

What is the idea behind nicotine replacemnet methods?

A
  • the problem with cigarettes is that you get a spike of nicotine -> this is the feeling that people want to replace, hence why they binge cigarettes
  • replacement methods don’t produce this high spike in nicotine because it is dangerous -> idea of replacement methods is to maintain a low level of nicotine in the blood to interfere with the craving to smoke
497
Q

What is the half-life of nicotine?

A
  • 1-4 hours -> therefore is a powerful reinforcing agent (like cocaine) as effects are lost quickly -> why people msoke lots of cigarettes a day
498
Q

Describe the metabolism/pharmacokinetics of nicotine.

A
  • vast majority (70-80%) of nicotine is broken down in the liver VERY QUICKLY by hepatic CYP2A6
  • major metabolite is cotinine -> it is an inactive and inert metabolism
499
Q

What are the pharmacodynamics of nicotine?

A
  • nicotine binds to the nicotinic acetylcholine receptor
  • NICOTINIC RECEPTORS ARE FOUND ALL THROUGHOUT THE AUTONOMIC NS ->, nicotine can interfere with most autonomic functions
500
Q

How does nicotine bring around euphoria?

A
  • is a stimulant -> acts directly on the neurone itself to stimulate it -> are lots of nACh receptors on the cell body of reward dopaminergic neurones
  • when the neurone is heavily stimulated, you get dopamine release in the nucleus accumbens -> feeling of reward
501
Q

What are the lung cancers and emphysema associated with smoking due to?

A
  • predominantly the volatile matter
502
Q

What are the CVS effects of nicotine?

A
  • nicotine leads to consistent autonomic stimulation (in the CNS and adrenal glands) -> increased HR and SV (sympathetic)
  • reduced blood flow/vasoconstriction of coronary and skin arterioles
  • vasodilation in the skeletal muscle arterioles
  • nicotine is pro-atherogenic -> has a negative effect on the lipid profile -> increases thromboxane (promotes platelet aggregation) and decreases NO

o can all build up to major CVS events such as MI (unlike cocaine its a build up and unlikely to get acute events)

o effects MAY be mitigated by exercise and diet

503
Q

What is nicotine effct on metabolic rate?

A
  • is a stimulant -> associated with weight loss
  • when people undergo smoking cessation they put on weight
504
Q

Describe nicotine impact on neurodegenerative disorders.

A

o Parkinson’s disease -> long-term nicotine use increases the number of brain cytochromes (CYPs) -> these cytochromes metabolise toxins (neurotoxins) -> suggests that chronic nicotine has a positive impact on Parkinson’s disease because it decreases neurotoxins that are present and contribute to disease development

o Alzheimer’s disease -> chronic nicotine use is associated with decreased beta-amyloid toxicity, and decreased amyloid precursor protein (APP) -> nicotine has a positive effect on the progression of Alzheimer’s

505
Q

How does caffeine bring about euphoria?

A
  • is a stimulant and should cause euphoric effects
  • adenosine activates adenosine receptors, which tend to decrease the euphoric effect -> adenosine therefore downregulates D1 function and decreases dopamine secretion
  • caffeine is an adenosine receptor antagonist -> increases dopamine release and enhances tissue response to dopamine
506
Q

Why are the euphoric effects of caffiene less obvious than other stimulants?

A
  • is orally administarted so there is a time delay, has a lower bioavailability and is released in small doses over a loner period of time
507
Q

How is absolute amount (of alcohol) calculated?

A

% ABV (alcohol by volume) x 0.78 = grams of alchol/100ml

508
Q

How is a unit of alcohol calculated?

A

% ABV x actual volume (ml) / 1000

  • 1 unit = 10ml or 8g of absolute alcohol
509
Q

What is considered to be a ‘low-risk’ amount of alcohol consumption?

A
  • 14 units or less per week
510
Q

How is binge drinking defined?

A
  • 8 or more units in one sitting
  • 18% of 16-24 do this on a weekly basis -> this number is falling
511
Q

Where is alcohol absorbed in the body?

A
  • 20% in the stomach
  • 80% in the intestines
512
Q

How does drinking on a full stomach influence blood alcohol levels?

A
  • alcohol is far more effectively absorbed from the small intestine -> therefore in order to absorb alcohol quickly, it needs to get into the intestine as quickly as possible
  • a method to speed up alcohol getting to the intestines is to drink on an empty stomach - fluid stimulates gastric emptying
  • drinking on a full stomach delays gastric emptying - houses the alcohol in the stomach, where it is far less effectively absorbed
513
Q

Describe the pharmacokinetics/metabolism of alcohol.

A

o 90% of alcohol dose is metabolised (85% of this is done in the first place it goes - the liver) the remaining 10% isn’t metabolised -> excreted as pure alcohol

o some excretion occurs through the lungs unchanged (e.g. breath test assesses amount of alcohol)

o 2 major groups of enzymes that predominate alcohol metabolism

  • alcohol dehydrogenase (75%)
  • mixed function oxidases (25%) – includes CYP450 -> its upregulation leads to alcohol tolerance
514
Q

Of the 90% of alcohol which is metabolised where is the remaining 15%, which isn’t metabolised in the liver, metabolised?

A
  • predominatly in the stomach -> does contain some alcohol dehydrogenase
  • females tend to have around 50% less alcohol dehydrogenase than males so this arm of metabolism is less effective
515
Q

If a man and a woman of similar height and weight equally share a bottle of wine, how will their blood alcohol levels compare?

A

o blood alcohol levels in the woman are higher

  • females metabolise it less effectively due to having less alcohol dehydrogenase in the stomach -> more alcohol gets across into the bloodstream
  • once in the blood, alcohol is less well diluted and distributed in females because men have more body water (59%) compared to females (50%)
516
Q

Describe the process/steps of alcohol metabolism.

A

o STEP 1: alcohol dehydrogenase and mixed fucntion oxidases convert alcohol into acetaldehyde -> acetaldehyde is a very toxic, so we don’t want it building up in the blood

o STEP 2: aldehyde dehydrogenase converts acetaldehyde to acetic acid which is inert and inactive -> this reaction occurs in the liver and the stomach

517
Q

What is a common regional problem with alcohol metabolism?

A
  • there is a very common genetic polymorphism in the aldehyde dehydrogenase enzyme -> very common in Asian populations (Asian flush)
  • results in ineffective metabolism of acetaldehyde -> builds up in the blood -> toxicity (nausea)
518
Q

What is disulfiram?

A
  • a drug that inhibits aldehyde dehydrogenase -> is used in alcohol aversion therapy
  • if given to alcoholics, whenever they drink alcohol, acetaldehyde builds up -> toxicity/nausea -> puts them off alcohol
519
Q

Is it possible to identify a pharmacological target for alcohol?

A
  • not really -> it has many targets
520
Q

Predict the affinity and efficacy for this target?

A
  • weak for both -> is not a very potent drug
  • large amount smust be consumed for it to have an effect
521
Q

What kind of drug is alcohol?

A
  • is primarily a depressant
  • CNS agitation/excitement may occur at a very low level -> self-confidence
  • CNS excitement is very dependent on your personality and environment (social/non-social)
522
Q

What systems/receptors does alcohol act on?

A
  • alcohol may directly affect the GABA receptor itself -> increase function - DIRECT EFFECT
  • is evidence for alcohol acting pre-synaptically, to increase allopregnenolone - INDIRECT EFFECT
  • alcohol may decrease NMDA receptor activation by binding to the receptor via allosteric modulation
  • alcohol may interfere with calcium channel opening -> impacts general neurotransmitter release
523
Q

How does alcohol bring about euphoria?

A
  • behaves a bit like heroin when it comes to euphoria
  • alcohol binds to the opioid receptors in the CNS -> switches off the GABA receptor via the opioid receptor (as opposed to cannabinoid receptors in cannabis) -> GABA is switched off -> increased firing rate of dopaminergic neurones -> euphoria
524
Q

What brain areas/tissue does alcohol interfere with, and therefore what process are impaired?

A
  • corpus collosum -> passes info from left brain (rules, logic) to right brain (impulse, feelings) and vice versa -> loss ofconnection occurs
  • hypothalamus -> loss of control of appetite, emotions, temperature, and pain sensation
  • reticular activating system -> loss of consciousness
  • hippocampus -> loss of memory
  • cerebellum -> lack of movement and coordination
  • basal ganglia -> lack of perception of time
525
Q

What are alcohols effects on the CVS?

A
  • vasodilation = acetaldehyde interferes with smooth muscle function in the arterioles -> calcium entry is impaired and prostaglandins are promoted -> cutaneous vasodilation -> redness -> is the principle behind Asian flush
  • tachycardia = depresses baroreceptor activity -> no stimulation of the PNS and a loss of inhibition on the SNS -> increased heart rate
526
Q

How does alcohol effect endocrine systems of the body?

A
  • increased diuresis (polyuria) is related to increased acetaldehyde -> acetaldehyde inhibits vasopressin production -> less AQPs in the collecting ducts -> LOSS OF FLUID
  • alcohol increases cortisol production (can produce ‘Cushing’s like’ syndrome) -> alcohol increases ACTH
  • has a negative effect on testosterone secretion (can have feminisation symptoms)
527
Q

How does chronic alcohol use cause effects on the brain?

A
  • alcoholics recieve less carbohydrates becuase large amount of calories comes from alcohol -> do not obtain enough thiamine -> neurones required thiamine to function -> brain regions with high metabolic demand become impaired
  • impaired metabolism -> reactive oxygen species are formed -> these can form mitochondrial damage -> apoptosis
528
Q

Describe the neurological disease heavily linked to chronic alcohol abuse.

A

o Wernicke-Korsakoff syndrome caused by thiamine deficiency

  • Wernicke’s encephalopathy (reversible part) -> 3rd ventricle & aqueduct -> cortical atrophy occurs/loss of cerebral white matter -> cauyses confusion, oculmotor symptoms and gait issues -> can progress to dementia (no longer reversible)
  • Korsakoff’s psychosis (irreversible) -> dorsomedial thalamus -> loss of memory due affects on areas of the deep brain - hippocampus
529
Q

How does alcohol affect the liver, what biological systems does it interfere with?

A

o a consequence of alcohol dehydrogenase is the depletion of NAD+ -> drinking too much alcohol leads to lots of NAD+ being used up -> NAD+ is needed for many stages in aerobic metabolism:

  • > pyruvate starts getting converted to lactate
  • > acetyl CoA starts getting converted to ketones
  • the liver is now exposed to lactic acid (acidosis) and ketones (ketosis) -> the ability of the liver to metabolise fats and lipids is impaired -> build up of fat in the liver and if the mixed function oxidase system is used a lot, free oxygen radicals are released into the blood
530
Q

Describe fatty liver.

A
  • acutely, after going on an alcohol binge, fatty liver is seen
  • is related to the inability to sufficiently metabolise (beta-oxidation) fats and lipids -> are stored in the liver as triacylglycerols in the liver
  • iis reversible, and not particularly dangerous in isolation
531
Q

What happens to the liver if alcohol is abused/binged on for a while?

A
  • the Krebs cycle is being permanently disrupted -> is a permanent generation of acidosis, ketosis and oxygen free radicals in the liver -> creates a very pro-inflammatory environment
532
Q

What is hepatitis?

A
  • the changes associated with chronic alcohol use - increased ketones, lactic acid, free radicals -> these all promote WBC influx
  • in hepatitis, we see both blood and hepatic cytokine changes -> increase in IL-6 and TNF alpha
  • hepatitis is still reversible if drinking is stopped
533
Q

Describe liver cirrhosis.

A
  • long term, if the inflammatory profile remains fibroblasts may begin to infiltrate the liver -> increase the amount of connective tissue being laid down, in place of active liver tissue -> decreased hepatocyte regeneration, increased fibroblasts and decreased active liver tissue
  • at some point, there is so little left of active liver tissue, that a liver transport is required -> liver failure
534
Q

What are the possible benficial effects of alcohol?

A
  • is evidence that low levels of alcohol semi-regularly protects against CVS -> e.g. a glass or red wine may have a protective effect against coronary artery disease -> though to be the polyphenols that are beneficial
  • the amounts that have to be drunk are likely to have negative affects to other systems, mainly the liver and brain
535
Q

What are the effects of alcohol on the gut?

A
  • ulceration is common in chronic alcoholics -> strongly associated with acetaldehyde
  • stomach cancer risk increases in alcoholics -> alcohol migh have carcinogenic effects
536
Q

Describe the causes for the components of a hangover.

A
  • nausea = alcohol is an irritant -> vagus -> vomiting centre
  • headache = vasodilation and acetaldehyde build up
  • fatigue = 1. sleep deprivation and 2. ‘Rebound’ - poor quality
  • restlessness and muscle tremors =‘Rebound’
  • polyuria and polydipsia = ↓ ADH secretion
537
Q

What is the classical presentation of a DVT?

A
  • immobile for 3 weeks after major surgery or long flight
  • calf of right leg is swollen and collateral superficial veins are present
  • palpation reveals localised tenderness and pitting oedema
  • normal BP, HR, RR and SpO2
538
Q

What is a two-level Wells score?

A
  • a score/test which predicts the likelihood of a DVT
539
Q

What would confirm the diagnosis of a DVT?

A
  • positive D-dimer test -> suggests more than confirms
  • ultrasound -> confirms
540
Q

What is the treatment of DVT?

A
  • positive D-dimer test suggests diagnosis of deep-vein thrombosis (DVT) -> given interim treatment with parenteral anticoagulant -> prevents anything worse from happening while further tests are ran to confirm the diagnosis
  • ultrasound scan confirms DVT -> given maintenance treatment with oral anticoagulant -> can be continued after discharged
541
Q

What drugs can be used as maintanence treatment for DVTs?

A
  • rivaroxaban -> MOST COMMON
  • apixaban
  • warfarin -> MOST COMMON
  • dabigatran

o work as maintanence because they all have oral administration

542
Q

How does dabigatran work?

A
  • inhibits factor IIa
543
Q

Why is dabigatran rarely used clinically?

A
  • can cause severe GI bleeding whihc can often lead to death
544
Q

How do rivaroxaban and apixaban work?

A
  • inhibit factor Xa
545
Q

What is the mechnism of action of warfarin?

A
  • vitamin K antagonist -> required for generation of factors II, VII, IX and X
546
Q

What drugs are usually used as interim treatment for DVTs?

A
  • heparin (IV or SC)
  • low-molecular weight heparins (SC) -> dalteparin -> MOST COMMON

o both have parenternal administrations

547
Q

How does the mechanism of heparin and low-molecular weight heparins differ?

A
  • both activate anti-thrombin (AT-III)
  • it is thought that low-molecular weight heparins may also affect Xa directly
548
Q

What are the advantages of rivaroxaban and warfarin?

A
  • warfarin is cheaper but has lots of drug interactions so need monitoring
  • rivaroxaban is better but effects cannot be reversed
549
Q

What is the major factor driving the treatment of DVTs?

A
  • trying to prevent pulmonary embolisms
550
Q

What are the presentations of a PE?

A
  • chest pain
  • dyspnoea
  • tachypnoea
  • low BP and SpO2 with a high HR
551
Q

How is the diagnosis of a PE made?

A
  • patient history - any known DVT etc
  • multiple-detector computed tomographic pulmonary angiography (CTPA)
552
Q

How does the pharmalogical treatment for DVT and PEs differ?

A
  • is mostly the same
  • interim will be given dalteparin or heparin -> heparin is given for PE and not for DVT because they are more severe -> heparin works faster but has a few more side effects
  • followed up with maintanence which is rivaroxaban or warfarin
553
Q

What is Virchow’s Triad?

A

o factors/elements that increase risk of thrombosis

  • rate of blood flow -> flow is slow/ stagnating -> no replenishment of anticoagulant factors and balance adjusted in favour of coagulation
  • consistency of blood -> natural imbalance between procoagulation and anticoagulation factors
  • blood vessel wall integrity -> damaged endothelia -> blood exposed to procoagulation factors
554
Q

What is the difference between NSTEMI and STEMI?

A
  • Non-ST elevated myocardial infarction (MI) -> white thrombus -> partially occluded coronary artery -> ischaemia
  • ST elevated myocardial infarction -> white thrombus -> fully occluded coronary artery -> not just reduced oxygen to cardiac muscle -> is now lack of oxygen -> leads to cell death and necrosis
555
Q

What is the presentation of a NSTEMI?

A
  • history of hypertension and hyperlipidaemia
  • shortness of breath, sweating, dizziness & chest pain
  • normal BP and HR with high RR and low SpO2
556
Q

What tests would be ran, and what would be the positive results if an NSTEMI was suspected?

A
  • no changes on the ECG
  • elevated troponin in the blood
557
Q

What is the difference between white and red thrombus?

A
  • white = occur in arteries, particularly coronary arteries -> are white due to foam cells caused by macrophages and are associated with atherosclerosis
  • red = form in deep veins (DVTs)
558
Q

What is the pharmalogical treatment of a NSTEMI?

A

o anti-platelet therapy

  • aspirin
  • clopidogrel
559
Q

What differs in the pharmalogical treatment of a NSTEMI and a STEMI?

A
  • NSTEMI = antiplatelets
  • STEMI = antiplatelets + thrombolytics
560
Q

What are the risk factors for acute coronary syndromes?

A
  • damage to endothelium
  • atheroma formation
  • platelet aggregation

o mainly associated with diet -> high fat/high LDL

561
Q

What is the role of thrombin (factor IIa) at a cellular level?

A
  • activates platelets -> changes there shape so they become sticky and attach to other platelets -> amplifies clot formation
562
Q

What is the mechanism of thrombin at a molecular level?

A
  1. thrombin binds to protease-activated receptor (PAR) on platelet surface -> PAR activation causes a rise in intracellular Ca2+ -> exocytosis of ADP from dense granules
  2. ADP activates P2Y12 receptors -> platelet activation/aggregation via auto/paracrine action
  3. PAR or P2Y12 activation liberates arachidonic acid (AA) -> Cyclo-oxygenase (COX) becomes activated and generates thromboxane A2 (TXA2) from AA
  4. TXA2 activation -> expression of GPIIb/IIIa integrin receptor on platelet surface

GPIIb/IIIa -> involved in platelet aggregation

563
Q

What is clopidogrel?

A
  • an oral antiplatelet drug
  • is an ADP (P2Y12) receptor antagonsit -> prevents platelet activation
564
Q

What is aspirin?

A
  • an oral antiplatelet drug
  • an irreversible COX-1 inhibitor -> prevents TXA2 formation -> lack of GPIIb expression -> no aggregation
565
Q

What are the actions of anti-thrombin?

A
  • inhibits factors IIa and Xa and to a lesser extent IXa and XIa
566
Q

What does the d-dimer test detect?

A
  • fibrin degradation products
567
Q

What is the diagnosis of someone presenting with a severe headache, dizziness, loss of coordination and numbness in the face, arms and legs?

A
  • stroke
568
Q

What investigation would differentiate between an ischaemic and a haemorrhagic stroke?

A
  • a CT scan
  • very important to know -> thrombolytic treatment for a ischamic stroke will kill someone with a haemorrhagic stroke
569
Q

What is the pharmacological treatment for an ischamic stroke?

A
  • thrombolytic therapy -> alteplase (tPA) -> given ASAP
570
Q

How does a clot which goes on to cause a stroke usually form?

