Peripheral Neural Transmission Flashcards

1
Q

What receptors does NA bind to

A

Binds to α&raquo_space;> β

But still activates β eg in SNS to heart

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

How are small molecule NTs acquired?

A

synthesised locally within presynaptic terminals

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

Where are the enzymes needed to locally synthesise neurotransmitters produced?

A

in the soma and then transported to the nerve terminus by slow axonal transport

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

Where do the precursor molecules for synthesising new small molecule NTs come from

A

may pre-exist within the nerve terminal but more commonly they are brought inside by some transporters found in the plasma membrane of the nerve terminus.

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

True or false:

small molecule NTs can be synthesised inside the vesicles

A

True
For some small-molecule neurotransmitters, the final steps of synthesis occur inside the vesicles where the necessary enzymes are presen

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

Where are neuropeptides usually synthesised

What happens next

A

in the soma where they are packaged into vesicles where further processing takes place

peptide filled vesicles are transported along the axon to the nerve terminal via fast axonal transport

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

What do fast and slow axonal transport in nerves transport?

A

fast: peptide filled vesicles (neuropeptides)
slow: small molecule neurotransmitters

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

True or false

all neurotransmitters are loaded into vesicles soon after synthesis

A

False
Gaseous transmitters eg NO are not

all others are

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

Briefly describe the membrane and the interior of the vesicles for NT transport

A

phospholipid bilayer

aqueous interior

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

What is the size of vesicles of small molecule transmitters

A

40-60nm

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

Describe the appearance of small molecule NT vesicles

Hence what are they known as

A

appear clear in EM

small, clear core vesicles

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

What is another name for small, clear core vesicles

Why?

A

synaptic vesicles

they are more abundant and cluster at the active zone

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

Where are neuropeptides loaded into vesicles

What size are these vesicles

A

in the soma

90-250nm in diameter

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

What are neuropeptide vesicles known as?

why

A

large dense core vesicles

they appear electron dense in EM

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

How are neuropeptide vesicles different to those of small molecule transmitters

A

neuropeptide - larger,
more e- dense
do not cluster at the active zone

small molecule:
small, clear-core vesicles,
gather at the active zone

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

How are vesicles released (as discovered by Katz)

A

in a quantal manner where 1 quantum is the content of a single vesicle (as we saw in HOM last year)

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

Which ion channels in the presynaptic nerve terminal open in response to depolarisation ?

What types of channel specifically?

A

voltage gated Ca2+ channels

Cav2 series: mainly P/Q and N type

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

How long does it take for Ca to trigger exocytosis

A

required conc of Ca is very quickly reached

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

Why is the required conc of Ca for exocytosis very quickly reached in the presynaptic terminal

A

Cavs are clustered around the active zone and some vesicles are already docked to release sites so release of contents can be v rapid

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

What is the releasable pool?

A

the presynaptic vesicles that are already docked to release sites in the terminal

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

How much of the total amount of vesicles is represented by the releasable pool

A

Only a v small amount

Their numbers can be enhanced on demand when vesicles from the reserve pool join them

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

What cell structure is the reserve pool associated with

A

cytoskeleton

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

What are the 6 steps of vesicle release and recycle

A

1) Transmitter loading
2) mobilisation of vesicles toward active zone
3) Docking
4) Priming
5) Exocytosis
6) recycling of vesicles

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

Describe the transmitter loading stage of the presynaptic vesicle life cycle

What precedes and follows it

A

NT imported using ATP
Proton pump acidifies interior to create an electrochemical gradient across vesicle membrane
Gradient provides energy for NT uptake by specialized transporters

Preceded by recycling of vesicle
Followed by mobilisation of vesicles

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

What protein family is important for mobilization of vesicles towards the active zone

What modification is important in this process

A

synapsins

phosphorylation

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

Describe vesicle mobilisation toward the active zone in presynaptic terminals

A

non phosphorylated synapsins reversibly bind to vesicles and actin to cross link them to each other, keeping the vesicles tethered to the reserve pool

Ca2+ increase (following depolarisation) activates CaMKII, which phosphorylates synapsins, allowing dissociation from vesicles.

The vesicles are now free to move

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

What is CaMKII

A

Ca2+ /calmodulin dependant protein kinase (type II)

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

Describe the docking stage of the presynaptic vesicle life cycle

What step precedes and follows it

A

At the active zone vesicles dock to nerve membrane via SNARE proteins

preceded by vesicle mobilization
followed by vesicle priming

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

What occurs at the priming stage of the vesicle cycle

A

Docked vesicles undergo ATP dependent prefusion that primes them for Ca induced release

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

Which type of protein is vvv important in vesicle priming

Are these the only proteins involved

A

SNARE

no - many others involved

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

What is the main purpose of priming vesicles

A

to organize SNAREs into correct conformation for membrane fusion

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

Describe the exocytosis step of vesicle release

What follows and precedes it?

A

Ca2+ (sensed by synaptotagmins) triggers complete fusion of vesicle with membrane in a rapid reaction

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

Which protein sense Ca2+ to initiate vesicle-membrane fusion

A

synaptotagmin

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

What are the 3 different routes that can be taken for vesicle recycling after release of the contents

A

reversible fusion pore
endocytosis
bulk retrieval pathway

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

Describe the reversible fusion pore route of vesicle recycling

A

vesicle does not completely fuse with nerve membrane - contents are released via a fusion pore instead

Vesicle is retrieved by closing the pore

Spent vesicles can either stay fused to membrane (kiss and stay) or relocate to reserve pool ( kiss and run)

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

How quickly can reversible fusion pore occur

A

vvv quickly

in tens to hundreds of milliseconds

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

What does ‘kiss and stay’ and ‘kiss and run’ refer to

A

reversible fusion pore

stay: spent vesicle remains fused to nerve membrane
run: spent vesicle relocates to reserve pool of vesicles

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

Describe the endocytosis pathway for vesicle return

What happens after the vesicle is recycled

A

vesicle coated in clathrin
the coat is later shed and the vesicle returned to interior of synaptic nerve terminal

empty vesicles either refill immediately with NT or pass through endosomal sorting

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

Describe the bulk retrieval pathway for vesicle return

A

excess membrane re-enters the terminal by budding from uncoated pits - not very common .

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

Why do many congenital myasthenic syndromes arise

A

defects in ACh release due to inadequate number of synaptic vesicles available for release.

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

What happens in familial infantile myasthenia

A

synaptic vesicles are much smaller

NOT a shortage of vesicles

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

What happens in Lambert Eaton myasthenic syndrome (LEMS)

What other disease can this syndrome be a complication of

A

ACh release is reduced due to autoimmune destruction of neuronal Cav channels in active zone

cancer

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

Why is vesicle- membrane fusion difficult

A

Because a membrane bilayer is a stable structure, fusion of the synaptic vesicle and plasma membrane must overcome a large unfavourable activation energy

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

What does SNARE stand for

A

soluble N-ethylmaleimide-sensitive factor attachment receptors

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

How conserved are SNARE proteins

A

SNAREs are universally involved in membrane fusion, from yeast to humans

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

How are SNAREs categorised

A

v- SNARE (v for vesicle)

t-SNARE - (t for target membrane)

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

What part of the SNARE protein is highly conserved

A

cytosolic domain:

the SNARE motif, that is 60-70 residues long and form coiled-coil alpha-helical structures

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

What SNAREs does each synaptic vesicle have

A

a single type of v-SNARE:

synaptobrevin (AKA VAMP)

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

What is another name for synaptobrevin

A

vesicle associated membrane protein or VAMP

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

What SNARE proteins are present in the target membrane of the presynaptic active zone

A

2 types:
syntaxin
SNAP25

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

What happens to the SNARE proteins during exocytosis

A

form a trans-SNARE complex

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

What does the trans-SNARE complex consist of

A

4 alpha helices - one each from synaptobrevin and syntaxin and two from SNAP-25

Other proteins involved are:
Munc 13, Munc 18, complexins, Rab3A

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

What must happen to the trans-SNARE complex after fusion

A

dissemble via the action of NSF and SNAP (an adaptor protein)

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

What proteins mediate dissembly of the SNARE complex

What is the action

A

cytoplasmic ATPase called NSF (N-ethylmaleimide-sensitive fusion protein) binds to SNARE complexes via an adaptor protein called SNAP

NSF and SNAP use the energy of ATP hydrolysis to dissociate SNARE complexes, thereby regenerating free SNAREs.

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

What ion is involved in vesicle fusion?

What does this mean?

A

Ca2+

process should implicate a Ca2+ sensor - a role fulfilled by synaptotagmins

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

How are synaptotagmins associated to the vesicle

Give another fact about their structure

A

anchored to synaptic vesicles via their N termini while their C-termini contain two C2 domains - C2A and C2B, that bind Ca2+.

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

What do the C2 and 3 domains of synaptotagmins do respectively

A

C2: when Ca2+ is bound, C2 binds sytaxin and SNAP 25, and also binds to membranes containing acidic phospholipids

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

Describe the Ca2+ binding of the C2 domain of synaptotagmins

A

low binding affinity so large [Ca2+] required for activation

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

How do nerves ensure C2 domains of synaptotagmins get enough Ca2+

A

by co-localisation of synaptotagmin with Cav channels that cluster the active zone

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

Where do Ca2+ rich microdomains develop in the presynaptic terminal

A

in the vicinity of Cav pores due to the poor diffusibility of Ca2+

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

Why is the high [Ca2+] near Cav channels important

A

high local [Ca2+] can then be sensed by the synaptotagmins present in close proximity with these channels

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

Name 2 toxins that target SNARE proteins

How they both work very generally

A

botulinum toxins (BoTx)

Tetanus (TeNT)

Both selectively cleave SNARES

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

What are the 2 types of target proteins on the postsynaptic membrane for released transmitters

A

ionotropic (ligand gated ion channels) receptors

metabotropic receptors (GPCRs)

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

How do most small molecule NTs interact with postsynaptic receptors

A

bind to their cognate LGICs to mediate a fast post-synaptic response which could be the generation of either excitatory or inhibitory post-synaptic currents

or could bind to GPCR for a slower response (catecholamines and neuropeptides also do this second one)

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

Which postsynaptic receptor family is important in synaptic plasticity

A

GPCR - modulate strength of synaptic transmission by altering function/availability of LGICs in the postjunctional membrane

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

Can neurotransmitters change K+ position

A

binding to post synaptic GPCRs can open K+ channels leading to K efflux and reduced excitability of post synaptic membrane

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

True or false
Very little neurotransmitter is released upon stimulation

What does this mean for removal

A

False
Neurotransmitters are released typically in large amounts and timely removal of transmitters from the synaptic cleft is critical to synaptic transmission

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

How can NT be removed from the synaptic cleft

A

reuptake (either into presynaptic terminal or into surrounding, non neuronal cells eg glia)
Destruction of neurotransmitter (eg proteolysis of neuropeptides)

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

How is ACh removed from the synaptic cleft

A

hydrolyzed by AChE

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

Which are removed more quickly from the synaptic cleft:

neuropeptides or small molecule transmitters

A

small molecule transmitters are removed faster than neuropeptides

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

What is homotypic interaction at synapses

What is this also called

A

When released transmitters modulate further release of their own from the same nerve terminals

Homotropic interaction

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

What is heterotropic interaction at synapses

AKA?

A

when released neurotransmitters modulate release of other transmitters from different nerve terminals through activating cognate receptors (GPCRs) in the presynaptic terminal membrane

AKA heterotypic interaction

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

What receptors regulate homotropic and heterotropic interactions?

A

GPCRs called presynaptic autoreceptors (if homotropic interaction)
or

presynaptic heteroreceptors (if bind NTs from other synapses)

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

What are presynaptic hetero/autoreceptors usually coupled to

A

inhibitory G proteins (Gi or Go) leading to reduced [cAMP]] and enhanced K+ efflux - ultimately reduces NT release

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

Give an example of one of the few cases where released NT acting on a presynaptic GPCR leads to enhanced NT released

A

motor neurons connecting the skeletal muscle fibre

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

Is it only neurotransmitters that can act on presynaptic GPCRs?

A

No
sometimes, chemicals that are not canonical transmitters (e.g. prostaglandins, endocannabinoids, NO etc.) and produced by the post synaptic neurons/ effector tissues, can also modify neurotransmission through this pre-synaptic routes.

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

Give 3 chemicals that are not canonical transmitters but can modify NT release via presynaptic GPCRs

A

prostaglandins,
endocannabinoids,
NO

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

Other than action on presynaptic GPCRs what can affect release of NT

A

anything that interferes with the generation of Ca2+ signals in the presynaptic terminal
eg by inhibiting Cav channels or by chelating Ca2+

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

Give 2 agents which inhibit Cav channels in the presynaptic terminal

A

verapamil

aminoglycoside antibiotics

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

Which drugs chelate Ca2+ in the presynaptic terminal

A

tetracyclines

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

Where are LDCVs located in the presynaptic terminal

A

away from the active zone

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

What does exocytosis of synaptic/ clear core vesicles result from

What can trigger this

A

transient but large and local increases in intracellular Ca2+

a single depolarising event is enough to provide such Ca2+ signals.

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

What do LDCVs require to by exocytosed? (compared to clear core vesicles)

How is this requirement often met

A

smaller but sustained rise in global Ca2+ concentration

by a rapid train of action potentials (= repetitive firing of the nerve).

