Theme II: Pharmacology in dental practice Flashcards

1
Q

What neurotransmitters are involved in parasympathetic & sympathetic autonomic system (post and pre ganglions), and the somatic system
What are 2 exceptions

A

1-Parasympathetic: ACh released from long pre & short postganglion.
2-Sympathetic: ACh in preganglion, and NA in postganglion
3-Somatic: ACh from neuron onto NMJ.

  • Sweat glands are controlled by sympathetic system but both ganglions release ACh.
  • For adrenal glands, neuron releases ACh and synapses onto adrenal medulla (not postganglion) which secretes adrenaline into blood (acts as a hormone)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Describe the mechanisms of acetylcholine neurotransmission (synthesis, storage, release, termination)

A

1-Synthesis: Choline is the precursor. It enters the preganglion and is converted to ACh by CAT enzyme
2-Stored in vesicles. Depolarisation and influx of Ca causes vesicle fusion with membrane and ACh spills out into synaptic cleft
3-Released by exocytosis.
-ACh binds to muscarinic or nicotinic receptors on post ganglion or target
4-ACh is metabolised in the synaptic cleft by ACh esterase enzyme into choline and can be reused.

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

Where are muscarinic receptors located, what type of receptor are they, what do they bind and what systems do they control

A
  • On target organs, at postganglionic parasympathetic synapses, controlling parasympathetic system.
  • Mainly in brain, also in heart, lungs, bladder, intestine
  • Bind acetylcholine
  • G protein coupled receptors so slow response. Metabotropic
  • M 1, 2 and 3 receptors
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

What do muscarinic agonists do. Give examples of their functions.

A

-Activate parasympathetic system = Parasympathomimetics

  • Treating glaucoma by contracting ciliary muscle and decreasing focal length to drain aqueous humour.
  • Treating xerostomia by increasing salivation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What do muscarinic antagonists do. Give specific examples

A

-Block muscarinic receptors so block parasympathetic activity= Parasympatholytic

  • For dilating pupils for eye surgery
  • Decreasing saliva for oral surgery, or for anaesthesia to prevent respiratory secretions getting into lungs due to lost cough reflex
  • Treating bradycardia by increasing cardiac output
  • Treating asthma by causing bronchodilation
  • Treating motion sickness by decreasing gastric motility.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

-What responses do neuronal type nicotinic agonists and antagonists cause.

A
  • these receptors are located on both sympathetic and parasympathetic postganglia so both systems are activated,
  • Both cause autonomic confusion. Therefore they are not used
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What do muscle type nicotinic agonists and antagonists do.

A

(Located on NMJ) Agonists and antagonist cause paralysis for surgery:

  • Agonists: increase effect of ACh neurotransmission so depolarisation which causes muscle contraction. As this synthetic agonist cannot be metabolised, it builds up and the fiber persistently depolarises and there is a loss of further excitability. Results in a depolarisation block.
  • Antagonists: Hyperpolarization so no excitability. It is a non-depolarising block.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Other than receptor interaction, how else can drugs affect acetylcholine

A
  1. ACh release from ganglion can be inhibited, causing autonomic and motor paralysis. Used in botox and to treat local muscle spasm.
  2. Metabolism can be inhibited. Anticholinesterases inhibits AChE enzyme which metabolises ACh, therefore transmission increases.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

How is noradrenaline (NA) made and how neurotransmission occurs

A
  • Tyrosine taken up into postganglion and is converted to DOPA, DA then NA.
  • Stored in vesicles to protect against interneuronal enzymes breaking it down.
  • Fuses with membrane and released into synapse by exocytosis in response to AP.
  • Acts on alpha and beta adrenoreceptors on tissues of sympathetic system
  • Termination: Uptake back into neuron and metabolised into amines by monoamine oxidase or is recycled.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What is different about a2 adrenoreceptors to the other types.

A

-a2 is a presynaptic receptor located on post ganglion. It turns off further release of NA, whereas the other receptors are located on tissues, control the sympathetic system

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

What are the functions of the different types of alpha and beta adrenoreceptors. Agonists.