A
  • clot forms within one of the atria of heart -> more common if someone has atrial fibrillation or flutter
  • clot embolises -> blocks one of the cerebral arteries -> necrosis
  • clot causing strokes occurs differently to DVT and atherosclerotic plaque
571
Q

What causes the formation of fibrin strands?

A
  • factor IIa binds to fibrinogen and converts it to fibrin strands
572
Q

What is the mechanism of thrombolytics?

A
  • convert plasminogen -> plasmin -> protease degrades fibrin
  • once the fibrin has broken down the clot will dissolve -> restores blood flow
573
Q

Why are thrombolytics not used to treat DVT or PE when they are very effective are breaking clots?

A
  • extremely dangerous drugs -> causes excessive bleeding
  • only used in emergency setting for ischaemic stroke and possible STEMI -> evidence suggests it is only really worth it in stroke patients over 80
574
Q

What is the good and bad lipoproteins?

A
  • good = HDL
  • bad = LDL
575
Q

What is the exogenous pathway of lipid metabolism?

A
  1. lipids enter the blood in two ways, one of which is absorption of fats from the diet -> amount of the cholesterol that enters the circulation from the diet is very small (other lipids (such as triglycerides) are certainly absorbed)
  2. triglycerides are absorbed as chylomicrons into the blood -> if you ingest a very fatty meal and then take a blood sample, chylomicrons are present in the plasma
  3. chylomicrons are broken down by lipases, into remnants -> some of this ends up as atheroma, in the vascular wall
576
Q

What is the endogenous pathway of lipid metabolism?

A
  • is a pathway in which the liver generates different lipoproteins, which are then broken down and converted -> some of the lipoproteins end up with the LDL receptor -> this endogenous pathway forms around 80% of the cholesterol in the body
  • most circulating lipids are endogenous
577
Q

Describe reverse cholesterol transport.

A

o is a process where cholesterol is taken OUT of blood vessels and foam cells

  • foam cells -> smooth muscle macrophages that are full of lipid (including cholesterol)
  • is a transformation of HDL (beneficial) back to LDL -> by cholesteryl ester transfer protein (CETP)
  • way to BLOCK THIS PROCESS exsists -> can block CETP and thus increase HDL and reduce LDL
578
Q

What are the stages of atherosclerosis?

A
  1. endothelial dysfunction
  2. fatty streak formation
  3. formation of the atherosclerotic plaque
579
Q

Describe endothelial dysfunction in atherosclerosis.

A
  • endothelium becomes dysfunctional -> problem because the endothelium is responsible for producing a lot of mediators
  • characterised by a series of early changes that precede lesion formation -> changes include greater permeability of the endothelium, up-regulation of leucocytes, endothelial adhesion molecules and migration of leucocytes into the artery wall
580
Q

Describe fatty streak formation in atherosclerosis.

A
  • fatty streak is the earliest recognisable lesion of atherosclerosis and is caused by the aggregation of lipid-rich foam cells -> derived from macrophages and T lymphocytes within the tunica intima
  • later on the lesions will also include smooth muscle cells
  • are usually formed in the direction of blood flow
  • most fatty streaks don’t actually develop into serious atherosclerosis, but it is an early stage and begins very early in human life
581
Q

Describe the formation of atherosclerotic plaques.

A
  • results from the death and rupture of the foam cells in the fatty streak
  • formation of necrotic core occurs
  • migration of smooth muscle cells into the intima and laying down collagen fibres results in the formation of a protective fibrous cap over the lipid core -> fibrous cap is extremely important because it separates the highly thrombogenic lipid-rich core from circulating platelets and coagulation factors
  • stable atherosclerotic plaques are characterised by a necrotic lipid core covered by a thick vascular smooth muscle-rich fibrous plaque
582
Q

What do complicated lesions often contain?

A
  • calcium
  • coronary artery disease can be detected by doing a CT scan of the heart -> detects any calcium -> believed that the more calcium you have in the plaque, the more likely you are to be symptomatic
  • higher calcium = greater risk of CVD
583
Q

What are remnant lipids?

A
  • remnant lipids come from the chylomicrons (part of the breakdown process of chylomicrons)
  • are important because they are quite atherogenic -> lots of remnant lipoproteins in the blood = higher risk of CHD compared to blood with fewer remnants
584
Q

Name some remnant lipids.

A
  • VLDL
  • chylomicron remnant
  • IDL
585
Q

Describe vunerable atherosclerotic plaques.

A

-vulnerable plaques are characterised by thin fibrous caps, a core rich in lipid and macrophages and less evidence of smooth muscle proliferation -> stable plaque has a very thick fibrous cap

586
Q

What are plaque ruptures associated with?

A
  • greater influx and activation of macrophages, accompanied by the release of matrix metalloproteinases that are involved in the breakdown of collagen
587
Q

Explain why stable and vunerable plaques are symptomatic and asymptomatic?

A
  • stable plaque = will probably have symptoms due to the thick cap obstructing blood flow to the heart -> PAIN
  • vulnerable plaque = only a very thin wall separating the lumen and lipid core -> no pain -> a surge in BP could lead to the breaking down of the thin fibrous cap-> THROMBOSIS
588
Q

What is LDL cholesterol linked with?

A
  • strongly associated with atherosclerosis and CHD events -> 10% increase results in a 20% increase in CHD risk
  • modified by other risk factors (all these things will aggregate the effects of LDL cholesterol) -> low HDL cholesterol, smoking, hypertension, diabetes
589
Q

Describe HDL cholesterol.

A
  • HDL cholesterol is protective for risk of atherosclerosis and CHD (promotes reverse cholesterol transport) -> lower the HDL cholesterol level, the higher the risk for atherosclerosis and CHD
  • HDL cholesterol tends to be low when triglycerides are high
590
Q

What lowers cholesterol?

A
  • HDL cholesterol is lowered by smoking, obesity and physical inactivity
591
Q

How do statins lower elevated LDL?

A

o act on the mevalonate pathway to inhibit HMG-CoA reductase

  • mevalonate pathway produces cholesterol as well as geranyl pyrophosphate and faeresyl pyrophospahe -> are extremely important in the cell function -> involved in the modification and activation of proteins
  • cell can’t function properly without these lipids but it can function properly without cholesterol -> cell must make these small lipids itself
  • by blocking cholesterol synthesis in the liver then the liver cells respond by making more LDL receptors -> bind to circulating LDL and lower it
592
Q

What is the Rule of 6?

A

regards statins -> doubling the dose of any of the statins, only achieves a 6% reduction in LDL cholesterol

  • applies to all statins and all doses -> effectiveness of statin treatment depends on the level of cholesterol that you achieve
593
Q

What is meant by the pleiotropic effects of statins?

A
  • effects that are not directly related to the reduction in cholesterol
594
Q

What are the pleiotropic effects of statins?

A
  • less platelet activation
  • lower thrombotic effect
  • increase plaque stability
  • reduced vascular inflammation
  • less SMC hypertrophy
  • reduced endothelial dysfunction
  • less SMC proliferation
  • reduced vasoconstriction

o some believe statins should be used when people have high CRP

595
Q

What are fibrates?

A
  • a lipid lower drug -> reduce plasma fatty acids and triglycerides
  • also reduces inflammation
596
Q

What is the main mechanism of fibrates?

A
  • activation of peroxisome proliferator activated receptors (PPAR) -> th ealpha receptors in particular
597
Q

When are fibrates often used?

A
  • diabetes with high glycerides
598
Q

Describe nicotinic acid.

A
  • lowers LDL, it increases HDL, it lowers triglycerides and increases fibrinolytic activity (clot dissolution) -> everything you could possibly want to lower CVD risk
  • isn’t used very much in clinical practice -> not well tolerated -> causes flushing, so it isn’t taken by patients
599
Q

Describe ezetimibe.

A
  • inhibits cholesterol absorption
  • reduce LDL cholesterol a reasonable amount (15-20%)
  • useful as an additional treatment to statins -> because of the rule of 6, you want to find something else that can be added to increase the effectiveness of statins at lowering cholesterol without just increasing the dose of the statin -> ezetimibe decreases LDL further when given with a statin
600
Q

Describe CETP inhibitors.

A
  • had a positive effect in increasing HDL and decreasing LDL -> however, when they expanded the clinical trial they found that it was ACTUALLY KILLING people
  • were ‘off-target’ (could’nt have predicted the effect) ADVERSE effects of torcetrapib (CETP inhibitor) -> might be due to activation of aldosterone synthesis leading to increased blood pressure
601
Q

Describe PCSK9 inhibitors.

A
  • PCSK9 is an inhibitor of the LDL receptor (breaks down the receptor) so it stops the LDL from reaching the LDL receptor

LDLR and PCSK9 gene expression are coinduced by statins

As a result, PCSK9 inhibition enhances the lipid-lowering effect statins

Monoclonal antibodies are the most advancedPCSK9 inhibitors currently in development

602
Q

What groups of patients gain the greatest effects from PCSK9?

A
  • familial hypercholestroleamia patients
603
Q

What are the properties of NSAIDs?

A

o analgesic properties -> relief of mild-to-moderate pain (toothache, headache, backache), post-operative pain (opiate sparing so people can take less opiates) and dysmenorrhea (menstrual pain)

o anti-pyretic -> reduction of fever -> influenza

o anti-inflammatory -> reduction of inflammation -> Rheumatoid arthritis, osteoarthritis (plus other musculoskeletal inflammation), soft tissue injuries (strains and sprains) and gout

604
Q

What is the mechanism of NSAIDs?

A

o inhibit COX enzymes -> inhibits prostanoid synthesis (e.g. prostaglandins, thromboxane, prostacyclin) -> inhibits inflammatory mediators

  • none of the factors are stored so NSAID effects are fast
605
Q

Describe prostanoid receptors.

A
  • are 10 known receptors and 5 prostanoids (agonists)
  • prostanoids have both G protein-dependent & independent effects (not all actions G-protein mediated) -> effects are extremely complex
  • side effects are both physiological and pro-inflammatory
606
Q

What is PGE2 and what receptors can it activate?

A
  • a prostanoid
  • can activate EP1, EP2, EP3 and EP4 -> causes G-protein (cAMP dependent and independent downstream actions)
607
Q

What are the unwanted actions of PGE2?

A
  • increased pain perception
  • increased body temperature
  • acute inflammatory response
  • immune responses – both positive and negative
  • tumorigenesis
  • inhibition of apoptosis
608
Q

What are the possible mechanism for why prostanoids lower pain threshold?

A
  • greater activation of P2X3 receptors
  • EP1 receptors and EP4 receptors (EP4 in periphery and spine)
  • endocannabinoids (neuromodulators in thalamus, spine and periphery)
  • decreasing beta-endorphin in spine

o not mutually exclusive

609
Q

How does PGE2 increase body temperature?

A
  • stimulates hypothalamic neurones initiating a rise in body temperature
610
Q

How does PGE2 bring about inflammation?

A
  • VERY complex process involving PGE2-EP3 signalling
  • EP3 works through multiple mechanism
611
Q

Name some of the desirable physiological actions of PGE2 (and other prostanoids)?

A
  • bronchodilation
  • renal salt and water homeostasis
  • gastro-protection
  • vaso-regulation (dilation and constriction depending on receptor activated)
612
Q

Why should asthamtics not take NSAIDs?

A
  • COX inhibition favours leukotrienes production -> these cause bronchoconstriction -> worsening of asthma symptoms
613
Q

How does PGE2 and COX regulate water homeostasis?

A
  • PGE2 = involved in increasing renal blood flow
  • COX 1 and 2 are involved in the production of products at multiple points in the nephron
614
Q

How can NSAIDs cause renal toxicity?

A
  • constriction of afferent renal arteriole
  • reduction in renal artery flow
  • reduced glomerular filtration rate
615
Q

What is the role of PGE2 in the stomach?

A
  • down-regulates HCl secretion -> reduces irritaion
  • stimulates mucus and bicarbonate secretion -> increases production of protective layer
616
Q

What gastric effects do NSAIDs have?

A
  • increased gastric and duodenal ulceration -> high load on the stomach when taken orally
617
Q

What are the CVS effects of NSAIDs?

A

o vasoconstriction

o salt and water retention

o reduce the effects of anti-hypertensive drugs

  • becoming acknowledged that use of NSAIDs poses a risk of hypertension, MI and stroke
618
Q

Why are COX-2 inhibitors more widely in use?

A
  • they do lower stomach and duodenal ulceration massively -> however, pose a higher risk of CVD
619
Q

Describe the unwanted effects of different NSAID target sites.

A
620
Q

Describe the risk vs rewards of NSAID useage.

A
  • analgesic use -> usually occasional and relatively low risk of side effects
  • anti-inflammatory use -> often sustained, higher doses and relatively high risk of side effects
621
Q

Name some strategies for limiting the GI effects of NSAIDs.

A
  • topical application
  • minimise NSAID use in patients with history of GI ulceration and treat H pylori if present (trigger factor for ulcers)
  • if NSAIDs are essential administer with omeprazole or other proton pump inhibitor
  • minimise NSAID use in patients with other risk factors and reduce risk factors where possible -> e.g. alcohol consumption or anticoagulant or glucocorticoid steroid use
622
Q

What is unique about aspirin compared to other NSAIDs?

A

o irreversibly binds to COX-1 enzymes

  • works by acetylating active sites of both COX enzymes -> mainly COX-1
623
Q

What are the effects of aspirin on platelet aggregation?

A
  • inhibits thromboxane (made by COX-1) more than PGI2 (made by COX-1 and 2)
  • platelts can’t make more thromboxane as they don’t have a nucleus -> required to make more enzymes as others are permenantly inhibited
  • reduced platelet aggregation
624
Q

What is the difference in affects between a high and low doses of aspirin?

A
  • low dose = platelets do not have a nucleus -> NO RE-SYNTHESIS of COX-1 in platelets à OVERALL REDUCED AGGREGATION OF PLATELETS
  • high dose = BARELY ANY ACTION ON PLATELET AGGREGATION -> endothelial cell COS enzymes will be in a permanent state of inhibition
625
Q

What are the major side effects of aspirin?

A
  • gastric irritation and ulceration
  • bronchospasm in sensitive asthmatics
  • prolonged bleeding times
  • nephrotoxicity
  • side effects likely with aspirin because it inhibits COX covalently, not because it is selective for COX-1
626
Q

What is the mechanism of paracetamol?

A
  • probably central AND peripheral mechanism
  • could be COX-3, cannabinoid receptors, endogenous opioids interaction, 5HT and adenosine receptor interaction
627
Q

What is the lethal cause of paracetamlo overdose?

A

o overdose can cause irreversible liver failure

  • paracetamol is normally metabolised by the cytochrome P450 complex
  • its metabolsim produces a toxic metabolite (NAPQI), which is highly reactive/electrophile ->at a therapeutic doset the NAPQI is produced is very small amounts and will be mopped up by glutathione
  • if glutathione is depleted (saturated with NAPQI) the metabolite oxidises thiol groups of key hepatic enzymes and causes cell death
628
Q

What is the antidote for paracetamole poisoning?

A
  • compounds with thiol groups -> usually intravenous acetylcysteine, occasionally oral methionine
  • if done early it can prevent liver damage
629
Q

Aspirin is unique amongst NSAIDS because:

a) It has no effect on COX-1
b) It has no effect on COX-2
c) It binds covalently to COX enzymes
d) It binds covalently to TP receptors
e) It causes gastric ulceration

A
  • c
630
Q

Inhibition of which enzyme will reduce platelet aggregation with fewest side effects?

a) COX-1
b) COX-2
c) Prostacyclin synthase
d) Prostaglandin E synthase
e) Thromboxane A2 synthase

A
  • e -> thromboxanes causes platelet aggregation, but not much else
631
Q

Assertion: Inhibition of PGI2 synthesis by low dose aspirin decreases the risk of stroke

Reason: Decreased PGI2 reduces platelet aggregation

a) Assertion true, reason true and explains assertion
b) Assertion true, reason true but does not explain assertion
c) Assertion true, reason false
d) Assertion false, reason true
e) Assertion false, reason false

A
  • e -> assertion false, reason false
  • synthesis of PGI2 (prostacyclin) is inhibited by low dose aspirin, but it is not this action which decreases the risk of stroke -> PGI2 actually reduces platelet aggregation -> stroke risk decrease is due to the inhibition of thromboxane synthesis
632
Q

What is the difference between an opiate and an opioid?

A
  • opiate = an alkaloid derived from the opium poppy
  • opioid = anything that has opiate like function, commonly synthetic
633
Q

What are the 4 (most common) natural opiates?

A
  • morphine
  • codeine
  • thebaine
  • papaverine
634
Q

Describe the structure-activity relationship of opiates, focus on morphine.

A
  • tertiary nitrogen = analgesia by permits receptor anchoring -> extending the side chain to 3+ carbons generates an antagonist
  • tertiary nitrogen allows the molecule to bind the receptor by giving it affinity and permits receptor anchoring -> side chain attached to the nitrogen determines the efficacy
  • 2 alcohol groups have a role in binding -> not as much as the tertiary nitrogen
635
Q

How do the structures of heroin and codeine differ to that of morphines?

A
  • heroin = diacetylmorphine
  • codeine = methylmorphine
636
Q

What is the significance of the structures of heroin and codeine?

A
  • they are both prodrugs of morphine which have increased lipid solubility
  • codeine -> morphine occurs outside the brain
  • heroin -> morphine occurs inside the brain
637
Q

How are opiods usually administarted?

A
  • orally
  • IV
638
Q

Describe the bioavailability of opioids.

A
  • opioids are weak bases -> pKa > 8 -> will be very ionised in the stomach -> poor absorption
  • once they enter the small intestines, they are well absorbed
  • opioids are heavily metabolised in the liver (first pass) -> this impacts their bioavailability -> all of the above are missed by giving via IV
  • pH of the blood is around 7.4, opioids are largely ionised in the blood (80%) -> only 20% of opioids remain unionised -> unionised form accesses tissues readily because it is more lipophilic
639
Q

What is the general rule about potency of opiods and their lipid solubility?

A

o more lipid soluble = more potent

  • exception to this is codeine
640
Q

Describe the metabolism of morphine.

A
  • morphine produces active metabolites -> morphine-6-glucoronide is the most potent active metabolite of morphine (10% of the blood levels of morphine at any one time) -> active metabolites are less likely to cause the negative (e.g. repsiratory depression) than morphine -> individuals who don’t metabolise morphine very well are more likely to have negative side effects
641
Q

Why is methadone often used to wean patients of heroin?

A
  • cleared very slowly -> is very lipid soluble so builds up in fatty tissue
  • effects therefore last long so it isnt as addictive
642
Q

Why is codeine less potent than morphine despite being more lipid soluble?

A
  • codeine is metabolised by two cytochrome P450 enzymes

o one enzyme activates codeine (CYP2D6) to morphine -> 5-10% of the codeine is converted via this route

o the other enzyme (CYP3A4) deactivates codeine

o poor metabolism -> codeien has little effect

  • is a very common polymorphisms in CYP2D6 enzyme -> people may have good metabolism of codeine -> more powerful effect (more morphine)
643
Q

What do opioids bind to?

A
  • opioid receptors
644
Q

What endogenous compunds act on opioid receptors?

A
  • endorphins
  • enkephalins
  • dynorphins/neoendorphins
645
Q

Name the 3 main opiate receptors.

A
  • Mu
  • delta
  • kappa
646
Q

Describe Mu opioid receptors.

