CNS Pharmacology Flashcards

1
Q

 Who was the first to demonstrate the usefulness of nitrous oxide as an analgesic in the process of tooth extraction?

A

Horace Wells

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

William Morton was known for his use of ether as a volatile anaesthetic during tooth extraction, T or F?

A

T

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

What was J. Simpson’s contribution to general anaesthesia?

A

He was the first person to demonstrate that chloroform could render patients’ unconscious. Was also deemed the co-discoverer of general anaesthetics

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

What are the two classes of chemical general anaesthetics?

A

Inhalational and intravenous

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

What type of general anaesthetic is NO2?

A

Inhalational chemical general anaesthetic

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

Give an example of some intravenous chemical general anaesthetics?

A

Halogenated hydrocarbons such as isoflurane. Also steroids and barbiturates such as thiopental are also intravenous

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

Give some examples of physical general anaesthesias?

A

Low pressure, hypothermia

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

What is meant by the term surgical anaesthesia?

A

The point at which patients become unresponsive and unable to sense pain

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

What evidence is there to suggest that anaesthetics aren’t agonists acting on a receptor?

A

Steep dose-response curve which isn’t seen in agonist binding. Low pressure can also induce anaesthesia

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

What is significant about the dose-response curve of general anaesthetics?

A

They have very steep dose-response curves

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

What property of general anaesthetics is very closely related to their potency?

A

Lipid solubility

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

Describe what is meant by a MAC value and how this relates to the potency of the general anaesthetic?

A

MAC value – minimum alveolar concentration of anaesthetic to alleviate a response to surgical incision in 50% of patients. The lower the MAC value the higher the potency of the general anaesthetic

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

What is meant by the oil:gas partition coefficient?

A

The ease with which a substance dissolves, or partitions into oil

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

Drugs with a low oil:gas partition coefficient will have a high potency and thus a lower MAC value, T or F?

A

F – drugs with higher oil:gas partition coefficients will have a high potency and lower MAC values

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

What is meant by the Meyer-Overton rule?

A

The effect of an anaesthetic is proportional to the molar concentration of the agent in lipid

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

What is the effect of general anaesthetics on the plasma membrane?

A

General anaesthetics lead to a volume expansion in the plasma membrane

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

What is the impact of general anaesthetics on lipid fluidity?

A

General anaesthetics increase lipid fluidity

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

What aspects of the general anaesthetic mechanism of action are explained by the lipid theory?

A

As antibiotics are thought to lead to a volume expansion of the plasma membrane and an increase in lipid fluidity the lipid theory of antibiotic action accounts for the meyer-overton rule. In addition, the effect of pressure is also explained as we know that a decrease in pressure results in a volume expansion of the lipid membrane and thus will give rise to anaesthesia. Finally this also accounts for the fact that such a diverse range of compound can have anaesthetic effects

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

What aspects of the general anaesthetic mechanism of action aren’t explained by the lipid theory?

A

The fact that decreasing temperature can have anaesthetic effects despite the fact that it leads to a decrease in membrane fluidity. It also doesn’t explain that the binding of anaesthetics is saturable, implying a specific interaction with a protein. Finally there is a loss of actions in some compounds that are extremely lipid soluble and being homologous to known anaesthetics

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

It in fact turned out that the mechanism of action of anaesthetics was a combination of the lipid theory and a protein binding theory, T or F?

A

T

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

What is significant about the binding sites of anaesthetics that have been proven to bind to proteins in the plasma membrane?

A

General anaesthetic binding sites tend to be in hydrophobic domains deep within the membrane

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

What type of proteins do general anaesthetics tend to bind to?

A

Ion channels

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

Which specific receptor is it known that some general anaesthetics bind to and how does this lead to anaesthesia?

A

GABAA receptors – inhibition of regions in the central nervous system leading to anaesthesia

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

Different anaesthetics all interact with the same subunits of the proteins to which they bind, T or F?

A

F – different anaesthetics bind to different subunits

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

Where to intravenous anaesthetics tend to bind on the GABAA receptors?

A

β subunit

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

Where in the GABAA receptor structure do volatile liquid general anaesthetics tend to bind?

A

The interface between the α and β subunits

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

What is the function of the two-pore-domain potassium channels?

A

Regulate the excitability of neurons

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

How do general anaesthetics that bind to the two-pore-domain potassium channels exert their effects?

A

Increase the excitability of the channels and make the firing of actions potentials less likely

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

Explain ketamine’s anaesthetic effect?

A

Ketamine is an NMDA receptor antagonist and is known as a dissociative anaesthetic which prevents the action of excitatory glutamate

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

What type of protein is targeted by isoflurane and how does this cause anaesthesia?

A

Voltage-gated Na+ channels – inhibition of these channels prevent depolarisation and action potential generation

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

What neurotransmitter is secreted at the giant synapse in the brain known as the Calyx of Held?

A

Glutamate

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

What is the effect of low concentrations of general anaesthetics?

A

Decreased synaptic transmission in the central nervous system

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

Decrease synaptic transmission at low concentrations of anaesthetics in which area of the brain are responsible for the amnesia experienced?

A

Hippocampus – new memory formation

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

Decrease synaptic transmission at low concentrations of anaesthetics in which area of the brain are responsible for the loss of consciousness?

A

Reticular formation

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

Decrease synaptic transmission at low concentrations of anaesthetics in which area of the brain are responsible for the analgesia experienced?

A

Thalamic sensory relay nuclei

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

What is the effect of high concentrations of general anaesthetics?

A

Decreased speed of conduction in the central and peripheral nervous system

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

What brain functions are lost at dangerously high anaesthetic concentrations?

A

Motor control, control of cardiovascular reflexes and respiration as well as autonomic regulation

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

What are the four stages of anaesthesia from lowest to highest drug concentration?

A

Analgesia, excitement, surgical anaesthesia, medullary paralysis

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

Why is the second stage of anaesthesia dangerous?