A
  • a1: Smooth muscle contraction & vasoconstriction. (Eg. treats anaphylactic shock by increasing BP and used with LA)
  • a2: on presynapse so inhibits further NA release so decreases sympathetic activity. (Eg. Vasodilation treats hypertension)
  • B1: Heart. Increases cardiac output to treat anaphylaxis or heart failure
  • B2: Lungs. Bronchodilation to treat anaphylaxis or asthma (salbutamol).
  • B3: increases lipolysis so used for fat loss and muscle gain
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Examples of noradrenic antagonists. Mention possible side effects too. Why a2 antagonist not used

A

-A1: vasodilation to treat hypertension
-A1: relaxes smooth muscle in bladder neck so increases urination
BUT can cause postural hypotension where low BP & dizzy when stand up

-A2: it stops the inhibition of NA release so can increase unwanted anxiety. So not used

  • B1: decreases cardiac output to treat hypertension ( BUT can cause rebound hypertension on withdrawal)
  • B2: ciliary muscle contraction and decreased intraocular pressure to treat glaucoma
  • B1 and B2 (non-selective): decreases cardiac output for hypertension but also causes bronchoconstriciton so shouldn’t be used for asthmatics.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Other than receptor interactions, how else can noradrenaline neurotransmission be increased or decreased.

A

1- Use a false substrate to inhibit enzyme activity for producing NA. Decreases transmission so can treat hypertension.
2-Inhibiting NA storage in vesicles decreases NA available for release. Less neurotransmission so can treat hypertension.
3-Inhibiting release also decreases neurotransmission.
4-Reptake into presynaptic ganglion can be blocked which prolongs the action of NA in the synapse. Antidepressant effect.
5-Inhibiting the metabolism enzyme (monoamine oxidase) increases NA available for release.

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

Difference between analgesics and anaesthetics. Difference between local and general anaesthetics

A
  • Anaesthesia: drugs used to prevent pain for a limited time for surgical procedures
  • Analgesics: drugs used to control pain. No total loss of feeling (eg. morphine)
  • Local: prevents pain in a localised area (eg. lidocaine)
  • General: loss of feeling and also loss of consciousness (eg. propofol)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What factors affect the efficiency of inhalation anaesthetics. How they are eliminated

A

1-Blood/ gas partition coefficient = blood solubility. Low solubility means rapid induction & recovery as less drug is needed to be inhaled to produce equilibrium. It means less retention of the drug following distribution.
2-Oil: gas partition coefficient= lipid solubility. High solubility means high potency
3-Vascularisation - Good blood flow in brain means high levels of drug. Poor blood flow in body fat means drug doesn’t accumulate.
4-Ventilation rate: high rate means high rate of removal.

Most drug is eliminated in the lungs, little is metabolised via hepatic metabolism as it is toxic (halothane is metabolised a lot by liver so toxic)

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

Side effects of inhaled anaesthetics

A
  • Malignant hyperthermia: hypermetabolism, increased sympathetic activity
  • Hypotension, vasodilation, decreased cardiac output
  • Respiratory depression
  • Hepatic toxicity
  • Decreased glomerular filtration and urine output (although not usually a problem due to the decreased cardiac output)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

How do local anaesthetics work.

A

-Block the electrical signalling in neurons by blocking voltage gated Na channels so no AP can be propagated. So no pain felt:

  • The weak base is in unionised form when it crosses the membrane as outside a cell the pH is 7.4. So it is lipid soluble.
  • Inside of the cell is slightly acidic so it becomes ionised which allows it to bind to the alpha subunit of the voltage gated sodium channel.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

What nerve fibres are more sensitive to local anaesthetics

A
  • Small myelinated fibres (nociceptive fibres) are blocked more effectively.
  • Then non-myelinated axons
  • Large myelinated fibres (motor axons) are less sensitive
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Unwanted side effects of local anaesthetics

A
  • Occur if drug escapes into systemic circulation
  • In the CNS it can cause confusion and agitation
  • In the cardiovascular system it causes hypotension, decreased cardiac output, inhibition of sodium conduction in cardiac tissue.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

Why are anaesthetics not very effective in infected or inflamed tissues

A

-The local tissue pH decreases in an infected area so when injected, the weak basic drug exists in an ionised form meaning it is less lipid soluble and difficult to penetrate the membrane.