A
  • are acted on by endorphins, heroin and morphine
  • loacted in the in the cerebellum, caudate nucleus, nucleus accumbens and PAG
  • important in pain and sensation
647
Q

Describe delta opioid receptors.

A
  • are acted on by enkephalins
  • located in the nucleus accumbens, cerebral cortex, hippocampus and putamen
  • important in motor and cognitive function
648
Q

Describe kappa opioid receptors.

A
  • acted on by dynorphins/neoendorphins
  • located in the hypothalamus, putamen and caudate
  • important neuroendocrine role (via the hypothalamus)
649
Q

What kind of drugs are opioids?

A
  • depressants
650
Q

What are the opiate receptors cellular mechanisms of action?

A
  • hyperpolarise cells -> hyperpolarisation (increase K+ efflux) -> nerves can’t fire
  • reduce the Ca2+ inward current ->massively impacts exocytosis and NT release
  • decrease adenylate cyclase activity within the cell
651
Q

What are the effects of opioids? Indicate the side effects specifically.

A
  • analgesia
  • euphoria
  • depression of cough centre (anti-tussive)
    • depression of respiration (medulla)*
    • stimulation of chemoreceptor trigger zone (nausea/vomiting)*
    • pupillary constriction*
    • GI effects*
  • (italics = side effects)*
652
Q

Describe the pain pathway.

A
  • pain is sensed peripherally, and is recognised by sensory neurones -> relay this information to the spinal cord -> spinothalamic neurones relay this information to the thalamus -> doesn’t process the information, but it directs it to the cortex
  • thalamus also directly activates the descending pain tolerance pathway -> simultaneously, the cortex has an additional impact on the pain tolerance pathway
  • the PAG region is the integrating centre for the pain tolerance pathway -> receives all information and determines output, it relays to the NRM (effector arm)
    • thalamus switches the PAG on automatically -> not long after, the cortex modulates this signal*
  • the NRM, descending neurones leave to suppress the feelings of pain that are reaching the brain
  • the NRPG is an auto-feedback element of the brain -> is away of switching on pain tolerance without any higher centre functioning at all -> isn’t as powerful as PAG, but is still an important
    • most of the information goes to the thalamus, but some filters through the NRPG and automatically switches on pain tolerance*
653
Q

What is the role of the thalamus in the pain pathway?

A
  • activates the descending pain tolerance pathway -> called PAG
  • directs information to the cortex
654
Q

What is the role of the NRM is pain?

A
  • effector arm of the PAG
655
Q

What is the role of the NRPG in pain?

A
  • auto-feedback element of the brain -> can switch on pain tolerance without higher centre involvement
656
Q

How does the hypothalamus affect pain?

A
  • hypothalamus constantly gives information to the rest of the brain about current state of health
  • in a poorer state of health, you are likely to be more sensitive to pain -> want sick people to be in pain so that they don’t expend their energy doing activities
657
Q

How does the locus coreleus affect pain?

A
  • is the sympathetic nervous system effector -> is largely independent of the analgesia system
  • if your stress pathway is activated, the LC is active -> automatic signal to dampen down pain
  • when you injure yourself playing sport, it doesn’t hurt very much at the moment in time -> makes sense in terms of flight or fight
  • if the SNS is active it suppresses pain information from the periphery to the brain
658
Q

How is pain modulated in the dorsal horn?

A
  • certain amount of signal comes down and reaches the dorsal horn, to suppress information moving from periphery to brain
  • substantia gelatinosa is a mini brain in the spinal cord -> processing and modification of the signal takes place
659
Q

How do opioids suppress pain?

A
  • act in the dorsal horn to prevent information relay from periphery to the spinothalamic tract (DEPRESSANT)
  • also act in the NRPG, PAg and NRM pathways -> mechanism here is disinhibition (i.e. switching off GABA) -> suppress the normal inhibitory signal in the pink regions -> more pain tolerance
660
Q

How do opioids cause euphoria?

A
  • opioids enter the brain and bind to their receptors -> depress the firing rate of GABA interneurons (much like cannabis)
  • means less inhibition on dopaminergic neurone -> increases its firing rate -> increased dopamine secretion into the nucleus accumbens -> feelings of reward
  • it is this mechanism that gives you a high after exercise
661
Q

Describe the cough pathway.

A
662
Q

How do opioids act as an anti-tussive (anti-cough)?

A

o ACh and neurokinin (NK) are the two NTs involved in coughing -> C-fibres send information up to the cough centre, via the vagus nerve

o 5-HT1A receptors are important -> have negative feedback receptors for serotonin -> serotonin is anti-cough

o opioids decrease the firing rate of C-fibres -> less information relay to the brain from irritation in the airways

  • also act in the cough centre, with a direct depressant effect on the cough centre, and inhibit 5-HT1A feedback receptors -> opioids prevent the 5-HT1A receptors from activating -> more serotonin available in the cough centre (anti-cough effect)
663
Q

How do opioids cause respiratory depression?

A
  • 2 important aspects of respiratory rhythm generation: central chemoreceptors and the Pre-Botzinger complex
  • central chemoreceptors are sensing the blood for carbon dioxide -> information is relayed to the medullary control centre -> provides a tonic drive (what level of respiration needed)
  • Pre-Botzinger complex is required for breathing within the medulla -> particularly inspiration
  • opioids inhibit both the chemoreceptors and the pre-Botzinger complex -> seen in overdose -> what kills people
664
Q

How do opioids cause nausea?

A
  • via a direct effect at the chemoreceptor trigger zone
  • GABA keeps the CTZ suppressed -> opioids switch this inhibition off -> CTZ signals to the medullary vomiting centre, and you feel nauseous
665
Q

Define miosis.

A
  • excessive constriction of the pupil
666
Q

How do opioids cause miosis?

A
  • opioid receptors are expressed in the Edinger-Westphal nucleus and therefore influence miosis
  • optic nerve is the sensory nerve that relays information to the pretectal nucleus -> signals are transmitted to parasympathetic nerves in the EWN -> opioids can influence the parasympathetic nerves here -> GABA suppress the parasympathetic nerves -> opioids switch inhibition off -> miosis
667
Q

If someone enters A&E passed out and has constricted pupils what will they have overdosed on?

A
  • an opioid -> heroin
  • normally when passed out pupils dilate due to lower brain activity but not with opioids
668
Q

How do opioids cause constipation?

A
  • opioid receptors everywhere within the enteric nervous system
  • opioids activation leads to less motility and fewer secretions
669
Q

How do opioids cause urticaria?

A
  • is NOT an allergic response
  • results from a direct interaction of opioids with mast cells -> causes histamine release -> seems to involve PKA, and doesn’t seem to involve opioid receptors
  • is caused by morphine and codeine mainly due to an OH in carbon position 6
670
Q

How do you become tolerant to opioids?

A
  • nothing to do with the liver and pharmacokinetics
  • comes about at the level of tissues -> RECEPTOR INTERNALISATION
  • arrestin is a very important protein in terms of driving receptor internalisation -> constant recycling
  • if opioids are constantly available, cells are suppressed -> opioid receptors are withdrawn from the cell -> arrestin protein concentration increases -> increased internalisation -> TOLERANCE
671
Q

Why are opioids so heavily associated with dependency?

A
  • withdrawal is associated with psychological craving and also physical withdrawal (flu-like)
  • cells try to upregulate activity to compensate for the opioids (increase in adenylate cyclase) -> once opioids are removed, adenylate cyclase is overactive
672
Q

What are the symptoms of a opioid/heroin overdose?

A
  • coma
  • respiratory depression
  • pin-point pupils
  • hypotension
673
Q

What is the treatment for an opioid overdose?

A
  • naloxene -> is an opioid antagonist
  • has 3 carbons chain on the tertiary nitrogen
674
Q

What are the 2 main forms of IBD?

A

o ulcerative colitis

o Crohn’s disease

  • 10% are indeterminate - features of both
675
Q

What are the risk factors for IBD?

A

o genetics

  • > 201 loci identified
  • > white, western europeans seem the most susceptible

o environmental

-> most important are smoking, diet and microbiome

676
Q

Describe Crohn’s disease as an autoimmune disease.

A
  • Th1-mediated e.g. IFN-gamma, TNF-alpha, IL-17, IL-23
  • florid T cell expansion
  • defective T cell apoptosis
677
Q

Describe ulcerative colitis as an autoimmune disease.

A
  • Th2- mediated .g. IL-5, IL-13
  • limited clonal expansion
  • normal T cell apoptosis
678
Q

What layers of the gut are affected by Crohn’s and ulcerative colitis?

A
  • Crohn’s = all layers
  • ulcerative colitis = mucosa and submucosa
679
Q

What regions of the gut are affected by Crohn’s and ulcerative colitis?

A
  • Crohn’s = any part of the GI
  • ulcerative colitis = rectum, spreading proximally
680
Q

Describe the inflamed areas in Crohn’s and ulcerative colitis?

A
  • Crohn’s = patchy
  • ulcerative colitis = continuous
681
Q

Are there abscesses/fissures/fistulae in Crohn’s and ulcerative colitis?

A
  • Crohn’s = common
  • ulcerative colitis = not common
682
Q

Is surgery curative in Crohn’s and ulcerative colitis?

A
  • Crohn’s = not always
  • ulcerative colitis = curative
683
Q

What are the symptoms of IBD?

A
  • ulcers
  • diarrhoea
  • abdominal pain
  • fevers
  • excessive sweating
  • anaemia
  • arthritis
  • weight loss
  • skin rashes

o doesn’t just affect the gut

684
Q

What are the symptomatic treatments for IBD?

A
  • aminosalicylates -> e.g. mesalazine, olsalazine
  • glucocorticoids -> e.g. Prednisolone
  • immunosuppressive agents -> e.g. Azathioprine
685
Q

Where are aminosalicylates absorbed?

A
  • olsalazine ( 2 linked 5-ASA) has to be activated/metabolised by the gut flora -> so only works in the colon -> if this is where the inflammation is most serious, it is better
  • mesalazine will be absorbed all the way through the gut
686
Q

What is the mechanism of action of mesalazine?

A
  1. absorbed in the gut and is metabolised in to N-acetyl 5-ASA by NAT-1
  2. N-acetyl 5-ASA binds to PPARy -> this triggers the formation of a transcription regulator (includes N-acetyl 5-ASA and PPARy)
  3. this downregulates inflammatory factors such as NF-KB/MAPK (less pro-inflammatory cytokines) and COX-2 (less prostaglandins)
687
Q

How are aminosalicylates used in ulcerative colitis treatment?

A
  • first line for both inducing and maintaining remission -> probably better than glucocorticoids
688
Q

How are aminosalicylates used in Crohn’s?

A
  • literature is unclear
  • is ineffective in inducing remission in Crohn’s
  • gGlucocorticoids are probably better in Crohn’s
  • may be effective in a subgroup of patients
689
Q

Name some glucocorticoids.

A
  • prednisolone
  • fluticasone
  • budesonide
690
Q

What is the mechanims of glucocorticoids in IBD?

A
  • glucocorticoids act at multiple points in the inflammation seen in bowel disease
  • can inhibit the production of IL-1 and TNF-alpha by dendritic cells. Dendritic cells are thought to have an important role in IBD
  • are very potent anti-inflammatory and immunosuppressive actions of GCs -> when given systemically, chronic glucocorticoid administration causes many unwanted effects
691
Q

What are the side effects of glucocorticoids?

A
  • hypertension
  • diabetes
  • osteoporosis
  • reactivation of infection (due to immunosuppression)
  • easy bruising
  • poor wound healing
  • thin skin

o similar to the symptoms of Cushing’s

692
Q

How are the side effects of glucocorticoids minimised?

A
  • topically administer glucocorticoid: fluid, foam enemas, suppositories and oral preparations
  • use a lower dose in combination with another drug (e.g. 5-ASA and glucocorticoids)
  • use an oral or topically administered drug with high hepatic first pass metabolism -> e.g. Budesonide -> little escapes into the systemic circulation
693
Q

What is the problem with using Budesonide to treat Crohn’s?

A
  • more side effects when we treat someone with prednisolone, compared to when we treat someone with budesonide -> problem with this is that the standard glucocorticoid (prednisolone) is actually BETTER than budesonide at inducing remission in active Crohn’s
694
Q

How are glucocorticoids using in ulcerative colitis?

A
  • use of glucocorticoids is in decline, due to evidence that aminosalicylates are superior
  • are best avoided due to their side effects
695
Q

How are glucocorticoids used in Crohn’s?

A
  • remain the drugs of choice for inducing remission in Crohn’s -> budesonide is preferred if the disease is mild
  • patients are likely to get side effects if GCs are used to maintain remission (long-term use)
696
Q

Describe azathioprine.

A
  • a pro-drug that has to be activated by the gut flora to 6-mercaptopurine -> can give 6-mercaptopurine directly
  • is a purine antagonist, and is therefore immunosuppressive
697
Q

What is the mechanism of azathioprine?

A
  • metabolised into 6-MeMPN -> inhibits de novo purine synthesis
  • metabolised into TGN -> acts as a flase purine molecule

o INTEREFERES WITH DNA SYNTHESIS AND CELL PROLIFERATION

698
Q

What are the effects of azathopprine on the immune system?

A

o impairs

  • > cell-mediated and antibody-mediated immune responses
  • > lymphocyte proliferation
  • > mononuclear cell infiltration
  • > synthesis of antibodies

o enhances

  • T cell apoptosis due to increased damage to the cells
699
Q

How is azathioprine used in ulcerative colitis?

A
  • used with some success in ulcerative colitis -> but there is no real reason to use it if the response is good with 5-ASA
700
Q

How is azathioprine used in Crohn’s?

A
  • has no benefit at all in active disease -> mainly used to maintain remission
  • Cochrane review shows it be glucocorticoid-sparing (co-administered to reduce steroid dose)
  • has a slow onset -. 3 to 4 months treatment for clinical benefit to be observed
701
Q

What are the side effects of azathioprine?

A
  • pancreatitis
  • bone marrow suppression

hepatotoxicity

  • increased risk (around 4-fold) of lymphoma and skin cancers

o nearly 10% of patients have to stop it due to side effects

702
Q

What are the 3 branches of manipulatin the microbiome?

A
  • nutrition-based therapies
  • faecal microbiota replacement therapies
  • antibiotic treatment with rifaximin
703
Q

What is the evidence surrounf nutritional-based therapies in IBD?

A
  • no evidence for probiotics in food stuffs have any impact at all in Crohn’s disease
  • some evidence for probiotics for maintenance of remission in Ulcerative Colitis
  • probiotics are as effective as 5-ASA in inducing and sustaining remission in ulcerative colitis
704
Q

What is the evidence for faecal microbiota replacement therapies?

A
  • re-populating a patient’s bowel with healthy gut flora may solve the problem
  • insufficient evidence for FMT-> but 2 out of 3 RCTs showed benefit in UC -> more studies are needed
705
Q

What is the evidence for antibiotic treatment with rifaximin?

A
  • interferes with bacterial transcription by binding to RNA polymerase
  • induces and sustains remission in moderate Crohn’s disease
  • may be beneficial in ulcerative colitis too
706
Q

Describe the current state of biological therapies for IBD.

A
  • are now a number of drugs that have been approved for use in IBD -> include anti-TNF alpha antibodies e.g. infliximab (iv)
  • other antibodies are effective, but some have more side-effects
  • new humanised antibodies are coming -> e.g. entanacept, which may have fewer side effects
707
Q

What is the mechanism of anti-TNF alpha antibodies?

A
  • anti-TNF alpha reduces activation of TNF a receptors in the gut -> reduces downstream inflammatory events
  • also binds to membrane associated TNF alpha -> induces cytolysis of cells expressing TNF alpha
  • promotes apoptosis of activated T cells
708
Q

Describe the pharmacokinetics of anti-TNF alpha antibodies.

A
  • infliximab is given intravenously and has a very long half-life (9.5 days)
  • most patients relapse after 8–12 weeks, therefore the infusion needs to be repeated every 8 weeks
709
Q

Describe the use of anti-tumour necrosis factor alpha in Crohn’s disease.

A
  • has been used successfully in the treatment of Crohn’s disease
  • 60% of patients respond within 6 weeks -> don’t understand the underlying pathology
  • potentially curative -> but many patients relapse with this treatment
  • successful in some patients with refractory disease and fistulae -> great for maintaining fistula closure

o no effect on ulcerative colitis -> expected as it Th-2 driven

710
Q

What are the problems with anti-TNF alpha antibodies?

A
  • emerging evidence that up to 50% lose response within 3 years time -> this is after only 60% responding to it -> due to production of anti-drug antibodies and increased drug clearance
711
Q

What are the side effects of anti-TNF alpha antibodies?

A
  • 4-5x increase in incidence of tuberculosis -> also a risk of reactivating dormant TB
  • increased risk of septicaemia
  • worsening of heart failure
  • increased risk of demyelinating disease (e.g. multiple sclerosis)
  • increased risk of malignancy
  • can be immunogenic -> azathioprine reduces risk, but raises the risk of tuberculosis and malignancy
712
Q

When and how should infliximab be used?

A
  • early use is better than using it as a last resort
  • combined infliximab and azathioprine therapy may be more effective than the antibody alone
713
Q

In IBD, budesonide causes fewer unwanted systemic effects than prednisolone because:

a. It can be administered topically
b. It can be co-administered with another drug
c. It has a higher potency at therapeutic doses
d. It has a lower potency at therapeutic doses
e. It is metabolised and inactivated locally

A
  • e
714
Q

The mechanism of action of Azathioprine in IBD:

a. Interferes with purine biosynthesis
b. Is a direct reduction of protein synthesis in the GI tract
c. Is blocked by co-administration with allopurinol
d. Means that it increases side-effects caused by infliximab
e. Needs activation of the drug by metabolism to 5-ASA

A
  • a
715
Q

Describe the movement of molecules in the proximal convoluted tubule.

A

o permeable to water -> can move across the tubule cells quite easily

  • throughout the kidney is an osmotic force, which tends to draw water out of the lumen, across into the blood -> glomerulus filters protein-free fluid but blood has LOTS of plasma proteins

o quite permeable to sodium

  • Na-K-ATPase channels are on the basal side of the cell -> works to retain the concentration gradient for sodium -> if sodium diffuses into the cell on the apical side, it needs to be removed on the other side to maintain a concentration gradient

o no HCO3- channels -> H+ actively pumped out of cell by H+-ATPase -> H+ + HCO3- = carbonic acid -> carbonic acid -> CO2 + water -> CO2 diffuses in and reforms H2CO3- befores it dissociates into HCO3-

  • don’t want H+ to accumulate -> Na-H-exchanger antiporter on membrane -> lots of Na reabsorbed in PCT so this is useful (Na also reabsorbed with glucose)
  • HCO3- exchanges out with Cl- via the bicarbonate/Cl- exchangers into blood

o lots of movement of amino acids and glucose across the cell, into the blood -> always coupled with sodium

o glucoronidation in phase II metabolism -> special transporters recognise the side chains and functional groups revealed in phase I metabolism -> kidney then moves the drug molecule into the lumen of the kidney for excretion in the urine

716
Q

Describe the movement of molecules in the collecting duct.

A

o descending limb

  • highly permeable to water -> cells have loose tight junctions
  • not many mitochondria -> don’t pump ions

o ascending limb

  • Na+ and Cl- are pumped out
  • very tight junctions -> impermeable to water
  • lots of mitochondria -> very high metabolic activity

o CREATES THE HEAVILY CONCENTREATED INTERSTITIAL SPACE FOR MATER REABSORPTION IN THE LoH, DCT and CD -> COUNTERCURRENT MECHANISM

717
Q

Describe the movement of molecules in the distal collecting duct.