A

In the excitement phase there is an exaggeration of reflexes (i.e. gagging) due to imbalances in excitatory and inhibitory neurons.

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

What are the four ideals for controlling anaesthesia?

A

Rapid induction and loss of consciousness, analgesia, muscle relaxation and rapid recovery

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

Which types of general anaesthetics are usually used to induce anaesthesia?

A

Intravenous anaesthetics

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

What are the advantages of intravenous anaesthetics?

A

Easy administration, less stressful for patients, rapid induction, metabolised well and quickly due to high lipid solubility

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

What are the disadvantages of intravenous anaesthetics?

A

Pain at site of injection, complex pharmacokinetics, cardiovascular and respiratory depression, control of concentration difficult and little can be done to resolve overdose

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

Ketamine administration leads to complete loss of consciousness, T or F?

A

F

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

Ketamine is a powerful analgesic, T or F?

A

T

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

What are some of the side effects of ketamine use?

A

Cardiovascular excitement, involuntary movements, euphoria, hallucinations and delirium

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

What is the role of inhalation drugs in anaesthesia and why are they more suited to this?

A

Inhalational anaesthetics are useful in the maintenance of surgical anaesthesia. It is much easier to control the levels of these drugs in the body

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

The reason inhalation anaesthetics are easier to control is because they are metabolised instantly by the body, T or F?

A

F – there is little to no metabolism of inhalational anaesthetics

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

What four factors are needed to be controlled when maintaining inhalation anaesthetic blood concentration?

A

Initial drug concentration, pulmonary ventilation, rate of transfer from alveoli to the blood and the rate of loss of the drug from arterial blood

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

What can be done if the state of anaesthesia becomes too deep?

A

Increase ventilation rate

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

Inhalational drugs are extremely lipid soluble, T or F?

A

T

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

Inhalation anaesthetics are redistributed into the fat rapidly, T or F?

A

F – they are redistributed slowly due to poor fat perfusion

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

What does the high lipid solubility of inhalation anaesthetics allow?

A

Rapid equilibration between body fluid concentration and alveolar space concentration

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

What is malignant hyperthermia?

A

Condition associated with the use of general anaesthetics that results in a rapid rise in temperature, huge increases in muscle contraction, hypertension and tachycardia

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

What causes malignant hyperthermia?

A

Mutations in the ryanodine receptor

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

Where is the mutated protein in malignant hyperthermia found?

A

Sarcoplasmic reticulum in skeletal (and some smooth) muscle

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

What happens in malignant hyperthermia in the presence of a general anaesthetic?

A

The ryanodine receptor has an increased sensitivity to anaesthetics and so opens allowing Ca2+ efflux from the sarcoplasmic reticulum. This leads to a oxidative burst by the mitochondria and a massive increase in ATP and intracellular Ca2+

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

How can malignant hyperthermia be treated?

A

Dantrolene is a ryanodine receptor inhibitor that is given in conjunction with the general anaesthetic to prevent the symptoms

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

What is meant by ataxia?

A

Irregular, uncontrolled muscle contractions

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

Recall some of the main symptoms of ataxia?

A

Ataxia/shaking, trunk instability, vertigo, nausea, vomiting, poor balance, nystagmus, tendency to falls

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

What inheritance pattern is seen in Type I episodic ataxia?

A

Autosomal dominant

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

What is the typical age of onset for type I episodic ataxia?

A

20-30

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

What can be said about the duration of attacks seen in patients with Type I episodic ataxia?

A

They are short/brief

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

What causes episodic ataxia type I?

A

Loss of function mutation in KCNA1

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

What type of protein in affected by the mutation that causes type I ataxia?

A

Voltage-gated K+ channel

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

What triggers attacks or episodes in type I ataxia?

A

What triggers attacks or episodes in type I ataxia? Physical or emotional stress, impacts to the vestibular system or sudden changes in position

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

Where is the channel protein that is mutated in Type I ataxia found?

A

In the cerebellum and neuromuscular junction

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

What can be said about the pattern of possible mutations in the protein that results in type I ataxia?

A

There is no clear mutation pattern, mutations occur throughout the protein in no specific areas

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

What is the impact on K+ currents as a result of the expression of mutant KCNA1 protein in animal models and what does this tell you about the impact of these mutations on protein function?

A

Mutant KCNA1 expression results in a decrease in the size of the K+ currents in the neuron thus indicating the mutations are loss of function

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

What part of the action potential is mediated by voltage-gated K+ channels?

A

Repolarisation

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

How does the mutation in KCNA1 lead to the increase excitability of neurons and thus inappropriate shaking movements?

A

Loss of function of the voltage-gated K+ channels compromises repolarisation and thus extends depolarisation. This results in the increased excitability of the neurons

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

Explain how the location of the KCNA1 channels accounts for the double impact of their mutation?

A

As KCNA1 is present both in the cerebellum and neuromuscular junction then mutations will result in problems with muscle contraction at the level of the neuromuscular junction but also problems with the signals coming from the cerebellum in the first place.

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

Why is it thought that acetazolamide is effective in treating ataxia?

A

Acetazolamide is a carbonic anhydrase inhibitor. By inhibiting carbonic anhydrase there is more HCO3- secretion in the kidney and thus more excretion in the urine. Loss of bicarbonate from the body results in a decrease in body fluid pH which in turn causes a decrease in the excitability of neurons

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

Phenytoin and carbamazepine are also effective medications for the treatment of type I ataxia. What class of compounds are they and how do they work to alleviate symptoms?

A

Na+ channel blockers that reduce neuronal excitability by preventing depolarisation

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

What is the inheritance pattern of type II episodic ataxia?

A

Autosomal dominant

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

How does the onset of type II episodic ataxia differ from that of type I?

A

Type II ataxia has an earlier onset typically in childhood to teenage years

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

How do the attacks in type II ataxia differ from that in type I?

A

The duration of the attacks are much longer from 30mins to 24hours

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

How do they symptoms of type II ataxia differ from type I?