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

Difference between pain and nociception

A
  • nociception is the neural encoding of a stimuli. It is the process whereby noxious stimuli are sensed by nociceptors in the peripheral tissues and transmitted to the CNS. Nociception leads to pain.
  • Pain is a subjective experience where it is how someone processes the stimulus which will be influenced by emotions, opinions, sensation and the circumstance.
  • Nociception is always consistent but pain will differ between people depending on the circumstance
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

Nociception pathway

A

1-Primary afferent neurons travel from peripheral tissue to CNS
- The nociceptor fibres are C, A beta and A delta which detect different types of pain and synapse in different lamina layers in the dorsal horn of the spinal cord.
2- Secondary afferents go to the brain stem
3-Neurones then go the thalamus and to different systems in the brain so that efferents can be activated
4- Efferents go from CNS to PNS, to allow the body to respond in a specific way.

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

Acute and chronic pain difference

A
  • Acute pain: Excessive mechanical/ thermal stimulation increases transmission and hypersensitizes the nerve fibres. Signals prompt action to relive the pain to prevent harm and to protect. Short-term.
  • Chronic pain is often a consequence of stimulation of chemical mediators. Stimuli not always there and nociception pathway not being signalled to but is still responding. Disordered pathway that continues when it shouldn’t.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

Difference between C, A delta and A beta fibres

A
  • A beta: Thick and myelinated. Mechanoreceptor - touch and pressure
  • A delta: Myelinated and rapid conduction. Sharp localised pain. Nociceptor, mechanoreceptors.
  • C fibers: Thinnest, non-myelinated so low conduction velocity. Dull and achy pain. Nociceptor, mechanoreceptor, thermoreceptor. Heat
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

What mediators stimulate nociceptive endings. What analgesics can inhibit these and therefore inhibit the nociceptive pathway

A
  • 5-HT, Kinins, metabolites of intermediary metabolism (eg. lactic acid), capsaicin, prostaglandin.
  • Opioids and NSAIDs decrease prostaglandin so reduces inflammation and reduces sensitivity of nociceptors to the mediators.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

How do NSAIDs work.

A
  • analgesics, reduce inflammation, anti platelet, antipyretic.
  • They irreversibly inhibit COX 2 which inhibits prostaglandin production, which are made at damaged sites to induce inflammation to deal with injury. Decreased prostaglandin means nociceptors are less sensitive to the effects of chemical mediators (5-HT, kinins)

-Drugs need to be selective for COX2 to prevent GI effects if COX1 was to be inhibited (this would decrease stomach mucous causing ulcers)

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

What are the NSAIDs aspirin, ibuprofen and paracetamol used for

A

-Aspirin: Analgesic and anti-inflammatory due inhibition of COX and prostaglandin which makes nociceptors less sensitive to mediators and inflammation.
Antipyretic as decreases prostaglandin that raises body temperature.
Antiplatelet as it acetylates platelet COX so less thromboxane A2, and epithelium makes more prostacyclin.
For mild analgesia.

-Ibuprofen: same 4 effects as aspirin, but maybe not anti platelet.
First choice in inflammatory joint disease. Less gastric irritation, more effective and better tolerated than other NSAIDs.

-Paracetamol: Analgesic and antipyretic effects, but not anti-inflammatory or platelet.
Effective in mild analgesia, less so in inflammatory conditions.
Overdose causes heptatoxicity

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

How do opioid analgesics work. Receptors. How are they administered and why

A
  • Agonists on mainly u opioid receptors which are G-protein couples receptors on neurons that regulate pain perception.
  • Binding inhibits adenylate cyclase, increasing potassium influx so decreases Ca and causes hyperpolarization which turns off neuronal excitability.
  • There are u, delta and k opioid receptors but u is the main target for opioid drugs
  • Causes euphoria and analgesia
  • Oral or rectal but usually IV or intramuscular due to unreliable gut absorption and extensive first pass metabolism
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

Side effects of opioid drugs

A
  • Drowsiness and sedation
  • Respiratory depression, cough suppressive, nausea
  • Tolerance: patient no longer responds to it in the same way so needs a higher concentration to get the same analgesic effect.
  • Dependance: sudden withdrawal after chronic treatment causes sweating or tremors for example
  • Constipation (inhibition of smooth muscle in gut)
  • Allergic reactions, urinary retention, pinhole pupils
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

Give examples of strong and weak opioids and when they are used. Which one is a naturally occurring opioid

A

1-Strong: used for moderate to severe pain.