A
  • Na-Cl transported on the tubular side (apical membrane) -> on the basal membrane is the Na-K-ATPase (ensures that the concentration gradient is maintained and that Na is reabsorbed into the blood)
  • potassium and chloride transporters are also present on the basal side.
718
Q

Describe the movement of molecules in the collecting duct.

A

o major hormones influencing the collecting duct are aldosterone and vasopressin

  • aldosterone is a mineralocorticoid -> binds to the MR receptor and influences nuclear transcription -> increases transcription of Na channels and Na-K-ATPase
  • vasopressin interacts with the V2 receptor, and sticks aquaporin channels into the apical membrane -> provides a mechanism for water to move across the cell
719
Q

What is aldosterones mechanism of action?

A

o steroid hormone -> diffuses into cell and binds with steroid hormone intracellular receptors bound to chaperone proteins -> chaperone protein released from receptor -> dimerization of steroid hormone-receptor complexes -> enters nucleus -> transcription of gene for Na-K-ATPase -> increased capacity to reabsorb sodium

720
Q

What is Liddle’s syndrome?

A
  • an autosomal dominant inherited disease of high bp
  • mutation in tha aldosterone activated sodium channel -> always switched on -> Na+ retention is increased -> hypertension
721
Q

What is the general mechanims of diuretics?

A
  • work by inhibiting the reabsorption of Na and Cl (i.e. increase excretion of Na and Cl) -> losing more ions to the urine means that water will follow
  • increase the osmolarity of the tubular fluid -> i.e. decrease the osmotic gradient across the epithelia
722
Q

What are the 5 major classes of diuretics with different mechanisms?

A
  1. osmotic diuretics -> e.g. mannitol
  2. carbonic anhydrase inhibitors -> e.g. acetazolamide
  3. loop diuretics -> e.g. frusemide (furosemide)
  4. thiazides -> e.g. bendrofluazide (bendroflumethiazide)
  5. potassium sparing diuretics -> e.g. amiloride, spironolactone
723
Q

Describe the mechanism of osmotic diuretics.

A
  • acts throughout the kidney
  • filtered by the glomerulus but NOT reabsorbed -> are pharmacologically inert -> increases the osmolarity of the tubular fluid -> less water leaves becuase of a lower osmotic drive so more is excreted
  • no action on Na reabsorption
724
Q

What are the clinical uses of osmotic diuretics?

A
  • pulmonary oedema
  • cerebral oedema
725
Q

Describe the mechanism of carbonic anhydrase inhibitors.

A
  • acetazolamide acts in the proximal tubule
  • inhibits both versions of the enzyme, apical membrane bound and cytoplasmic enzyme -> bicarbonate and hydrogen aren’t converted to carbon dioxide and water effectively
  • little CO2 and water that gets in won’t be converted back to bicarbonate and hydrogen ions -> less hydrogen ions means that there is less Na-H exchange at the apical membrane -> less Na enters the cells and is reabsorbed, so less water follows via osmosis -> more water excreted
726
Q

Which of the 5 diuretics aren’t classical used as diuretics?

A
  • osmotic diuretics
  • carbonic anhydrase inhibitors
727
Q

What are the most powerful diuretics?

A
  • loop diuretics -> can increases water excretion by 30%
728
Q

Where is the kidney do loop diuretics act?

A
  • ascending loop of Henle -> targeting the triple transporter
729
Q

Describe the mechanism of loop diuretics.

A
  • prevent Na from moving across the cell, and into the interstitium -> impacts the counter-current effect -> Na reabsorption is massively impaired
  • in the PCT and LoH, there is something called potassium recycling -> potassium is constantly being reabsorbed and lost-> constant movement of potassium replenishes a certain amount of positive charge to the lumen -> there is repulsion within the tubule -> if potassium recycling is interfered with potassium movement is reduced and the excess positive charge is diminished -> less Ca, Mg and Na move through this paracellular route -> lower interstitial concentration -> less osmotic movement
730
Q

Describe potassium recycling.

A
  • potassium is constantly being reabsorbed and lost-> constant movement of potassium replenishes a certain amount of positive charge to the lumen -> there is repulsion within the tubule -> if potassium recycling is interfered with potassium movement is reduced and the excess positive charge is diminished -> less Ca, Mg and Na move through this paracellular route
731
Q

What are the overall effects of loop diuretics?

A
  • increased water excretion
  • increased potassium loss
  • decrease reabsorption of calcium and magnesium
732
Q

What is the main use for loop diuretics?

A
  • oedema
733
Q

What are the unwanted effects of loop diuretics?

A
  • hypokalaemia
  • hyponatraemia
  • hypovolaemia
  • metabolic alkalosis
  • hyperuricaemia
  • hypotension
734
Q

Describe the mechanism of thiazide diuretics.

A
  • target the apical side -> act on Na-Cl transport in the distal tubule -> has its effect as it is being excreted
  • further along the kidney you get, the less powerful the diuretic effect becomes -> because there is less fluid and less sodium to impact -> maximum of 5-10% Na and water loss

o action on Na+ reabsorption -> inhibit Na+ and Cl- reabsorption in early distil tubule

o action on water reabsorption -> increased ­ tubular fluid osmolarity = decreased H2O reabsorption in the collecting duct

735
Q

What are the overall effects of thiazide diuretics?

A
  • increased water excretion
  • increased sodium delivery to distal tubule -> increased potassium loss (same as loop diuretics)
  • increased magnesium loss
  • increased calcium reabsorption
736
Q

What is the major problem with thiaziades and loop diuretics?

A
  • effect the renin secretion
  • over time, patients suffer a degree of hyponatraemia (due to Na loss) -> because low Na concentration in the kidney stimulates renin secretion
  • diuretics cause a lower sodium load to pass through the lumen, which stimulates renin secretion -> over time diuretics tend to directly promote renin secretion -> NOT HELPFUL because renin increases aldosterone, which promotes reabsorption
  • often, diuretics are given WITH ACE inhibitors, to reduce the effect
  • in the case of loop diuretics, they influence renin secretion indirectly (through hyponatraemia) and directly (blocking the triple transporter)
737
Q

What are the 2 classes of potassium-sparing drugs?

A
  • aldosterone receptor antagonists -> e.g. spironolactone
  • inhibitors of aldosterone-sensitive sodium channels -> e.g. amiloride
738
Q

What is the mechanism of aldosterone receptor antagonist diuretics?

A
  • are MR inhibitors -> decreases the ability of aldosterone to produce Na channels and Na-K-ATPase
  • increased tubular fluid osmolarity = less water reabsorption in the CD
739
Q

What is the mechanism of inhibitors of aldosterone-sensitive sodium channels?

A
  • Na blocking drug -> prevents Na from getting into the cell in the first place
  • increased tubular fluid osmolarity = less water reabsorption in the CD
740
Q

What class is the weakest diuretic?

A
  • potassium sparing -> only effect the CD and DCT where there isn’t much sodium
741
Q

What are the overall effects of potassium spairng diuretics?

A

o action on Na reabsorption: Inhibit Na reabsorption (and concomitant K+ secretion) in DCT -> 5%

o action on water reabsorption: ­ tubular fluid osmolarity = decreased H2O reabsorption in the collecting duct

  • other effects: decreased reabsorption Na+ to distal tubule and increased ­ H+ retention (deacreased Na+/H+ exchange)
742
Q

What are the unwanted effects of potassium sparing diuretics?

A
  • hyperkalaemia
743
Q

What are the unwanted effects of thiazides?

A
  • hypokalaemia
  • hyponatraemia
  • hypovolaemia
  • metabolic alkalosis
  • hyperuricaemia
  • hypotension
744
Q

How do diuretics cause hyperuricaemia?

A
  • diuretics directly affect the transporter that moves uric acid into the lumen
  • there is a transporter on the basal side of kidney cells that exchanges organic ions with uric acid -> diuretics compete for and interfere with this protein -> less exchange occurs, leading to less uric acid moving into the cell and tubule from the blood -> uric acid builds up in the blood -> can be associated with gout
745
Q

Name a disease associated with hyperuricaemia.

A
  • gout
746
Q

What are the clinical uses of diuretics?

A

o oedema

o heart failure

o thiazides are first line treatments for hypertension in Afro-Caribbean people and anyone over 55 years -> tend to have salt sensitive hypertension which is associated with low renin -> no point giving them ACE inhibitors

  • thiazides promots fluid loss and reduce bp
747
Q

Why are thiazides used over other diuretics in anti-hypertensive treatement?

A

o initial response is good (4-6 weeks) -> due to decreased plasma volume -> but, after 4-6 weeks, the plasma volume is restored = diuretic effect is lost

o thiazides chronic use = VASODILATORY EFFECT

  • decreased T.P.R., activation of eNOS (endothelium)
  • Ca2+ channel antagonism
  • opening of KCa channel (smooth muscle)
748
Q

How are adverse drug reaction classified by time?

A
  • acute = within 1 hour
  • sub-actue = 1-24 hours
  • latent = more than 2 days
749
Q

How are adverse drug reaction classified by severity?

A
  • mild = noticed but requires no change in therapy
  • moderate = requires a change in therapy, may require additional treatment, and possibly hospitalisation
  • severe = disabling, life-threatening, or ones that cause damage to the foetus
750
Q

How are adverse drug reaction classified by type of reaction?

A
  • Type A: Augmented pharmacological effect
  • Type B: Bizarre
  • Type C: Chronic
  • Type D: Delayed
  • Type E: End-of-treatment
751
Q

Describe Type A adverse drug reactions.

A
  • reaction is often an extension of the pharmacologic effect of the drug that we already know about -> therefore usually predictable and often dose dependent
  • responsible for at least two-thirds of ADRs
  • can either have a linear increase in toxicity (digoxin) or can have a tipping point which once over can have detrimental effect (paracetamol)
752
Q

Describe Type B adverse drug reactions.

A
  • tend to be more dramatic and are less understood
  • are idiosyncratic (particular to given individuals) or immunologic reactions -> include allergy and “pseudo-allergy”
  • are rare and unpredictable
  • exampels are chloramphenicol and aplastic anemia or ACEi and angioedema (pseudo-allergy)
753
Q

Describe Type C adverse drug reactions.

A
  • are associated with long-term use of drugs -> involve dose accumulation (total dose someone’s exposed to over a period of time)
  • examples are methotrexate (used in chemotherapy and as an immunosuppressant) and liver fibrosis or anti-malarial drugs and ocular toxicity
754
Q

Describe Type D adverse drug reactions.

A
  • often have delayed effects and are not strongly linked to dose - may happen at low dose
  • include carcinogenicity due to immunosuppressants or teratogenicity because of thalidomide
755
Q

Describe Type E adverse drug reactions.

A
  • withdrawal reactions = opiates, benzodiazepines, corticosteroids
  • rebound reactions = clonidine, beta-blockers, corticosteroids
  • “adaptive” reactions = neuroleptics (major tranquillisers)
756
Q

Use clonidine as an example of a rebound reaction.

A

o clonidine used to be used as an anti-hypertensive

o is an alpha 2 agonist, so it reduces the release of noradrenaline from sympathetic neurones -> reduction in sympathetic outflow leads to a drop in bp

  • missing one or two doses of clonidine, can lead to a substantial RISE in blood pressure -> long-term use of clonidine causes long-term suppression of peripheral noradrenaline production, which, in turn, leads to a compensatory upregulation in adrenergic receptors on the post-synaptic neurone -> this upregulation in receptors means that when the inhibition of NA release by clonidine is removed, NA starts being produced again and has more receptors to act on and can cause a much great effect =HUGE RISE IN BLOOD PRESSURE

o isn’t used an more because there are plenty of other anti-hypertensives

757
Q

What are the 4 classes of allergies?

A
  • Type I -> immediate, anaphylactic (IgE) -> e.g. anaphylaxis with penicillin
  • Type II -> cytotoxic antibody (IgG, IgM) -> e.g. methyldopa and hemolytic anemia
  • Type III -> serum sickness (IgG, IgM) antigen-antibody complex -> e.g. procainamide-induced lupus
  • Type IV -> delayed hypersensitivity (T cell) -> e.g. contact dermatitis
758
Q

Define pseudoallergy.

A
  • a condition which presents like an allergy but is pharmacologically mediated instead of immunologically
759
Q

How can aspirin cause pseudoallergies?

A
  • 5% of people who take these drugs get bronchospasm
  • aspirin inhibits COX enzymes -> a reduction in prostanoid/prostaglandin synthesis (bronchodilators)
  • instead, the body makes more leukotrienes which are pro-inflammatory and bronchoconstrictors
760
Q

How can ACEi cause pseudoallergies?

A

o 10-20% of people taking ACE inhibitors suffer from a chronic, dry cough

  • due to accumulation of inflammatory peptides (e.g. bradykinin) in the lung -> bradykinin triggers cough through sensory receptors in the lung

o in 1-5% of people taking the drug, people may suffer from angioedema

  • angioedema = has many of the same signs and symptoms of anaphylaxis, but much less severe
761
Q

What class of drugs commonly cause adverse drug reaction?

A
  • antibiotics
  • anti-neoplastic drugs
  • anticoagulants
  • cardiovascular drugs
  • hypoglycemic drugs
  • anti-hypertensive drugs
  • NSAID/analgesics
  • CNS drugs
762
Q

How are adverse drug reactions detected?

A

o subjective report -> e.g. patient complaint

o objective report -> direct observation of events and abnormal findings

  • physical examination
  • laboratory test
  • diagnostic procedure
763
Q

What is the yellow card scheme?

A
  • a public way of detecting adverse reactions, which was introduced in 1964 after thalidomide -> before which there was NO SYSTEM of checking whether drugs that were marketed were effective or safe

o ADR suspected -> ADR confirmed (high probability) -> frequency estimated -> prescribers informed

764
Q

What are the 3 types of drug interactions?

A
  • pharmacodynamic interactions = related to the drug’s effects in the body -> receptor site occupancy
  • pharmacokinetic interactions = related to the body’s effects on the drug -> due to absorption, distribution, metabolism and elimination (ADME)
  • pharmaceutical interactions = drugs interacting outside the body -> most commonly found with IV infusions
765
Q

Describe pharmacodynamic drug interactions.

A
  • additive -> put two drugs together, they produce an effect that is the sum of the drugs -> overlapping toxicities e.g. ethanol & benzodiazepines
  • synergistic actions of antibiotics -> two drugs potentiate each others’ actions to get a greater effect than expected
  • antagonistic effects -> drugs that antagonise each others’ actions e.g. anticholinergic medications (amitriptyline and acetylcholinesterase inhibitors)
766
Q

Give an example of altered absorption of drugs.

A

o chelation -> irreversible binding of drugs in the GI tract

  • tetracyclines and quinolone antibiotics can be involved in chelation with ferrous sulfate (Fe2+), antacids (Al3+, Ca2+, Mg2+) and dairy products (Ca2+)
  • form a stable chelate -> don’t absorb the mineral ion or the antibiotic
767
Q

How can protein binding interactions cause pharmacokinetic drug interactions?

A
  • competition between drugs for protein or tissue binding sites increases free (unbound) concentration due to displacement -> enhanced pharmacological effect
  • PB interactions are not usually clinically significant -> warfarin is an exception -> is very tightly bound to proteins, so even displacing a small amount can have drastic effects
768
Q

How can drug elimination cause adverse drug reactions?

A
  • probenecid and penicillin = good -> reduces elimination of penicillin -> makes it last longer
  • lithium and thiazides = bad -> thiazides reduce clearance of lithium -> can lead to fatal toxicity
769
Q

Define pharmacogenetics and pharmacogenomics in order to differentiate the 2.

A
  • pharmacogenetics = study of genetically determined inter-individual differences in therapeutic response to drugs and susceptibility to adverse effects
  • pharmacogenomics = use of genome-based techniques in drug development
770
Q

How is psychoses split?

A
771
Q

What are the symptoms of depression?

A
  • emotional -> misery, apathy, pessimism, low self-esteem, loss of motivation, anhedonia (inability to enjoy pleasureable activities)
  • biological -> slowing of thought and action, loss of libido, loss of appetite, sleep disturbance
772
Q

What are the two main groups of depression?

A
  • unipolar
  • bipolar
773
Q

Describe unipolar depression.

A

o mood swings are in same direction -> feel down

o relatively late onset (adulthood)

o can be split into reactive depression (75%) and endogenous depression (25%)

  • reactive depression: depression in response to stressful life events, that is non-familial
  • endogenous depression: depression that is unrelated to external stresses, with a familial pattern

o drug treatment -> is the same treatment for reactive and endogenous

774
Q

Define reactive depression.

A
  • depression in response to stressful life events
  • is non-familial/inherited
  • 75% of unipolar depression
775
Q

Define endogenous depression.

A
  • depression that is unrelated to external stresses
  • has a familial/inheritable pattern
  • 25% of unipolar depression
776
Q

Describe bipolar depression.

A
  • oscillating depression and mania (hyper-excitability with symptoms opposite to depression)
  • less common than unipolar depression
  • tends to have an early adult onset
  • a strong hereditary tendency
777
Q

What is the treatment for bipolar depression?

A
  • lithium -> not a conventional anti-depressant -> more of a mood stabilising drug
778
Q

What is the monoamine theory of depression?

A
  • depression = a functional deficit of central monoamine transmission
  • mania = a functional excess of central monoamine transmission
  • this hypothesis is based on pharmacological evidence, but the biochemical evidence inconsistent
779
Q

What are the 2 main monoamines in the monoamine theory of depression?

A
  • noradrenaline
  • serotonin/5-HT
780
Q

How fast do anti-depressants work and what is the theory behind this?

A
  • several weeks
  • with anti-depressants there is a down-regulation of a2, beta and 5-HT receptors (monoamine receptors)
781
Q

What is the principal action of tricyclic anti-depressants?

A
  • blocks NA and 5-HT re-uptake
  • positive effect on mood
782
Q

What is the principal action of MAO inhibitors?

A
  • increases stores of NA and 5-HT
  • positive effect on mood
783
Q

What is the principal action of reserpine?

A
  • inhibits NA and 5-HT storage
  • negative effect on mood
784
Q

What is the principal action of alpha-methyltryosine?

A
  • inhibits NA synthesis
  • negative effect on mood
785
Q

What is the principal action of methyldopa?

A
  • inhibits NA synthesis
  • negative effect on mood
786
Q

What is the principal action of electroconvulsive therapy?

A
  • increases CNS responses to NA and 5-HT
  • positive effect on mood
787
Q

Describe the structure of TCAs.

A
  • have 2 main chemical groups -> deibenzazepines and dibenzcycloheptenes
  • all have a 3 ring structure
788
Q

What is the mechanims of TCAs?

A

o are neuronal monoamine re-uptake inhibitors

o prevents NA and 5-HT uptake much more so than dopamine

  • other receptor actions on α2 receptors, muscarinic AchRs, histamine receptors and 5-HT receptors -> some of these are important to the action of TCA other contribute to the side effects of TCAs
  • alpha 2 receptors -> if TCAs bind to and antagonise these receptors, they enhance the release of NA, block the inhibitory control over NA, and allow a greater increase of NA into the synaptic cleft
  • is a delayed down-regulation of β-adrenoceptors & 5-HT2 receptors
789
Q

Describe the pharmacokinetics of TCAs.