A

Type II episodic ataxia is more likely to lead to trunk instability, vertigo, nausea, vomiting and headaches

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

What triggers the onset of symptoms in type II ataxia?

A

Physical or emotional stress, impacts to the vestibular system and sudden changes in position

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

Episodic ataxia type II is caused by mutations in which protein?

A

CACNA1A

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

What type of protein is mutation in type II ataxia?

A

Voltage-gated Ca2+ channel

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

Where is the mutant protein found in type II ataxia?

A

In the cerebellum

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

Describe the structure of the CACNA1A protein?

A

Membrane protein with 24 transmembrane domains in 4 blocks of 6. Each block of 6 contains a pore region and the 4th TMD is the voltage sensor

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

CACNA1A is regulated by cGMP, T or F?

A

F – it is highly regulated by β accessory proteins

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

What are the two types of mutations seen in CACNA1A that result in episodic ataxia type II?

A

Substitution mutations and truncation mutations

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

Which type of type II ataxia mutation elicits the most severe symptoms in patients?

A

Truncation mutations

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

What can be said about the mutation patterns seen in CACNA1A?

A

There is no clear mutation pattern however there is some indications of clustering of mutations

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

A mutation in CACNA1A will cause episodic ataxia type II, T or F?

A

F – mutations in CACNA1A can cause one of three different allelic disorders. Missense mutations tend to cause familiar hemiplegic migraine whereas a repeat expansion of the c-terminus will result in spinocerebellar ataxia type VI

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

Episodic ataxia is a progressive neurodegenerative disease, T or F?

A

T

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

In which cells of the cerebellum is CACNA1A found?

A

Purkinje and granule cells

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

CACNA1A is found in the axons of the cells in the cerebellum, T or F?

A

F – it is more commonly found on the somas

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

What is the endogenous role of CACNA1A?

A

What is the endogenous role of CACNA1A? Mediates exocytotic neurotransmitter release

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

What is seen in CACNA1A knockout model mice?

A

They develop normally up until 10 days however after that they begin to show problems with balance, ataxia and twisting motions, mimicking the symptoms in human patients. They usually die after 3-4 weeks, relatively earlier than equivalent in humans

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

The G239R mutation is a common type II episodic ataxia mutation. What does this nomenclature represent?

A

What does this nomenclature represent? This means that it is a glycine (G) to arginine (R) transition mutation that occurs at position 239 in the protein

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

When measuring Ca2+ in cells, why are barium ions used instead of Ca2+?

A

Barium is in the same group as calcium and thus also forms a 2+ ion. Calcium currents are difficult to measure because during Ca2+ influx there is a feedback mechanism whereby Ca2+ influx results in the closure of the channels through which they move through. By using Ba2+ this effect is prevented

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

What are the results of the G239R mutation on Ca2+ currents and the downstream effects on action potential generation?

A

There is a decrease in the size of the Ca2+ currents and thus there is less Ca2+ coming into the cell. This means that there is a decrease in the amount of neurotransmitter released into the synaptic cleft and therefore less likely action potential generation in the postsynaptic cells

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

In contrast to type I episodic ataxia, where there is an increase in neuronal excitability, the mutation that causes type II ataxia leads to a decrease in excitability of cells in the cerebellum, T or F?

A

T

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

Explain the double effect of CACNA1A mutations?

A

As a result of a CACNA1A mutation there is a loss of function of the voltage-gated Ca2+ channels. This causes a decrease in the Ca2+ currents generated in the neurons in the cerebellum and thus less neurotransmitter release and less neuronal excitability. In addition to the smaller Ca2+ currents there is a change in the voltage dependency of the CACNA1A channel so that its highest activity is seen at a more positive membrane potential. This increases the time taken until the voltage-gated Ca2+ channels are activated thus decreasing the overall effectiveness of the channel

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

Explain the overall summary of effects of CACNA1A mutations in episodic ataxia type II?

A

Decreased Ca2+ influx, less neurotransmitter release leading to problems with the control of skeletal muscle

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

Name an effective treatment for type II episodic ataxia?

A

Acetazolamide

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

What are seizures?

A

Episodic neuronal discharge

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
102
Q

What are the four main causes of epilepsy?

A

Tumour, trauma, genetics/inherited, infection

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

What are the two main types of epilepsy and how can they be further subdivided?

A

Partial epilepsy – where only one area of the brain is affected. Generalised epilepsy – where the whole brain is affected and symptoms are more severe. These can be subdivided into simple epilepsy where the patient doesn’t lose consciousness and complex epilepsy where they do

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

Complex generalised epilepsy is the most severe type, T or F?

A

T

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

Epilepsy can be acquired, T or F?

A

T – can be acquired as a result of neuronal damage or in autoimmune disease

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

What causes seizures at the neuronal level?

A

Increase excitability of neurons

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

Epilepsy is usually only caused by mutations in ion channels of excitatory neurons that leads to an increase in excitability, T or F?

A

T or F? F – mutations in ion channels that cause epilepsy have been found in excitatory and inhibitory neurons

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

Inhibitory neurons modulate the levels of excitation in the excitatory neurons, T or F?

A

T

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

Increased excitability of the neurons can happen via two mechanisms, what are these?

A

Mutations that lead to a direct increase in excitation of the excitatory neurons or that lead to a decrease inhibition of the excitatory neurons by the inhibitory neurons

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

Which phase of the action potential is mediated by the voltage-gated Na+ channels?

A

Depolarisation

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

Which phase of the action potential is mediated by the voltage-gated K+ channels?

A

Repolarisation

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

What aspect of electrical excitation in the nervous system is mediated by the voltage-gated Ca2+ channels?

A

Release of neurotransmitters from the presynaptic neurons in synaptic transmission

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

On the membrane of which type of neurons are AchRs found?

A

Postsynaptic excitatory neurons

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

What type of receptor is the AchR?

A

Ligand-gated ion channel

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

Activation of AchRs causes what in the neuron?