  • Morphine-for severe pain relief, terminal care
  • Pethidine- more lipid soluble, rapid onset, short duration, less constipation.

2-Weak: used for mild to moderate pain.

  • Dextroproxyphene
  • Dihydrocodeine- cause nausea and constipation

Morphine is naturally occurring.

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

What does chemotherapy and effective chemotherapeutic agents mean. What is selective toxicity

A

-Chemotherapy is the elimination of invading cells or microbes

  • Effective Chemotherapeutic agents are selectively toxic, meaning the drug is toxic to the invading cells but non-toxic to the host cells
  • Exploitable differences between invading species and host such as differences in metabolism and cell structure. Drug targets uniques properties to invading species
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

What are the 4 major mechanisms of antibacterial action and the drugs used

A

1-B lactam ring of penicillins target peptidoglycan. Inhibits transpeptidase which is used in cross-linking
2-Ribsomes are targeted to inhibit protein synthesis. Tetracycline binds to 30S subunit which stops initiation of synthesis. Macrolides bind to 50S to block translocation
3-DNA gyrase used for preventing supercoiling is inhibited by fluoroquinolone, so DNA replication is inhibited
4-Sulphonamides and trimethoprim inhibit folate metabolism so inhibit DNA synthesis.

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

How does acyclovir treat viral infection

A
  • Treats recurrent human herpes simplex type 1 and 2
  • phosphorylated in infected cells by thymidine kinase to the active triphosphate form
  • This is a nucleoside analogue so terminates the chain, inhibits DNA polymerase and viral DNA synthesis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

Difference between anxiolytics, sedatives, hypnotics and general anaesthetic

A
  • Anxiolytics alleviate fear and anxiety
  • Sedatives alleviate fear and anxiety. Also produce a degree of amnesia and analgesia
  • Hypnotics reduce peripheral awareness and used to treat insomnia. Cause drowsiness and induce sleep
  • General anaesthetics is sleep. Unconscious.

All can cause similar effects depending on the dose

35
Q

What is anxiety

A
  • Feeling of uneasiness, apprehension and tension about what is to come.
  • Increased sympathetic activity (palpitations, sweating, flushed, dizzy)
  • Excessive anxiety becomes disabling for the patient.
36
Q

What is the similarities and differences between barbiturates and benzodiazepines

A
  • They are anxiolytics. CNS depressants.
  • They are both allosteric modulators of GABA receptors which is the principal inhibitory neurotransmitter of the CNS. Increase Cl.
  • Benzodiazepines bind to gamma and alpha subunit of GABA. They increase GABA’s affinity for the receptor which increases frequency of channel opening so increase conductance. BDZ doesn’t open the channel itself. They are safer
  • Barbiturates bind to the Beta subunit and increase the duration of chloride ion channel opening, so can become toxic.
37
Q

How do benzodiazepines work. What are they used for

A
  • Bind to allosteric site on gamma and alpha subunit of GABA receptor. This increases the receptor’s affinity for GABA and increases the opening frequency of the chloride channel
  • Increased conductance of Cl so increases the inhibitory neurotransmission process, hyperpolarizing the postsynaptic cell.
  • Decrease sympathetic activity so treat anxiety, panic disorders, phobias and insomnia.
  • Causes sedation, decreases time taken to sleep and increases sleep duration. Decreases dreaming and deep sleep
  • Muscle relaxation and anticonvulsant effects for epilepsy
38
Q
  • Difference between short and long lasting benzodiazepines.

