SAQ's Flashcards

1
Q

Evaluate the relevance of mouse models for understanding 3 conditions

A
  1. NTD allowed the identification of mutations of proteins in planar cell polarity pathway - different labs found NTD in their mice when mutationswere here + allows us to get an idea of development of anencephaly:
    2 stages: failed cranial neuralation -> exencephaly -> anencephaly
  2. Prion - strain typing
  3. DS -> developmental stages of DS
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Briefly explain the variation in clinical phenotype based on the processes of normal neurulation in the embryo

A

1o neurulation defects:
• craniorachischisis, anencephaly, open spina bifida

The whole process aims at wrapping up this plate of cells into a tube which will fuse and separate – be covered
by surface layer that will eventually fuse so that we get a tube. It follows that the problem here is it leads to an
open defect because this fusion never finally occurs.

2o neurulation defects:
• closed spina bifida, often with lipoma

Half of neural tube is formed by secondary neuralation. - no closure process - problem with separation since
neural tube here forms by canalisation. The process can be interrupted by tethering.

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

Explain the pathogenesis underlying anencephaly

A

Two step process:

  • anerior neuropore fails to close - skull doesn’t form
  • exencephaly - then amniotic fluid degrades brain
  • anencephaly -> still birth
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Define anencephaly and spinal bifida; explain the role of planar cell polarity signalling in their formations.

A

If the anterior neuropore fails to close, then we anencephaly. It’s the zippering down that doesn’t finish to completion and we get open spina bifida (sack contains CSF) Planar cell polarity signalling:

  • mutations in proteins of this pathway lead to NTD
  • they all have different functions but act together for closure 1 to occur
  • > closure 1 is needed or whole body will not close i.e. craniorachisis
  • e.g. Scrb1 -> cellular localisatiton
  • e.g. Vngl2 - tail closure in mice + frogs
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Outline the evidence for hypoxia playing a role in MS

A

If we inject liposaccharide which induces hypoxia inducible factor 1

  • a pattern III MS lesion appears after 10 days
  • Lesion will always appear at base of DH
  • > vascular watershed
  • Cerebral angiograms - we know that arteries run over the surface then branch inwards
  • as fine vessels go deeper - less O2 in blood
  • FAD fluoresce in mitochondria also elude to hypoxia since FAD can only be made in the presence of O2
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What are the main pathological features of MS?

A

Demyelination - conduction block
Inflammation - conduction block
Mitochondria - lack of ATP -> Na+/K+ pump less efficient -> RMP setting
Degeneration -> loss of function

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

How does the pathophysiology of MS dictates some of the symptoms experienced?

A

Uhthoff’s - Cooling increases safety factor – kinetics of sodium channels
-> warm up proteins they work faster – sodium channels open and close faster if they get warmer – if normal is green – the duration shortens in demyelinated preparation – AP just about making it across – AP shorter – may go into conduction block

Tingling: ectopic activity -> generator potential generating AP -> more regular depolarizing potential generating each individual action potential
– inward K and inward Na develop at demyelinated state
If the brain is receiving burst of impulses
– pin prick going to interpret as a tingling sensation.

Permanent symptoms: continuing outflow of axoplasm has caused transected axons. CNS axons don’t regenerate these are permanently functionless

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

Describe the common types of TBI

A

Focal Damage: characterised by fractures, intercerebral contusions, bleeding: subarachnoid haemmorhage, extradural and subdural haematomas and intracerebral haemmorhage
- CT better to identify

Diffuse axonal injury: damage to connections between regions - MRI better to identify - clearer (SWI)

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

Describe the cognitive deficits following network disruption due to TBI

A
  • DAI leads to disruption of large-scale brain networks
  • Leads to typical pattern of cognitive deficits
  • Poor attention / working memory
  • Executive dysfunction
  • Behavioural issues
  • Social cognition difficulties
  • Psychiatric problems, criminality, relationship/job breakdown, substance abuse and risk taking behaviour
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What are the current methods used to assess TBI

A

Neuropsychological tests to assess executive dysfunction, lack of insight, central cognition problems, impulse issues, attention and memory, intelligence

Executive e.g. Stroop/ Trail making test

Insight from employer + friends may be more useful since Neurpsych tests: removed from day to day function, lacks sensitivity for high level deficits, poor detection of social cognitive deficits, mood/somatic symptoms can affect results

Paedeatric/autism tests might be useful

Validated - criminals may abuse tests

MRI can also be used to predict outcome
- Gliosis and scarring
– brain volume loss

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

What is the potential role of advanced imaging in TBI

A

Serial MRI - ? progressive atrophy
- Gliosis and scarring
– brain volume loss - longitudinal volumetric imaging
- yearly – if you see atrophy by more than 1% -> even
after a single concussion there seems to be some atrophy 6 months later