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

Why are the different requirements for exocytosis of LDCVs and clear core vesicles useful

A

specific patterns of electrical activity in a neuron may lead to the preferential release of a neuropeptide or a small-molecule NT

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

What are the 3 main broad sites of pharmacological intervention at the synapse

A

presynaptic nerve terminal,

sypatic/ junctional cleft

the post-synaptic/post-junctional target membrane

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

What are the 2 ways a pharmacological agent can modulate peripheral neurotransmission

A

direct (binds to postsynaptic target as an agonist, antagonist or pore blocker)

indirect (reduces/ enhances amount of NT released)

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

What was the first neurotransmitter to be discovered

A

ACh (1914)

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

Where is ACh found in the peripheral nervous system (5)

A
  • motor nerves that innervate the skeletal (voluntary) muscles
  • all autonomic pre-ganglionic nerves
  • nerves terminating at the adrenal medulla
  • postganglionic parasympathetic nerves at neuro-effector junctions
  • postganglionic sympathetic nerves innervating the sweat glands
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
89
Q

The availability of what chemical is vital for ACh synthesis?

How do we acquire it?

A

choline

primarily from diet as there is little de novo synthesis in neurons
can be recycled after ACh breakdown

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

What is ACh hydrolysed to

A

acetate and choline

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

What happens to the products of AChE

A

much of the choline is taken up by ChT1 for ACh synthesis

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

What is usually the rate limiting step in ACh synthesis

A

reuptake and availability of choline

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

What mediates choline reuptake into the presynaptic terminal

A

ChT1

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

Describe ChT1

What is it a family of

what can it transport

what inhibits it?

A

high affinity choline transporter
a member of the solute carrier family of proteins

transports Na+ and can be inhibited by hemicholinium

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

Is hemicholinium used clinically?

A

no

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

How is ACh formed from choline

A

Within the cytoplasm of the pre-synaptic nerves, choline is acetylated to form ACh by choline acetyltransferase (ChAT or CAT)

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

Where does the acetyl group for choline in ACh synthesis come from

A

acetyl CoA from the Mt

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

What is Triethylcholine

A

competes with choline as a substrate for CAT and is converted to acetyltriethylcholine which is in turn released instead of ACh

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

What does the release of acetyltriethylcholine mean

A

it is far less potent than ACh so acts as a false transmitter

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

How is ACh stored in the presynaptic terminal

A

in cholinergic vesicles

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

How does ACh enter the vesicles in the presynaptic terminal

A

by vesicular ACh transporters (VAChTs)

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

How do VAChTs work

A

high H+ gradient to allow exchange of H+ for ACh

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

What provides the H+ gradient across the ACh vesicle wall

A

V- type ATPase

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

What kind of vesicle is ACh stored in

A

clear core generally

contain ATP also at a ratio of 10:1

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

Do cholinergic presynaptic vesicles ever have dense core vesicles?

A

yes - some have vesicles containing VIP

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

Why do cholinergic vesicles sometimes contain ATP and VIP

A

ATP and VIP function as co transmitters at some synapses

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

What drug targets ACh storage

A

vesamicol

an experimental compound, is a non-competitive and reversible blocker of VAChT.

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

What is vesamicol

A

an experimental compound, is a non-competitive and reversible blocker of VAChT.

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

Which pharmacological agents target ACh release?

A

Botulinum (BoTXs/BoTNs)
β bungarotoxin (β-BuTx)
α- latrotoxin (α-LTX)
Tetanus neurotoxin (TeNT)

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

What produces botulinum

A

Clostridium botulinum
a Gram-positive bacterium responsible for botulism that may happen through food poisoning, colonisation of the gut in an infant or via exposed wounds

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

What do botulinum toxins target

A

cholinergic peripheral synapses (NMJ)

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

Describe the action of BoTX (think about their structure)

A

heavy chains bind to some gangliosides specific to the target nerves which allow them to be endocytosed specifically into these nerves.

C termini are proteases that are selective for SNARE

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

What are symptoms of botulism (2)

A

skeletal muscle weakness (which may lead to respiratory paralysis)

autonomic signs that would be associated with loss of cholinergic activity (e.g. constipation, blurred vision, dry mouth, difficulty in swallowing, urinary retention etc.).

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

Where is β - BuTx found

A

Venom of branded kraits

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

What is the action of β BuTx

A

Causes irreversible damage to pre-synaptic terminals which then lose synaptic vesicles

ACh release block is probs via its phospholipase A2 action - degrades phospholipids in active zone

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

Does β BuTx attack all nerves

Why

A

Only cholinergic motor nerves

May stem from its interaction with the substrate phospholipids that are unique to these nerves

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

How does β BuTx kill you

A

Respiratory failure

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

Where is α latrotoxin found

A

Black widow spider venom

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

How does α LTX cause paralysis

A

Causing a massive release and subsequent depletion of the ACh at the NMJ

ACh release in absence of depolarisation/ Ca entry

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

How do we think α LTX works

A

As a tetramer, It can form a calcium permeable ion channel

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

What produces TeNT

How does it enter the nervous system

A

Clostridium tetani (another Gram-positive bacterium)

enters the nervous system at the NMJs

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

How can TeNT enter the nervous system via the NMJ

A

the C-terminal heavy chain of TeNT binds to some gangliosides that are enriched on the peripheral terminals of motor neurons and this binding enables endocytosis.

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

What happens once TeNT enters through the NMJ

A

trapped in endosomes, TeNT progresses towards soma via retrograde axonal transport and is then discharged into the intersynaptic space within the spinal cord

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

What happens after TeNT is released into the inter synaptic space in the spinal cord

A

binds to presynaptic membrane of inhibitory interneurons to be endocytosed into the synaptic vesicles of these neurons

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

What happenns to TeNT after it is endocytosed into the synaptic vesicles of presynaptic inhibitory neurons of the spinal cord

A

acidic nature of vesicles causes insertion of N terminal light chain into cytoplasm and cleaves synaptobrevin

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

What is the result of TeNT’s N terminal cleaving synaptobrevin on the synaptic vesicles in the inhibitory presynaptic neurons in the spinal cord

A

the inhibitory interneurons are unable to release glycine, causing disinhibition of the motor neuron which then results in violent (tetanic) contractions of skeletal muscle.

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

True or false

ACh is a physiological inhibitor of itself

A

true
At postganglionic parasympathetic nerve endings, inhibitory muscarinic M2 receptors participate in auto inhibition of ACh release;

other mediators, such as noradrenaline (NA), also inhibit the release of ACh.

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

How does ACh modulate its own release at NMJ

What is the purpose

A

pre-synaptic nicotinic receptors (nAChRs) facilitate ACh release

allows synapse to function reliable during prolonged high frequency activity

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

What are cholinoceptors

A

cholinergic receptors that ACh binds to after its release

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

Where are cholinoceptors found and how can they be classified

A

post synaptic membrane

muscarinic and nicotinic

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

How can you experimentally determine a nicotinic and muscarinic AChR

A

on the basis of their different affinities for agents that mimic the action of ACh (cholinomimetic agents)

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

What are nAChRs

A

LGIC belonging to the ‘Cys Loop’ family

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

Where are nAChRs found

A

at the NMJs, autonomic ganglia, adrenal medulla, the CNS and some other non-neuronal (notably various immune and epithelial) cells

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

Describe the structure of a functional nAChR

A

a pentamer consisting of α,β, γ, δ and ε subunits with at least 2α subunits present. Each subunit consists of four transmembrane (M1-M4) helices and the M2 helix from all five subunits together form the pore. ACh binds to the interface between the α and neighbouring δ or γ subunits and minimally 2 molecules of ACh are required for the activation of the channel leading to the opening its pore.

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

What are most isoforms of the nAChR permeable to

Is this always the case?

A

Na and K

No- one of the CNS isoforms namely the (α7)5 channel, is highly Ca2+ permeable.

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

What does activation of nAChRs cause

A

depolarisation and therefore an EPP (muscle) or EPSP (peripheral ganglia)

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

What happens if an EPp is strong enough

A

An action potential is generated

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

True or false

nAChRs can be desensitised

A

True

Usually by prolonged or much stronger agonist stimulation

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

How can nicotinic AChRs be divided based on location

A

Nm (muscle) and Nn (neuronal)

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

What is the major isoform of the Nn nAChR

Where is it found

A

(α3)2(β2)3

Autonomic ganglia

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

What is the major variant of Nm nAChR?

Where is this found

A

(α1)2(β2)δε

Adult skeletal muscle membrane

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

What are the predominant Nm isoforms of the nAChR in the CNS

A

(α4)2(β2)3

(α7)5

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

True or false

All nAChRs are metabotropic

A

False

All mAChRs are metabotropic (GPCRs)

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

Why are they called muscarinic AChRs?

A

in addition to binding ACh, also recognize muscarine, an alkaloid that is present in certain poisonous mushrooms.

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

How do AChRs respond to nicotine?

A

nAChR: affinity
mAChR: low affinity

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

What do mAChRs mediate

A

postganglionic transmission in the peripheral nervous system, being present on the membranes of effector tissues including heart, tissue-specific smooth muscles (lung, some blood vessels, bladder, gut etc.) and glands (salivary, lacrimal etc). They also occur in the CNS

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

Where is the M2 mAChR found

What is its action here

A

pre-synaptically on the endings of noradrenergic nerves and cholinergic nerves

its activation causes a decrease in transmitter release

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

How many subtypes of mAChRs are there

Which are the most important in the periphery

A

5: M1-5

M1, 2, 3

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

Are M1, 2, 3 mAChRs found in autonomic ganglia

A

. Some of these isoforms may be present in autonomic ganglia but they are mostly present in the effector tissue membrane and they mediate the signalling leading to the biological functions of cholinergic stimulation.

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

Is AChE usually floating about?

A

no it is tethered to the post synaptic membrane

a soluble AChE is found in the cytoplasm of the presynaptic neuron (obvs doesnt affect life span of released ACh)

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

What does neurotransmission in autonomic ganglia involve

A

release of ACh by pre-ganglionic fibres and the rapid depolarization of post-ganglionic nerve membranes via the activation of neuronal nAChRs (Nn).

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

How is the structure of cholinergic autonomic ganglia different to the NMJ

A

Unlike the NMJs, ganglia do not have discrete end plates with focal localization of receptors; rather, the dendrites and nerve cell bodies contain the receptors

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

Which Nn is insensitive to alpha bungarotoxin

What can inhibit these

A

(α3)2(β2)3

trimetaphan (a competitive antagonists) and hexamethonium

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

What are the 4 different changes in membrane potential of postganglionic nerves from cholinergic stimulation

A
  • an initial EPSP (via Nn receptors) that may result in an action potential
  • an IPSP mediated by muscarinic M2 receptors
    (Gi/o-coupled)
  • a secondary slow EPSP mediated by muscarinic M1 receptors (Gq/11-coupled)
  • a late, slow EPSP mediated by various neuropeptides
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
155
Q

What is the issue with agents acting at the autonomic ganglia

A

lack of selectivity between sympathetic and parasympathetic ganglia and few can also have effects on the CNS. Their peripheral action is largely governed by the extent of the sympathetic and parasympathetic control of the tone of the effector systems.

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

What are the 2 categories in which ganglionic stimulants can be grouped

A

Nn agonists

mAChR agonists

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

Name a member of the Nn receptor group of ganglionic stimulants

A

nicotine

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

What is nicotine

A

a tertiary amine found in tobacco leaves

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

What does nicotine do at low and high concentrations

A

stimulates Nn channels whilst at higher doses, it desensitizes the channel (typical of for LGICs)

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

Name some products that use nicotine as an aid for smoking cessation

A

gums, lozenge, e-cigarettes (‘vapes’) and transdermal patches as part of the nicotine replacement therapy (NRT)

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

What is varenicline used for

A

used as a smoking cessation ‘aid’ along with counselling and behavioural therapy to help people stop smoking

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

What drug is used to aid smoking cessation in conjunction with therapy

Describe its action

A

varenicline

high affinity partial agonist for the α4β2 receptors (mainly implicated in nicotine addiction) and also a full agonist on α7 receptors

both are in the CNS

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

What receptor is mainly implicated in nicotine addiction

A

α4β2 Nn receptor

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

Name 2 ganglionic stimulants that act as Nn agonists

Are these drugs

A

tetramethylammonium (TMA)

dimethylphenylpiperazinium (DMPP)

not drugs but useful for experiments

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

What are mAChR agonists blocked by

A

atropine

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

What do mAChR agonists mimic

A

the slow EPSP

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

How does nicotine act as an Nn receptor blocking agent

A

initially stimulates the ganglia by an ACh-like action and then blocks them by causing a persistent depolarization

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

Do all ganglionic blocking agents act in the same way as nicotine

A

no other ones
impair transmission either by competing with ACh for binding at ganglionic Nn receptors (e.g. trimethaphan) sites or by blocking the channel pores (hexamethonium).

equivalent to non depolarising blockers at NMJ

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

What does trimethaphan do

A

competes with ACh binding at ganglionic Nn receptors

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

Which drug blocks channel pores at ganglionic cholinergic synapses

What is important to remember when prescribing this drug

A

hexamethonium

Blockade of ganglionic Nn channels by hexamethonium manifests use dependence.

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

What were the earliest antihypertensive agents

Are they still used?

A

ganglionic blocking agents

no - clinically obsolete by now due to numerous side effects, largely coming from their inability to discriminate between sympathetic and parasympathetic ganglia.

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

How many nerves innervate one muscle fibre

A

only one nerve fibre supplies each muscle fibre - the end plate

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

What happens at the NMJ structurally

A

the pre-synaptic axon terminal boutons synapse with the muscle fibre membrane (sarcolemma) that is deeply-infolded as ‘junctional folds’

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

Where are Nm nAChRs present in the sarcolemma

What else is near them

A

on the crest of the junctional folds

volatage gated Na channels are deep in the folds and in the non junctional part of the sarcolemma

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

What happens when ACh binds to Nm receptors

A

the nAChRs open to allow Na influx and K efflux

generates the EPP and if threshold for Nav channels is reached an AP is elicited

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

Nm channels are equally permeable to Na and K - does this mean equal amounts of Na enter and K leave?

A

No
the prevailing electrochemical gradients for Na+ is much greater than that of K+, so more Na+ enters than K+ leaves the cytoplasm

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

How is ACh removed from the NMJ

A

hydrolysed by AChE present in the basal lamina within the junctional cleft.