A

Opening of the channel and Na+ influx

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

On the membrane of which type of neurons would you find GABAA receptor?

A

Postsynaptic membrane of inhibitory neurons

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

What type of receptor is the GABAA receptor?

A

Ligands gated ion channel

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

Activation of the GABAA receptor causes what?

A

Opening of the channel and Cl- influx shifting the membrane potential more negative towards hyperpolarisation

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

What is the role of the HCN or hyperpolarisation-activated cyclic nucleotide gated channel?

A

The HCN channels is a voltage-gated channel that becomes activated when the membrane is hyperpolarised. As the membrane potential shifts more negative the open probability of the channel increases and its action is increased, shifting the membrane potential back towards depolarisation. This channel is regulated by cyclic nucleotides such as cAMP and cGMP and is important in determining the timing of action potential firing.

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

Explain how mutations in ion channels present in inhibitory neurons can lead to epilepsy?

A

Loss of function mutations in ion channels in the inhibitory neurons can lead to less GABA release at the synapse and thus a decrease inhibition of the excitatory neurons effectively increasing its excitability

121
Q

What two channels can be affected by mutations that lead to loss of inhibition by inhibitory neurons?

A

Voltage-gated Ca2+ channels at the presynaptic terminal when mutated can have a loss of function results in less GABA release in the cleft and thus less inhibition of the excitatory neurons. Voltage-gated Na+ such as Nav1.1 present in axon can also have loss of function mutations. These channels mediate depolarisation and a loss in function will lead to decreased depolarisation and less action potential generation. This in turn will decrease the frequency of GABA release into the synapse thus resulting in a decrease in inhibition of the excitatory neuron

122
Q

As well as mutations on the postsynaptic membrane and axon of excitatory neurons, mutation in ion channels in the dendrites can also lead to an increase in excitation, T or F?

A

T

123
Q

Explain how mutations in ion channels on the postsynaptic membrane of excitatory cells leads to an increase in excitation of the excitatory neurons seen in epilepsy?

A

Loss of function mutations in the GABAA receptors will lead to a decrease in Cl- influx and as a result less inhibition of the excitatory neurons causing overexcitation.

124
Q

How can mutations in ion channels present in the axons of excitable neurons lead to epilepsy?

A

The Nav1.2 ion channel mediates depolarisation in the excitatory neurons. A gain of function mutation in this ion channel would cause overexcitation and thus epilepsy. This is due to increased depolarisation in the excitatory neurons.

125
Q

Explain how mutations in the AchRs can cause overexcitation which may lead to epilepsy?

A

he AchRs are found on the postsynaptic membrane of excitatory neurons. Gain of function mutations in these receptors will lead to increased Na+ influx and thus increased depolarisation in excitatory neurons which as a result will show increased excitability

126
Q

Describe the classic voltage-gated Na+ channel structure, as shown by Nav1.1?

A

24 transmembrane domain protein in 4 blocks of 6. Each block contains a pore region and a voltage sensor which is the 4th transmembrane domain in each.

127
Q

What two types of mutation are known to cause epilepsy?

A

Missense and truncation mutations

128
Q

Both types of mutations cause the same types of epilepsy, T or F?

A

F – missense and truncation mutations cause different types of epilepsy with different symptoms

129
Q

There is a clear pattern of mutations in the Nav1.1 protein that result in epilepsy, T or F?

A

F – there is no clear pattern

130
Q

Depending on where in the protein each type of mutation is determines the severity and the types of symptoms shown in the patients, T or F?

A

T

131
Q

Why are knockout models for epilepsy often heterozygotes?

A

This mimics what is seen in the patients where the epilepsy is caused by a dominant mutation and thus patients only have one faulty copy

132
Q

What is seen in the inhibitory neurons of the Nav1.1 knockout mice in comparison to wild type?

A

Knockouts still showed some action potential generation but in a much less regular pattern. Sometimes the neurons fired, sometimes they did not and on occasions where they did not, there was a loss of inhibition of the excitatory neurons

133
Q

How can the knockout data be used to explain the fact that patients with epilepsy don’t show symptoms all of the time?

A

Nav1.1 knockouts still retained some ability of the inhibitory neurons to fire action potentials, thus inhibition did still occur some of the time. Where action potentials weren’t generated is where there was overexcitation and explains what is seen in humans where seizures come on almost spontaneously

134
Q

Missense mutations in Nav1.1 can have varying degrees of severity from mild symptoms to severe, T or F?

A

F

135
Q

What type of mutation leads to the most severe types of epilepsy?

A

Loss of function mutations as a result of truncation

136
Q

What is meant by pain?

A

Unpleasant sensory and emotional experience associated with actual or potential tissue damage

137
Q

Why is pain useful?

A

Important protective mechanism to tell you something is wrong

138
Q

A loss of function in what channel is responsible for a diminished or lack of pain perception?

A

Nav1.7

139
Q

What is erythromalgia and what causes it?

A

Erythromalgia is a heightened sense of pain that can be triggered by non-noxious stimuli. It is caused by a gain of function mutation in Nav1.7

140
Q

What is the purpose of anxiety?

A

A fear response that acts as a perception of danger

141
Q

Why can small levels of anxiety be good?

A

Anxiety acts as a motivational response to a negative emotion

142
Q

What are the two different types of anxiety and how to they differ?

A

Generalised anxiety is where there is a negative feeling all of the time with no actual trigger. Fear-induced anxiety is an irrational response to a potential fearful stimulus.

143
Q

Give some examples of fear-induced anxiety?

A

Phobias, PTSD, OCD and panic disorders

144
Q

What two means are there for treating clinical anxiety?

A

Psychiatric, pharmacological

145
Q

What are the two types of drugs used to treat anxiety?

A

Antidepressants, antiepileptics and antipsychotics. Otherwise drugs that interact with the GABA receptors

146
Q

What type of receptor are the GABAA receptors?