- Unwanted side effects of benzodiazepines

A
  • Short lasting (temazepam) are metabolised by glucuronidation (phase 2) to inactive compounds. Short half life
  • Long lasting are metabolised to active metabolites. Long half life so cause hang over effect (Diazepam)
  • Unwanted effects include drowsiness, confusion, amnesia. Interact with alcohol to cause respiratory depression. Tolerance and dependance. Sexual fantasies.
  • Short acting ideal for old people as they lack oxidation for phase 1 reactions
39
Q

How do antipsychotics work, and their side effects

A
  • Treat schizophrenia/ psychosis (disorganised behaviour, hallucinations, paranoia, blunted emotions, loss of energy, seeing/ believing things that are not real)
  • Dopamine is increased in these patients so these drugs are D2 receptor antagonists to counteract the increase in DA function.
  • It is the mesocortical/ mesolimbic pathway that is dysregulated which is involved in mood, reward, addiction.
  • BUT inhibition of other pathways will increase prolactin and cause tremors.
  • Non-selective drugs can also bind to non-dopaminergic receptors (H1, M1 and a1) which cause side effects such as sedation
40
Q

What are 1st and 2nd generation antipsychotic drugs

A
  • 1st generation (or typical antipsychotics) are D2 receptor antagonists to cause the therapeutic effect of reducing reward etc. BUT the other DA pathways are inhibited causing side effects such as EPS, tardive dyskinesia and hyperprolactinemia.
  • These drugs also have affinity for non-dopaminergic receptors (H1, M1 and a1) causing sedation, dry mouth and postural hypotension for example.
  • These drugs also have limited efficacy against negative symptoms of psychosis and patients can relapse.

-2nd generation: better because they have fewer side effects as they dissociate faster from the D2 receptor, and they are better at treating negative symptoms (blunted emotions, loss of energy, social withdrawal). And no affinity for M1, M1, a1.

41
Q

How do antidepressants work. Describe what tricyclic antidepressants (TCAs), Selective serotonin/ noradrenaline reuptake inhibitors (SSRIs, SNRIs), and monoamine oxidase inhibitors do

A

-Treat reduced levels of 5HT and NA in depression.

  1. TCAs inhibit the reuptake of 5HT and NA by blocking transporters so it increases neurotransmission.
    - However, have affinity for the H1, M1 and a1 receptors so cause side effects such as sedation, dry mouth, postural hypotension. And effects are delayed so feel worse before better.
  2. SSRIs have same efficacy as TCAs but are selective so don’t bind to the M1 etc. receptors so no severe side effects. Block 5HT and NA transporters and reuptake, increasing neurotransmission.
  3. Monoamine oxidase inhibitors block the metabolism of 5HT and NA so more is recycled so neurotransmission increases
42
Q

What is the neurotransmission process of dopamine. What are the 3 main pathway in the brain and what it controls

A

-Tyrosine enters presynapse and converted to DOPA then DA in 2 enzymatic steps. Stored in vesicles and released by exocytosis. Acts on 4 dopaminergic receptors. Recycled or metabolised in the presynaptic ganglion.

1- Nigrostriatal pathway: controls fine movements. Reduced DA (in Parkinson’s) causes tremors, muscle rigidity and loss of facial expression
2-Mesocortical/ mesolimbic pathway: controls mood and reward so increased DA here causes psychosis
3-Tuberonifundibular: DA also acts as a hormone when released form hypothalamus and acts on ant pituitary. Inhibits prolactin

43
Q

What is the neurotransmission process of 5HT and what does it control

A
  • Happy hormone
  • The precursor tryptophan enters presynapse and converted to 5HT in 2 enzymatic steps. Stored in vesicles and released by exocytosis. Acts on 5HT receptors on postsynaptic ganglion. Taken back up into the preganglion by serotonin transporter and is either recycled or metabolised.
  • Low levels are linked with depression
44
Q

What is the difference between neurotransmitters and hormones (where they are made and where they go)

A
  • Neurotranmitters: chemical substances synthesised in neurones and secreted directly into adjacent neurons or tissues
    -Hormones: made by endocrine glands and secreted into the blood stream so is carried to non-adjacent sites in the body.
    [And only cells with specific receptors will respond]
45
Q