A
  • rapid oral absorption
  • highly plasma protein bound (90 – 95% of TCAs are PPB) -> important in terms of interactions
  • have hepatic metabolism -> TCAs are metabolised in the liver and generates active metabolites which are weaker but still functioning
  • undergo renal excretion (as glucuronide conjugates)
  • plasma t1/2 = 10-20 hours -> relatively long half life, so can be given once a day due to the long duration of action
790
Q

What are the unwanted effects of TCAs at a therapeutic dose?

A
  • atropine-like effects -> dry mouth, blurred vision, constipation, urinary retention -> amitriptyline seems to have more muscarinic antagonist activity than some of the other TCAs so it is worse with it
  • postural hypotension (mediated through the vasomotor centre) -> is a CENTRAL EFFECT
  • sedation (TCAs cause H1 antagonism) -> patients feel drowsy during the day -> many depressed patients have trouble sleeping, so this can be taken advantage of -> ask the patient to take the TCA just before bed
791
Q

What are the unwanted effects of TCAs at a toxic dose (over dosage)?

A
  • CNS ->: excitement, delirium, seizures -> coma and respiratory depression
  • CVS -> cardiac dysrhythmias -> ventricular fibrillation and sudden death
  • TCAs that are muscarinic antagonists also affect vagal input to the heart

o MUST BE HANDLED WITH CARE -> commonly utilised in attempted suicide

792
Q

State the drug reaction which occur with TCAs.

A

o many exist

  • increased TCA effects when co-administration with aspirin and phenytoin (anti-convulsant)
  • warfarin can displace TCAs from their binding sites -> moves plasma levels of TCA into a toxic range
  • hepatic microsomal enzymes metabolise TCAs, so TCA effects increase if co-administered with drugs that are metabolised by the same enzyme system -> e.g. neuroleptics and oral contraceptives
  • potentiation of CNS depressants with TCAs (alcohol in particular)
  • an interaction with antihypertensive drugs (monitor BP closely) -> difficult to predict
793
Q

Give an example of a TCA.

A
  • amitriptyline
794
Q

Give an example of a MOAi.

A
  • phenelzine
795
Q

What are the 2 classes of MAOs?

A
  • MAO-A -> preference for NA and 5-HT
  • MAO-B -> preference for dopamine
796
Q

When might you want a MAOi to be specific?

A
  • in Parkinson’s -> MAO-B inhibitors are used to increase dopamine levels in the brain
797
Q

Are MAOi used to treat depression selective?

A
  • no -> inhibit both MAO-A and MAO-B as well as other enzymes
798
Q

What is the mechanism of MAOi’s?

A
  • irreversibly bind to MAO enzymes and therefore blocks the breakdown of NA, 5-HT and dopamine
  • all of these chemicals therefore build up and have a larger effect on the brain -> occurs very fast
  • causes a down-regulation of β-adrenoceptors and 5-HT2 receptors so the clinical effects aren’t seen for a while
799
Q

Describe the structure of MAOi’s.

A
  • all MAOi’s have got a single ring structure
  • are many different classes -> phenelzine is a hydrazine
  • phenelzine has a single ring with a carbon side chain -> on the end is a hydrazine functional group which is VERY REACTIVE -> forms the covalent bonds with the MAO enzyme -> irreversible inhibition
800
Q

Describe the pharmacokinetics of MAOi-‘.

A
  • rapid oral absorption
  • short plasma t1/2 (few hours) but longer duration of action due to irreversible inhibition -> taken either once a day or every other day
  • metabolised in the liver and excreted in the urine
801
Q

What are the unwanted effects of MAOi?

A
  • atropine-like effects (much less than with TCAs)
  • postural hypotension (common) -> probably a vasomotor mediated effect
  • sedation with long-term usage -> becomes seizures in over dose
  • weight gain (possibly excessive) -> associated with increase appetite with MAOIs
  • hepatotoxicity (due to hydrazines) -> is rare (e.g. 1 in 10,000 patients)
802
Q

What drug interactions occur with MAOi?

A
  • tyramine is metabolised by MAOs -> if they are being inhibited, we may encounter problems -> ‘Cheese reaction’ -> tyramine-containing foods + MAOI leads to a hypertensive crisis (throbbing headache, increased blood pressure and intracranial haemorrhage -> patient must avoid foods high in tyramine (mature cheese, red wine etc.)
  • react with TCAs to produce hypertensive episodes (avoid)
  • react with pethidine to cause hyperpyrexia, restlessness, coma & hypotension
803
Q

What is tyramine?

A
  • an indirectly acting sympathomimetic amine (has actions similar to activation of the SNS)
  • under normal circumstances, there are not sufficient levels of tyramine in the body to cause problems
804
Q

How can the multiple drug interaction of MAOi be avoided?

A
  • using reversible MAO-A inhibitors (RIMA) such as moclobemide -> are slective for MAO-A
  • has fewer interactions but has a shorter duration of action -> must be taken 2-3 times a day
  • aren’t as effective anti-depresants as traditional MAOi
805
Q

Give an example of a selective serotonin reuptake inhibitor.

A
  • fluoxetine
806
Q

What is the mechanism of SSRI?

A
  • selective 5-HT re-uptake inhibition (SSRIs selectively inhibit the reuptake of 5-HT)
  • less troublesome side effects than other anti-depressants, so they are safer in the case of overdose
  • BUT are less effective against severe depression
807
Q

Describe the pharmacokinetics of SSRIs.

A
  • oral administration of SSRIs
  • plasma t1/2 (18-24 hours) -> given once a day, orally
  • is a delayed onset of action (2-4 weeks before we see the optimal anti-depressant action of SSRIs)
  • fluoxetine competes with TCAs and for the hepatic enzymes -> co-administration must be avoided (same with MAOi as they interact)
808
Q

Describe the structure of SSRIs.

A
  • a couple of ring structures with an aliphatic side chain
809
Q

What are the unwanted effects of SSRIs?

A
  • fewer unwanted effects than TCAs/MAOIs -> massive advantage -> makes them the frontline treatment (providing its not severe depression)
  • 10% of patients complain of GI side effects (nausea, diarrhoea)
  • some people complain of insomnia
  • 30% of patients complain of loss of libido
810
Q

What is venlafaxine?

A

o a SNRI -> shows dose-dependent Reuptake inhibition of both 5-HT and NA

  • 5HT > NA (SNRI) -> increasing the dose increases NA re-uptake inhibition
  • a very high dose of venlafaxine inhibits the dopamine transporter too

o is 2nd line treatment for severe depression

811
Q

What is mirtazapine?

A
  • an α2 Receptor antagonist -> increases NA and 5-HT release in the brain by blocking the inhibitory control over NA and 5-HT -> increases NA & 5HT release in the brain
  • other receptor interactions may contribute to antidepressant activity (sedative - histamine antagonism)
  • useful in SSRI-intolerant patients
812
Q

Tricyclic antidepressant drugs (TCAs) work largely by:

A: Antagonism at 5HT receptors

B: Inhibiting central DA reuptake

C: Blocking VSCCs

D: Inhibition of central NA & 5HT reuptake

E: Enhancement of the action of GABA

A
  • D: Inhibition of central NA & 5HT reuptake
813
Q

The ‘cheese reaction’ is most likely to be caused by:

A: Tricyclic antidepressants (TCAs)

B: Selective serotonin reuptake inhibitors (SSRIs)

C: Monoamine oxidase inhibitors (MAOIs)

D: Reversible MAO-A inhibitors (RIMAs)

E: α2-Adrenoceptor antagonists

A
  • C: Monoamine oxidase inhibitors (MAOIs)
814
Q

How does cisplatin bring about nausea and vomiting?

A

o TOXIC to the lining of the stomach

o cisplatin affects enterochromaffin cells (toxic to them) and causes destruction of these cells -> causes the release of free radicals -> EXCESSIVE 5-HT RELEASE from the stomach

o 5-HT goes on to act on 5-HT3A receptors, located on:

  • nerve fibres to the nucleus tractus solitarius (NTS) (cpnnected to the VC via nerves)
  • nerve fibres to the vomiting centre (VC)
  • nerve fibres to chemoreceptor trigger zone (CTZ)

o as a result of increased serotonin, there is increased activity of the solitary tract -> increased activity of the VC -> NAUSEA AND VOMITING.

o also a direct link between cell bodies in the stomach and the vomiting centre

o nerve fibres stretching from the stomach to the chemoreceptor trigger zone (outside the BBB so detects things in blood an feeds this information back to the vomiting centre) -> increases the chances of nausea and vomiting

815
Q

What is treatment for chemotherapy triggered nausea and vomiting?

A

o ONDANSTERON -> a 5-HT3A receptor antagonist -> less 5-HT binding -> less nausea and vomiting

  • given with two other things:
  • glucocorticoids -> reduces free radical production -> remove effects of free radicals
  • aprepitant -> neurokinin-1 receptor antagonist -> dampens down the trnsmission from the NST to the VC
816
Q

Where are neurokinin 1 receptors found?

A
  • in the connection between solitary tract and the vomiting centre
817
Q

How can motion sickness be pharmacologically treated?

A
  • hyoscine -> non-selective muscarinic receptor antagonist -> most effective treatment for motion sickness
  • promethazine -> H1 receptor antagonist
818
Q

How does a H1 receptor antagonist treat motion sickness?

A
  • blocks the histamine H1 receptors -> receptors may be pre-synaptic or post-synaptic

receptors are somehow involved in the signalling process from the hypothalamus to the CTZ

  • blocking histamine receptors reduces the likeliness of vomiting being triggered
819
Q

What is motion sickness?

A
  • a mismatch between the labyrinth and the vestibular system
820
Q

How do non-selective muscarinic receptor antagonists prevent motion sickness?

A
  • blocks all 5 of the muscarinic receptors
  • no one is entirely sure how the cholinergic system plays a role in vomiting -> but because hyoscine is such an effective agent against motion sickness, we know ACh is involved
  • muscarinic receptors are most likely to be involved in signalling from labyrinth to vestibular system
821
Q

What is gastroparesis?

A
  • delayed emptying of the stomach due to it being unable to contract -> triggers the vomiting reflex
  • symptoms are abdominal pain and bloating
  • often linked to T2DM
822
Q

How does gastroparesis bring about vomiting?

A
  • delayed stomach emptying due to reduced stomach contraction
  • 5-HT release and direct stimulation of the VC by nerves -> involves dopamine2 receptors
  • activation of 5-HT receptors on the VC and the chemoreceptor trigger zone
823
Q

What is the treatment for nausea and vomiting in gastroparesis?

A
  • metoclopramide -> dopamine D2 receptor antagonist
  • 5-HT receptor antagonist
824
Q

How do D2 receptor antagonists prevent nausea and vomiting in gastroparesis?

A
  • prokinetic -> stimulates gastric emptying by acting on the stomach itself (stimulates contractility)
  • inhibits D2 receptors in the vomiting centre -> blocks likelihood of vomiting from D2 stimulation
  • is a 5-HT3A receptor antagonist -> inhibits activation of CTZ
825
Q

Describe the physiology of nausea and vomiting.

A

o vomiting centre (area postrema) -> innervated by the nucleus of the tractus solitarius

  • chemoreceptor trigger zone communicates with the vomiting centre
826
Q

Name 3 mechanistic trigeers of nausea and vomiting.

A
  • cytotoxic drugs
  • motion sickness
  • gastrointestinal problems
827
Q

Identify the 4 main classes of anti-emetic drugs.

A
  • 5-HT3A receptor antagonists
  • histamine H1 receptor antagonists
  • muscarinic receptor antagonists
  • dopamine D2 receptor antagonists
828
Q

What is the principle clinical use for 5-HT3A receptor antagonists?

A
  • chemotherapy induced N&V
829
Q

What is the principle clinical use for histamine H1 receptor antagonists?

A
  • motion sickness
830
Q

What is the principle clinical use for muscarinic receptor antagonists?

A
  • motion sickness
831
Q

What is the principle clinical use for dopamine D2 receptor antagonists?

A
  • gastroparesis induced N&V
832
Q

What are the main side effects of 5-HT3A receptor antagonists

A
  • headaches
  • constipation
833
Q

What are the main side effects of histamine H1 receptor antagonist?

A
  • drowsiness
834
Q

What are the main side effects of muscarinic receptor antagonists?

A
  • drowsiness, dry mouth and a bit of constipation
835
Q

What are the main side effects of dopamine D2 receptor antagonists?

A
  • extra-pyramidal side effects
  • galactorrhoea
836
Q

What are the main risk factors of Alzheimer’s disease?

A

o MAIN = AGE

o are genetic components too -> APP, PSEN, ApoE

  • APP = mutation in amyloid precursor protein and/or presenilin gene increased risk of early onset Alzheimer’s disease
  • ApoE = Apo lipoprotein E miutation -> increased risk of late onset Alzheimer’s
837
Q

What are the clinical symptoms of Alzheimer’s?

A

- memory loss -> especially recently acquired information = MOST PROMINENT SYMPTOMS

  • disorientation/confusion -> forgetting where they are
  • language problems -> stopping in the middle of a conversation
  • personality changes -> becoming confused, fearful, anxious
  • poor judgement -> such as when dealing with money
838
Q

What is the physiological process behind APP breakdown?

A
  1. amyloid precursor protein (APP) is cleaved by a-secretase
  2. sAPPa released, but the C83 fragment remains
  3. C83 is then digested by g-secretase, and the products removed
839
Q

Describe the beta amyloid hypothesis of Alzheimer’s?

A
  1. APP cleaved by b-secretase
  2. sAPPb released, but the C99 fragment remains
  3. C99 is then digested by g-secretase, releasing b-amyloid (Ab) protein
  4. Ab forms toxic aggregates à formation of beta-amyloid plaques which associate with neuronal cells and increase the likelihood of their death
840
Q

What is the tau protein?

A
  • soluble protein present in neuronal axons that form the internal skeleton of neuronal cells
  • is important for assembly & stability of microtubules
841
Q

Describe the Tau hypothesis of Alzheimer’s?

A
  • hyper-phosphorylated tau is insoluble -> self-aggregates to form neurofibrillary tangles -> are neurotoxic -> results in microtubule instability -> neuronal cell death
  • correlates with symptoms very well -> more tau hyper-phosphorylation the worse the disease
842
Q

Describe the inflammation hypothesis of ALzheimer’s.

A

o caused due to inappropriate activation of microglial cells

  • increased release of inflammatory mediators & cytotoxic proteins
  • increased phagocytosis
  • decreased levels of neuro-protective proteins
843
Q

What are the 4 drugs licensed for UK use in the treatment of Alzheimer’s?

A
  • donepezil - GOLD STANDARD -> anticholinesterase
  • rivastigmine -> anticholinesterase
  • galantamine -> anticholinesterase
  • memantine -> NMDA receptor blocker
844
Q

What is donezepil and how effective is it in the treatment of Alzheimer’s?

A
  • reversible acetylcholinesterase (found only in the NS) inhibitor
  • effective uin treating symptoms for around a year
  • long-plasma half-life so only 1 tablet a day is required
845
Q

What is rivastigmine and how effective is it in the treatment of Alzheimer’s?

A
  • pseudo-reversible AChE/BChE inhibitor -> inhibits acetylcholinesterase and butyrylcholinesterase
  • 8 hour half-life (relatively short) -> needs to be administered 2-3 times a day but it has be reformulated as transdermal patch formulation which require less applications
  • -nhibition of butyrylcholinesterase enzyme leads to more side effects (liver problems)
846
Q

What is galantamine and how effective is it in the treatment of Alzheimer’s?

A
  • reversible cholinesterase inhibitor with a 7-8 hour half-life
  • a7 nAChR agonist -> agonist of specific isoforms (neuronal included in this) of the nicotinic ACh receptor (in the CNS)
847
Q

What is memantine and how effective is it in the treatment of Alzheimer’s?

A
  • -* use-dependent non-competitive NMDA receptor blocker with low channel affinity -> more the NMDAr is activated, the more likely memantine is to have an inhibitory effect -> occurs with a lot of neuron degeneration such as in moderate and severe Alzheimers -> only licensed for moderate to severe/late Alzheimers
  • long plasma half-life
848
Q

Name 3 high profile drug failures in the treatment of Alzheimer’s.

A

1. gamma-secretase inhibitors -> tarenflurbil and semagacestat

2. beta-amyloid antibodies -> bapineuzumab and solanezumab -> aducanumab is in clinical trials and is looking psitive at the moment

3. tau inhibitors -> methylene blue

849
Q

What is GABA?

A
  • most important inhibitory NT in the brain
850
Q

What is GABA synthesised from?

A
  • glutamate
  • a bit strange seeing that glutamate is the most important excitatory NT in the brain
851
Q

What are GABA-A and GABA-B receptors?

A
  • GABA-A = sit on the post-synaptic membrane and are chloride ionphores -> type I receptors
  • GABA-B = sit on pre-synaptic membrane and are G-protein coupled -> type II -> have a regulatory effect on GABA secretion and production
852
Q

What is the mechanism of GABA?

A
  • GABA binds to GABA-A receptors which are ion channel linked
  • when stimulated by GABA, the ion channel changes conformation to transiently become a chloride ion -> chloride is negative, so they hyperpolarise the post-synaptic cell -> cell has a very negative potential -> harder to excite/less likely for a action potential to occur
853
Q

How is GABA deactivated?

A

o GABA taken up by the surround glial cells OR back into the pre-synaptic terminal

854
Q

Describe GABA metabolism.

A

- GABA transaminase (GABA-T) is the enzyme in the first step - GABA -> succinic semialdehyde

  • second enzyme is succinic semialdehyde dehydrogenase (SSDH) to produce succinic acid
  • succinic acid goes back into the TCA cycle in the cells and as glutamate also arises from the TCA cycle -> a GABA shunt is formed
  • around 10% of the activity of the TCA cycle is involved in generating GABA as a transmitter
855
Q

Where are GABA-T and SSDH found?

A
  • both are mitochondrial enzymes
856
Q

How can GABA metabolism be manipulated pharmacologically?

A
  • GABA-T and SSDH can be unhibited -> increases GABA levels in the brain -> enhances GABA in the CNS -> anticonvulsant/anti-epileptic effects
857
Q

Name 2 anticonvulsant and antiepileptic drugs that work by inhibiting GABA metabolsim.

A

- sodium valproate -> GABA-T and SSDH inhibitor and Na blocker

- vigabatrin -> GABA-T inhibitor

858
Q

What are the 4 main proteins making up the GABAA receptor complex?

A
  • GABA receptor protein
  • benzodiazepine receptor protein
  • barbiturate receptor protein
  • chloride channel protein
859
Q

What happens to the GABA-A receptor complex when GABA binds to it?

A
  • GABA binds to the GABA-A receptorat the GABA receptor protein
  • this causes the GABA receptor protein and benzodiazepine receptor protein link together -> mediated by a peptide called GABA modulin
  • result is a momentary opening of the chloride channel protein -> hyperpolarisation
860
Q

What is bicuculline?

A
  • a competitive antagonist for the GABA-A receptor -> binds to the GABA receptor protein
861
Q

Where do benzodiazepines bind?

A
  • benzodiazepine receptor protein
862
Q

What are the 2 main effects of benzodiazepines on GABA neurotransmission?

A
  • facilitate GABA mediated opening of the Cl- channel
  • facilitate GABA binding to its own receptor
863
Q

What is flumezanil?