A

Ligand-gated ion channels – ionotropic

147
Q

What type of neurotransmission are the GABAA receptors involved in?

A

Fast inhibitory neurotransmission

148
Q

Where abouts at the synapse are GABAA receptors found?

A

Postsynaptically on the dendrites and cell bodies

149
Q

As GABA is an inhibitory neurotransmitter, binding to GABAA receptors causes influx of which ions?

A

Cl-

150
Q

How many different subunits make up the GABAA receptor?

A

5

151
Q

How does subunit composition vary?

A

Subunit vary depending on the location, function and intended pharmacology of the receptor

152
Q

GABAA receptors have a single binding site, T or F?

A

F – they have several

153
Q

What is the name given to the GABA binding site on the GABA receptor?

A

Orthosteric site

154
Q

Name a compound that is an agonist of the GABA binding site on the GABAA receptor?

A

Muscimol

155
Q

Name an antagonist of the GABA binding site on the GABAA receptors?

A

Picrotoxin, bicuculline

156
Q

What is the result of antagonism of the GABA binding site?

A

Seizures

157
Q

What is the name of the site on the GABAA receptors where benzodiazepines bind?

A

Allosteric site

158
Q

What is the role of the site which benzodiazepines bind to?

A

? The allosteric site is responsible for the regulation of channel activity

159
Q

Name an agonist of the site in which benzodiazepines bind?

A

Diazepam

160
Q

Name an antagonist of the site in which benzodiazepines bind?

A

Flumazenil

161
Q

What other compounds also bind to the site on the GABAA receptor that benzodiazepines bind to?

A

Barbiturates, neurosteroids and general anaesthetics

162
Q

What class of receptors are the GABAB­ receptors?

A

GPCRs

163
Q

What are the proteins coupled to the GABAB receptors?

A

Gi or Go

164
Q

What type of neurotransmission is mediated by the GABAB­ receptors?

A

Slow transmission – they modulate neurotransmission

165
Q

GABAB receptors are only found postsynaptically, T or F?

A

F – they are found both presynaptically and postsynaptically

166
Q

Activation of the GABAB receptors at the axon terminals causes what?

A

Inhibition of voltage-gated Ca2+channels and neurotransmitter release

167
Q

Activation of postsynaptic GABAB receptors on the soma and dendrites causes what?

A

Activation of GIRK K+ channels and the inhibition of depolarisation, action potential generation and excitability

168
Q

Name a classic agonists and selective antagonist of the GABAB­ receptor?

A

Agonist – baclophen. Antagonist – phaclophen

169
Q

What is the standard subunit composition of a GABAA receptor?

A

2 α subunits, 2 β subunits and 1 γ subunit

170
Q

Where is the orthosteric site found in the GABA receptor?

A

β subunits

171
Q

Where is the allosteric site found in the GABA receptor?

A

At the interface between the α and γ subunits

172
Q

Different genes code for the different GABA receptor subunits, T or F?

A

T

173
Q

Which GABA receptor subunit is responsible for the sedative effect caused by benzodiazepines?

A

α1 subunit

174
Q

Which effect of benzodiazepines are the α2, α3 and α5 subunits responsible for?

A

Anxiolytic effect

175
Q

GABAA receptors are part of the nicotinic receptor superfamily, T or F?

A

T

176
Q

How can anxiety be studied in animals?

A

Small rodents tend to dislike being in bright light for fear of predation. If you shine a bright light on a mouse its blood cortisol levels will increase as it is stressed and the mouse will begin to look for shade. Administration of diazepam removes the bright light avoidance behaviour as the mouse is less anxious

177
Q

Describe how a conflict test can be useful in studying the effects of anxiolytics on anxiety?

A

If you train an animal to press a lever in order to receive a reward it will continue to elicit this behaviour. If you occasionally provide an electric shock after pulling the lever the animal will reduce the amount of lever pulling as it is anticipating an electric shock. Applying analgesics doesn’t reduce this behaviour despite the animal no longer being able to sense pain. However, administration of anxiolytics does result in no decrease in lever pulling after occasional electric shocking as the anticipation has been removed.

178
Q

What is the overall effect of benzodiazepine at the level of neurotransmission in the brain?

A

Benzodiazepines increase the level of inhibitory neurotransmission in the brain

179
Q

What are the five main physiological effects of benzodiazepines?

A

Sedation and reduced anxiety, hypnosis, anterograde amnesia, anticonvulsant, reduced muscle tone

180
Q

Sedation of patients occurs with the lowest doses of benzodiazepines, T or F?

A

T

181
Q

What are the effects of benzodiazepines on sleep?

A

Hypnosis - reduced latency of sleep onset, increased stage 2 non-REM sleep, decreased duration of REM and slow-wave sleep

182
Q

What are the effects of benzodiazepines on memory?

A

Benzodiazepines result in anterograde amnesia that results in short term memory loss and the prevention of memory formation for the duration of the benzodiazepines effects

183
Q

Benzodiazepines given at high concentrations acts as anticonvulsants, T or F?

A

T

184
Q

Explain the allosteric regulation of the GABAA receptors by benzodiazepines?

A

Benzodiazepines increase the activity of GABAA receptors increasing Cl- influx and a greater inhibition of postsynaptic activity

185
Q

The binding of benzodiazepines to the GABAA receptors is irreversible, T or F?

A

F – its reversible

186
Q

Name an antagonist of the allosteric site of the GABAA receptor?

A

Flumazanil

187
Q

How do benzodiazepines lead to an increase in the GABA receptor activity?

A

Benzodiazepines increase the frequency of opening of the GABA receptors rather than changing the conductance or mean open time

188
Q

Why are benzodiazepines when taken alone, not very dangerous?

A

They don’t directly activate channels by themselves, they only increase the activity of channels already open

189
Q

At high concentrations barbiturates are also positive allosteric modulators of the GABAA channels, T or F?