How is cortisol made and what are its effects on the body. What do glucocorticoids do

A
  • Stress causes release of CRH from hypothalamus
  • This causes release of ACTH from anterior pituitary
  • Cortisol is then released from the adrenal cortex
  • Increases blood glucose (gluconeogenesis) Less stored in tissues.
  • Suppresses immune system so glucocorticoids (increase cortisol) are used for treating inflammatory conditions such as asthma.
46
Q

What is Cushing’s syndrome. Symptoms. Causes. How is it treated

A
  • hypercortisolemia
  • Caused by tumour in adrenal or pituitary gland, or is a side effect from prolonged glucocorticoid therapy
  • Symptoms include fat deposition in shoulders, waist and face. Thinning of skin and stretch marks. Bruising. Depression.
  • Treated by removal of tumour, or metyrapone which inhibits cortisol synthesis by blocking the 11B hydroxylating enzyme
47
Q

How and where is insulin made. What are its effects

A

-Made in Beta cells of the islets of Langerhans in the pancreas

1 -When blood glucose is high, glucose is taken up into the B cell and enters glycolysis to make ATP.
2-ATP blocks potassium channels, depolarising the cell.
3- Influx of Ca causes an increase in insulin release from storage granules
4-Insulin lowers blood glucose (glycogenesis)
-Lipolysis, protein synthesis also

48
Q

What is type 1 and 2 diabetes. Difference between the two.

A
  • consequence of low insulin, increasing blood glucose as it cannot be stored. 2 types:
  • Type 1: Insulin dependant diabetes. The body attacks B cells. Loss of these cells means hypo secretion of insulin. [AUTOIMMUNE]

-Type 2: Non-insulin dependant. Insulin receptors become hypersensitive so don’t work properly. Enough is made it is just not having the right effect. (caused by obesity) [METABOLIC]

49
Q

How is type 1 & 2 diabetes treated

A
  • Type 1 treated with exogenous insulin
  • Type 2 treated with sulphonylureas which block potassium channels of pancreatic B cells which depolarises it. Increases Ca influx which stimulates insulin secretion, (independent of glucose levels).
50
Q

How are oestrogen and progesterone synthesised. What are their effects

A
  • Hypothalamus releases GRH.
  • Anterior pituitary releases FSH which causes secretion of oestrogen from Graafian ovarian follicles. Oestrogen sensitises LH in pituitary which then causes release of progesterone
  • The 2 hormones can negatively feedback on the cycle.
  • Oestrogen: causes proliferation of endometrium to prep it for implantation
  • Progesterone: further preps the endometrium and maintain it during pregnancy.
51
Q

How do contraceptives work.

A
  • exogenous oestrogen and progesterone taken to target the negative feedback system of their production so secretion and ovulation is inhibited. Mimics the pregnant state so no fertilisation
  • Oestrogen inhibits FSH release from pituitary so prevents ovarian follicle development
  • Progesterone inhibits LH from pituitary and thickens the cervical mucus so passage of sperm to ovum is difficult
  • Combined pill most effective
52
Q

What is the process of haemostasis/ thrombosis. Difference between the two.

A

stopping blood flow by vasoconstriction, platelet aggregation (plug) and coagulation:
1-Constriction reduces blood flow and pressure
2-Plug forms when collagen is exposed. Platelets adhere to collagen by binding to vWF
3-Plug converted to clot when fibrin is made from fibrinogen in the blood clotting cascade

-Haemostasis is a normal process for wound healing and to limit damage. However thrombosis is the unwanted formation of a thrombus (eg. in atherosclerosis) It occludes the arteries and can lead to a heart attack or pulmonary embolism.