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

where is ACh found in the NMJ

A

in the basal lamina within the junctional cleft.

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

hat do directly acting agents modulating NMJ transmission act on?

A

directly on the Nm receptors on the sarcolemma

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

How do the subunits of the Nm receptor at the NMJ vary between the adult and foetus

A

The adult Nm is represented by (α1)2β1δε

the foetal form has gamma in place of the ε subunit

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

What is the adult Nm receptor irreversibly antagonized by

A

α-bungarotoxin

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

What is the main clinical use of neuromuscular blocking agents

A

as an adjuvant in surgical anaesthesia to obtain relaxation of skeletal muscles.

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

How do muscle relaxants improve the safety of anaesthesia ?

A

less anaesthetic is required, minimizing respiratory and cardiovascular depression and allowing rapid post-anaesthetic recovery

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

What is used in conjuction with GA during laryngoscopy, bronchoscopy, and esophagoscopy

Why do we not have to worry about these drugs having a wide spread effect

A

Short-acting drugs are used to facilitate endotracheal intubation

charged so don’t cross BBB and action remains peripheral

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

What are the 2 types of NMJ blockers

A

Depolarising and • non-depolarizing neuromuscular blocking agents

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

How do depolarising NMJ blockers work

A

Nm agonists so open Nm channels but are not readily hydrolysed by AChE

depolarisation last longer leading to some fasciculations,
followed by blocking of neuromuscular transmission and flaccid paralysis (called phase I block).

The depolarisation from the Nm channels leaves Nav channels in a inactivated state

with increasing [depolarising blocker] over time the block converts from phase 1 to phase II (membrane gradually repolarizes but now the Nm channels become desensitized)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
187
Q

How can a depolarising blockade be potentiated

A

Anti-AChE agents

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

Name depolarising NMJ blocker

A

Suxamethonium (succinylcholine)

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

Why is succinylcholine considered unique

A

Suxamethonium (succinylcholine) represents the only depolarizing blocking agent being used in the clinics at present.

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

What is phase I and II of the depolarising block at the NMJ

A

Phase I: extended depolarisation leads to fasciculations before flaccid paralysis because the open Nm receptors maintain the depolarized state of the sarcolemma and thus leaving all the Nav channels in inactivated state

Phase II: agent increases and membrane gradually repolarizes but now the Nm channels become desensitized.

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

When is succinylcholine used despite its side effects

A

still in conditions where fast onset and brief duration of action are required, e.g. with tracheal intubation

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

Why does succinyl-Ch have a v brief duration of action

A

due to its rapid hydrolysis in the plasma by butyrylcholinesterase (BuChE, also known as pseudocholinesterase).

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

Where is BuChE synthesised

What does this mean when administering NMJ blockers?

A

liver

patients with liver disease or genetic deficiency of this enzyme therefore run the risk of prolonged action of suxamethonium (which is hydrolysed by BuChE)

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

Whyy must you check a patient doesnt have electrolyte imbalance before administering succinyl-Ch

A

suxamethonium is a depolarising NMJ blocker and causes

K+ loss from the muscles into blood, which may lead to hyperkalemia and can be life-threatening for patients with electrolyte imbalance

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

What are non depolarising NMJ blockers essentially

A

competitive antagonists of Nm channels

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

true or false

non-depolarising NMJ blockers lead to flaccid paralysis

A

true
they prevent depolarization of the sarcolemma and inhibit muscular contraction, leading to a flaccid paralysis

fasciculations are not seen with these

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

How can the competitive action of non depolarising NMJ blockers be overcome

Why is this useful clinically?

A

by administration of anti-cholinesterases (e.g. neostigmine), which increase the [ACh] in the NMJ.

gives anaesthesiologists an option to shorten the duration of the neuromuscular blockade.

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

What was the first known non depolarising block

A

curare - a cocktail of natural alkaloids used as arrow poison by South American Indians to paralyze prey

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

What is the major constituent of curare

Is it used clinically?

A

d-tubocurarine

no due to poor selectivity between the ganglionic and NMJ-specific nAChRs

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

What are synthetic non-depolarising blocking drugsdivided into

A

the aminosteroid group (the ones with uronium suffix) such as pancuronium, rocuronium etc.
and
the benzylisoquinolinium group (the ones with the urium suffix) such as atracurium

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

Describe the aminosteriod group of drugs

Give 2 examples

A

synthetic NMNJ non-depolarising blocking drugs

Ends in uronium

pancuronium, rocuronium

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

Describe the benzylisoquinolinium group of drugs

eg?

A

synthetic non-depolarising blocking drugs

(the ones with the urium suffix) such as atracurium, mivacurium

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

How does the onset of rocuronium compare to succinyl choline

A

succinyl Ch has a faster onset

non-depolarising neuromuscular blocking drugs have a slower onset of action than suxamethonium

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

How can non-depolarising neuromuscular blocking drugs be divided based on action?

A

by their duration of action as short-acting (15–30 minutes), intermediate-acting (30–40 minutes), and long-acting (60–120 minutes), although duration of action is dose-dependent

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

Which kinds of non-depolarising neuromuscular blocking drug is used more commonly in practice

A

Drugs with a shorter or intermediate duration of action, such as atracurium and rocuronium, are more widely used than those with a longer duration of action, such as pancuronium

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

Name 2 non depolarising NMJ drugs which have an intermediate duration of action

Which has a longer duration

A

atracurium and rocuronium

pancuronium

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

What is common about NMJ non depolarising blocker structure

A

quaternary ammonium (=charged) compounds therefore poorly absorbed (cannot be given orally) and rapidly excreted.

For the same reason, they do not cross the placenta (which allows them to be safe in obstetric anaesthesia) as well as the blood-brain barrier.

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

What do anesthetists use to monitor postoperative recovery of neuromuscular transmission?

A

tetanic fade

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

What is tetanic fade

A

non-depolarising blocking agents also block facilitatory pre-synaptic autoreceptors and thus inhibit the release of ACh during repetitive stimulation of the motor nerve

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

What are some of the clinical responses to NMJ blocking agents caused by

A

release of histamine

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

Name 4 NMJ blocking agents that cause histamine release

A

Suxamethonium, mivacurium,
atracurium

to a lesser extent than d-tubocurarine unless administered rapidly

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

Which NMJ blockers are less likely to cause histamine release

A

amino steroids such as pancuronium and rocuronium

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

Why is histamine released by muscle relaxants?

A

Histamine release typically is a direct action of the muscle relaxant on the mast cell rather than an outcome of individual’s allergic hypersensitivity

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

How does alpha- bungarotoxin work

What is it often used in

A

irreversibly binds to the ACh binding site on the adult Nm receptors at the NMJ and thus block the transmission.

a- BuTx, specially its various antibody/fluorophore conjugates are widely used in biochemical and structural studies of Nm receptors and NMJs.

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

How are distinct bungarotoxins different

A

α-Bungarotoxin inhibits the binding of acetylcholine (ACh) to nicotinic acetylcholine receptors; β- and γ-bungarotoxins act presynaptically causing excessive acetylcholine release and subsequent depletion

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

How is BoTX used clinically

A

BoTX-A
injected locally for cosmetics

can treat certain ophthalmic conditions associated with spasms of eye muscles (e.g., blepharospasm).

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

how do anti-AChEs at on NMJ transmission

A

act indirectly by prolonging the life time of release ACh within the junctional cleft.

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

Why must patients with myasthenia gravis be careful with certain antibiotics

A

members of the aminoglycoside antibiotics (e.g. streptomycin, neomycin etc.), in large doses inhibit Cav channels and thus can produce neuromuscular blockade, especially in patients with myasthenia gravis

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

Which antibiotics can block Cav channels at the NMJ

Give 2 examples

A

aminoglycoside antibiotics

e.g. streptomycin, neomycin

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

What is the effect of aminoglycoside antibiotics on patients treated with a NMJ blocker

What other drug class has a similar effect

A

in normal or low doses can unpredictably prolong muscle paralysis

tetracyclines

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

Where are mAChRs primarily found

What is a key exception

A

present in autonomic ganglia and vascular endothelial cells

also on autonomic effector cells innervated by post-ganglionic parasympathetic nerves

exception: postganglionic sympathetic nerves to sweat glands are cholinergic and the released ACh acts via mAChRs present in the gland

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

What is unusual about mAChRs being in vascular endothelial cells

A

they receive little or no cholinergic innervations

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

In the majority of cases how many types of mAChR are present in the effector tissue

A

at least two muscarinic receptor subtypes (typically M3 and M1/M2)

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

What is the ‘neural’ subtype of mAChR

A

M1

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

Where are M1 mAChRs mainly found

A

on CNS, autonomic ganglia and on gastric parietal cells

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

How to remember what the different classes of mAChR are coupled to

A

all odd ones (M1, 3, 5); coupled to Gq/11

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

what does agonist binding to mAChR M1 lead to

What other mAChR types would behave in the same way

A

activation of PLCβ -> hydrolyses PIP2 to IP3 and DAG
IP3 mediates intracellular Ca2+ release
DAG activates PKC

M3 and M5

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

Why can IP3 readily diffuse through the cytosol

What do the characteristics of DAG mean for it respectively

A

it is polar and activates the ER membrane-bound IP3 receptors which then mediate intracellular Ca2+ release

DAG being lipophilic, stays in the membrane and activates PKC

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

Does DAG only activate PKC ?

A

No
also activates several members of the TRP family of ion channels which are mainly involved in sensory perception and several of them are also Ca2+ permeable, mediating Ca2+ influx.

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

What else (other than PLC) mediates [DAG]

A

DAG lipase to produce arachidonic acid (AA).

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

What is the importance of the production of AA from the action of DAG lipase on DAG

A

may trigger a distinct Ca2+ influx pathway in some cells

most importantly, it is the precursor of biologically active lipid-derived molecules eg prostaglandins and leukotrienes .

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

What do M1 receptors in neurons inhibit

How does it do this (2)

What is the purpose?

A

K+ conductance (the m channel)

through the depletion of the PIP2 in the postganglionic nerve membrane. In certain case, the Ca2+ signals via calmodulin (CaM) and PKC, also contributes to the process

brings about a late depolarisation in the autonomic ganglion which occurs many seconds after the initial, fast EPSP induced by Nn receptors

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

What are M channels that conduct K+ known to be composed of

A

subunits of the Kv7/ KCNQ channel family.

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

Describe the M2 mAChR receptor

What are they coupled to

What does this mean they activate

A

cardiac subtype

Gi/o proteins

the inhibition of adenylyl cyclase (AC) which results in decreased cAMP production and thus decreased activation of protein kinase A (PKA)

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

What does M2 activation ultimately lead to

A

in decreased phosphorylation of a number of proteins that notably include the voltage-gated Ca2+ channels (CaVs), reducing probability of opening and Ca2+ influx

This decreases NT release from neurons

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

What is the predominant mAChR subtype in the heart

How are they distributed

A

M2

largely confined to the SAN and AVN to some extent in the atria but much less in the ventricles.

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

What does stimulation of the M2 receptors in the heart result in

A

negative chronotropic effect

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

How does activation of M2 channels in the heart

A

reduced Ca2+ current via the L and T-type Cavs due to reduced PKA-mediated phosphorylation of these channels

βγ opens g protein coupled inwardly rectifying K+ channels in the nodal pacemaker cells. This causes hyperpolarisation by K+ efflux

threshold for opening of the HCN channels which mediate the slowly-depolarizing inward Na+ current (the If current), is shifted towards a more negative value.

Reduces atrial contractility via triggering I(K-ACh)

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

What are the inwardly rectifying K+

A

GIRK1

Kir3.1

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

What is the K+ efflux current following M2 stimulations in the heart called

A

I (K-ACh)

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

What causes the threshold opening of the HCN channels which mediate the slowly-depolarizing inward Na+ current (the If current), to shift towards a more negative value.

A

reduced [cAMP]

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

What can the M3 mAChR subtype be classed as

A

glandular subtype

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

What are M3 receptors coupled to

What is usually the result of stimulation of these mAChRs

A

Gq and G11

stimulation of glandular secretions, e.g. saliva, sweat etc

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

What doe activation of M3 receptors in smooth muscle lead to

A

contraction because of the increase in cytosolic free [Ca2+]i

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

What causes [Ca2+]i to increase when M2 mAChRs are activated in smooth muscle

Why is the effect different in vascular smooth muscle

A

IP3 invoked Ca2+ release from ER

increased endothelial [Ca2+]i binds to CaM
Ca2+-CaM then activates eNOS to produce NO leading to smooth muscle relaxation

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

What are the following:
CaM
eNOS
EDRF

A

CaM = calmodulin
eNOS: endothelial nitric oxide synthase
EDRF: ‘endothelium-derived relaxation factor’ ie NO

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

What is the effect of CaM-Ca2+ activating eNOS in smooth muscle

A

NO activates soluble guanylyl cyclase, produces cGMP and ultimately relaxes the vascular smooth muscle

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

How can pharmacological agents modulate muscarinic cholinergic synapses (2)

A

the activity of mAChRs by physically binding to these receptors (directly-acting agents)
- the amount/life-time of ACh released from the post-gangilonic nerve terminal (indirectly-acting agents)

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

What happens to vascular smooth muscle if you rub away the endothelium and add ACh

A

no relaxation until the ACh is washed away

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

Which mAChR is mainly present in the eye and which parts of the eye

A

mainly M3 - constrictor pupillae; ciliary muscles; lacrimal glands

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

What does stimulation of M3 receptors in the constrictor pupillae lead to

A

contraction
shortens pupil diameter (miosis)
lowers intraocular pressure

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

What does stimulation of M3 receptors in the ciliary muscles lead to

A

contraction to reduce focal length of lenses, allowing accommodation

253
Q

What does stimulation of M3 receptors in the lacrimal glands lead to

A

increased secretion

254
Q

What is the distribution of mAChR in the detrusor muscle and mucosa/submucosa of the bladder

A

detrusor: M2=80%
M3=20%
(some beta 3 ARs present too)

mucosa and submucosa: M2 and M3

255
Q

What is the PNS supply to the bladder

A

via pelvic nerve

256
Q

What is the distribution of mAChRs throughout the lungs

A

pre-junctional terminal: M2 to reduce ACh release

postganglionic cell body: M1

airway smooth muscle cells: M2 and 3 for bronchoconstriction

at submucosal glands: M1, M3, to increase mucus secretion

257
Q

How does the Vagus affect the lungs

A
tonically fires to
contract (mainly via M3-
PLC(beta)-IP3 mediated) airway
smooth muscle during
normal breathing - vagal tone
258
Q

What effect does the vagus nerve have on the lungs when irritants are present

Who is this dangerous for

A

irritants stimulate sensory vagal afferents, activating a reflex PNS activity via the vagal efferents causing bronchospasm

it is major
bronchoconstricting factor for COPD patients

259
Q

True or false:

It’s relatively easy to develop directly acting mAChR ligands that are subtype selective

A

False
No it is very challenging

the classical (orthosteric) binding site (= where ACh binds to) is highly conserved across mAChR subtypes

260
Q

What have we used to discover that the ACH binding site is conserved across mAChRs

A

Analysis of amino acid sequence and recent X-ray crystallographic studies

261
Q

What have we used to overcome the problems of the conserved binding site of mAChRs

A

these receptors possess distinct allosteric sites located often within the extracellular loops or the outer segments of different transmembrane (TM) helices.