A

F – at low concentrations barbiturates are positive allosteric modulators of GABAA however, at high concentrations they directly cause an opening of the channels

190
Q

Describe how barbiturates lead to an increase in activation of the GABAA receptors?

A

Barbiturates increase the mean open time of the receptors this giving rise to bigger Cl- currents

191
Q

What is meant by the background constitutive activity of the GABA receptor?

A

The GABA receptors are capable of spontaneously adopting an active conformation in the absence of an agonist

192
Q

What is the role of GABA receptor inverse agonists?

A

Inverse agonists stabilise the GABA receptor in the closed or inactive conformation, stopping it from opening by itself

193
Q

Name a family of compounds that are inverse agonists of the GABA receptors?

A

β-carbolines increase memory and learning

194
Q

What three aspects of the pharmacokinetics of benzodiazepines are influenced by lipid solubility?

A

Determines the onset of actions, redistribution and accumulation

195
Q

What factors determine the benzodiazepine half-life in the body?

A

Renal function, age and microsomal enzyme activity induction

196
Q

How can benzodiazepine metabolism actually lead to an increase in duration of action?

A

First phase metabolism of some benzodiazepines can produce other active metabolites

197
Q

How can plasma protein binding influence a benzodiazepines effects?

A

Binding to plasma proteins may increase the time it takes for the drug to get into the central nervous system. Competitive binding of multiple drugs for plasma proteins can result in wild fluctuations in drug concentrations in the blood

198
Q

Define epilepsy?

A

Unprovoked seizure activity in the brain characterised by high frequency discharge as a result of an imbalance between excitatory and inhibitory neurotransmission

199
Q

What are the two treatment options for epilepsy and which neurotransmitters do these target?

A

? Increase inhibitory neurotransmission by increasing GABA or GABAA activity. Decrease excitatory neurotransmission by decreasing glutamate levels or receptor activity

200
Q

What are the two main types of epilepsy and how do they differ?

A

Partial and generalised epilepsy. Partial epilepsy has its effects localised to only one hemisphere of the brain whereas generalised effects both

201
Q

List some of the causes of epilepsy?

A

Head trauma, infection, tumour, local lesions, genetics

202
Q

Mutations in which classes of proteins are often responsible for epilepsy?

A

Voltage-gated Na+ channels, GABAA receptors and nAchRs

203
Q

Seizure activity in the motor cortex results in convulsions in regions of the body innervated by that region of the brain, T or F?

A

T

204
Q

What is the result of seizure activity in the hypothalamus?

A

Autonomic discharge e.g. salivation and increase secretion

205
Q

Seizure activity in which region of the brain is responsible for the loss of consciousness sometimes seen in epilepsy?

A

Reticular formation

206
Q

Which technique is often used to diagnose epilepsy?

A

Electroencephalogram

207
Q

What is the main difference seen in the EEGs of patients with generalised and partial epilepsy?

A

General epilepsy EEGs will show high frequency brain activity all over the brain picked up by all the electrode, whereas partial epilepsy patients EEGs will only show abnormal electrical activity picked up by some electrodes due to the limited spread

208
Q

How do absent type epilepsy EEGs differ from tonic-clonic types?

A

he EEGs show slow synchronised oscillatory behaviour whereas the tonic-clonic type shows rapid high frequency activity followed by rhythmic wave type activity

209
Q

What type of protein is usually mutated in patients with absent epilepsy?

A

Ca2+ channel

210
Q

Where is absence type epilepsy more commonly seen and how is it characterised?

A

Absent epilepsy is more often seen in children and is characterised by a brief loss of attention

211
Q

What are the three types of targets for antiepileptic’s at the inhibitory synapses?

A

Increase GABAA activity and increase inhibitory neurotransmission. Block GABA uptake back into presynaptic terminal to increase the duration of GABAs effects and this the duration of inhibition. Decrease GABA metabolism by inhibiting enzyme responsible for its breakdown

212
Q

Which target for the treatment of epilepsy gives specificity?

A

GABAA receptor activation

213
Q

How do benzodiazepines work? Benzodiazepines are allosteric regulators of the GABAA receptors. They increase the activity of the GABAA receptors by increasing the frequency of opening of the GABA channels. This increases Cl- currents across the membrane and thus the inhibition of the neuron

A

Benzodiazepines are allosteric regulators of the GABAA receptors. They increase the activity of the GABAA receptors by increasing the frequency of opening of the GABA channels. This increases Cl- currents across the membrane and thus the inhibition of the neuron

214
Q

Give some examples of benzodiazepines?

A

Diazepam, clonazepam, clobazam

215
Q

What are the problems of benzodiazepine use for the treatment of epilepsy?

A

Sedation, tolerance and withdrawal

216
Q

How do barbiturates work?

A

At low concentrations they are positive allosteric modulators of GABAA receptors. They increase the mean open time of the channels to give rise to bigger Cl- currents thus resulting in greater inhibition. At high concentrations barbiturates can directly cause opening of GABAA channels and leads to a massive inhibition of brain activity.

217
Q

Give some examples of barbiturates?

A

Phenobarbitone, primidone

218
Q

What are the problems of barbiturate use in the treatment of epilepsy?

A

Low therapeutic index, sedation and complex pharmacokinetics

219
Q

In which metabolic process is GABA a by-product of?

A

Kreb Cycle

220
Q

What is the name of the enzymes that converts glutamate to GABA?

A

Glutamic acid decarboxylase

221
Q

How does valproate act to treat epilepsy?

A

Increases the level of GABA in the brain most likely at due to working at the level of transcription controlling enzymes involved in GABA synthesis

222
Q

What is the main target for the treatment of epilepsy at the excitatory synapses/neurons?

A

Inhibition of glutamate receptors and decrease excitation

223
Q

What class of channels can also be targeted to decrease excitation and treat epilepsy?

A

Inhibition of voltage-gated Na+ that drive depolarisation and the rising phase of the action potential will lead to less excitatory neurotransmission

224
Q

How do Na+ channel blockers work?