53
Q

What are the main factors in the blood clotting cascade. What is produced

A
  • Intrinsic pathway initiated by exposed collagen. Factors 12, 11, 9.
  • Extrinsic pathway is initiated by damaged tissue. Factors 3 & 7.
  • Common pathway includes factor 10. Prothrombin makes thrombin (active 2). Thrombin converts fibrinogen to fibrin.
  • Fibrin is the clotting agent which forms next to platelets to form the clot.
  • Factor 13 cross links the fibrin
54
Q

How does heparin work. Overdose effect and treatment

A
  • It is an anticoagulant. It binds to antithrombin III which inactivates thrombin and clotting factors, so no clot forms.
  • IV or subcutaneous
  • Overdose causes bleeding which is treated by heparin antagonist (protamine) to dissociate the heparin-antithrombin complex
55
Q

How does warfarin work. Overdose treatment

A
  • It is a vitamin K antagonist. It degrades clotting factors dependant on vitamin K as they cannot be carboxylated and activated. (2,7,9,10)
  • Taken orally
  • 1 to 2 day lag period as high plasma bound, so takes time to have full effect
  • Overdose causes bleeding. Treated by taking vitamin K
56
Q

Why warfarin and heparin used in combination to start with

A
  • Warfarin has a lag time of 1-2 days so heparin covers the lag period as it is fast acting. Heparin can then be withdrawn after this period, and then the oral anticoagulant can be taken.
57
Q

Role of vitamin K in coagulation

A
  • Needed for gamma carboxylation of glutamic acid residues of prothrombin
  • Many clotting factors are dependent on K (2,7,9,10) so it is essential for the blood clotting cascade to form fibrin and a clot
58
Q

How aspirin interacts with warfarin

A
  • It increases risk of bleeding as it is an anti-platelet agent.
  • It inhibits COX involved in prostaglandin production so blocks synthesis of prostaglandin and so inhibits platelet-derived thromboxane A2 (used for aggregation) and endothelium-derived prostacyclin (inhibits aggregation)
  • Endothelium can make new COX but platelets cannot so net effect of prostacyclin increase which inhibits aggregation.
59
Q

Interaction of anticoagulant and antibiotics

A
  • Metranizadole, trimoxazole and tetracycline inhibits P450 so inhibits metabolism of anticoagulants so enhance anticoagulant activity so increase risk of bleeding.
  • Rifampicin induces P450 so decreases its activity
60
Q

Interaction of antiplatelet drugs (aspirin) and NSAID

A
  • NSAID interacts with the antiplatelet function of aspirin
  • aspirin increases the risk of mucosal bleeding after minor dental surgery so using in combination with NSAIDs are contradictive in inflammation management after treatment as it will increase risk of bleeding.
61
Q

target of tricyclic antidepressants (eg. amitriptyline)

A

-Blocks reuptake of 5HT and NA, so increases levels

62
Q

How do macrolide and tetracycline antibiotics work.

A
  • Target ribosomes and protein synthesis
  • Tetracycline - binds to small 30S ribosome subunit so stops initiation of protein synthesis
  • Macrolide- binds to 50S and blocks translocation
63
Q

How do fluroquinolones work

A
  • Target nucleic acid synthesis and DNA replication

- Inhibit DNA gyrase so supercoiling is not prevented and DNA can no longer be made

64
Q

How do sulphonamides and trimethoprim work

A
  • Target folate metabolism and so DNA synthesis
  • Sulphonamides: inhibit dihydropteroate
  • Trimethoprim: inhibits dihydrofolate reductase
65
Q

What is the method of action polyenes for treating fungal infection. Give examples of drug names.

A
  • Polyenes have high affinity to ergosterol in fungal cell membrane so bind and forces the membrane apart causing it to die
  • Nystatin, Amphotercin B
66
Q

What is the method of action of azole antifungals. Give example of drug names

A

-Fungal cell membranes contain ergosterol (instead of cholesterol in eukaryotes) so this is a target.
-Azoles block CYP450 in synthesis of ergosterol so inhibits membrane synthesis and so no replication
Eg-Ketoconazole, miconazole, clotrimazole

67
Q

What are cholinergic receptors

A
  • receptors that get activated when they bind acetylcholine.
  • There are 2 types: nicotinic (muscle & neuronal type) and muscarinic receptors
68
Q

Where are nicotinic receptors located, what type of receptor are they, what do they bind and what systems do they control

A
  • Ligand gated ion channels so fast response.
  • Bind ACh so causes channels to open and depolarisation for example.
  • Neuronal-type receptors: found in brain and on sympathetic and parasympathetic postganglia.
  • Muscle-type receptors: located on NMJ in somatic system
69
Q

Do nicotinic or muscarinic receptors have a faster response

A

-Nicotinic ACh receptors have a faster response. They are ligand gated ion channels (ionotropic) whereas muscarinic are G protein coupled.