Because these regions show a considerable degree of sequence variation across M1–M5 receptors, considerable progress has been made in developing subtype specific allosteric modulators.

262
Q

How do allosteric modulators affect mAChRs

A

by altering the affinity or efficacy of orthosteric muscarinic ligands.

263
Q

What are PAMs in relation to mAChRs

What is another related drug type

A

Positive allosteric modulators (PAMs)

enhance orthosteric activity,

negative allosteric modulators (NAMs) inhibit it

264
Q

allosteric modulators have lead to development of new therapeutic agents with increased efficacy and reduced side effects. Name 2 diseases where this research is focussed

A

several severe disorders of the CNS, including Alzheimer disease and schizophrenia.

265
Q

Other than allosteric modulators, what is another potential strategy for mAChR subtype selectivity

A

development of hybrid, bitopic ligands that interact with both the orthosteric binding cavity and an allosteric site

266
Q

Why are hybrids that bind to both the orthosteric and allosteric sites of mAChRS being developed?

A

By targeting orthosteric and allosteric sites simultaneously, bitopic ligands can potentially achieve both high affinity and receptor subtype selectivity.

267
Q

How can directly-acting parasympathomimetic or cholinomimetic agents (muscarinic agonists) be classified?

A

1) choline esters

2) naturally occurring cholinomimetic alkaloids

268
Q

Name some choline esters

A

include ACh and synthetic esters of choline, such as carbachol (carbamylcholine) and bethanechol

269
Q

Name 2 synthetic choline esters

A

carbachol (carbamylcholine) and bethanechol

270
Q

name a cholinomimetic alkaloid

A

pilocarpine

271
Q

True or false

. All of the direct-acting cholinergic drugs have a longer duration of action than ACh.

A

True

All of the direct-acting cholinergic drugs have a longer duration of action than ACh.

272
Q

What is Bethanechol

What does it activate and where does it not have action?

A

chemically a hybrid of methacholine and carbachol

non-selectively activates mAChRs
lacks nicotinic action

273
Q

What is the effect of bethanechol on the bladder

A

stimulates the detrusor muscle of the bladder (via M3 receptor), whereas the trigone and sphincter muscles are relaxed

ie produces urination

274
Q

What is bethanechol licensed for

Is it very common?

A

for acute postoperative, postpartum and neurogenic urinary retention

its use has largely been superseded by catheterisation

275
Q

What is pilocarpine used for

A

to treat glaucoma

276
Q

What is the drug of choice for emergency lowering of intraocular pressure for glaucoma patients?

How is it administered?

A

pilocarpine

by instillation as eye drops and it is readily absorbed across the conjunctival membrane

277
Q

How is pilocarpine absorbed

A

administered by instillation as eye drops and it is readily absorbed across the conjunctival membrane

278
Q

What is the action of pilocarpine once it has been absorbed across the conjunctiva

A

contracts the ciliary muscle to produce traction on the trabecular meshwork around the Schlemm canal, causing an immediate drop in intraocular pressure as a result of the increased drainage of aqueous humour

279
Q

How quickly does pilocarpine act

How long does it last

Is it repeatable?

A

action occurs within a few minutes

lasts 4 to 8 hours

can be repeated.

280
Q

Why may patients who have taken pilocarpine struggle to focus their eyesight?

A

pilocarpine causes miosis, which is experienced as a spasm of accommodation and vision is fixed at a random distance

281
Q

What is the effect of atropine on the eye

A

opposes the effects of pilocarpine as atropine is a muscarinic blocker

282
Q

Name 2 diseases where pilocarpine is used to increase secretion

A

xerostomia

Sjögren’s syndrome

283
Q

What is Sjögren’s syndrome

A

syndrome which is an autoimmune disease of glands that secrete fluid

284
Q

What is xerostomia

A

chronic dry mouth problem, most common complications associated with radiotherapy of head and neck cancer)

285
Q

How do pharmacological antagonists of mAChRs work

A

directly acting anti-cholinergic or cholinolytic agents

286
Q

What site do competitive inhibitors of mAChRs block

Where are these receptors found?

A

the othosteric site

on effector cells at parasympathetic neuroeffector junctions in the peripheral ganglia (as well as the CNS).

287
Q

True or false

mAChR competitive inhibitors can block SNS transmission

A

True - mostly acts on PNS but can act on cholinergic junctions eg sweat and salivary glands

288
Q

Do mAChR competitive inhibitors affect AChRs

What does this mean

A

no

little or no action at NMJs or autonomic ganglia.

289
Q

Describe the chemical structure of anti-muscarinic agents

A

contain an ester and basic groups like that of ACh but bulky aromatic group replacing the acetyl moiety

290
Q

What kind of drug is scopolamine

Name another of this type

A

a naturally occurring alkaloid mAChR inhibitor

atropine (you need to know this one)

291
Q

WHat are the 3 different types of anti-muscarinic agent

Name one of each

A

naturally occurring alkaloid (atropine)

natural alkaloid semisynthetic derivative (ipratropium)

synthetic agents (solifenacin)

292
Q

Name 2 semi synthetic anti muscarinic agents

A

homatropine,
methscopolamine,
ipratropium

293
Q

Name 5 synthetic muscarinic agents

A
tropicamide, 
cyclopentolate, 
pirenzepine, 
darifenacin, 
solifenacin
294
Q

Give example of some relatively selective mAChR blockers (4)

What are most mAChR antagonists?

A

pirenzepine = M1
MT7= M1
darifenacin and solifenacin =M3

most are non selective

295
Q

What is MT7

A

A toxin in the venom of the African green mamba that selectively targets M1.

296
Q

What are mAChR blockers used for clinically

A

to inhibit effects of parasympathetic activity in the respiratory tract, eye, urinary tract, GI tract and heart

297
Q

Which muscarinic blockers are used on the eye clinically

A

topically for mydriasis and cycloplegia in ophthalmic examinations

298
Q

Would you want to use atropine for an ophthalmic examination?

A

Short-acting, relatively weak mydriatics, such as tropicamide (action lasts for 4–6 hours) and cyclopentolate (action up to 24 hours), are typically preferred over atropine (action may last up to 7 days).

299
Q

What drugs would you use to make funduscopy easier (2)

What effects do they cause that make this beneficial

A

tropicamide
cyclopentolate

mydriasis
cycloplegia

300
Q

What is used as the standard treatment of poisoning

A

atropine in large doses by irreversible, long lasting inhibition of AChE

301
Q

What is the main effect of antimuscarinic agents on the heart

A

main effect is on rate - dominant effect= tachycardia

clinical doses may lead to transient bradycardia

302
Q

How does the effect of atropine on the heart change as its dose changes

A

Larger doses of atropine cause progressive tachycardia by blocking M2 receptors on the SA nodal pacemaker cells, thereby antagonizing parasympathetic (vagal) tone to the heart

303
Q

in which type of patient is the effect of atropine more profound

A

healthy young adults, in whom the vagal tone is considerable

304
Q

What is atropine useful for clinically

A

to manage bradycardia associated with the excessive use of the ‘beta blockers’ and acute myocardial infarctions.

305
Q

Which mAChR is involved in bronchoconstriction

What happens if these receptors are activated on lung epithelia

A

M3 on airway smooth muscle

mucous secretion

306
Q

Describe a ‘vagally-mediated reflex bronchospasm’

A

lung M3 mAChR activation happens normally as a ‘vagal tone’ but many irritants (dust, allergens, histamine, some gases etc.) via local sensory nerves can make it more prominent in the form of a ‘vagally-mediated reflex bronchospas

307
Q

What is the relevance of vagal tone in COPD

Is this the same in asthma

A

it is the major reversible brochoconstrictive component

no

308
Q

What does M3 blockade in bronchial smooth muscle result in?

How does the effect of these blockers differ from a normal patient to a COPD patient

A

bronchorelaxation/ bronchodilation due to decreased muscle tone

minimal effect in normal airways but significant in COPD

309
Q

What are the 2 categories of mAChR blockers used in COPD

Give an example of each

A

LAMA (long acting muscarinic antagonist) - tiotropium

SAMA (short acting muscarinic antagonist) - ipratropium

310
Q

Name 2 LAMA drugs

A

tiotropium and glycopyrronium

311
Q

True or false

All drugs in both SAMA and LAMA categories have higher selectivity for one type of mAChR

A

True
Higher selectivity for M3 than M2

LAMA also dissociate much faster from M2 than 3

312
Q

Where does the longevity of LAMA action come from

A

their slow dissociation from M3 receptors

313
Q

What kind of anti muscarinic agent can be used if asthma is not being adequately controlled by beta-AR stimulants

A

LAMA as additional bronchodilators

314
Q

What do you use to treat urinary incontinence

Give 2 examples

How does this work

A

M3 selective antagonists

eg solifenacin and darifenacin

selective inhibition of M3 receptors that mediate contraction of bladder’s detrusor muscle

315
Q

Which anti-muscarinic drug can be used to reduce excessive salivation

Which feature allows this

A

glycopyrronium

cannot cross BBB

316
Q

Why might someone have the symptom of excessive drooling

A

drug induced or associated with heavy-metal poisoning and Parkinson’s disease

317
Q

What are the 2 types of cholinesterase

Describe the specificity of each

A

BuChE (AKA pseudocholinesterase)
AChE

AChE = only ACh
BuChE= much broader
318
Q

Name 2 drugs that BuChE can break down

A

procaine (a local anaesthetic)

suxamethonium (depolarising NMJ blocker)

319
Q

Where is BuChE made

Where can it be found in the body (4)

A

liver

mostly blood
traces in: skin, gut, and brain

320
Q

Where is AChE found (2) and what form does it take in these locations

A

both in the presynaptic nerve ending (as a soluble form in the cytoplasm) and in the synaptic cleft (as tethered to the postsynaptic neuronal membrane/junctional fold).

321
Q

What is the presynaptic purpose of AChE (2)

A

guards against the accumulation of cytoplasmic ACh outside storage vesicles

termination of ACh action

322
Q

How is AChE attached to the post synaptic terminal

A

3 tetramers of AChE are attached by disulphide bonds to a long collagenous tail which tethers the enzyme to the postsynaptic/post-junctional target membrane.

323
Q

Where in the synpase is the AChE primarily responsible for ending ACh action ?

A

membrane-bound oligomeric form of AChE

324
Q

What is the turnover of AChE

A

> 10,000 mol per active site per second

325
Q

What kind of enzyme are the cholinesterases?

A

serine hydrolases

326
Q

What part of the Ser group in Ser hydrolases performs the nucleophilic attack

What is the purpose of this attack

A

hydroxyl (-OH) group

An intermediary adduct with the substrate is formed, which then undergoes hydrolysis

327
Q

What is the 3D structure of AChE

Where is the catalytic site? Give measurements and a specific name

A

hourglass

at the base of a deep (~about 20Å from the surface) and narrow (~ 5Å wide) tunnel named the aromatic gorge.

328
Q

What is the peripheral anionic site?

What is the purpose of this?

A

the entrance too the aromatic gorge of AChE

recognition of ACh

329
Q

What is one of the key interaction s of the peripheral anionic site

A

a weak cation-pi interaction between the heteroaromatic ring of a particular tryptophan residue (W279 for mammalian AChE) and the charged quaternary nitrogen of ACh

330
Q

What happens after the peripheral anionic site recognizes ACh

A

transferred down the aromatic gorge to the active site

331
Q

How is the ACh helped down the aromatic gorge after being recognised by the peripheral anionic site?

A

14 conserved aromatic residues line the gorge and help in pushing the ACh downwards towards the catalytic centre through cation-pi interactions with ACh. An electrostatic gradient running down the gorge also favours such movement of ACh.

332
Q

What happens to ACh when it reaches the end of the aromatic gorge

A

to the ‘catalytic anionic site’ (CAS) which helps in properly positioning ACh against the catalytic triad consisting of specific glutamate, histidine and serine

333
Q

Describe the tetrahedral intermediate formed by nucleophilic attack of Ser on ACh in AChE (a Ser hydrolyase)

A

has a charged oxygen

intermediate is stabilised by backbone amide groups (oxyanion hole)

from this the ACh bond is broke, releasing choline, and leaving acetyl-serine behind

334
Q

After choline has been released from AChE, an acetyl serine is left. What happens next

A

The serine¬-acetate bond is then spontaneously hydrolysed by water to release acetate and regenerate the enzyme

335
Q

How fast is the action of AChE

A

one of the fastest known : >10^4 molecules of ACh are hydrolyzed per second by a single AChE molecule, requiring only about 100microsec.