Investigation of advanced imaging (DTI, Tau-PET)
and novel CSF markers (CCL11)
PET – ligand that binds to tau in brain -> red is tau binding
-> more controls were done and it’s not very specific
– lots of people have tau in their brain

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

Briefly explain the relationship between TBI and neurodegeneration

A

if you have mTBI – you’re 4 times more likely to develop AD and a younger age
– if you are hospitalised for your TBI – 6 times more likely for psychiatiric disorders + early death
Scott et al., 2018 - found chronic microglial activation associated with degeneration but turning off micro glia with minocycline worsens degeneration
Cole et al., 2018 found progressive brain atrophy over time
Anne McKee 2009 -> abnormal tau deposition in 47 cases of athletes

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

Key features of Down syndrome clinically

A

Invariant features:

  • Cognitive impariment
  • Hypotonia - lack of muscle tone
  • AD-like pathology

Variable features:

  • Spontaneous abortion (2% of all)
  • Heart defects
  • Autoimmune disorders
  • Leukaemia
  • Dementia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Key features of Down syndrome genetically

A

French group 1959 – year that the first chromosomal disorders were
really pinned down in humans
– it was very difficult to look at chromosomes before this
– by the late 50’s it became clear that humans have 48 chromosomes
– Gautier learned how to make chromosomes from myocytes
-> chromosome 21 had 3 chromosomes in down syndrome -> trisomy 21

47 mega bases of extra DNA - Hsa21 ~230 protein coding genes +
29 miRNA’s with lots of non-coding elements
Not due to mutant gene due to a dosage problem
– genes like to be in 2 doses not 3

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

List mechanism and therapies for neurological consequences of DS

A

in vitro – if you take pluripotent stem cells and try to grow neurospheres
these also develop reduced neuronal number -> slower cell cycling
going on -> excessive GABA transmission

-> DS cognition -> 40% IQ 50-60

ACEI not worked in mice or in humans – lots of other trials in place
– anti-inflammatories – anti depressants - -> trial
– pregnant women Prozac fluoxetine – affect neurogenesis
-> DS fetuses + fluoxetine

• Other trials NMDA antagonists, anti-inflammatories
(damp down effects of microglia),
antidepressants (affecting neurogenesis).
• Excess GABA inhibition, some data from mouse on a GABAA antagonist,
large well-controlled Roche trial. Not worked.
Inverse agonist of alpha5 subunit of the GABA A receptor

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

Discuss the relation between DS and AD

A

DS is greatest known genetic risk factor for Alzheimer’s disease.

  • ~100% DS AD-like pathology by age 40
  • APP lies on Hsa21
  • APP triplication alone causes early onset AD (rare families)

Three copies APP sufficient to cause AD in DS, but likely effects from other Hsa21 genes in DS….

– that’s the gene that churns out amyloid beta – sits on chromosome 21
– there are rare families around with a copy of chromosome 21.
- 2 copies of APP and normal chromosome
– so in those individuals they cause APP – 3 normal
- Likely that there are other Ch 21 genes

17
Q

List and briefly describe the mechanisms affecting structure and function of the neuro-muscular junction in myasthenia gravis

A
  1. Compliment binding and activation at the NMJ
    - compliment cascade punches a hole in membrane damaging the
    membrane. This leads to the membrane attack complex
    -> compliments eventually generates a big channel
    -> muscle fibres are very big they’re generated by a fusion of many cells
    – muscle fibres don’t die but you do get damage and although it
    repairs itself you get inflammation leading a loss of junctional fold
    and chronically lose this space
  2. Antigenic Modulation
    - Crosslinking of AchR’s
    - internalisation
    - AchR degradation -> lysosomic degradation
  3. Functional AchR block
    - Autoab to AchR - blocks activity
18
Q

How is MG associated with thymic pathology?

A

Thymus gland is implicated in this complicated two-way relationship of myasthenia gravis. In people with myasthenia. they can have a thymic hyperplasia ~80% of young patients. Thymus cells start exposing muscle
antigens
– myoid cells express skeletal muscle antigens. MR/CT
mandatory in AChR-positive MG because of association with thymoma.

~30% of patients with thymoma have MG
- Also associated with acquired neuromyotonia and other paraneoplastic syndrome

19
Q

Describe 3 types of MG, how they are tested and treated?