A

Na+ channel blockers stabilise the voltage-gated Na+ channels in the inactivate state thus preventing the neuron from depolarising.

225
Q

Na+ channel blockers are non-sedative, T or F?

A

T

226
Q

Give examples of Na+ channel blockers used in the treatment of epilepsy?

A

Phenytoin, carbamazepine

227
Q

What is the most widely used anti-epileptic drug?

A

Carbamazepine

228
Q

What type of Ca2+ channels are implicated in absence type seizures and what is unusual about them?

A

T-type Ca2+ channels – can mediate the rising phase of the action potential in some brain regions instead of Nav channels

229
Q

Name some examples of T-type Ca2+ channel blockers used in the treatment of epilepsy?

A

Ehtosuximide, GABApentin – binds to accessory α2δ subunit that controls T-type Ca2+ channel trafficking to the membrane

230
Q

Levetiracetam is a new drug found to be effective in treating epilepsy, how does it work?

A

Decreases the exocytosis and release of glutamate containing vesicles by binding to synaptic vesicle protein-2A

231
Q

Which conditions is depression commonly associated with?

A

Insomnia, anxiety and addiction

232
Q

What are the two categories of depression and how do they differ?

A

Bipolar disorders – where patients alternate between low and over-exuberant moods. Unipolar disorders – where patients have consistent low mood

233
Q

What is meant by endogenous and reactive depression?

A

Endogenous depression is where there is a constant low mood that doesn’t seem to have a clear cause or trigger. Reactive depression is usually caused by a stressful event

234
Q

Describe the peripheral aspect of depression?

A

Increase in cortisol levels as wells as gherlins and leptins associated with feeding behaviour

235
Q

Describe the classical symptoms of depression?

A

Low mood (anhedonia), negative thoughts, pessimism and misery, diminished ability to think/concentrate, loss of libido, rapid increase or decrease in weight, apathy, low self-esteem

236
Q

Depression is more common in males than female, T or F?

A

F – twice as likely in females

237
Q

What is the recommended treatment for depression?

A

Antidepressant drugs in combination with counselling

238
Q

After the cause of depression has been alleviated, a patient’s depression will not subside, why is this?

A

Depression causes changes in brain chemistry that can only be reversed though medication

239
Q

What are the three main classes of antidepressants and how do they differ?

A

MOAIs -inhibit the breakdown of monoamine neurotransmitters and increase the levels in the synapse. Tricyclic antidepressants – generally inhibit monoamine neurotransmitter reuptake with no selectivity. Selective reuptake inhibitors – selectively inhibit the uptake of monoamine transmitters into the neurons

240
Q

Give an example of an SSRI?

A

Fluoxetine/Prozac

241
Q

What additional class of molecules are being investigated as antidepressants?

A

Monoamine receptor antagonists

242
Q

Which type of MOA is found in the central nervous system and is responsible for the metabolism of 5-HT and noradrenaline thus making it the main target of antidepressant drugs?

A

MAO-A

243
Q

Explain the side effect of MAOIs known as the cheese effect?

A

Tyramine is an amine derivative of tyrosine that is also broken down by MAO. Inhibition of MAO leads to a build-up of tyramine which triggers the release of monoamines in the peripheral nervous system thus resulting in hypertension

244
Q

Give an example of an MAOI?

A

Moclobemide

245
Q

How do tricyclic antidepressants (TCAs) act?

A

Non-selectively inhibit the reuptake of monoamines back into the neurons

246
Q

What are the side effects of TCA use and why is this?

A

TCA side effects are due to the result of their antagonism of the mAchRs leading to a dry mouth, blurred vision and constipation

247
Q

Give an example of a tricyclic antidepressant?

A

Amitryptyline, clomipramine

248
Q

SSRIs have far fewer side effects than TCAs, T or F?

A

T – due to their selectivity

249
Q

Give an example of an SNRI?

A

Reboxetine

250
Q

Where is the main source of noradrenergic neurons located in the brain?

A

Locus serealus

251
Q

The function of noradrenaline can be remembered using the acronym P.A.A.M, what does this stand for?

A

Pain, attention, arousal, mood

252
Q

Recall the monoamine biosynthesis pathway?

A

Tyrosine à L-Dopa à Dopamine à Noradrenaline à Adrenaline

253
Q

Which enzyme is responsible for the breakdown of noradrenaline?

A

MAO-A

254
Q

Which enzyme is responsible for the breakdown of noradrenaline in the cytoplasm as well as terminating its action?

A

Catechol-O-Methyltransferase

255
Q

What is the rate limiting step, and its accompanying enzyme, in the monoamine neurotransmitter synthesis pathway? Conversion of tyrosine à L-Dopa – Tyrosine hydroxylase

A

Conversion of tyrosine à L-Dopa – Tyrosine hydroxylase

256
Q

Stimulation of the α1 adrenoceptors has been known to stimulate 5-HT neurons in the raphe nucleus, T or F?

A

T

257
Q

Where are the majority of serotonergic neuronal cell bodies found in the brain?

A

Raphe nucleus

258
Q

What are some of the roles of 5-HT?

A

Sleep and wakefulness, mood and emotion, feeding behaviour

259
Q

How can 5-HT actions be linked to anhedonia?

A

5-HT has a role in regulating limbic processing, thus decreased 5-HT levels may result in decreased emotion to normally pleasurable experiences

260
Q

What is the precursor in 5-HT synthesis and where is it obtained?

A

Tryptophan amino acid precursor obtained in the diet from chocolate and protein rich foods

261
Q

What is the rate determining step and the enzyme involved in the synthesis of 5-HT?

A

Conversion of tryptophan to 5-hydroxytryptophan – catalysed by tryptophan hydroxylase

262
Q

Serotonin has a huge array of receptors, but what classes of receptors are these?

A

GPCRs and ligand-gated ion channels

263
Q

The 5HT1A receptors are inhibitory receptors, what type of G protein are they coupled to?