336
Q

What are anticholinesterases also referred to as

A

as indirectly acting cholinergic/cholinomimetic agents because they generally do not activate receptors directly

337
Q

Why are anti-AChE agents often used as toxins

Give a famous example of how they were used recently

A

cholinergic synapses are widely distributed throughout the body

deployment of nerve gas (novichok) in an attempt to assassinate victims in Salisbury in 2018

338
Q

What is a necessary feature of an anti-AChE agent used to treat Alzheimer’s

Give 2 examples

A

can cross BBB

rivastigmine
donepizil

339
Q

How can you categorise anti-AChE agents by:

a) mechanism of inhibition
b) duration of action

Give examples

A

a) • Reversible: a) non-covalent (edrophonium) b) covalent inhibitors (physostigmine, neostigmine etc)
• Irreversible - organophosphates such as dyflos, parathion, ecothiophate

b) • Short acting (edrophonium)
• Intermediate acting (the ‘stigmines’)
• Long acting (the organophosphates

340
Q

What is edrophonium

A

Non-covalent, reversible, short acting anticholinesterase

341
Q

Describe the structure of edrophonium

Where can it act

A

a quaternary ammonium compound

restricted to the periphery

342
Q

Name a non-covalent, reversible, short acting anticholinesterases and describe how it interacts with AChE

A

edrophonium

interacts only to the catalytic anionic site (CAS) of the enzyme, primarily through a cation- π interaction between its quaternary nitrogen and the aromatic (indole) ring of a tryptophan (W84) residue of AChE

the reversible nature of this interaction explains the short duration of edrophonium action

343
Q

What is the duration of edrophonium action

A

2-10 min

344
Q

What is edrophonium used for

A

the tensilon test -
in order to diagnose myasthenia gravis

HOWEVER:
Edrophonium is discontinued in the UK as its use as a diagnostic tool in myasthenia gravis has been largely superseded by antibody testing and the use of electrical conduction studies.

345
Q

What does a positive result in the tensilon test look like?

Mention drug used and illness diagnosed

A

A myasthenia positive individual characteristically feels temporary improvement in the facial weakness and ptosis within 5-10min of edrophonium injection

346
Q

Describe Covalent, reversible, medium acting anti-cholinesterases as a group

Give 4 examples

A

esters of carbamic acid (instead of acetic acid as with ACh) and have the suffix ‘stigmine’ and include physostigmine, neostigmine, pyridostigmine and rivastigmine.

All except rivastigmine are charged and therefore act only peripherally.

347
Q

How does neostigmine interact with AChE?

What kind of drug is this

A

With their basic group, they interact with the anionic site of AChE and transfer carbamyl group to the hydroxy group of Ser 203. Carbamylated AChE is more stable than acetylated AChE and takes minutes to hydrolyze. Hence these are medium acting agents.

Covalent, reversible, medium acting anti-cholinesterases

348
Q

How and when is neostigmine administered

A

intravenously to reverse neuromuscular blockade after surgery and orally to treat myasthenia gravis

349
Q

How is pyridostigmine administered

When

Why this drug

A

orally

myasthenia gravis

longer action than neostigmine

350
Q

How is ACh relevant to Alzheimer’s

A

there is significant loss of cholinergic neurons in the basal forebrain and the resultant reduced cholinergic neurotransmission is considered to underlie, at least in part, the dementia, intellectual deterioration, and personality changes associated with AD.

351
Q

Describe irreversible, long acting anti-cholinesterases as a chemical group

A

organophosphorous compounds containing a pentavalent phosphorous atom connected to three oxygen atoms (sometimes two oxygen and one sulfur atoms) along with a labile group, such as fluoride (dyflos or DFP) or an organic group (ecothiophate, parathion)

352
Q

How do irreversible, long acting anti-AChE work

A

labile group released and Ser’s -OH phosphorylated

this is v stable and lasts hundreds of hours

353
Q

How is the AChE-organophosphate complex following administration of irreversible long acting anti-AChE drugs

A

subject to a process known as ‘aging’, in which oxygen–phosphorus bonds within the inhibitor undergo spontaneous rearrangement in favour of stronger bonding between the enzyme and the inhibitor. Once aging occurs, the duration of AChE inhibition is increased even further.

Thus, organophosphate inhibition is essentially irreversible, and the body must synthesize new AChE proteins to restore AChE activity.

354
Q

How are irreversible, long acting anti-AChE used clinically

Use specific examples

What other uses do they have

A

limited clinical uses

Ecothiophate- used locally in glaucoma

insecticides
chemical warfare nerve gases eg sarin and novichok

355
Q

Name 4 nerve gases

What kind of drug is this

A

sarin, tabun, soman, and novichok

irreversible, long-lasting anti-AChE

356
Q

What does acute poisoning by irreversible anti-AChEs result in

When does it become fatal

A

excessive salivation, severe bradycardia, hypotension and difficulty in breathing

when combined with a depolarising neuromuscular block and central effects

357
Q

How do you treat anti-AChE poisoning

A

large doses of atropine as it can reach brain and periphery

358
Q

What is the effect of atropine of peripheral NMJ compromise

what other drug is used in a compromised NMJ

A

no effect

pralidoxime (2-PAM, a AChE reactivator) must be used instead

359
Q

What is 2-PAM used for

How does it work

A

to reactivate AChE

cationic group of pralidoxime interacts with the anionic site of AChE, which brings the oxime group into close proximity with the phosphorylated serine. The oxime group is a very strong nucleophile and lures the phosphate group away from the Ser hydroxyl of the enzyme

360
Q

How is the AChE reactivation by 2-PAM limited

Give the chemistry behind this

A

effectiveness is limited to within a few hours of exposure

phosphorylated AChE undergoes ageing, rendering phosphorylated groups no longer susceptible to nucleophilic attack

361
Q

Name 3 catecholamines

Why are they known as catecholamines#?

A

noradrenaline
adrenaline
dopamine

they possess a catechol and amine connected by a 2 carbon linkage

362
Q

How is the 2 carbon linkage in dopamine different to the other catecholamines

A

NA, Adr, and ISO all have an alcoholic -OH on the beta C (the C closest to the catechol)

DA does not

363
Q

What is a catechol

What is an amine group

A

catechol: aromatic ring with two proximal hydroxyl groups
amine: -NHR, where R = H or alkyl group

364
Q

What is the difference between noradrenaline and Adr

A

NA is effectively Adr lacking the methyl on the amine

365
Q

What is the main NT in the SNS

A

NA

366
Q

What is the precursor of NA?

Where does this precursor have important action

A

dopamine

mainly in the CNS and dopaminergic nerves

367
Q

How does Adr mainly act

Where is it made

A

as a hormone

adrenal glands

368
Q

True or false

NA is effectively the N-methylated version of Adr

A

False

Adr is effectively N-methylated NA

369
Q

How could you convert NA to Adr

Where does this take place

A

methylation

place in the chromaffin cells of adrenal medulla in presence of the enzyme PNMT (phenylethanolamine N-methyltransferase

370
Q

How much of the catecholamines in the adrenal medulla are Adr

A

80%=Adr

20%= NA

371
Q

What is ISO

A
isoprenaline
a synthetic (= N-isopropyl) derivative of NA, often used experimentally.
372
Q

How does ISO differ from NA

A

amine group on NA is just -NH2

ISO: -NH-CH-(CH3)2

373
Q

What is the starting material for biosynthesis of catecholamines? (Give the specific type)

How is this taken to to form catecholamines?

A

L-tyrosine

present in bodily fluid and taken up from the circulation by catecholaminergic nerves and converted into DOPA (dihydroxyphenylalanine) by tyrosine hydroxylase (TOH)

374
Q

What is the rate limiting step in catecholamine biosynthesis

A

conversion of L-tyrosine to DOPA by tyrosine hydroxylase (TOH)

375
Q

Where is TOH found

A

ONLY in catecholaminergic nerves

376
Q

Give each step of Adr synthesis with the enzyme for each equation (5)

A
  1. L-tyrosine
    enzyme: TOH
  2. DOPA
    Enzyme: DOPA decarboxylase
  3. Dopamine
    enzyme: Dopamine beta-hydroxylase
  4. NA
    enzyme: PNMT

5.Adr

377
Q

what is DOPA decarboxylase also known as?

Why?

A

L-aromatic acid decarboxylase

it’s relatively non-selective and catalyzes decarboxylation of other L-aromatic acids such as L-histidine or L-tryptophan to produce histamine and serotonin (5-HT), respectively

378
Q

What is the effect of L-aromatic acid decarboxylase on the following:

a) L-histidine
b) L- tryptophan

A

converts them to

a) histamine
b) 5-HT

379
Q

What happens to dopamine after it is formed (not conversion to NA)

What drives this process

A

stored into vesicles via the vesicular monoamine transporter (VMAT)

transvesicular H+ gradient

380
Q

What happens to dopamine inside the vesicles

A

hydroxylated at its β-carbon to form NA and this is catalyzed by dopamine β-hydroxylase (DBH).

381
Q

Where can DBH be found

How do you stop DB being released with NA when NA vesicles fuse with presynaptic membrane

A

exclusively within noradrenergic nerves

you don’t - small amounts of DBH are co-released with NA

382
Q

What happens to released DBH

What can we use this as an indicator for

A

not rapidly degraded or susceptible to any uptake mechanism, so its concentration in plasma and body fluids can be used as an index of overall sympathetic nerve activity.

383
Q

Where is PNMT mainly expressed in the adrenal medulla

A

in the adrenaline-releasing (A) cells that are distinct from the smaller proportion of noradrenaline-releasing (N) cells

384
Q

What controls PNMT expression

A

glucocorticoids eg cortisol from adrenal cortex

385
Q

How does cortisol affect catecholamine production (2)

A

Reduction in cortisol levels causes a decrease in production of PNMT and, hence, in the Adr:NA ratio.

also increases activity of dopamine β-hydroxylase (DBH).

386
Q

Which pharmalogical agent is used experimentally to block NA production

A

α-Methyltyrosine -

a competitive inhibitor of TOH

387
Q

What is the importance of blocking TOH as a way of reducing amount of NT produced?

A

it is the rate limiting step using α-Methyltyrosine is the only effective way to reduce amount of catecholamine produced

388
Q

Give an example of the clinical use of α-Methyltyrosine

A

in the past was used pre-op before treatment for pheochromocytoma (adrenal gland tumour, increasing Adr and NA secretion)

389
Q

What is the most effective way to increase production of catecholamines pharmacologically

A

bypass the step catalysed by TOH through using substrate for one of the later enzymes

390
Q

What happen in Parkinson’s disease

A

dopaminergic neurons in the substantia nigra pars compacta (SNc) are selectively degenerated

391
Q

Why can we not just use exogenous DA to replace that which is lost by destruction of dopaminergic neurons in Parkinson’s?

What is used instead?

A

DA does not cross the BBB

L-DOPA, DA’s natural precursor, can cross
Therefore L-DOPA is used to boost DA synthesis in the Parkinsonian brain

392
Q

What accounts for the unwanted peripheral effects of L DOPA therapy

A

A substantial fraction of the administered L-DOPA undergoes decarboxylation in the periphery by peripheral decarboxylase to produce DA and NA

393
Q

How to overcome the peripheral side effects of L-DOPA therapy

A

use in combination with carbidopa

carbidopa cannot cross the BBB but can inhibit peripheral decarboxylases

this ensures L-DOPA conversion to DA only occurs in CNS

394
Q

Which drug is used to inhibit DBH

How is it used clinically

A

Disulfiram

as an adjunct in the treatment of alcohol dependence

395
Q

How does disulfiram work

What is the basis of its action in dealing with alcoholism?

A

act by chelating the Cu2+ ion which is an essential co-factor of DBH or it may attack the sulphur-handling system for the methyl donor, S-adenosyl methionine

inhibition of aldehyde dehydrogenase

396
Q

How selective are the enzymes in the biosynthesis of catecholamine biosynthesis

What does this mean

A

TOH- selective
DDC and DBH - non selective

can result in the production and storage of a variety of other amines in the synaptic vesicle

397
Q

What happens when α-Methyldopa is taken into noradrenergic nerve terminals

A

converted successively into α-methyldopamine (α-methyl DA), and α-methylnoradrenaline (α-methyl NA)

398
Q

What happens to α-methyl NA in nerve terminals

A

stored within synaptic vesicles and released with NA as a false transmitter

399
Q

Describe the action of α-methyl NA compared to NA

A

functional but less active than NA at alpha 1 ARs and more selective towards alpha 2

400
Q

Name 2 drugs that target alpha 2 more selectively than NA does normally

What do they both result in? How?

A

clonidine
α-methyl NA

decrease BP
inhibition of NA release (via presynaptic alpha2 AR stimulation) and central action

401
Q

What kind of vesicle is NA usually stored in

What is often stored with it

A

clear core

ATP

402
Q

what transports NA and DA into vesicles

What family is it part of

What does it use to power the active transport?

A

VMAT2 (vesicular monoamine transporter)

SLC protein family

trans-vesicular H+ gradient

403
Q

How many H+ are transported per amine by the VMAT2

A

2H+ extruded per amine imported into the vesicle

404
Q

How is the H+ gradient in catecholamine vesicles set up

A

up by an ATP-dependent proton pump (a v-ATPase).

405
Q

What is VMAT1 also known as

Where is it found

A

chromaffin granule amine transporter

in non-neural tissues including the chromaffin cells of the adrenal medulla and few other cells of the GI tract.