A

“MuSK MG

  • Generalised -> AchRb -, MuSKab +
  • Pronounced bulbar weakness and may have tongue and facial atrophy.
  • Neck, shoulder and respiratory involvement without ocular weakness.
  • Less likely to respond to acetylcholine esterase (AChE) inhibitors, and their symptoms may actually worsen with these medications

Seronegative MG
- 50% of patients with ocular MG (majority seroconvert and generalize within 18 months)
• Some seronegative patients have anti-AChR detectable with cell-based assay
• 10-50% of persistently AChR seronegative patients are positive for MuSK ab
• ~10% positive for LRP4 ab
• Antibodies to Agrin or ColQ have been reported in some “triple-negative” patients

LRP4 MG
- Lipoprotein-related protein 4 is present on the postsynaptic membrane and is a coreceptor for agrin and is essential for for agrin-induced activation of MuSK in concert with Dok-7.
Full activation of MuSK results in activation of rapsyn, which then induces clustering of AChRs by binding them to the post-synaptic scaffold.
Antibody to this protein is present in 9.2% of double seronegative myasthenics (absence of AChR and Anti-MuSK antobodies).”

20
Q

Briefly describe two presynaptic disorders of the NMJ

A
  1. Lambert–Eaton myasthenic syndrome

If you have a low release probability – PPF -> insufficient calcium
-> stimulate several times and it gets stronger
– LEMS have a very strong association with SCLC
– lung cancer about 50% of people with limbs have SCLC
– if you diagnose LEMS they may have SCLC
-> sometimes its perinoplastic and sometimes it’s not

  1. Botulinism
  • Wound (skin-popping)
  • Ingestion (home preserves)
  • Neonatal/infantile (contaminated honey)
  • Cosmetic/iatrogenic
  • Presents with bulbar palsy and descending paralysis
  • Bioassay for diagnosis
  • Supportive treatment (~6 weeks) +/- antitoxin

Botulism is caused by Botulinum toxin – all have this property shred in common with tetanus toxin -> descending paralysis

21
Q

Normal brain function depends on a high energy supply. How does removal of this supply lead to stroke?

A

The brain uses 20% resting energy, so it is filled up with this mesh of vessels bringing blood to the tissue. Blood carries oxygen and glucose -> glucose is
transformed by respiration -> ATP -> There are a lot of processes which use
ATP – when an AP comes into the presynaptic cell, sodium concentration increases for a depolarization. Pumps are really important for maintaining
ion concentration gradients.
They’re powered by sodium which is pumped out by sodium pumps which uses ATP

22
Q

What are the causes and clinical response to loss of blood flow in stroke?

A

Fatty depositis leading to loss of blood flow downstream of Circle of Willis.
Stroke incidence correlates with the gradual rise in BP and deposition of fat in blood vessels. Most (87% of) strokes involve a clot blocking an artery. In an Ischemic stroke, if you have a block in the blood vessel, a clot forms and gradually builds up and occludes the vessel then you cut off blood supply downstream. A rarer form is hemorrhagic stroke where you actually get a bursting of a blood vessel -> you lose the energy supply to the brain – this can occur because a blood vessel pops – 10% of all strokes is due to that happening inside the brain and 3% is because it bursts just under brain surface

Chemical clot removal - enzyme tissue plasminogen activator dissolves clot
- ineffective long distance

Surgical removal with stent + wire mesh -> only large vessels

23
Q

Why does blood flow remain low after a clot is removed in stroke?

A

Pericytes form long processes along blood vessels; they’re designed to constrict blood vessels. Peppiatt and colleagues (2006) did a simple experiment to test this. They put a patch pipette near the capillary and then depolarized the pericyte. They found that the capillary constricts to nearly 0 in diameter and this effect is dependent on calcium entry. During stroke, pericytes constrict vessels. normally calcium is get low in pericyte due to ATP pumping. If you lose ATP supply when you have a stroke that will stop all the pumping and calcium will rise to activate myofilaments which will squash the blood vessel.

In ischaemia pericytes constrict capillaries and die in rigor. they constrict over 50 minutes – then they die – constricted in rigor and capillary is squashed to have a lower diameter – 90% of pericytes die in white matter -> calcium dependent excitotic death”

24
Q

What role does glutamate play in stroke?

A

NMDAR’s are the Achilles heel of strokes – in gray matter – glutamate kills neurones during stroke – dennis choi (1987). 5 minutes of glutamate at 100 microM kills 50% of neurones – during synaptic transmission glutamate goes up to 1 or 2 miliM just for a millisecond to activate AMPAR – it’s the duration of the glutamate application which is triggering the death.

Ca2+ goes into mitochondria through a calcium uniporter -> voltage is -200 for ATP production – calcium goes in -> cytochrome C – apoptotic factor = cell death.

Cation influx through glutamate-gated channels depolarizes neurons. This leads to anion influx via a transporter that acts as an anion channel followed by water influx for osmotic reasons – the cell pops
-> microglia phagocytosis”

25
Q

Which signalling pathways lead to neuronal death in stroke?

A

Neuronal activity evokes capillary dilation that is dependent on:
1. AMPA/KA receptors (presumably postsynaptic)
2. Astrocyte Ca2+ signalling
3. PLD2 and DAG lipase mediated AA production
4. PGE2 activation of EP4 receptors
all of these greatly reduce dilation so it’s a signaling pathway where glutamate act through astrocytes to work on pericytes via prostaglandins.