A
  • a competitive benzodiazepine antagonist -> competes with benzodiazepine for its binding site
864
Q

Where do barbiturates bind?

A
  • barbiturate receptor protein
865
Q

What are the 3 main effects of barbiturates on GABA neyrotransmission?

A
  • facilitate GABA mediated opening of the Cl- channel
  • facilitate GABA binding to its receptor but this is not reciprocated
  • at higher concentrations, barbiturates can have a direct action on the chloride channel
866
Q

How do benzodiazepines and barbiturates mechanisms differ?

A
  • benzodiazepines = increase the FREQUENCY of chloride channel opening
  • barbiturates = increase the DURATION of the chloride channel opening
867
Q

Compare the selectivity of barbiturates and benzodiazepines.

A

o barbiturates are LESS selective than benzodiazepines

  • barbiturates also reduce excitatory transmission via some antagonist action at glutamate receptors -> contributes to their anxiolytic and hypnotic effects
  • also have other membrane effects -> e.g. direct action on Cl channel at higher concentrations
  • their reduced selectivity may explain: induction of surgical anaesthesia (we can’t do that with benzodiazepines) and the fact that they are less safe than the benzodiazepines
868
Q

What are the clinical uses of benzodiazepines?

A
  • anti-spastics -> diazepam -> their action is at the spinal cord and reduces AP propagation in alpha motor neurones
  • antiolytics
  • sedatives/hyponotics
869
Q

What are the clinical uses of barbiturates?

A
  • anaesthetics -> thiopentone -> generally used to induce anaesthsia -> maybe swapped to a different anaesthic for longer operations
  • anticonvulsants
  • anti-spastics
  • anxiolytics
  • sedatives/hypnotics
870
Q

What is a antiolytic?

A
  • a drug which removes anxiety without impairing mental or physical activity (“minor tranquillisers”)
871
Q

What is a sedatives?

A
  • a drug which reduces mental and physical activity without producing loss of consciousness
872
Q

What is a hypnotic?

A
  • a drug which induces sleep
873
Q

Describe the structure of barbiturates.

A
  • barbiturates all have a six-membered ring (4 carbons and 2 nitrogens) in the middle -> differ in their substituent groups
874
Q

What is amobarbital?

A
  • a barbiturate useful in the severe intractable insomnia
  • has a half-life of 20-25 hours
875
Q

Why are barbiturates no longer the frontline sedatives and hypnotic drugs?

A

o they have many side effects

  • possess low safety margins -> depress respiration -> overdosing is lethal
  • alter natural sleep by decreasing REM sleep -> leads to hangovers and irritability
  • enzyme inducers -> barbiturates induce microsomal enzymes -> must avoid co-administration (e.g. warfarin)
  • potentiate effect of other CNS depressants (e.g. alcohol)
  • tolerance occurs in the tissue and pharmokinetically
  • dependence -> withdrawal syndrome = insomnia, anxiety, tremor, convulsion and death
876
Q

Describe the structure of benzodiazepines.

A
  • tricyclic -> classic three-ring structure (2 six-membered and 1 seven-membered)
  • are around 20 available – but they all act at GABAA receptors
  • all have similar potencies & profiles, and have the same mechanism
877
Q

How are benzodiazepines administrated?

A
  • well absorbed per orally -> peak plasma concentration is reached after about 1 hour
  • sometimes given IV in the treatment of status epilepticus (continuous seizure activity) -> is a medical emergency unlike other seizures
878
Q

Where are benzodiazepines metabolised?

A
  • the liver
879
Q

How are benzodiazepines excreted?

A
  • in the urine as glucuronide conjugates
880
Q

What are the 2 classes of benzodiazepines?

A
  • short-acting
  • long-acting -> have a slower metabolism or generates active metabolites
881
Q

Name 2 short-acting benzodiazepines and explain how they are metabolised?

A
  • oxazepam -> metabolised in the liver to its inactive glucuronide
  • temazepam -> metabolised initially to oxazepam, which is then converted to the glucuronide conjugate -> this occurs pretty fast though
  • both have a half-life of around 8 hours
882
Q

Name 1 long-acting benzodiazepine and explain how it is metabolised?

A
  • diazepam -> metabolised via temazepam and oxazepam to the glucuronide
  • some diazepam is metabolised through nordiazepam then oxazepam
  • diazepam has slower metabolism than the other two and it also generated active metabolites
  • diazepam has a half-life of around 32 hours
883
Q

What are the clinical uses of benzodiazepines?

A

o anxiolytics -> generally the longer acting benzodiazepines

- diazepam

  • chlordiazepoxide
  • nitrazepam

o sedatives/hypnotics -> generally the shorter acting benzodiazepines

- temazepam

- oxazepam

  • exception = nitrazepam (t1/2 = 28 hours) -> can be used as a sedative/hypnotic (it also has a daytime anxiolytic effect
884
Q

What are the advantages of the benzodiazepines over the barbiturates?

A
  • wider margin of safety -> overdose = prolonged sleep which is rousable -> also have an intravenous shot of flumazenil (competitive antagonist) to overcome this
  • mild effect on REM sleep -> do not induce the hangover effect
  • do NOT induce liver enzymes
  • have less tolerance and dependency than barbiturates
885
Q

What are the unwanted effects of benzodiaepines?

A
  • sedation -> when being used as anxiolytics patients will feel a little drowsy
  • confusion, ataxia -> impaired manual skills
  • potentiate other CNS depressants (e.g. alcohol and barbiturates)
  • tolerance -> less than barbiturates because of tissue tolerance only - no pharmacokinetic tolerance
  • dependence -> associated with withdrawal syndrome but less than barbiturates
  • free plasma concentration of benzodiazepines can be increased by giving aspirin and heparin or other plasma protein binding drugs
886
Q

What is zopiclone?

A
  • a short acting cyclopyrrolone -> acts at benzodiazepine receptor but it is NOT a benzodiazepine
  • enhances GABA-mediated neurotransmission by binding to the BZ binding site
  • similar efficacy to benzodiazepines
  • minimal hangover effects but dependency still a problem
887
Q

Name some other drugs (that arent barbiturates or benzodiazepines) that are antiolytics.

A
  • some antidepressant drugs -> SSRIs -> less sedation and dependence however they have delayed response/long-term treatment
  • some anti-epileptic drugs -> e.g. Valproate, tiagabine
  • some antipsychotic drugs -> e.g. Olanzapine, quetiapine
  • propranolol -> non-selective beta-blocker -> improves the physical symptoms of anxiety -> e.g. Tachycardia (b1) and Tremor (b2)
  • buspirone -> 5-HT1A agonist -> has fewer side-effects e.g. less sedation compared to benzodiazepines however has a slow onset of action (maximal anxiolytic effect may be seen in days/weeks)
888
Q

Benzodiazepines are used to treat ‘panic attacks’ and other anxiety states. By what mechanism do they produce their anti-anxiety effects?

A: Inhibition of GABA breakdown

B: Activation of 5-HT1A receptors

C: Enhancement of the action of GABA at GABA-A receptors

D: Inhibition of GABA reuptake

E: Enhancement of the action of GABA at GABA-B receptors

A
  • C
889
Q

Which of the following drugs is commonly used in the treatment of insomnia?

A: Thiopental

B: Phenytoin

C: Baclofen

D: Sodium valproate

E: Temazepam

A
  • E -> temazepam
890
Q

Describe the synthesis of dopamine.

A
  • production of dopamine = L-tyrosine -> L-DOPA -> dopamine
  • dopamine is stored in vesicles at the synaptic terminal
  • process utilises the enzymes -> tyrosine hydroxylase (rate limiting) and DOPA decarboxylase
891
Q

How is dopamine removed from the synapse?

A
  • dopamine transporter
  • noradrenaline transporter
892
Q

What 3 enzymes breakdown the catcholamine neurotransmitters and which enzymes have preferences?

A
  • MOA-A = metabolises DA, NE & 5-HT
  • MAO-B = metabolises DA only
  • catechol-O-methyl transferase (COMT) = metabolises all catecholamines -> more widely distrubuted than the other pre-synaptic intracellular MAO enzymes
893
Q

What are the 4 main dopaminergic pathways?

A
  • nigrostriatal pathway -> important in Parkinson’s
  • mesolimbic pathway/reward pathway -> important in schizophrenia
  • mesocortical pathway -> important in schizophrenia
  • tuberoinfundibular pathway
894
Q

Describe the nigrostriatal pathway.

A
  • cell bodies of the nerve are present within the substantia nigra pars compacta (SNc), and the nerves project up to the striatum
  • inhibition of these neurones results in movement disorders
  • MPORTANT IN Parkinson’s
895
Q

Describe the mesolimbic pathway.

A
  • cell bodies are located in the ventral tegmental area (VTA), and the axons project up to the Nucleus Accumbens (NAcc)
  • BRAIN REWARD PATHWAY
  • IMPORTANT IN SCHIZOPHRENIA
896
Q

Describe the mesocortical pathway.

A
  • cell bodies start in the VTA, but this time they project to the cerebrum (various areas of the cortex)
  • is important in executive functions and complex behavioural patterns
  • activation associated with positive schizophrenia symptom while inhibition associated with negative schizophrenia symptoms -> IMPORTANT IN SCHIZOPHRENIA
897
Q

Describe the tuberoinfundibular pathway.

A
  • cell bodies in the arcuate nucleus project to the median eminence
  • regulates prolactin secretion
  • inhibition results in hyperprolactinaemia
898
Q

What are the risk factors for Parkinson’s?

A
  • age
  • genetics can increase the chance of early onset but not the normal form -> SNCA and LRRK2
899
Q

Describe the pathophysiology of Parkinson’s.

A
  • severe loss of dopaminergic projection cells in SNc (nigrostriatal tract) -> patients have lost 80% of their dopaminergic cells before symptoms
  • Lewy bodies & neurites are found within neuronal cell bodies & axons -> consist of abnormally phosphorylated neurofilaments, ubiquitin & a-synuclein -> no-one quite knows how they are involved in the pathology
900
Q

What are the clinical presentations of Parkinson’s?

A
  • motor symptoms = CARDINAL SYMPTOMS = resting tremor, bradykinesia, rigidity, postural instability
  • autonomic nervous system effects = olfactory deficits, orthostatic hypotension, constipation
  • neuropsychiatric = sleep disorders, memory deficits, depression, irritability
901
Q

What is levodopa?

A

o dopamine replacement therapy for Parkinson’s

  • converted to DA by DOPA decarboxylase (DOPA-D)
  • can cross the blood-brain barrier (BBB)
902
Q

What are the problems of levodopa?

A
  • cannot simply give DA because it will enter the periphery -> acts of CTZ and cause nausea and vomiting due to its breakdown by peripheral DOPA-D (not just found in the brain)
  • has long-term side effects -> dyskinesias and ‘on-off’ effects
  • is not disease modifying, it just reduces the symptoms of PD
903
Q

How is nasuea and vomiting prevented from occuring due to levodopa?

A
  • by giving levodopa in adjunction with carbidopa and benserazide which are DOPA-D inhibitors
  • don’t cross the BBB -> prevent peripheral conversion but not in the CNS
  • reduces the required dose of levodopa and takes away some of the symptoms
904
Q

How can entacapone and tolcapone be used in the treatment of Parkinson’s?

A
  • both are COMT inhibitors -> reduces the breakdown of levodopa in the brain
  • can only be given with levodopa -> doesn’t work by itself
905
Q

What are the 3 different classes/approaches to Parkinson’s treatment?

A
  • dopamine replacement -> levodopa = GOLD STANDARD
  • dopamine receptor agonists
  • monoamine oxidase B (MOAB) inhibitors
906
Q

Describe dopamine receptor agonists as a treatment for Parkinson’s.

A

o artificial stimulation of the dopamine receptors

  1. ergot derivatives = bromocriptine and pergolide
    - act as potent agonists of dopamine D2 receptors (are effectibe) however, these are associated with cardiac fibrosis and increase likelihood of valvular disease
  2. Non-ergot derivatives = ropinirole and rotigotine
    - ropinirole is also available as extended-release formulation and rotigotine is also available as a patch
907
Q

What are the advantages of using monoamine oxidase B inhibitors as a firstline treatment for Parkinson’s?

A
  • reduce the amount of levodopa required as well as increasing the amount of time before levodopa is needed
  • the longer someone is on levodopa the higher the chance of the dyskinesias and ‘on-off’ effects are to occur -> when they do the drug has stopped working
  • give longer controlled Parkinson’s
  • is associated with the cheese reaction
908
Q

Name some MAOB inhibitors used in the treatment of Parkinson’s.

A
  • selegiline (deprenyl)
  • rasagiline
909
Q

Which neuronal dopaminergic pathway extends from the ventral tegmental area to the nucleus accumbens?

a. Nigrostriatal tract
b. Mesolimbic pathway
c. Mesocortical pathway
d. Spinothalamic tract
e. Tuberoinfundibular pathway

A
  • b
910
Q

Which PD medication increases the amount of DA produced in nerve terminals?

a. Bromocriptine
b. Entacapone
c. Levodopa
d. Rasagiline
e. Ropinirole

A
  • c
911
Q

Why are non-ergot derivative D agonists recommended over ergot derivatives?

a. Ergot derivatives cause psychosis
b. Ergot derivatives are less efficacious
c. Ergot derivatives are costlier
d. Ergot derivatives are associated with cardiac fibrosis
e. Ergot derivatives can cause tardive dyskinesias

A
  • d
912
Q

Which gene is known to be involved in schizophrenia?

A
  • DISC1
913
Q

Which pathways are thought to be involved in schizophrenia?

A
  • mesolimbic and mesocortical
914
Q

At what age does schizophrenia seem to onset?

A
  • 15-35
915
Q

Which group of people are more liekly to suffer from schizophrenia?

A
  • ethnic minorities
916
Q

How are schizophrenic patients life expectancy altered?

A
  • 20-30 years less than average -> probably due to likelihood to abuse drugs to control symptoms
  • is a link between schizophrenia and cannabis use
917
Q

What are the symptoms of schizophrenia and what are they associated with?

A

o positive = increased mesolimbic dopaminergic activity -> hallucinations, delusions/paranoia, thought disorder/denial about oneself

o negative = decreased mesocortical dopaminergic activity -> affective flattening/lack of emotion, alogia/lack of speech, avolition/apathyloss of motivation

918
Q

Before the discovery of first generation antipsychotics what was the treatment for schizophrenia?

A
  • labotomy or electroconvulsive therapy
919
Q

Name 2 first generation antipsychotic drugs.

A
  • chlorpromazine
  • haloperidol
920
Q

What is the mechanism of chlorpromazine?

A
  • probaly a D2 receptor antagonist
  • not known exactly -> was discovered as a treatment while looking a=for a new antihistamine -> is also an effective antihistamine
921
Q

What are the side effects of chlorpromazine?

A
  • high incidence = anti-cholinergic effects, especially sedation
  • low incidence = extrapyramidal side-effects (EPS)
  • drug is also a muscarinic receptor antagonist -> this causes the side effects
922
Q

What is the mechanism of haloperidol?

A
  • a very potent D2 antagonist -> 50 times more so than chlorpromazine
  • therapeutic effects develop over 6-8 weeks
  • little impact on negative symptoms -> maybe even worsening - causes Parkinson’s like movement due to effects on the mesocortical pathways
923
Q

What are the side effects of haloperidol?

A
  • high incidence = EPS (extra-pyramidal side effects)
924
Q

What is the difference betwene the first and second generation antipsychotics?

A
  • second generation targets serotonin rather than dopamine
925
Q

Name 4 second generation antipsychotics.

A
  • clozapine
  • risperidone
  • quetiapine
  • aripiprazole
926
Q

What is the mechanims of clozapine?

A
  • very potent antagonist of 5-HT2A receptors -> inhibits these receptors
  • also has actions on M1, H1 receptors and alpha-1 receptors and dopamine D2 receptor
927
Q

What is the most effective antipsychotic currently on the market?

A
  • clozapine -> only drug to show efficacy in treatment of resistant schizophrenia and negative symptoms -> no-one knows how it does so but it does
928
Q

Seeing that clozapine is the most effective antipsychotic, why is it only used as a last resort treatment for schizophrenia?

A

o has a severe side effect profile

  • can cause potentially fatal neutropenia, agranulocytosis, myocarditis and weight gain
929
Q

What is the mechanism of risperidone?

A
  • very potent antagonist of serotonin and D2 receptors
930
Q

What are the side effects of risperidone?

A
  • more EPS & hyperprolactinaemia (medication is inhibiting dopamine receptors in the tuberoinfundibular pathway) than other atypical antipsychotics, weight gain
931
Q

What is the mechanism of quetiapine?

A
  • very potent H1 receptor antagonist as well as affecting the dopamine receptors
932
Q

What is the mechanims of aripiprazole?

A

o partial agonist of D2 & 5-HT1A receptors

  • in theory = when there is too much activity, the partial agonist acts as an inhibitor and reduces positive symptoms and when there is little activity, the partial agonist increases activity and reduces negative symptoms
  • however in practice, it is no more efficacious than typical antipsychotics
933
Q

What are the side effects of aripiprazole?

A
  • reduced incidences of hyperprolactinaemia and weight gain than other antipsychotics
934
Q

Haloperidol is not effective at treating which symptom of schizophrenia?

a. Affective flattening
b. Auditory hallucinations
c. Delusions
d. Thought disorders
e. Visual hallucinations

A
  • a
935
Q

Which antipsychotic drugs is associated with neutropenia and agranulocytosis?

a. Aripiprazole
b. Chlorpromazine
c. Clozapine
d. Quetiapine
e. Risperidone

A
  • c
936
Q

How does aripiprazole differ from other antipsychotic drugs?

a. It has an immediate onset of action
b. It has no effect on D2 receptors
c. It has none of the common side-effects
d. It is a partial agonist at D2 and 5-HT1A receptors
e. It inhibits histamine H1 receptors

A
  • d
937
Q

What do general anaesthetics cause?

A
  • loss of consciousness -> ALL the general anaesthetic agents can do this at low concentration
  • suppression of reflex responses -> ALL the general anaesthetic agents can do this at high concentration
  • relief of pain (analgesia)
  • muscle relaxation
  • amnesia
938
Q

What are the two routes of administration of general anaesthetics?

A
939
Q

Describe the Meyer/Overton correlation theory.

A

o anaesthetic potency increases in direct proportion with oil/water partition coefficient -> in other words anaesthetic potency is directly correlated with lipid solubility

o TWO main problems with that explanation:

  • at therapeutic doses, changes to the lipid bilayer were minute
  • how would the change in the membrane impact on membrane proteins anyway?

o rise in temperature seemed to have the same effect on the membrane as the general anaesthetic so this was a poor explanation

940
Q

What is thought to be the real mechansim of general anaesthetics nowadays?

A
  • redcued neuronal excitability

or

  • altered synaptic function
941
Q

What is the mechanism of intravenous general anaesthetic agents?

A
  • intravenous GA agents alter synaptic function, primarily through enhancement of GABA-A receptors -> POTENTIATION
  • IV GAs are slightly more selective -> they target the GABA-A receptor
  • GABA-A receptor is made up of 5 subunits -> subunit composition is different in different brain regions
942
Q

What will a patient feel just before being knocked out by an IV GA?

A
  • euphoria due to GABA-A importance in bring about euphoria
943
Q

How can IV GA bring about different effects?