406
Q

What is reserpine

What does it do

A

– a naturally occurring alkaloid (not used clinically),

binds to the amine site of the VMAT2 with high affinity and irreversibly blocks the uptake of monoamines (DA, NA and 5-HT) into vesicles

407
Q

What does reserpine’s action eventually lead to

Why does this happen

A

long lasting depletion of stored NA (and 5-HT in the CNS)

vesicles do allow leakage of the stored catecholamines into the cytoplasm where it is metabolised

408
Q

What was reserpine used for originally

Why was it discontinued?

A

as an anti-hypertensive

lead to profound psychological depression (due to decreased central 5-HT)

409
Q

Have any VMAT2 inhibitors been approved by the FDA

What are they approved for

A

a few - namely tetrabenazine and valbenazine

to manage abnormal involuntary movements associated with Huntington’s disease, tardive dyskinesia etc

410
Q

What do the 2 VMAT2 inhibitors have in common

A

both pro-drugs

upon in vivo metabolism, produce the active metabolite alpha-dihydrotetrabenazine (DTBZ)

411
Q

How does DTBZ differ from reserpine, meaning it is safer

Which monoamine is most affected

A

DTBZ REVERSIBLY inhibits VMAT2 unlike reserpine

DA

412
Q

How can you alter release of catecholamines

A

1) direct blockade of catecholaminergic neurons
2) triggering release in absence of any depolarisation (indirectly-acting and mixed-acting sympathomimetic drugs)
3) through stimulating pre-synaptic receptors.
4) alter storage

413
Q

Give an agent that directly blocks adrenergic neurons by acting on vesicles in the presynaptic terminal

A

Guanethidine

414
Q

How does guanethidine enter noradrenergic neurons

A

accumulates selectively into noradrenergic nerves by Uptake 1/NET

415
Q

Is guanethidine selective?

A

yes - complex, selective action on noradrenergic nerves - not fully understood

416
Q

How does guanethidine action vary based on dosage

A

At low doses, guanethidine blocks impulse conduction in these nerves, like the local anesthetics. It also accumulates into the synaptic vesicles via VMAT2, causing gradual, long-lasting depletion of NA (like reserpine).

At a very high dose, it irreversibly damages the nerve, effectively serving as a neurotoxin selective to noradrenergic neurons

417
Q

What happens to guanethidine at low doses

A

guanethidine blocks impulse conduction in these nerves, like the local anesthetics. It also accumulates into the synaptic vesicles via VMAT2, causing gradual, long-lasting depletion of NA (like reserpine).

418
Q

What happens to guanethidine at very high doses

A

irreversibly damages the nerve, effectively serving as a neurotoxin selective to noradrenergic neurons

419
Q

What was guanethidine used treat

A

uncontrolled hypertension

no longer recommended in the UK

420
Q

What is tyramine (2)

A

an indirectly acting sympathomimetic

a dietary amine

421
Q

Name an Indirectly acting sympathomimetic (NOT tyramine)

How does it act generally

A

dexamphetamine

capable of releasing stored transmitter from noradrenergic nerve endings by a Ca2+-independent process.

422
Q

Are tyramine and dexamphetamine AR agonists ?

A

NO - they are substrates for monoamide transporters

423
Q

How are tyramine and dexamphetamine taken up into nerves

What happens to them inside the nerve

How is this helpful?

A

are avidly taken up into noradrenergic nerve endings by Uptake1/NET

loaded into vesicles by VMAT2, displacing NA

NA is instead expelled into synaptic cleft by reverse NET action

424
Q

When can a tyramine rich diet become a serious problem

A

Tyramine-riched food intake in presence of MAO inhibition can lead to severe hypertensive crisis

425
Q

What kind of drugs are MDMA and ecstasy

A

Indirectly acting sympathomimetics

they are CNS stimulants, often abused for recreational purposes

426
Q

Name a mixed acting sympathomimetic amine

How do they work generally

A

ephedrine

indirectly release NE like tyramine and dexamphetamine but they also can directly activate adrenoreceptors (ARs)

427
Q

How is ephedrine used usually

A

as a nasal decongestant as it results in NA-mediated vasoconstriction in some blood vessels to the nose

428
Q

What does ephedrine act on

A

some direct action on bronchial β2-ARs and thus can help in relaxing the airways.

Also used to treat urinary incontinence in female dogs after the surgical removal of their reproductive organs.

429
Q

What effect do tyramine, ephedrine, amphetamine, and related drugs cause?

How does this differ from reserpine action

A

a relatively rapid and brief liberation of the transmitter, producing a sympathomimetic effect indirectly

reserpine, by irreversibly blocking the loading of monoamines into synaptic vesicles, produces a slow, prolonged depletion of NA (and other monoamines) from the presynaptic nerves.

430
Q

Through which presynaptic receptor is NA release most prominently regulated

A

α2-adrenoceptors (coupled to Gi/o) which, when stimulated, decrease the amount of NA released

431
Q

Is it only presynaptic α2-adrenoceptors that modulate NA release

A

no

there can be pre-synaptically located β2-ARs (coupled to Gs) which upon stimulation, enhance NA release

432
Q

How does cAMP affect NA release

A

increased cAMP production increases Ca2+ channel activating phosphorylation by PKA - more NA released

433
Q

Other than decreasing AC activity via the Gi/o, how does activating presynaptic alpha2-ARs on noradrenergic nerves decrease NA release

A

beta-gamma subunit, also results in the opening of K+ channels (GIRK)s which causes hyperpolarisation of the nerve terminal membrane and reduces its excitability

434
Q

Are the presynaptic the predominant α2-ARs and the occasional β2-ARs autoreceptors?

A

yes usually as released NA interacts with them

However, In some cases, there can be pre-synaptic heteroreceptors (e.g. muscarinic M2 receptors and δ-opioid receptors) which upon activation by cognate agonists, decrease NA release.

435
Q

How are catecholamines inactivated?

What are the 2 processes involved?

Which process is more important pharmacologically

A

removal from cleft

one into the presynaptic neuron (Uptake 1) and the other into postjunctional effector cells (Uptake 2).

Uptake 1: inhibiting this process enhances and prolongs the actions of released NA whereas inhibiting Uptake 2 does not affect the response.

436
Q

Describe Uptake 1 in noradrenergic synpases

A

This is a high affinity, low capacity system mediated by a transporter called NET (norepinephrine transport protein).

437
Q

What is NET

A

a member of the solute carrier (SLC) family of transporters. It is Na+-dependent, and blocked by many drugs including cocaine

438
Q

What kind of drugs block NET

A

cocaine

tricyclic antidepressants eg imipramine

439
Q

Name a tricyclic antidepresant

A

imipramine

440
Q

How much of the NA released from SNS neurons is recaptured by NET

A

> 75%

441
Q

VMAT2 has a much higher affinity for NA than does MAO. What does this mean for NA storage

A

most of the recaptured NE is resequestered into storage vesicles.

442
Q

What is in charge of transmission from noradrenergic neurons in the periphery

A

Uptake 1

443
Q

What is 6-OHDA

A

6-Hydroxydopamine (AKA oxidopamine)

a synthetic neurotoxic organic compound that is experimentally used (injected into brain areas) to selectivity destroy dopaminergic (and noradrenergic) neurons in the brain.

444
Q

How do you induce Parkinson’s disease in animals

A

inject oxidopamine which induces nigrostriatal damage

445
Q

How selective is 6-OHDA

Where does this come from

A

selective for catecholaminergic

its selective uptake into these neurons via NET

446
Q

What happens to oxidopamine once it has been taken up into the noradrenergic neuron via NET

A

it is readily oxidized into reactive metabolites including 6-OHDA quinone and hydrogen peroxide that damage to the neurones

447
Q

Describe Uptake 2 at noradrenergic synapses

A

lower affinity, higher capacity system of uptake of NA that hasnt been collected by Uptake 1

448
Q

What transporter mediates Uptake 2

A

ENT: extraneuronal amine transporter

449
Q

What transporter family does ENT belong to

A

to a largely and widely distributed family of Organic Cation Transporters (OCTs).

450
Q

Which of Uptake 1 and 2 is Na+ dependent

What does this mean

A

Uptake 1

ENT in Uptake 2 is not Na+ dependent (unlike NET)

they have different pharmacological profiles

451
Q

What inhibits ENT

A

the irreversible α-AR blocking agent - phenoxybenzamine (which will also block NET at much higher concentrations).

normetanephrine, a metabolite of NA, corticosteroids (such as corticosterone and hydrocortisone)

452
Q

Name a irreversible α-AR blocking agent of ENT

What is its effect on NET

A

phenoxybenzamine

will also block NET at much higher concentrations

453
Q

Where does most metabolism of catecholamines take place?

Why?

A

in the same cells where the amines are synthesized and stored

There is ongoing passive leakage of catecholamines from vesicular storage granules of sympathetic neurons and adrenal medullary chromaffin cells

454
Q

What happens to most of the catecholamines that leak into the neuron cytoplasm?

What happens to the rest

A

VMAT2 effectively sequesters 90% of the amines leaking back into storage vesicles

10% escapes sequestration and is metabolized.

455
Q

Which 2 enzymes are responsible for catecholamine metabolism

A

monoamine oxidase (MAO) and catechol O-methyltransferase (COMT)

456
Q

How should MAO and COMT work in theory vs practice

A

Although these enzymes in principle can act on the catecholamines in any order and the final metabolites should be the same but, in practice, two different metabolites (VMA and MOPEG) are found in the urine

VMA= from periphery
MOPEG= from CNS
457
Q

What do the following stand for:
VMA
COPEG

A

vanillylmandelic acid

3-methoxy-4-hydroxyphenylethyleneglycol

458
Q

How does MAO work

A

removes the amine group from the catecholamines and converts them into corresponding aldehydes, which, in the periphery, are rapidly metabolised into the corresponding carboxylic acids.

459
Q

Where does MAO action occur

A

in the outer membrane of mitochondria and it is abundant in noradrenergic nerve terminals but is also present in liver, intestinal epithelium and other tissue

460
Q

What is the main role of MAO in

a) nerve terminals
b) liver

A

a) degrade NA and DA leaking from vesicles

b) inactivates circulating monoamines such as tyramine

461
Q

What are the 2 isoforms of MAO

How do they differ

A

MAO-A and -B

MAO-A degrades 5-HT, NA and DA, while MAO-B degrades DA more rapidly than the other monoamines.

462
Q

How does pharmacological treatment aimed at MAO differ between isoforms

A

Non-selective or selective MAO-A inhibitors are used in depression and anxiety disorders whilst selective MAO-B inhibitors is used in the treatment of Parkinson’s disease.

463
Q

Why might there be an increase in blood pressure if you are eating lots of cheese and wine while on MAO inhibitors

What is this known as

A

Cheese and red wine contain tyramine, which is usually metabolised by MAO in liver/gut before reaching circulation

a large amount of tyramine-rich food is ingested, then significantly higher amount of tyramine reaches the blood and then can trigger release of NA. The latter can be sufficient to cause widespread vasoconstriction and a fatal hypertension

The cheese effect

464
Q

What other foods can the ‘cheese effect’ result from

A

tyramine rich foods: red wines, pickled herring, yeast extracts, soya beans

465
Q

Where is COMT found

A

in cytosol, primarily liver and chromaffin cells (adrenal medulla)

ABSENT FROM NORADRENERGIC NERVE CELLS

466
Q

is COMT free in the cytosol of chromaffin cells

A

no - it is membrane
bound.
This is also true of COMT in the liver cells

467
Q

what does COMT do chemically ?

A

methylates one of the aromatic hydroxyls (at position 3 of the aromatic ring) in the catecholamines (or their deaminated metabolites produced by MAO).

468
Q

What is the main end product of catecholamine metabolism by MAO then COMT

How is it excreted

How is this fact used clinically

A

VMA

via urine

in phaeochromocytoma VMA excretion is markedly increased

469
Q

What is used as an inhibitor of COMT

What is this used for

A

Entacapone

used in Parkinson’s disease in combination with L-DOPA and Carbidopa

470
Q

Why is entacapone used with L-DOPA to treat Parkinson’s disease

A

. It reduces metabolism of L-DOPA in the periphery so that more L-DOPA could reach the CNS where it is needed to produce DA for the Parkinson’s patient and less side effects occur from activation of dopamine receptors present in various peripheral tissues.

471
Q

How does ISO affect the different ARs differently?

A

it barely activates α adrenoceptor subtypes but it possesses significantly higher potency at β adrenoceptor subtypes compared to Adr and NA.

472
Q

Give 9 major effects of stimulation of α1-ARs

A
  • Dilatation of pupils (mydriasis)
  • Constriction of blood vessels supplied to vicera, brain and skin
  • Salivary secretion
  • Decreased gastrointestinal motility and tone (due to relaxation of smooth muscles)
  • Localized (e.g. in palms of hands) secretion of sweat
  • Piloerection (goose bumps)
  • Contraction of trigone and sphincter muscles of urinary bladder
  • Ejaculation of sperms
  • Increased glycogenolysis and gluconeogenesis
473
Q

Give 4 effects of α2 AR stimulation

A
  • Inhibition of neurotransmitter release (mentioned earlier)
  • Platelet aggregation
  • vasoconstriction
  • Inhibition of insulin release from pancreatic beta cell
474
Q

Give 2 results of β1 AR stimulation

A
  • Increased force of myocardial contraction

* Increased heart rate

475
Q

Give 4 results of β2 AR stimulation

A
  • Relaxation of tracheal and bronchial smooth muscle
  • Relaxation of smooth muscle of blood vessels supplied to heart and skeletal muscle
  • Relaxation of uterine smooth muscle (for non-pregnants)
  • Hepatic glycogenolysis
476
Q

Give 2 actions of β3 AR stimulation

A
  • lipolysis
  • thermogenesis

This is the least studied thus far

477
Q

What are the key effects of NA

Via which receptor?