Dilation in arterioles is totally blocked by NMDA and NOS – glutamate activates interneuornes to release NO which is a dilator that controls arterioles. Therefore, there are 2 pathways controlling CBF. Arterioles dilate less than capillaries, and the signalling pathway is different.

26
Q

What is a seizure and what are the different types of seizures?

A

ILAE definition of seizures: A transient occurrence of signs and/or symptoms due to abnormal excessive or synchronous neuronal activity of the brain

Focal seizures: – originating within networks limited to one hemisphere. These may be discretely localised or more widely distributed. Burst then tangential spread.

Generalised seizures – originating at some point within, and rapidly engaging, bilaterally distributed networks…can include cortical and subcortical structures, but not necessarily include the entire cortex. Generalised seizures can present themselves as convulsions (maximal expression – seizure spreads through whole brain) or absence seizures (activity stops). Generalised seizures may begin in one part of the brain but they rapidly engage both hemispheres and subcortical structures

Generalised Absence Seizures: very brief – the EEG (scalp) spiking wave goes to about 3/s then it goes stops – the patient is completely unaware for that period then activity goes straight back to normal. This is a seizure that involves thalamocortical structures. These regions are typical for generating sleep -> sensory information comes in and the thalamus gates information into the cortex so the thalamus is switching off the signal to the cortex and then normal functioning returns after the signal is switched on again”

27
Q

What is the difference between seizures and epilepsy?

A

ILAE definition of seizures: A transient occurrence of signs and/or symptoms due to abnormal excessive or synchronous neuronal activity of the brain

Epilepsy - reduced threshold -> more likelty to have a seizure. Epilepsy is idiopathic, symptomatic and cryptogenic. Its occurrence lies on a spectrum between genetic and acquired

28
Q

Briefly describe 3 genetic and 3 environmental factors that may influence the development of epilepsy.

A

Genetic: Mendelian - single gene change/ polygenic/ absence seizures are genetically determined - happen in childhood

Environment: TBI, alcohol, stroke, dementia, tumour

-> second hit hypothesis: it can take years before epilepsy manifests itself following insult.

29
Q

What is migraine? Briefly outline the different phases and associated symptoms

A

Headache :

  • 4-72 hours
  • 2 of these: one sided pain, moderate/ severe pain – aggravated by movement
  • Photophobia / phonophobia/ osmophobia
 Premonitory phase: 
 • Tiredness 
 • Yawning 
 • Concentration impairment 
 • Neck Stiffness 
 • Mood Change 
 • Polyuria/Polydipsia
 • Food Cravings 
 • Cranial Autonomic Symptoms 

Pain phase: Nausea, photophobia, phonophobia

Postdrome: Tiredness, concentration, neck discomfort

30
Q

How is tiredness mediated in migraine - what are potential therapies?

A

OX1 + OX2 -> DORA -> filorexant

31
Q

How is yawning mediated in migraine- what are potential therapies?

A

DA from A11 nucleus - naratriptan - modulate descending DA pathways

32
Q

How is craving mediated in migraine- what are potential therapies?

A

NPY -> Hypothalamic nuclei - Basal ganglia - Limbic system -> antagonist. NPY dose-dependently inhibits nociceptive trigeminovascular transmission

33
Q

Are migraines genetic?

A

about 55% of patients with migraine aura have a mutation in the P/Q voltage gated calcium channel on Chr 19 (FHM-I CACNA1A) ,a smaller group have ATP1A mutation on Na +/K + ATPase chr 1q23 (FHM-II ATP1A2) and an even smaller group have SCN1A mutation on chr 2. If you take this mutation and knock it into a mouse – you can reduce the threshold for inducing CSD in a mouse.

TWIK-related SC K+ (TRESK) Wood, 2010 - dominant mutation - reduce channel activity -> familial migraine w/aura

34
Q

What are the different types of human prion disease?

A

Sporadic - Creutzfeltd-Jakob Disease

Genetic - Gertsmann-Straüssler-Scheinker syndrome, Fatal familial insomnia, CJD

Acquired - Iatrogenic (cadiver growth hormone), Kuru (ritualistic endocannibalism), vCJD (bovine spongiform encephalopathy transmission)

35
Q

What is the molecular basis of prion strains?

A

• distinct isolates that can be serially passaged in mice of identical prion protein genotype and genetic background
• distinguished by biological properties: incubation period (ip) and distinct patterns of neuropathology and PrP deposition in brain
-Multiple prion strains can be propagated in the same host expressing the same PrP

36
Q

What is the moleuclar basis of prion therapeutics?

A
  • Definition and validation of PrP C as the key therapeutic target
  • Development of antibodies to block or prevent prion replication
  • Small molecules as PrP C ligands to block replication
  • Establish experimental medicine infrastructure and protocol