A

o GABA-A is a type 1 ionotropic receptor so it is comprised of 5 subunits -> subunit combination is different in different parts of the brain

o IV agents seem to target specific subunits in the GABA-A receptor:

  • beta-3 = suppression of reflex responses (important at the synaptic level)
  • alpha-5 = amnesia (extra-synaptic)
944
Q

What is the mechanism of inhalational general anaesthetics?

A
  • inhalational GA agents alter synaptic function and have an affect on GABA -> probably 50% less powerful (other targets are involved): GABA-A Receptors and Glycine Receptors (glycine is an inhibitory neurotransmitter)
  • are far less selective for GABA-A than the IV agents
  • seem to be more selective for alpha-1 containing GABA -> important in suppression of reflex responses
945
Q

What class of GA is nitrous oxide?

A
  • inhalational
946
Q

What is the mechanism of nitrous oxide (N2O)?

A
  • blocks the NMDA-type glutamate receptors to induce its anaesthetic effect -> less GABA specific and more glutamate specific
  • competes at the binding site for glycine on the glutamate receptor -> glycine is an important CO-AGONIST of NMDA receptors which allows the full receptor response to be transduced -> interfering with glycine will interfere with NMDA
947
Q

How are neuronal nicotinic acetylcholine receptors involved in GA mechanism?

A

o important for the analgesic effects and amnesia but don’t influence the loss of consciousness or hypnotic effects of inhaled agents

  • increasing the concentration of the inhalational agent leads to a reduction in nerve conduction via an effect on nicotinic acetylcholine receptors -> INHIBITION
  • agent enters the brain, binds to and blocks the ACh receptor -> ANALGESIA
948
Q

How do inhaled GA agents reduce neuronal excitability?

A

o enhancing the opening of TREK (background leak K+ channels) -> hyperpolarisation -> reduced neuronal excitability

  • important in terms of suppression of reflex responses
949
Q

Compare the selectivity of IV and inhaled GAs.

A
  • IV agents mainly affect GABA-A -> seem to be a lot more selective
  • inhalation agents are far less selective
  • therefore a GENERAL RULE is that a higher dose of inhalation agents is needed compared to IV
950
Q

Describe the neuroanatomy which leads to the lose of consciousness with GAs.

A
  • unconscious = massive decrease in cortical activity

o thalamus is a relay station for information going between the cortex and the rest of the CNS -> depressing the excitability of thalamocortical neurones -> disconnection of the periphery from brain -> mediated by background leak K+ channels leading to hyperpolarisation and enhanced GABA function -> both occur due to GA

o reticular activating system also affects consciousness -> thalamocortical neurones respond to sensory information and also impacted by the RAS -> ghe greater the firing of the RAS, the greater the level of arousal -> anaesthetics interfere with this process by blocking the RAS neurones

951
Q

Describe the neuroanatomy which leads to suppression of reflex responses with GAs.

A

o location = depression of reflex pathways in the DORSAL HORN OF THE SPINAL CORD

  • GABA and glycine receptors are important targets here
  • anaesthetic agents that enhance GABA and glycine function in the dorsal horn will decrease the activity of the dorsal pathways -> suppresses the reflex responses
952
Q

Describe the neuroanatomy linked to the amnesic effects of GAs.

A
  • GAs tend to cause amnesia at a pretty LOW DOSE -> amnesia tends to be the first effect
  • are a lot of GABA receptors in the hippocampus that have the alpha 5 subunit -> GAs suprress these subunits -> decrease in synaptic transmission in the hippocampus (where most memory formation occurs) -> loss of memory
953
Q

What is the difference between inhalational and IV GA agents in terms of excretion and the consequences of this?

A
  • IV agents are injected into the blood, travel to the brain and have their anaesthetic effects -> excretion rate is difficult to control if it has been injected straight into the blood
  • inhaled gaseous agent enters the blood but a large amount is excreted -> gives good control over the agent -> can control inhalational agents better
954
Q

Explain the blood:gas partition co-efficient and how it effects the level of control and anaesthetist has?

A
  • if you fill the lungs with anaesthetic agent, this anaesthetic will diffuse across into the blood
  • if it dissolves POORLY in the blood (has a low blood:gas partition coefficient) a fairly large proportion remains in gaseous form within the blood -> agent transfers into the brain very effectively
  • component that is dissolved in the blood (in a liquid form) will diffuse poorly into the brain

o for control you want an anaesthetic agent that has a low blood:gas partition coefficient, because this makes it MORE available to the brain and because if it is POORLY DISSOLVED IN THE BLOOD (low blood:gas partition coefficient), it is EASIER to clear it from the brain - simply remove the source and it will quickly diffsue out

955
Q

What are the different roles of GAs in the induction and maintanence of anaesthia?

A
  • induce with the intravenous -> often propofol
  • maintain with the inhalational -> allows us to have control -> often enflurane
956
Q

What are the positive about both inhaled and IV GA agents?

A
  • inhaled = rapidly eliminated and rapid control of the depth of anaesthesia
  • IV = fast induction and less coughing and excitatory phenomena (seizures)
957
Q

State a mjor complication when deciding how much GA to use?

A
  • obesity -> GA are very lipid soluble -> why 24 hrs after surgery you may feel drowsy again -> leaking back into the blockstream
  • had to balance how much GA is needed to maintain anaethesia and not cause lots of longer-term effects
958
Q

What agents are used to cause a loss of consciousness in clinical practice?

A
  • induction = IV GA agents
959
Q

What agents are used to cause a suppression of reflex responses in clinical practice?

A
  • maintenance = inhalaed GA agents
960
Q

What agents are used to cause a analgesia in surgery, in clinical practice?

A
  • opiods -> e.g. IV fentanyl
961
Q

What agents are used to cause muscle relaxation in surgery, in clinical practice?

A
  • neuromuscular blocking drugs -> e.g. suxamethanium
962
Q

What agents are used to cause amnesia in surgery, in clinical practice?

A
  • benzodiazepines -> e.g. IV midazolam
963
Q

Describe/Define local anaesthetics.

A
  • definition = drugs that reversibly block neuronal conduction when applied locally
  • all local anaesthetics are weak bases and sodium ion channel blockers
964
Q

Describe the generation of a neuronal action potential.

A
  • Phase I = Depolarisation: when neurones are depolarised due to a stimulus VGSCs open -> sodium rushes into the neurone -> generates a rapid depolarisation phase
  • Phase II = Repolarisation: within a millisecond the sodium channels closeand VGKCs open -> potassium leaves the neuronal cell -> allows the neurone to enter a repolarisation phase
  • Pase III = Refractory Period: sodium channels have been restored to their resting state, however potassium channels remain open -> it isharder for a cell to generate another AP but it can
  • Phase IV = RESTING STATE: both sodium and potassium channels have restored to their resting state -> cell responds normally to depolarising stimuli

o action potentials are all or nothing

965
Q

What are the common structures to all local anaesthetics?

A
  • aromatic region
  • basic amine side chain
  • linked by an ester or amide bond
966
Q

Give an one example of each of the two different groups of local anaethetics.

A
  • ester LA = cocaine
  • amide LA = lidocaine
967
Q

Which LA doesn’t follow the structural rules of LAs?

A
  • benzocaine -> doesn’t have the basic amine side chain -> just has an alkyl group on the side
  • has weak LA properties, and is lipid soluble so is used as a surface anaesthetic -> is useful in throat lozenges
968
Q

What is the main mechanism of LAs?

A

o hydrophilic pathway

  • inject a local anaesthetic close to sensory, pain-conducting neurones -> unionised form of the LA is lipid soluble and is able to pass through the connective tissue sheath, to gain access to the sensory axons inside the neurone
  • once the lipid-soluble form of the LA is inside the axon, an equilibrium is established (unionised and ionised versions of the LA)
  • the ionised form of the LA has the anaesthetic property -> ionised form binds to the inside of the VGSCs and stereochemically hinders the influx of Na ions
  • in order for the ionised form of the LA to be able to bind to its target site, the VGSCs must be open
969
Q

Explain why LAs are use dependent drugs.

A
  • the more the neurones are active, the more the VGSCs will be opening and closing -> the more time they are spending in the open state
  • means that the LAs can bind to their target sites more effectively and gives LAs a degree of selectively -> pain neurones are firing rapidly so these will be blocked more commonly/often than motor neurones
970
Q

Where are LAs administrated?

A
  • close to the nociceptive neurones
  • if introduced close to motor neurones, we will see weakness and relaxation of skeletal muscles
971
Q

Describe the minor pathways which has a small effect on the mechanism of action of some LAs.

A

o the hydrophobic pathway -> is more important for the more lipid-soluble LAs

  • as the unionised form crosses the axonal membrane, some can drop into the ion channel and convert into the cation ionised form to block the ion channel -> means that, by the hydrophobic route, the LAs can drop into a closed channel as well as the open channels in the hydrophilic pathway

o may also influence channel gating -> influences mechanism -> some LAs prefer the inactive state of the sodium channels -> by binding this holds them open for longer

972
Q

Why is infected tissue harder to locally anaethetise compared to healthy tissue?

A
  • infected/damaged tissue tends to be more acidic -> more LA is is in the unactive unionised form in the acidic conditions (LAs afre weak bases)
973
Q

What are the effects of LAs?

A
  • prevent the generation and conduction of action potentials because they block the VGSCs
974
Q

How are LAs selective for pain neurones/nociceptive neurones?

A

o selectively block small diameter fibres and non-myelinated fibres

  • pain impulses are conducted by narrow nonmyelinated fibres
975
Q

What are the 6 routes of administration of LAs?

A
  • surface
  • infiltration
  • IV regional
  • nerve block
  • spinal
  • epidural
976
Q

Describe surface anaesthesia.

A
  • applied to mucosal surface -> mouth, bronchial tree, eyes, nose and throat
  • administrated in spray form or powder form
  • problems is that high concentrations of the LA are needed to gain an effective anaesthetic action -> high concentrations can lead to systemic toxicity
977
Q

Describe infiltration anaesthesia.

A
  • LA is injected subcutaneously into tissues -> accesses the sensory nerve terminals directly
  • is the applications in minor surgery (suturing and stitching)
  • adrenaline co-injection often given too -> because adrenaline causes vasoconstriction (not extremities) -> increases the duration of action of the local anaesthetic, reduces the incidence of systemic side effects (thus limits toxicity) -> adrenaline is not given in the extremities because it could lead to ischaemic damage
978
Q

Describe IV regional anaesthesia.

A
  • given distal to a pressure cuff which acts to shut off the blood supply
  • has applications in limb surgery (more major surgery)
  • systemic toxicity may occur if there is premature cuff release -> rushes straight to the heart and then possibly the CNS -> should keep the cuff on for 20 minutes minimum
979
Q

Describe nerve block anaesthesia.

A
  • local anaesthetic is injected close to the nerve trunks e.g. dental nerves
  • low doses can be used however there is a relatively slow onset (minutes)
  • vasoconstrictor co-injection with nerve block anaesthesia often occurs
980
Q

Describe spinal anaesthesia.

A
  • LA is injected into the sub-arachnoid space -> has an action on the spinal roots
  • useful in abdominal, pelvic and lower limb surgery when GA is too dangerous -> anaesthetic mixes with the CSF (injected at L3/L4)
  • decreases blood pressure, and may produce a prolonged headache (accesses brain) -> decreased bp due to LA aacting on small-diameter pre-ganglionic sympathetic ANS neurones
  • glucose can be mixed with the LA -> increase in specific gravity -> allows level/area of control by adding glucos and tilting the table
981
Q

Describe epidural anaesthesia.

A
  • injected into fatty tissue of epidural space (not as deep as spinal anaesthesia) -> action on the spinal roots
  • used for abdominal, pelvic and lower limb surgery (same as spinal) but also for painless childbirth
  • disadvantages compared to spinal are a slower onset, and higher doses required -> higher doses = more likely to experience systemic side effects
  • advantage compared to spinal is are more restricted action and less effects on blood pressure
982
Q

State the pharmacokinetic properties of lidocaine.

A
  • absorption through muscous membranes = good
  • plasma protein binding = 70%
  • metabolism = hepatic N-dealkylation
  • plasma half-life = 2 hours
983
Q

State the pharmacokinetics properties of cocaine.

A
  • absorption through muscous membranes = good
  • plasma protein binding = 90%
  • metabolism = hepatic and plasma non-specific esterases
  • plasma half-life = 1 hour
984
Q

What are the unwanted effects of lidocaine?

A

o CNS = stimulation, restlessness, confusion and tremor -> PARADOXICAL EFFECTS

-> due to inhibitory GABAergic neurones in the CNS being more sensitive to local anaesthetics, than other receptors -> removes the inhibitory effect

o CVS = myocardial depression, vasodilatation, decreased BP -> due to Na channel blockade

985
Q

What are the unwanted effects of cocaine when being used as a LA?

A

o CNS euphoria and excitation

o CVS = increased cardiac output, vasoconstriction and increased BP -> can cause cardiac arrythmias

  • both due to the sympathomimetic action of cocaine
986
Q

Lidocaine:

A: Inhibits reuptake of 5-hydroxytryptamine

B: Blocks voltage-gated K+ channels

C: Is a competitive muscarinic cholinoceptor antagonist

D: Is a weak base

E: Is a general anaesthetic

A
  • D
987
Q

Which ONE of the following statements about local anaesthetics is INCORRECT? They:

A: Cause blockade of voltage-sensitive sodium channels

B: Block rapidly firing neurones more readily than more slowly firing neurones

C: Enhance action potential propagation

D: Are largely ionised at physiological pH

E: Have their durations of action increased if injected with adrenaline

A
  • C
988
Q

What are the presentating features of peptic ulcers?

A
  • epigastric pain
  • burning sensation that occurs after meals
989
Q

What investigations are done to confirm the diagnosis of a H pylori positive uncomplicated peptic ulcer?

A

o carbon-urea breath test -> give the patient a lot of urea and as H. pylori metabolises urea to nitrogen, if you get increased levels of nitrogen, this means the H. pylori infection is present

o stool antigen test -> involves testing for H. pylori antigens within the stool of the patient

o history of the presenting complaint -> pain and burning only comes after eating food

990
Q

What is the treatment for a H. pylori positive uncomplicated peptic ulcer?

A
  • amoxicillin and clarithromycin/metronidazole -> antibiotics to treat H. pylori infection -> once infection is gone the stomach heals itself
  • proton pump inhibitor for a week -> reduces acid production to alleviate symptoms
991
Q

Describe helicobacter pylori.

A
  • a gram negative, motile, microaerophilic bacterium
  • is a commensal bacteria in the vast majority of people
992
Q

How does H. pylori cause peptic ulcer formation?

A

o ulcer formation

  • increased gastric acid formation due to increased­ gastrin and/or decreased somatostatin
  • gastric metaplasia -> cell transformation due to excessive acid exposure
  • downregulation of defence factors -> less epidermal growth factor and bicarbonate production

o virulence

  • urease -> catalyses urea into ammonium chloride and monochloramine which both damage epithelial cells and urease is also antigenic so evokes an immune response
  • certain virulent strains produce CagA (antigenic) or VacA (cytotoxic) -> more intense tissue inflammation
993
Q

What is the presentation of a H pylori positive complicated peptic ulcer?

A
  • constant epigastric pain
  • constant burning sensation
994
Q

What investigation are doen to confirm the diagnosis of a H pylori positive complicated peptic ulcer?

A

o carbon-urea breath test -> give the patient a lot of urea and as H. pylori metabolises urea to nitrogen, if you get increased levels of nitrogen, this means the H. pylori infection is present

o stool antigen test -> involves testing for H. pylori antigens within the stool of the patient

o history of the presenting complaint -> pain and burning which is more constant in nature

o endoscopy -> look at the ulcer in the stomach - GOLD STANDARD

995
Q

What is the treatment for H pylori positive complicated peptic ulcers?

A
  • amoxicillin and clarithromycin/metronidazole -> all 3 can be given in the case of an individual with recurrent peptic ulcers
  • next consider adding quinolone or tetracycline in addition
  • addition of BISMUTH -> protects the mucus layer
  • always a proton pump inhibitor (omeprazole) for 4-12 weeks -> reduces acid production to alleviate symptoms and also reduce further damage to the ulcerated/vunerable areas
996
Q

If a patient presents with epigastric pain and a burning sensation but has negative carbon-urea breath test and stool antigen test, what is the most likely cause of the symptoms?

A
  • H pylori negative peptic ulcer caused by prolonged NSAID use -> most likely to be aspirin
997
Q

What is the pathophysiology of a peptic ulcer caused by NSAID use?

A
  • NSAIDs can be directly cytotoxic and can also reduce mucus production -> breakdown of barrier -> epithelia exposure -> ulceration
  • NSAIDs may increase the likelihood of bleeding by acting as an anti-platelet agent -> H. pylori does NOT have this effect -> symptoms of NSAID caused peptic ulcers can be worse
  • increased acidity -> peptic ulcer
998
Q

What is the treatment for peptic ulcers caused by NSAID usage?

A
  • removal of NSAID drugs -> can’t always do this -> e.g. in individuals with other co-morbidities
  • proton pump inhibitor or histamine H2 receptor antagonist (Ranitidine) for 4-8 weeks
999
Q

Name and describe the 4 pathways which can lead to an increase in peptic acid secretion.

A
  1. Acetylcholine (ACh) released from neurones (vagus or enteric) acts on muscarinic (M3) receptors -> increased intracellular Ca2+
  2. Prostaglandins (PGs) released from local cells act on EP3 receptors -> increased ­cAMP
  3. Histamine released from enterochromaffin-like cells (ECL) act on H2 receptors -> increased cAMP
  4. Gastrin released from blood stream acts on cholecystokinin B receptors -> increased intracellular Ca2+

o all cause extra secretion by either increasing intracellular calcium or cAMP which causes translocation of secretry vesicles to parietal cell apical surfaces -> increased H+ secretion

1000
Q

What are the 3 membrane classes of bacteria?

A

o gram positive -> prominent, thick peptidoglycan cell wall

  • e.g. Staphylococcus Aureus

o gram negative -> outer membrane with lipopolysaccharide

  • e.g. Escherichia Coli

o mycolic bacteria -> outer mycolic acid layer

  • e.g. Mycobacterium Tuberculosis
1001
Q

Describe prokaryotic nucleic acid synthesis.

A

- dihydropteroate (DHOp) is produced from paraaminobenzoate (PABA) by DHOp synthase, and converted into dihydrofolate (DHF)

- tetrahydrofolate (THF) is produced from DHF by DHF reductase -> THF IS IMPORTANT IN DNA SYNTHESIS

1002
Q

Describe prokaryotic DNA replication.

A

- DNA gyrase (aka topoisomerase) releases tension from the DNA molecules and therefore allows the process of DNA replication to take place

1003
Q

Describe prokaryotic RNA synthesis.

A

- RNA polymerase produces RNA from a DNA template -> prokaryotic RNA polymerase differs from eukaryotic RNA polymerase

1004
Q

Describe prokaryotic protein synthesis.

A

- ribosomes -> RNA molecules go through the prokaryotic ribosomes, which produce protein from RNA templates

  • prokaryotic ribosomes = 70S -> 30s and 50s
  • eukaryotic ribosomes = 80S -> 40s and 60s
1005
Q

-> prevents What is the mechanism of the sulphonamide class?

A
  • antibiotics that inhibits DHOp synthase -> prevents prokaryote nucleic acid synthesis
  • most bacteria are resistant to sulphonamides
1006
Q

What is the mechanism of the trimethoprim class?