A

Through α1 AR stimulation, Increases TPR due to vasoconstriction in most vascular beds (including kidney)

Both diastolic and systolic pressures increase enough to trigger reflex bradycardia which decreases pulse rate

478
Q

Which receptors does adrenaline activate and what is this attributed to

A

α and β -ARs

The present of a missile group at the amino N atom

479
Q

What are the 7 broad types of reaction caused by catecholamines or sympathomimetics

A
  1. A peripheral excitatory (= contractile) action on certain types of smooth muscle, such as those in blood vessels supplying skin, kidney, and mucous membranes; and on gland cells, such as those in salivary and sweat glands.
  2. A peripheral inhibitory (= relaxing) action on certain other types of smooth muscle, such as those in the wall of the gut, in the bronchial tree, and in blood vessels supplying skeletal muscle (we need to breathe more, our leg muscles need to have more blood supply to have energy for ‘fight or flight’).
  3. A cardiac excitatory action that increases heart rate and force of contraction. (remember what happens if we are exposed to stress/danger or we simply do some exercise)
  4. Metabolic actions that include enhanced glycogen breakdown in liver and muscle and liberation of free fatty acids from adipose tissue (remember the benefits of exercise which corresponds to increased sympathetic activity, we also need more energy for ‘fight or flight’).
  5. Endocrine actions, such as modulation (increasing or decreasing) of the secretion of insulin, renin, and pituitary hormones.
  6. Actions in the central nervous system (CNS), such as respiratory stimulation, an increase in wakefulness and psychomotor activity, and a reduction in appetite.
  7. Prejunctional actions that either inhibit or facilitate the release of neurotransmitters, the inhibitory action being physiologically more important
480
Q

How does NA predominantly act (ie which receptors)

Why is this

A

NA predominantly acts via apha-ARs

and this is attributed to the presence of a methyl (-CH3) group at the amino N atom.

481
Q

What is the effect of Adr when it activates β1-ARs

A

Positive inotropic and chronotropic affects (thus increase pulse rate)

482
Q

What is the effect of Adr stimulating β2 ARs

A

Compensates for constriction of arterioles in skin, mucus membranes, and viscera by Faisel dilation of skeletal muscle vascular bed which also reduces TPR

483
Q

What are the α1 mediated effects of Adr

A

Construction of arterioles in the skin, mucus membranes, and viscera

484
Q

What is the net effect of adrenaline stimulating ARs

A

Increase in systolic pressure coupled with a slight decrease in diastolic pressure

485
Q

True or false

Isoprenaline only manifests α1 AR mediated effects

A

False
Only manifests β-AR effects

This selectivity towards β adrenoreceptors stems from
A bulkier substituent (isopropyl) at the amino N atom

486
Q

What are the effects of isoprenaline? Describe the effects through the receptors which are stimulated

What is the net effect

A

β1 AR: Intense stimulation of the heart, increasing rate, contractility, and cardiac output

β2: Delete arterioles of the skeletal muscle, resulting in marked decrease in TPR

Because of its cardiac stimulatory action, may increase systolic pressure slightly, but this is outweighed by reduction in TPR with a net reduction in mean arterial and diastolic blood pressures

487
Q

What are adrenoceptor agonists also known ask

A

Directly acting sympathomimetic agents

488
Q

What do non-selective AR agonists act on

A

Both α and β ARs

489
Q

Give a non-selective adrenoreceptor agonist

When is this used

A

Adrenaline

Acute cardiac failure, acute severe asthma or anaphylactic shock. In all these cases it can be life-saving
Carried in an Epipen

Adr is also used by anaesthetists

490
Q

What is status astmaticus

A

Acute severe asthma

491
Q

How is adrenaline given in cases of anaphylactic shock

What does the Adr do (2)

A

Intramuscularly,

To counteract the systematic vasodilation and reduction in test you perfusion that is largely caused by massive histamine release

Also acts on bronchial smooth-muscle to relieve bronchospasm

492
Q

How do you anaesthetists use adrenaline

A

Local anaesthetic preparations often contain low concentrations of adrenaline which greatly increases the duration of local anaesthesia by producing vasoconstriction at the sight of injection

Allows local anaesthetic to persist at the injection site before being absorbed into systemic circulation

Keeping anaesthesia local avoids systemic effects

493
Q

What do selective α-AR agonists do

A

Selective for α ARs over β

But poorly discriminate between α-ARs

494
Q

Name a selective α AR agonist

How is it administered

What is it used for

A

Xylometazoline

Topically

To relieve nasal congestion

495
Q

How does xylometazoline relieve nasal congestion

A

Causes vasodilation in nasal mucosa – dilated blood vessels in nasal mucosa is an underlying feature of stuffy nose

496
Q

Name a selective α1 agonist

What is it used for (3)

A

Phenylephrine (oxymetazoline is also one)

To treat nasal congestion, to produce mydriasis, and treat acute hypotension (eg in septic shock)

497
Q

Name 2 α2 selective AR agonists

What are they used for

A

Clonidine and α-methylNA (from α-methyldopa)

Anti-hypertensives

498
Q

How do clonidine and α-methylNA reduce BP

A

Partly by inhibiting NA release from peripheral nerve endings

Also on CNS neurons that reduces SNS discharge to periphery

499
Q

What receptor, other than α2-AR, does clonidine bind to

What is the consequence

A

I1 receptors

Unknown

500
Q

What is an example of an α2-AR agonist used in veterinary medicine

How is it used

A

Xylazine

As a sedative with analgesic and muscle relaxant properties

501
Q

What is a non selective β AR agonist

What is it used for

What was the issue

What has replaced it

A

ISO

In asthma to relax bronchi through actions on β2- adrenoreceptors but

But increased heart rate from β1- AR stimulation was a major problem

β2-AR selective agonists

502
Q

Name a β1 AR selective agonist

What is it used for

What is the issue

A

Dobutamine

In cases of cardiogenic shock to increase cardiac output

All β1-AR agonists can cause cardiac arrhythmias

503
Q

Name 2 short acting β2 AR selective agonists (SABA)

What are they used for

A

Salbutamol (need to know)
Terbutaline

Mainly for bronchodilator effects in asthma

504
Q

What is salbutamol

How is taken usually

How long does it take for maximum effect

How long does it last

What else can it be used for

A

The blue puff asthma inhaler

Inhaled as needed

Within 30 mins

3-5 hours

Prophylactically against allergen and exercise induced bronchospasm

505
Q

Name 2 long acting β2 agonists
Which do you NEED to know

How long counts as long lasting

How are they used

A

Salmeterol (need to know)
Formoterol

8-12 hours

Used prophylactically in chronic asthma

506
Q

What is clenbuterol

A

A horse LABA

507
Q

What kind of drug that acts on ARs is used to treat COPD

Eg

A

LABAs

Especially very long lasting ones like indacaterol

508
Q

What is mirabegron

A

A β3- AR agonist that relaxes detrusor smooth muscle to increase bladder capacity

509
Q

Who would take mirabegron

A

Patients with an overactive bladder

510
Q

Give 2 other names for AR antagonists

How are they classified

A

Adrenergic blockers or sympatholytics

Based on relative affinity to α or β ARs in the SNS

511
Q

Name 2 α-AR antagonists that do not discriminate between α subtype (which do you need to know✅)

What did they treat but are now obsolete for? Why!

A

Phentolamine
Phenoxybenzamine ✅

Anti hypertensives (largely replaced by α1 antagonists) - as they may reduce blood pressure so much that it triggers reflex tachycardia

512
Q

What does phentoxybenzamine do chemically

What is its specific clinical use therefore

A

Covalently binds to alpha-AR, causing long lasting inhibition

Preparing patients with phaeochromocytoma for surgery since surgical manipulation tends to cause massive catecholamine release

513
Q

What is prazosin

What does it do

So what is it used for

A

Selective and competitive α1-AR antagonist

decreases TPR and BP by relaxing arterial and venous smooth muscle

as an anti-hypertensive without causing the same degree of reflex tachycardia observed with the non-selective α-AR antagonists

514
Q

What are the negative side effects of α1-AR antagonists

Give an explanation

A

postural hypotension and incontinence, the latter occurring because α1-ARs mediate contraction of the smooth muscle of the bladder, which is now inhibited

515
Q

Name 2 α1-AR antagonists (need to know both)

A

prazosin

tamsulosin

516
Q

How are α1-AR antagonists used to treat some prostate pathologies

A

cause relaxation of the prostate capsule and inhibit prostate hypertrophy, which has lead to the development of drugs like tamsulosin. These are selective α1A-AR antagonists that allow better bladder emptying and thus reducing urinary retention associated with benign prostatic hypertrophy

517
Q

What is tamsulosin

A

a selective α1A-AR antagonists

518
Q

What is the benefit of selective α1A-AR antagonists

A

Since α1B-ARs largely mediate vasoconstriction, these α1A-AR antagonists produce much less postural hypotension which is a common problem with non-selective and α1-selective AR antagonists.

519
Q

What is Yohimbine

What is it useful for

A

a naturally occurring alkaloid, is a selective α2-AR selective antagonist

no clinical use for the humans (but is in animals), being only a useful experimental tool to determine α-AR subtype response in a given tissue

520
Q

Name a a naturally occurring alkaloid, is a selective α2-AR selective antagonist

A

yohimbine

521
Q

True or false

All of the clinically available β-blockers are competitive antagonists for the beta-AR

A

true

522
Q

What do cardioselective beta blockers primarily block

A

β1 -ARs.

523
Q

What is the clinical use of β2 -AR blockers

A

there arent any

524
Q

Do beta blockers cause postural hypotension?

What does this mean?

A

no
because the α ARs remain unaffected.

Therefore, normal sympathetic control of the vasculature is maintained

525
Q

What do all beta blockers result in

What is the effect on blood pressure

A

negative inotropic and chronotropic effects (block β1-ARs)

decrease blood pressure in hypertensive patients, but have no effect in normotensive individuals

526
Q

What contributes to the antihypertensive effects of beta blockers

A

decreases in peripheral vascular resistance and cardiac output both contribute to the antihypertensive effect of these drugs observed in patients with high blood pressure.

527
Q

Name 7 general conditions that beta blockers can be used in

A

primarily indicated in hypertension and angina but can be useful in cardiac arrhythmias, myocardial infarction, heart failure, hyperthyroidism, and glaucoma

528
Q

How to recognise a beta blocker name

exceptions?

A

ends in -olol

except for labetalol and carvedilol

529
Q

What is the prototypical β blocker

Describe its selectivity

What is the effect

A

Propranolol

Non selective among β ARs.

Reduces hypertension

530
Q

What are the mechanisms of propanolol that decrease hypertension (4)

A

Decreased cardiac output

Inhibition of renin release

Decrease TPR with long-term use

Decrease SNS outflow from CNS

531
Q

What do the undesirable side-effects of propanolol and other non-selective β- blockers stem from

Give examples

A

Blockade of β2

β2 blockade in COPD and asthma patients can exacerbate their condition and lead to a serious crisis

β2 blockage also causes loss of vasodilation to cutaneous blood vessels leading to coldness in extremities

532
Q

What are 2nd generation β blockers

Eg? (You need to know this drug)

A

β1 selective antagonists
AKA cardioselective β blockers

Atenolol

533
Q

What is the benefit of 3rd generation β blockers

Eg

A

Polypharmacological benefits

Nebivolol - selectively blocks β1 But also offers additional benefit as vasodilator and cardioprotective agent

534
Q

How is Nebivolol cardioprotective

A

Through promoting NO release from endothelium and in myocardium

535
Q

What is carvedilol

A

A mixed α1/β-AR antagonist with anti-oxidant and anti - inflammatory properties

536
Q

What are NANC transmitters

Eg

A

Non-adrenergic, non-cholinergic transmitters

Eg purines: ATP+ derivatives
Neuropeptides
Volatile substances: NO

537
Q

What are small molecule NANC NTs stored in

What about neuropeptides

A

Clear core vesicles

LDCVs

538
Q

Where are neuropeptides synthesised and stored

A

Synthesised, processed and packaged into large dense core vesicles in the cell body and then move down along the axon towards the nerve terminal via axonal transport

539
Q

How does storage of ATP in adrenergic nerves compare to cholinergic nerves

Release?

A

ATP and ACh coexist in cholinergic vesicles whilst ATP, NPY, and catecholamines are found within storage granules in adrenergic nerves and the adrenal medulla

similarly, ATP is released along with NTs

540
Q

Which 2 peptides are mostly implicated in ANS transmission?

Name 9 other notable neuropeptides or groups of peptides

What do all these work on

A

Vasoactive intestinal peptide (VIP) and neuropeptide Y (NPY)

the enkephalins, 
substance P and other tachykinins, 
somatostatin, 
gonadotropin-releasing hormone (GnRH), 
cholecystokinin (CCK), 
calcitonin gene–related peptide (CGRP), 
galanin 
chromogranins

their cognate GPCR

541
Q

How are NANC transmitters often released in the periphery

eg?

A

concurrently with the classical neurotransmitters as the nerves are depolarised

eg: ATP is released with ACh and NA in the urinary bladder and vas deferens, respectively.

542
Q

what is suramin

A

ATP antagonist (blocks P2X and some P2Y subtypes)

543
Q

If you have a typical biphasic or twin-peaked contractions of the guinea pig vas deferens measured in response to electrical stimulation of sympathetic nerves innervating the tissue, how will the peaks be affected by suramin and prazosin

A

suramin: ATP antagonist
prazosin: α1-adrenoceptor antagonist

suramin alone abolishes early peak
prazosin alone abolishes late peak
response is completely eliminated with both S+P

544
Q

How do neurons that release many NTs, do so?