70
Q

What inhibits metabolism of ACh in the synaptic cleft and what effect does it have on the autonomic and somatic nervous systems.

A
  • Anticholinesterases inhibits AChE enzyme involved in metabolism, so ACh builds up.
  • Increased transmission at muscarinic receptors at parasympathetic postganglionic synapses increases saliva, bradycardia, vasodilation, pupil constriction etc.
  • On nicotinic receptors in NMJ, it causes increased muscle twitching and at large dose causes a depolarising block so induces paralysis for surgery.
71
Q

Difference in metabolism of ACh and NA after release into synapse

A
  • NA is taken up back into the postganglion and metabolised into amines or is recycled.
  • ACh is metabolised in the synapse cleft by ACh esterase
72
Q

What does metyrapone treat and how

A

-Treats cushings syndrome by inhibiting the 11B hydroxylating enzyme involved in cortisol synthesis, so blocks cortisol synthesis

73
Q

What measures the time for a clot to form

A

Partial thromboplastin time (PTT)

74
Q

What molecules induce and inhibit platelet aggregation

A
  • Induced by serotonin, ADP, thromboxane A2

- Inhibited by Nitric oxide and prostacyclin

75
Q

List the drug classes can be used for analgesia

A
  • NSAIDs
  • Opioids
  • General & local anaesthetics
  • Anxiolytics
  • Non-medical= alcohol, nicotine, caffeine, cocaine

[-They inhibit the process of pain by inhibiting the nociceptive pathway]

76
Q

What do cyclooxygenase 1 & 2 do.

A
  • COX1 = stimulate prostoglandins that secrete mucous in the stomach. So inhibition of COX1 causes unwanted ulcers.
  • COX2 = stimulate prostaglandins that activate inflammatory cells, so inhibition reduces inflammation, pain, fever, platelets.
77
Q

What are prostaglandins. How are they produced

A
  • They are a group of lipid compounds called eicosanoids which are made at damaged sites to induce inflammation to deal with injury. And they also increase stomach mucous.
  • Arachidonic acid is converted to prostaglandins by COX. Thromboxane (platelet aggregation) and prostacyclin (platelet inhibition) are produced in the pathway too
  • made form fatty acid metabolism
78
Q

what are the opioid neurotransmitters

A

enkephalins, endorphins, dynorphins

79
Q

What channels do general anaesthetics target

A
  • Ligand gated ion channels
  • Excitatory, so antagonists used= NMDA, ACh, 5HT receptors.
  • Inhibitory= GABA, glycine. So agonists used.
80
Q

What is the structure of a voltage gated ion channel

A
  • beta 1 & 2 subunits anchor the alpha subunit in the lipid membrane.
  • The a unit is a single polypeptide with the channel in the centre. It has voltage sensors that change orientation when voltage varies, controlling opening and closing.
81
Q

What are the 3 components of the structure of local anaesthetics and their function

A
  • Aromatic lipophilic group = ensures lipid solubility
  • Ester/ amide bond =makes duration limited as hydrolysed.
  • Amine side chain = the basic element, so ensures the molecule is ionised in the acidic cell to bind to the receptor
82
Q

What is the pH inside and outside of a cell

A
Outside = 7.4
Inside = 7 or slightly acidic
83
Q

What are GABA receptors

A
  • they are the principal inhibitory neurotransmitter and receptor in the CNS
  • made of alpha, beta and gamma subunits with a chlorine channel complex.
  • when open, influx of chlorine causes hyper polarisation.
84
Q

which inhaled general anaesthetic has most hepatic toxicity.

A
  • Halothane

- Used for induction and maintenance of general anaesthesia