A

Gi­

264
Q

How do acute and chronic increases in 5-HT levels differ at the level of the 5HT1A receptor?

A

Acute increases in 5-HT leads to increase activation of the 5HT1A receptors and a decrease in neuronal firing and subsequent 5-HT release. Chronic increase in 5-HT levels leads to a desensitisation to the neurotransmitter. This occurs by the internalisation and degradation of the 5HT1A receptors leading to a loss of inhibition and more action potential firing

265
Q

Serotonin is also known to have an effect on neurotrophins, explain this effect?

A

5-HT neurons regulate the expression of brain-derived neurotrophic factor (BDNF) which is responsible for synaptic stabilisation, particularly in the dendritic spines. Phosphorylation of CREB by PKA leads to increased translocation to the nucleus and increased BDNF transcription. Activation of the 5HT1A receptors leads to a decrease in PKA and thus a decrease in CREB activation

266
Q

How does the neurotrophin hypothesis link to depression?

A

The decreased levels of 5-HT seen in in depression leads to a decrease in BDNF and thus a reduced synaptic stability therefore results in depression

267
Q

What class of molecules are used to treat psychosis?

A

Dopamine antagonists

268
Q

What is Schizophrenia?

A

A psychotic disease characterised by disturbances in area of the brain associated with thought perception, attention, motor behaviour and emotion

269
Q

Give some examples of the negative symptoms of Schizophrenia?

A

Blunting of emotions, withdrawal from social contact, flattening of emotional responses, reluctance to perform everyday tasks

270
Q

Give some examples of the positive symptoms of Schizophrenia?

A

Hallucinations, delusions, thought disorders, delusions of grandeur, bizarre behaviours and stereotypes movements

271
Q

List some of the causes of Schizophrenia?

A

Genetic and environmental factors, can also be associated with drug use (methamphetamine, cannabis, cocaine and opiates)

272
Q

Glutamate and serotonin receptors have also been implicated in Schizophrenia, T or F?

A

T

273
Q

What are the four dopamine pathways in the brain?

A

Nigrostriatal pathway, mesocortical and mesolimbic pathway, tuberohypohyseal pathway and the medullary trigger zone

274
Q

Loss of dopamine neurons in the SN is associated with Parkinson’s Disease, T or F?

A

T

275
Q

What is the link between dopamine and psychotic disorders?

A

Excess activity of the dopamine system is associated with Schizophrenia, ADHD and drug dependence

276
Q

What is the precursor of dopamine?

A

Tyrosine

277
Q

Recall the synthesis of dopamine from its precursor?

A

Tyrosine à L-Dopa (catalysed by tyrosine hydroxylase). L-Dopa à Dopamine (catalysed by dopa-decarboxylase)

278
Q

Which step is the rate limiting step in dopamine synthesis?

A

Conversion of tyrosine to L-Dopa

279
Q

Dopamine receptors are of the GPCR and ligand-gated ion channel varieties, T or F?

A

F – dopamine receptors are only GPCRs

280
Q

How many subtypes of dopamine receptors are there?

A

5

281
Q

All dopamine receptors are excitatory, T or F?

A

F – D2 and D4 are inhibitory

282
Q

What is the effect on stimulation of dopamine receptors on cAMP levels?

A

Stimulation of D1, D3 or D5 receptors leads to an increase in cAMP levels whereas stimulation of D2 or D4 receptors results in a decrease in cAMP levels

283
Q

Explain how the dopamine receptors can autoregulate?

A

Explain how the dopamine receptors can autoregulate? The D2 receptors control the function and release of other dopamine releasing neurons

284
Q

Which enzyme is responsible for the metabolism of dopamine in the mitochondrial membrane?

A

MAO-B

285
Q

Which enzyme is responsible for the metabolism of dopamine in the cytosol?

A

Catechol-O-Methyltransferase

286
Q

Give an example of a dopamine receptor agonist?

A

Apomorphine

287
Q

Which dopamine receptor is the most suitable target for antipsychotics?

A

? D­2 receptors

288
Q

D2 receptor agonists are used in the treatment of Schizophrenia, T or F?

A

F – D2 antagonists are used

289
Q

Give an example of a classical antipsychotic used in the treatment of Schizophrenia and that is a member of the phenothiazine compounds?

A

Chlorpromazine

290
Q

What are the side effects of first generation antipsychotics?

A

Motor disturbances, prolactin secretion

291
Q

What is meant by extrapyramidal effects of antipsychotics? ­

A

Effects on pituitary hormone secretion such as prolactin due to their interference with the tuberohypophyseal dopamine pathway

292
Q

Atypical antipsychotics have a lower incidence of extrapyramidal effects, name one?

A

Clozapine, sulpiride

293
Q

Explain why prolonged use of antipsychotics leads to neurotoxicity?

A

Prolonged antipsychotic drugs use leads to accumulation of toxic metabolites within the neurons resulting in the degeneration of aminergic neurons

294
Q

Stimulation of which dopaminergic pathway leads to the euphoria and excitement associated with dopamine receptor antagonists?

A

Mesocortical and mesolimbic pathway

295
Q

Explain how acute use of antipsychotics can lead to increased blood pressure and decreased gut motility amongst other side effects?

A

Dopamine antagonists can displace noradrenaline in the peripheral nervous system leading effects of sympathetic nervous system stimulation

296
Q

Which dopaminergic pathway is the target of antipsychotic drugs?

A

Mesocortical and mesolimbic pathway

297
Q

Only a small number of D2 receptors need to be occupied in order to alleviate the symptoms of Schizophrenia, T or F?

A

F – 80% occupancy is required in order to effectively remove symptoms

298
Q

Sedation is another common side effect of dopamine receptor antagonist use, what is the hypothesised cause of this?

A

Interferences with other amine systems such as histamine receptors

299
Q

How can D2 antagonists lead to abnormal breast development in males?

A

Stimulation of the tuberohypophyseal pathway leading to hyperprolactin secretion