How do they interact with the postsynaptic membrane

A

Many other neurons contain and release more than one transmitter, sometimes from the same vesicles and sometimes from different ones

each NT has its own receptor

545
Q

Which NTs are commonly released alongside NA in the SNS

A

ATP and NPY

546
Q

What does co-transmission allow

Elaborate

A

greater complexity of signalling

NT time course may vary
Often NTs are released independently of each other

547
Q

What does the different time course of NTs in co-transmission allow

A

allow different types of responses to be elicited in the postsynaptic cell. Transmitters with a prolonged action, because of slow removal from the synaptic cleft, can also act on more than one target group of cells

548
Q

Give example of NTs being released independently of each other from same presynaptic neuron

A

e.g. higher concentrations of Ca2+ are required to elicit release of peptide transmitters (discussed early in this handout), so they are preferentially released at high rates of neural stimulation (i.e depolarisation).

549
Q

What does a substance have to meet to be considered a neurotransmitter

A

Dale’s criteria

550
Q

How is the action of ATP and its derivatives mediated

A

through purinergic receptors or purinoceptors:
Adenosine receptors
Ionotropic P2X receptors
Metabotropic P2Y receptors

551
Q

What are the 3 types of purinergic receptors?

Elaborate briefly on each

A

a) Adenosine receptors – A1, A2A, A2B and A3 (all are GPCRs)
b) Ionotropic P2X1-7 receptors – all are homo/heterotrimeric ATP-gated cation channels
c) Metabotropic P2Y1,2,4,6,11-14 receptors – all are GPCRs whilst few still remain as ‘orphan’ i.e. the natural agonists are yet to be known.

552
Q

What is an orphan receptor

A

one where the natural agonists are yet to be known

553
Q

How is ATP stored in nerve terminals (2) and how may it be released (3)

A

may be present in the cytosol or stored into vesicles by the vesicular nucleotide transporter (VNUT)

can be released by exocytosis in Ca2+-dependent way or through large membrane channels called the pannexins or via the nucleotide transporters (NtT).

554
Q

How does ATP enter vesicles

A

VNUT: vesicular nucleotide transporter

555
Q

When may cytoplasmic ATP cause neurotoxicity

A

released from dying or necrotic cells including degenerating neurons and excessive release in this way could lead to further cellular damage/neurotoxicity

556
Q

What happens when ATP is released into the extracellular space

A

broken down into ADP, AMP and adenosine by action of ecto-nucleotidases

557
Q

How is ATP converted to ADP etc in the extracellular space

What does this lead to

A

by action of ecto-nucleotidases

termination of transmission at the P2X and P2Y receptors

558
Q

Conversion of ATP to its derivatives by ecto-nucleotidases ceases transmission at P2Y and P2X receptors. Does this mean transmission at the synapse has ceased

A

no adenosine can still act ?

  • maybe ask Ritter about this not sure
559
Q

Where is adenosine present

A

in the cytosol of all cells and is taken up and released via a specific membrane-bound nucleoside transporter(s) (NsT).

560
Q

How is adenosine taken up and released from cells

A

via a specific membrane-bound nucleoside transporter(s) (NsT).

561
Q

How can you get inosine?

A

hydrolyze adenosine by adenosine deaminase

562
Q

What are P2X receptors

A

non- selective cation channels (i.e. equally permeable to Na+ and K+ and with significant Ca2+ permeability.

563
Q

What is the permeability of P2X channels

A

equally permeable to Na+ and K+ and with significant Ca2+ permeability.

564
Q

when do P2X channels open

A

open within milliseconds of the binding of extracellular ATP

565
Q

How many P2X receptor isoforms are there in mammals

A

7

assemble as homo or heterotrimers to form a functional channel

566
Q

Where are P2X receptors found

A

have a widespread tissue distribution, being expressed on both central and peripheral neurons, where they are involved in synaptic and/or neuromuscular transmission.

567
Q

What are the dominant P2X receptor types in neurons

A

2, 4, 6

568
Q

What is the dominant P2X subtypes in smooth muscle

A

P2X1

569
Q

What is the importance of P2X receptors in pathology

A

Changes in the expression of P2X receptors have been characterized in many pathological conditions of the cardiovascular, gastrointestinal, respiratory, and urinogenital systems and in the brain and special senses

570
Q

In the vas deferens and blood vessels, which NT produces the fast and slow response from SNS innervation

A

ATP produces fast responses mediated by P2X1 receptors, followed by a slower component mediated by G protein-coupled alpha-ARs.

571
Q

Give an experiment demonstrating the importance of P2X receptors

A

Male mice lacking the P2X1 receptor gene have a drastically reduced fertility because of much reduced sperm count in the ejaculate

572
Q

What receptor could be targeted for a male contraceptive

A

P2X1

573
Q

How is adenosine an atypical NT

A
  • it is neither stored in vesicles nor released by a Ca2+-dependent process
574
Q

Where is adenosine found

A

found in the cytosol as well as in the extracellular fluid throughout the body, including the central and peripheral nervous system`

575
Q

How is adenosine produced extracellularly

How is it then taken up

A

adenosine is produced following ectonucleotidase-catalyzed hydrolysis of ATP and then taken up by cells through a nucleoside transporter (NsT).

576
Q

What does dipyridamole do

A

blocks NsT to increase [adenosine]e

577
Q

What is methotrexate

What does it do and what is the mechanism

A

an anticancer and immunosuppressant drug, also appears to increase the extracellular concentration of adenosine, through mechanism(s) not very clear to date

578
Q

What does adenosine boosting do

A

believed to contribute to its immunosuppressive action significantly.

579
Q

What kind of range are [adenosine]i and e kept within

A

submicromolar

580
Q

What are the 2 ways adenosine can be metabolised

A

to inosine (by adenosine deaminase) or to adenine nucleotides (by adenosine kinase)

581
Q

Why can [adenosine]e increase under stressful conditions

A

large amount of ATP can be released from the injured cells (e.g. endothelial cells, neutrophils, cardiomyocytes, smooth muscle cells, glial cells etc.) via large pore-containing channels such as pannexins or connexins (or directly after necrotic cell death) and subsequently dephosphorylated to adenosine by ecto-nucleotidases

intracellular production may also increase

582
Q

What is a autacoid

A

locally acting hormone

583
Q

How are all biological actions of adenosine mediated

A

through adenosine receptors

through the activation (A2A and A2B) or inhibition (A1 and A3) of adenylyl cyclase and thus cAMP signalling

584
Q

Which adenosine receptors are activatory and which are inhibitory

Do they have any other effects (3)

A

A2a and A2b activated AC and increase [cAMP]
A1 and A3 inhibit AC

Some, if not all, of these receptors have also been reported to activate phospholipase C (and thereby trigger intracellular Ca2+ release) and mitogen-activated protein kinase (MAPK).
Also modulate Cav and K+ channel function

585
Q

How do adenosine receptors affect ion channels at the PM

A

modulate function of voltage-gated Ca2+ channels (via influencing their level of phosphorylation) and K+ channels (via βγ subunit and/or altering phosphorylation states).

586
Q

What is the effect of adenosine on the heart

What the receptor mediating

A

negative inotropic and chronotropic effects (A1 receptor)

587
Q

How can the effects of adenosine on the heart be used therapeutically

How is it administered

Is it safe? why?

A

to terminate supraventricular tachycardia

an intravenous bolus injection

due to its short duration of action (destroyed/taken up in seconds of intravenous administration) it is considered safer than other alternative drugs

588
Q

What is the effect of adenosine on smooth muscle?

Which receptors mediate this effect and what are they coupled to

A

relaxation

A2A and A2B receptors that are Gs coupled

589
Q

What is the effect to adenosine pre-synaptically

Via which receptor?

A

exerts pre-synaptic inhibitory effects on the release of excitatory transmitters in the CNS and periphery

presynaptic A1

590
Q

How does caffeine work

A

blocks inhibitory action of adenosine on presynaptic A1 receptors

591
Q

Name a methylxanthine agent

What kind of drug is it

A

caffeine

competitive antagonist of A1 receptors.

592
Q

Name an adenosine derivative drug

What kind of drug is it

what effect is it used for

A

Regadenoson (Trade names: Lexiscan or Rapiscan)

selective A2A adenosine receptor agonist

coronary vasodilatory activity

593
Q

What has the FDA approved regadenoson for?

A

for diagnostic purpose only in a type of heart scan called ‘radionuclide myocardial perfusion imaging’

594
Q

What diagnostic procedure can regadenoson be used for?

What does this procedure assess?

Why is regadenoson useful?

A

‘radionuclide myocardial perfusion imaging’

to see the blood flow in the heart muscle

it acts as a stress agent that has a similar effecton the heart as exercise

595
Q

How big is the neuropeptide family

A

> 100 members

596
Q

How big are neuropeptides

A

3-36 amino acids

597
Q

Where are neuropeptides made

A

soma

598
Q

How are neuropeptides made

A

synthesis is a multistep process that takes places in the soma and begins with the transcription of the gene that encodes one or more prepropeptides in the nucleus, splicing of the resulting primary RNA transcript to produce a mRNA

Prepropeptide mRNA is exported to cytoplasm for translation, which makes the prepropeptide. This can become several different neuropeptides

599
Q

Name a prepropeptide

A

pre-POMC

600
Q

What do prepropeptides contain characteristically

A

contain an N-terminal signal sequence (or “pre” sequence) that directs the newly synthesized protein into the lumen of the rough ER and thus into the proper, regulated secretory pathway

601
Q

What does remval of the pre sequence from a prepropeptide result in

What happens to it

A

in a propeptide (e.g. POMC)

released from the ribosome after translation is complete, transferred to the Golgi complex, and subsequently packaged within large dense core vesicles (LDCVs)

602
Q

What happens to the propeptide within the golgi and LDCVs

A

undergoes additional posttranslational processing, which involves additional cleavages and covalent modifications.

603
Q

What is the main neuropeptide at cholinergic synapses

A

VIP

604
Q

Why do neuropeptides produce a longer response

What terminates their action

A

because they are not rapidly removed from the ECF

response is terminated either by diffusion or by extracellular peptidases

605
Q

What receptors do neuropeptides activate

how can they thus be described

A

GPCRs

neuromodulators (more appropriate than neurotransmitter)

606
Q

What do neuropeptides account for in autonomic ganglia

be specific

A

released neuropeptides (for e.g. GnRH) account for the late slow EPSP.

607
Q

True or false
Receptors for neuropeptides bind their ligands with greater affinities than do the receptors for small-molecule transmitters

A

true
. This high affinity implies that neuropeptides can act at low concentrations, which is what would be expected if they often diffuse for great distances.

608
Q

What is needed to make NO normally

How does this differ in pathology

A

nitric oxide synthase (NOS) which converts L-arginine to NO and L-citrulline

cells eg vascular smooth muscle can generate NO independent of Ca2+ after stress

609
Q

What are the different isotypes of NOS

A

nNOS (NOS I)
iNOS (NOSII)
eNOS (NOS III)

610
Q

Where is nNOS expressed

A

neuronal NOS (nNOS, or NOS I) expressed in the CNS and NANC nerves

611
Q

Where is NOS II expressed

A

inducible NOS (iNOS, or NOS II) is usually absent but its expression is induced in macrophages and other cells by bacterial lipopolysaccharide and/or inflammatory cytokines, notably interferon-γ.

612
Q

Which cells produce iNOS when induced to do so

A

fibroblasts, vascular smooth muscle cells, endothelial cells, Kupffer cells, neutrophils

613
Q

Which inflammatory cytokine induces iNOS expression in some cells

A

interferon-γ.

614
Q

Where is eNOS expressed

A

expressed in platelets as well as endothelial cells

615
Q

Where does NO go after produced

A

diffuses all directions - across the presynaptic membrane and the membranes of non-neuronal cells producing it.

616
Q

What does NO activate

A

sGC (soluble guanylyl cyclase)

617
Q

How does NO activate sGC chemical

A

through interacting with the haem moiety of the enzyme

618
Q

What does the cGMP produced by NO action do

A

can act directly on effector proteins eg ion channels but more commonly through the activation of (PKG). PKG phosphorylates various proteins and leads to relaxation of smooth muscle, particularly in the blood vessels and this accounts for the term endothelium-derived relaxation factor (EDRF) that is used to denote NO

619
Q

what are nitrergic nerves

A

NO releasing

620
Q

Where are innervations with nitrergic (i.e. NO releasing) postganglionic nerves present

A

in several peripheral tissues that notably include upper airways, gastrointestinal tract (the enteric neurons) and male sexual organs

621
Q

What kind of NOS do enteric nitrergic neurons express

What role does the released NO play in the GI tract (6)

A

nNOS

NO regulates the muscle tone of the sphincter in the lower esophagus, pylorus, sphincter of Oddi, and anus.

regulates the accommodation reflex of the fundus and the peristaltic reflex of the intestine

622
Q

What is reduced nNOS associated with in the GI tract

eg

A

GI tract motility disorders

nNOS-/- mice had ~50% loss of inhibitory NANC transmission in small intestine and showed distended stomach and pyloric hypertrophy

623
Q

How is nNOS expression associated with diabetes (use a mouse experiment)

A

Diabetic mice often show delayed gastric emptying that has been attributed to negligible expression of nNOS protein and thus inadequacy in NO-mediated NANC transmission.

Insulin promotes nNOS protein expression and thereby restored NANC-mediated pyloric relaxation

624
Q

How is insulin associated with nNOS

A

promotes nNOS protein expression

625
Q

What happens to nitrergic neurons in achalasia

A

, there is selective loss of the nitrergic neurons supplied to oesophageal sphincter muscles, which incapacitates oesophagus to move food along.

626
Q

What is the blood pressure like in mice who lack the gene coding NOS III?

Why?

A

they are hypertensive

The L-arginine/NO pathway is tonically active in resistance vessels, reducing peripheral vascular resistance and hence systemic blood pressure

627
Q

How is Nebivolol associated with NO

A

the selective 1-AR blocker Nebivolol (more correctly speaking, its metabolite) also triggers NO production

628
Q

What class of drug use NO to vasodilate

A

nitrovasodilators

donate NO to relax blood vessels