A
  • antibiotic which inhibits DHF reductase -> prevents prokaryote nucleic acid synthesis
1007
Q

What is the mechanism of the fluoroquinolones class?

A
  • antibiotics that inhibit DNA gyrase and topoisomerase IV -> prevents prokaryote DNA replication
1008
Q

What is trimoxazole?

A
  • a combination of trimethoprim and sulphonamide
1009
Q

What is the mechanism of the rifamycins class?

A
  • antibiotic that inhibits bacterial RNA polymerase -> reduces subsequent protein production -> bacteria cell death
1010
Q

Name a common use for rifampicin, part of the rifamycin class.

A
  • mycobacterium infection -> TB
1011
Q

Name some classes of antibiotics that target bacterial ribosomes.

A
  • aminoglycosides (e.g. Gentamicin)
  • chloramphenicol -> not a class but a single drug

- macrolides (e.g. Erythromycin)

  • tetracyclines
1012
Q

Describe bacterial wall synthesis.

A

1. Peptidoglycan synthesis

  • a pentapeptide is created on N-acetyl muramic acid (NAM) -> N-acetyl glucosamine (NAG) then associates with NAM forming peptidoglycan

2. Peptidoglycan transportation

  • peptidoglycan is transported across the cell membrane by a bactoprenol molecule -> once it is in the periplasm, it undergoes a few steps before being incorporated into the bacterial wall

3. Peptidoglycan incorporation

  • is incorporated into the cell wall -> transpeptidase enzyme cross-links peptidoglycan pentapeptides
1013
Q

What is the mechanism of glycopeptide class?

A
  • binds to pentapeptide -> prevents peptidoglycin synthesis
  • class includes vancomycin
1014
Q

What is the mechanims of the bacitracin class?

A
  • inhibits bactoprenol regeneration -> prevents transportation of peptidoglycan
1015
Q

What is the mechanism of the beta-lactam class?

A
  • binds covalently to transpeptidase inhibiting peptidoglycan incorporation into the cell wall
1016
Q

What class of antibiotics is vancomycin?

A
  • glycopeptide
1017
Q

What are the 3 classes with in the beta-lactam class of antibiotics?

A
  • carbapenems
  • cephalosporins
  • penicillins
1018
Q

What is the mechanism of lipopeptides and polymyxins?

A
  • lipopeptides -> disrupt gram-positive cell membranes
  • polymyxins -> binds to LPS & disrupts gram-negative cell membranes
1019
Q

What are the main causes for the development of resistance?

A
  • unnecessary and inappropriate prescription -> 50% of antibiotic prescriptions not that bad in the UK
  • livestock farming -> 30% of UK antibiotic use is in livestock farming
  • lack of regulation -> over-the-counter availability in Russia, China, India
  • lack of development -> very few new antibiotics have been developed in recent years
1020
Q

What are the 5 major antibiotic resistance mechanisms?

A
  1. beta-lactamase
  2. production of additional targets
  3. alteration in the target enzymes
  4. hyper-production
  5. alteration in drug permeation
1021
Q

How does beta lactamase lead to antibiotic resistance?

A
  • hydrolyse the C-N bond of the beta-lactam ring of beta lactam antibiotics
1022
Q

What makes beta-lactamase resistant antibiotics resistant?

A
  • beta lactamases target the beta lactam ring -> the resistant antibiotics tend to have molecules around the beta lactam ring (shields it – steric hindrance)
  • prevents the beta lactamase accessing the ring
  • examples include flucloxacillin and temocillin
1023
Q

How must amoxicillin be administrated for it to be effective?

A
  • must be co-administrated with clavulanic acid -> confers beta-lactamase resistance
1024
Q

How does the production of additional targets lead to antibiotic resisitance?

A
  • bacteria can introduce an additional target on top of the current target -> this new target is unaffected by the drug
  • e.g. =E Coli -> produces a different DHF reductase enzyme, making them resistant to trimethoprim
1025
Q

How does the alteration of a target enzyme lead to antibiotic resistance?

A
  • alteration of the enzyme targeted by the drug -> enzyme is still effective, but the drug is now ineffective
  • e.g. = S Aureus -> mutations in the ParC region of topoisomerase IV confers resistance to quinolones
1026
Q

How does hyper-production lead to antibiotic resistance?

A
  • bacteria significantly increase levels of DHF reductase -> antibiotic is overwhelmed an cannot keep up with the additional production
  • e.g. = E Coli -> produces additional DHF reductase enzymes making trimethoprim less effective
  • this resistance mechanism isn’t utilised by bacteria much as it puts a lot of strain on the bacteria itself
1027
Q

How does alteration in drug permeation lead to antibiotic resistance?

A
  • is a very commonly utilised mechanism of resistance
  • alterations in drug permeation = reductions in aquaporins so a harder route for the antibiotic to enter the cell and/or increased efflux systems
  • the bacteria reduces methods of influx and increase efflux mechanisms to limit drug interaction
  • e.g. = primarily of importance in gram-negative bacteria
1028
Q

How can fungal infections be classified in terms of tissue and organs?

A
  • superficial = outermost layers of skin
  • dermatophyte = skin, hair or nails
  • subcutaneous = innermost skin layers
  • systemic= primarily respiratory tract
1029
Q

What are the 2 most common categories of anti-fungals?

A
  • azoles
  • polyenes
1030
Q

What is the mechanism of azoles?

A
  • azole anti-fungal drugs inhibit cytochrome enzymes that are present in the fungal cells -> inhibition of cytochrome P450-dependent enzymes involved in membrane sterol synthesis -> is normally important in ergosterol production which is incorporated into the fungal cell wall
1031
Q

What is the mechanism of polenes?

A
  • punches holes into the cell membrane of the fungal cells
  • interacts with cell membrane sterols forming membrane channels -> cells burst
  • can also punches holes in the cell membranes of all cells -> BAD SIDE EFFECTS -> is more selective for fungal cells
1032
Q

Describe the structure of a virus.

A
  • viruses have nucleic acid (genetic material), which is either RNA or DNA -> this is protected by a coat of proteins (capsid)
  • bigger viruses also have a lipid envelope and envelope proteins
1033
Q

What hepatitis viruses need treatment?

A
  • only chronic infection requires treatment
  • hepatitis A can only be an acute infection -> patient is poorly for a month, but then gets better so there is no need for treatment
  • hepatitis B and C can become chronic infections -> is associated with chronic inflammation at the liver -> cirrhosis and hepatocarcinoma may result -> vaccination is important for hepatitis B
1034
Q

What is the treatment for hepatitis B?

A
  • daily tenofovir for the rest of there life -> a nucleotide analogue
  • is a reverse transcriptase inhibitor
  • sometimes given with Peginterferon alfa
1035
Q

What is the major difference between hepatitis B and C viruses?

A
  • B = DNA
  • C = RNA
1036
Q

Define virion.

A
  • the complete, infective form of a virus outside a host cell, with a core of RNA and a capsid
1037
Q

What is the treatment for hepatitis C?

A
  • ribavirin and peginterferon alfa -> ribavirin is a nucleoside analogue -> prevents viral RNA synthesis
  • boceprevir is a protease inhibitor -> most effective against hepatitis C genotype 1
  • usually cured with 3 months -> can take longer with advanced disease with cirrhosis
1038
Q

Describe the HIV life cycle.

A

o attachment of the virus to the host cell and entry into host cell

  • viral proteins must merge with host cell receptors on the cell surface and then interact with leukocyte membrane receptors à viral capsid endocytosis

o replication and Integration

  • within the cytoplasm, the reverse transcriptase enzyme converts viral RNA into DNA -> DNA is transported into the nucleus, and is integrated into the host DNA
  • takes place over a couple of days -> once this happens, there is nothing we can do to cure it -> all that can be done is try to inhibit viral replication

o assembly and release

  • host cell’s ‘machinery’ is utilised to produce viral RNA and essential proteins
  • virus is assembled within cell -> mature virion is released out of the cell
1039
Q

How many drugs are given in the treatment of HIV?

A
  • is ALWAYS A COMBINATION THERAPY -> usually 2 or 3 antiretroviral drugs
1040
Q

What are the targets for antibiotics?

A
  • cell wall
  • DNA synthesis
  • protein synthesis
1041
Q

What are the targets for antiretroviral drugs used in the treatment of HIV?

A
  • HIV entry inhibitors
  • HIV replication inhibitors
  • HIV integrase inhibitors
  • HIV protease inhibitors
1042
Q

How where the 2 people cured of HIV cured?

A
  • chemotherapy and bone marrow transplant due to leukaemia
  • the donated bone marrow had the CCR5-delta 32 mutation -> provides immunity to HIV strains as the virus cannot enter the cell
1043
Q

Describe in detail the process of HIV attachment and cell entry?

A
  • HIV glycoprotein GP120 attaches to the CD4 receptor which are found mainly on WBCs
  • GP120 also binds to either CCR5 or CXCR4 which act as co-receptors
  • GP41 penetrates host cell membrane
  • viral capsid enters the cytoplasm of the cell
1044
Q

State some HIV entry inhibitor antiretroviral drugs.

A
  • enfuvirtide binds to HIV GP41 transmembrane glycoprotein -> prevents HIV fusion and entry
  • maraviroc blocks the CCR5 (the co-receptor) chemokine receptor -> if GP120 doesn’t find CCR5, the HIV capsid cannot enter the cell -> is very well tolerated
1045
Q

Name some HIV replication inhibiting antiretroviral drugs.

A

o nucleoside reverse transcriptase inhibitors -> e.g. zidovudine (AZT)

  • activated by a 3-step phosphorylation process

o nucleotide reverse transcriptase inhibitors -> e.g. tenofovir

  • bind to the active site of the RT enzyme -> therefore compete

o non-nucleoside RT inhibitors -> e.g. efavirenz = most used antiretroviral drug globally

  • no phosphorylation required
  • binds to allosteric sites on the RT enzyme
1046
Q

What is the role of viral integrase?

A
  • insert viral DNA into host DNA
1047
Q

Name a HIV integrase inhibiting antiretroviral drug.

A
  • raltegravir (anothe 2 are now available)
  • integrase inhibitors block the viral integrase enzyme, normally responsible for integration of HIV DNA into the genome
1048
Q

Name a HIV protease inhibiting antiretroviral drug.

A
  • saquinavir
  • usually also administrated with ritonavir which reduce the inhibitors first pass hepatic metabolism by CYP450
1049
Q

Describe, in detail, HIV virus assembly and release.

A
    • gag* precursor encodes all viral structural proteins
  • HIV protease cleaves Gag precursor protein -> allows the proteins to become functional -> without this step, the virus would be ineffective
1050
Q

What class of drugs are tenofovir and ribavirin (hepatitis treatments)

A
  • nucleoside analogues
1051
Q

What is the treatment of herpes simplex virus?

A
  • nucleoside analogues = Acyclovir
  • blocks the activity of enzymes that copy DNA into RNA
1052
Q

Describe the herpes simplex virus.

A
  • double stranded DNA virus surrounded by tegument and enclosed in a lipid bilayer
1053
Q

`Describe the influenza virus.

A
  • multipartite single stranded RNA virus containing envelope proteins called neuraminidase and hemagglutinin -> neuraminidase cleaves the sialic acid on the host cell allowing release while hemagglutinin is important in cell entry
1054
Q

What is the treatment of influenza?

A
  • neuraminidase inhibitor call oseltamivir
  • problem is that it must be inhaled as soon as you are infected with the influenza virus -> owever, by the time symptoms become apparent, it is too late
  • BEST way to stop influenza is by yearly vaccination
1055
Q

What is epilepsy?

A
  • epilepsy is a neurological condition causing frequent seizures
  • seizures are “sudden changes in behaviour caused by electrical hypersynchronization of neuronal networks in the cerebral cortex” -> over-excitation due to too much glutamatergic activity
  • glutamate is the main excitatory NT within the CNS -> over-activity causes hyper-excitability
1056
Q

How is epilepsy diagnosed?

A
  • electroencephalography (EEG) -> more constant, less regular firing
  • magnetic resonance imaging (MRI)
1057
Q

What are the different types of seizures seen in epilepsy?

A

o generalised

  • tonic-clonic seizures
  • absence seizures
  • tonic-atonic seizures
  • myoclonic seizures
  • status epilepticus

o partial/focal seizures

  • simple
  • complex
1058
Q

What are generalised seizures?

A
  • the increased synchronization begins simultaneously in both hemispheres of brain
1059
Q

What occurs in tonic-clonic seizures?

A
  • loss of consciousness -> muscle stiffening -> jerking/twitching -> deep sleep -> wakes up
  • MOST COMMON MANIFESTATION OF EPILEPSY
1060
Q

What occurs in absense seizures?

A
  • breif staring episodes with behavioural arrest
1061
Q

What occurs in tonic/atonic seizures?

A
  • sudden muscle stiffening (tonic)/sudden loss of muscle control (atonic)
  • similar to tonic-clonic without the clear different phases
1062
Q

What occurs in myoclonic seizures?

A
  • sudden, brief muscle contractions -> see lots of muscle movement
1063
Q

What occurs in status epilepticus?

A
  • over 5 minutes of continuous seizure activity
  • LEAST COMMON BUT, MOST DANGEROUS
1064
Q

What occurs in partial/focal seizures?

A
  • simple = retained awareness/consciousness
  • complex = impaired awareness/consciousness
1065
Q

What are partial/focal seizures?

A
  • seizures which begins within a particular area of brain, and may spread out
1066
Q

What are seizures due to?

A
  • increase in excitatory neurotransmitter -> most commonly glutamate
  • decrease of inhibition -> most commonly GABA
1067
Q

Describe neurotransmission at glutamatergic synapses.

A
  1. action potential arrives at the pre-synaptic terminal
  2. voltage-gated Na+ channel (VGSC) opens -> membrane depolarisation
  3. voltage-gated K+ channel (VGKC) opens à membrane repolarisation
  4. Ca2+ influx through VGCCs à vesicle exocytosis
  5. synaptic vesicle associated (SV2A) protein (docking protein) allows vesicle (containing glutamate) attachment to the presynaptic membrane
  6. glutamate is released into the synapse
  7. glutamate activates excitatory post-synaptic receptors (e.g. NMDA, AMPA & kainate receptors)
1068
Q

What are the 3 types of anticonvulsants that target glutamatergic synapses?

A
  • voltage-gated sodium channel blockers
  • voltage-gated calcium ion blockers
  • glutamate exocutosis and receptors
1069
Q

What characteristics are shared by all anticonvulsants used in epilepsy?

A
  • fast onset with a long half-life/time of action
1070
Q

Name some voltage-gated sodium channel blockers used in epilepsy.

A
  • carbamazepine
  • lamotrigine
1071
Q

State the mechanism of action, uses and possible issues with carbamazepine.

A
  • mechanism = stabilises the inactive state of the Na+ channel -> more likely to remain in an inactive state -> reduced neuronal activity
  • uses = tonic-clonic and partial seizures
  • issues = enzyme inducer of the cytochrome P450 pathways -> reduces the activity of a number of other drugs and also can cause potentially lethal skin side effects (Steven Johnson Syndrome & toxic epidermal necrolysisEN) in individuals with HLA-B*1502 allele -> more common in Chinese people
1072
Q

State the mechanism of action, uses and possible issues with lamotrigine.

A
  • mechanism = directly inactivates Na+ channels -> reduces glutamate neuronal activity
  • uses = tonic-clonic and absence seizures
  • issues = no notable issues
1073
Q

State and describe a voltage-gated calcium ion channel.

A

- ethosuximide is a T-type calcium channel (predominantly found in neurones) antagonist -> T-type and not L-type as targetted by hypertensive treatments

  • mechanism = T-type Ca2+ channel antagonist -> reduces activity in relay thalamic neurones -> prevents the propagation of the AP -> decreased neurotransmission
  • uses = absence seizures
1074
Q

Name some glutamate exocytosis and receptor targetting anticonvulsants.

A
  • levetiracetan
  • topiramate
1075
Q

State the mechanism of action and uses of levetiracetam.

A
  • mechanism = binds to synaptic vesicle associated protein (SV2A) -> prevents and reduces glutamate exocytosis
  • uses = myoclonic seizures
1076
Q

State the mechanism of action and uses of topiramate.

A
  • mechanism = inhibits NMDA and kainate receptors and also affects VGSCs & GABA receptors -> affects a few receptors so a bit of a ‘dirty drug’
  • uses = myoclonic seizures -> MOST COMMON USES IS NEUROPATHIC PAIN AND NOT EPILEPSY
1077
Q

Describe GABA neurotransmission at the gabaergic synapse.

A
  1. GABA can be released tonically (released naturally without stimulation) or following neuronal stimulation
  2. GABA activates inhibitory post-synaptic GABAA receptors
  3. GABAA receptors are chloride (Cl-) channels -> leads to membrane hyperpolarisation
  4. GABA is taken up by the GAT transporter
  5. GABA is then metabolised by GABA transaminase (GABA-T)
  6. GABA is broken down back into glutamate
1078
Q

What is the mechanism and uses of diazepam?

A

o mechanism = a positive allosteric modulator of GABA-A receptors -> doesn’t activate the channel alone, but it increases the activity of the GABA that binds to the channel - increases GABA activity, but doesn’t increase the amount of GABA in the synapse

  • GABA receptor activated more -> PAM -> increases GABA-mediated inhibition

o uses = status epilepticus -> administrated as a rectal gel

1079
Q

What is the mechanism of action and uses of sodium valporate?

A

o mechanism = inhibits GABA transaminase enzyme -> increases GABA-mediated inhibition

  • has dual mechanism of action:
  • > inhibits GABA-T -> increases the amount of GABA
  • > reduces the amount of glutamate being formed

o uses = all forms of epilepsy apart from status epilepticus due to diazepam being so more effective

1080
Q

What are the pharmacological monotherapy treatments for tonic-clonic seizures?

A
  • carbamazepine
  • lamotrigine
  • valproate
1081
Q

What are the pharmacological monotherapy treatments for absence seizures?

A
  • ethosuximide
  • lamotrigine
  • valproate
1082
Q

What are the pharmacological monotherapy treatments for tonic/atonic seizures?

A
  • valproate
1083
Q

What are the pharmacological monotherapy treatments for myoclonic seizures?

A
  • levetiracetam
  • valproate
  • topiramate
1084
Q

What are the pharmacological monotherapy treatments for status epilepticus?

A
  • diazepam
1085
Q

What are the pharmacological monotherapy treatments for partial/focal seizures?

A
  • carbamazepine
  • lamotrigine
  • levetiracetam
  • valproate
1086
Q

What type of seizure is characterized by sudden muscle stiffening?

a. absence
b. atonic
c. tonic/atonic
d. tonic-clonic
e. myoclonic

A
  • c
1087
Q

What type of seizures are characterized by breif staring episodes with behavioural arrest?

a. absence
b. atonic
c. tonic/atonic
d. tonic-clonic
e. myoclonic

A
  • a
1088
Q

Name the anticonvulsant that will increase the synaptic GABA levels.

a. ethosuximide
b. diazepam
c. carbamazepine
d. lamatrigine
e. valproate

A
  • e
1089
Q

What anticonvulsant would you prescribe for a patient suffering from atonic seizures?

a. ethosuximide
b. diazepam
c. carbamazepine
d. lamatrigine
e. valproate

A
  • e