Module 2: Neurological and Psychiatric Disorders of the Central Nervous System Flashcards

1
Q

What are the different categories of acute stroke?

A
  • TIA
  • Cerebral ischaemic stroke (CI) roughly 80%
  • Primary intracranial cerebral haemorrhage (ICH)
  • Sub-arachnoid haemorrhage (SAH)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Wha are current treatments for CI to improve blood flow?

A
  • tPA
  • mechanical thrombectomy
  • aspirin
  • anti-platelet drugs
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What are some examples of prophylaxis for acute stroke?

A
  • Statins
  • ACE Inhibitors
  • Anti-platelets
  • Anti-hypertensives
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

What is the penumbra?

A
  • An area where tissue viability may be sustained
  • A realistic target for treatment
  • Penumbra represents tissue at risk of infarction where perfusion is adequate to maintain cell viability but not adequate for normal neuronal function
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What is the blood flow in Cerebral Ischaemia?

A
  • Normal: >50ml/100g/min
  • Olighaemia: 22 - 50ml/100g/min
    (Hypoperfusion but likely to survive due to factors such as collateral blood vessels)
  • Ischaemic penumbra: < 22ml/100g/min
    (Misery perfusion likely to progress to infarction)
  • Rapid cell death: <10ml/100g/min
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What is the therapeutic window for stroke?

A

3 hours

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

Describe energy failure in stroke

A
  • Reduced blood flow
  • ATP reduced (20% of total O2 consumption used by the brain which is ~ 2% body weight)
  • Ion gradients, Na+ pump fails and hence membrane potential NOT maintained
  • Extracellular glutamate (GLU) elevated
  • Energy dependent GLU transporters inactivated
  • Acidosis
  • Na+ and Cli- entry accompanied by H20 (passive) leads to oedema
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Describe the mechanisms of calcium overload in stroke

A
  • Caused by NMDA receptor activated calcium entry and depolarisation
  • Leads to activation of:
    • Proteolytic enzymes (actin degradation)
    • Phospholipase A2 and Cyclo-oxygenase (free radical generation)
    • Nitric oxide synthase (NO generation)
  • Calcium causes mitochondrial swelling, reduced oxidative phosphorylation (loss of mitochondrial trans-membrane potential-proton motive force), cytochrome c loss (mitochondrial transition pore) —> APOPTOSIS
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What are the different types of nitric oxide synthase?

A
  • nNOS: retrograde messenger
  • eNOS: vasodilator
  • iNOS: immune mediator
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What does each NOS do?

A
  • nNOS: Causes toxic levels of NO free radicals- neuronal lesion
  • eNOS: improves cerebral blood flow
  • iNOS: Enhances toxic effects in ischaemia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What are some examples of exogenous antioxidants and free radical scavengers?

A
  • Superoxide dismutase
  • Catalase
  • Alpha-tocopherol
  • Glutathione peroxidase
  • Ascorbic acid
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Describe severe insult due to NMDA receptor mediated neurotoxicity

A
  • Ca2+ entry
  • Ca2+ uptake into the mitochondria
  • Free radical generation
  • Severe ATP depletion
  • Mitochondrial swelling

—> NECROSIS

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

Describe mild insult due to NMDA receptor mediated neurotoxicity

A
  • Transient depolarisation
  • ATP levels reduced
  • Ca2+ loaded mitochondria
  • Cytochrome c release from mitochondria

—> APOPTOSIS

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

What is the experimental evidence that NMDA receptors mediate tissue damage?

A

NMDA Receptors:

  • NR2A KO decreases infarct size (focal ischaemia)
  • Interruption of signalling using a 2B subunit antibody affecting PSD95 interaction reduces ischaemic damage
  • NR1 antibody given at 4h after MCAO reduces infarct size from 25% to 15% (+/- tPA) (Macrez et al)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What is the experimental evidence that AMPA receptors mediate tissue damage?

A
  • GluR2 antisense knockdown increases injury (global)- AMPA receptor more Ca2+ permeable.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

What are limitations of NMDA and AMPA antagonists?

A
  • HIGHLY effective up to ~2h after insult BUT have psychotomimetic (NMDA) and respiratory depressive properties
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

What is the ischaemic cascade?

A
  • A cascade of reactions which are self-perpetuating and no longer subject to physiological regulation (‘vicious cycle’) leading to cell death initially necrotic and later apoptotic
  • In parellel, neuroprotective mechanisms are activated and balance between the two mechanisms determines the fate of the new cell
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

What example of early response genes does glutamate activate?

A
  • Inducible transcription factors (IEGs) which activate/repress other genes
  • Enzymes such as COX-2 which underlie developmental and behavioural responses
  • Neuroprotective mechanisms e.g. HSPs which counter damaging effects
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

How does glutamate activate transcription?

A
  • NMDA Receptor Ca2+ entry —> Ca2+-calmodulin kinase IV pathway (CAMKIV) —> Phosphorylation of cAMP-response element binding protein —> CREB/CREB binding protein complex activates transcription (transcription factors and neurotrophic factors)
  • This pathway mediates an injury response that can contribute to cell survival or cell death
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

What are the penumbra/peri-infarct effects of glutamate?

A
  • Elevated extracellular K+ and glutamate depolarisation in penumbra
  • Upregulation in injury response genes (e.g. c-jun, ATF3 and HSPs)
  • Extends area of infarct
  • Sensitive to glutamate antagonists
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Is chronic treatment with COX2 selective inhibitors a viable treatment for Stroke?

A
  • No
  • COX2 selective inhibitors, whilst lacking gastric toxicity, decrease prostacyclin (vasdilator) and lack COX1 anti-thrombotic properties which potentiates cardiovascular events
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What are heat shock proteins?

A
  • Act as protein chaperones facilitating the transfer of proteins between subcellular compartments
  • Following a noxious stimulus (heat, ischaemia) HSPs are induced which target abnormal proteins for degradation
  • HSPs are also anti-apoptotic and antioxidant (HSP27)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What does Sulindac do? (Stroke)

A

It’s an NSAID which increase HSP27 and decreases infarct size

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

What is ischaemic pre-conditioning (IPC)?

A
  • IPC is a process in which brief exposure to ischaemia provides robust protection/tolerance to subsequent prolonged ischaemia (~TIA)
  • HSP involvement in IPC has been demonstrated in cardiac and cerebral ischaemia (Sun et al 2010) mediated through the NF-kB pathway(Tranter et al 2010).
  • Ischaemic preconditioning reduces infarct size in mouse
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

How does inflammation play a part in stroke?

A
  • Neutrophils enter the brain parenchyma (30 min) and later, lymphocytes and macrophages (5-7 days) (iNOS elevated).
  • Enabled by the disruption in the Blood brain barrier
  • Production of mediators of inflammation:
    • TNF alpha
    • Platelet activating factor
    • Interleukin 1 beta
    • Adhesion molecules on endothelial cell surface (ICAM-1, p and E-selectins)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

Describe the cellular inflammatory response of stroke in more detail

A
  • Neutrophils accumulate within 30 minutes on vascular endothelial cells
  • Cell adhesion molecules (Selectins, Integrins, Immunoglobulins) promote adherence leading to infiltration of cells into the brain parenchyma.
  • Neutrophils cause tissue damage by releasing O2 free radicals & proteolytic enzymes
  • Other cells entering the tissue e.g. lymphocytes promote tissue damage (24h)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

Describe the role of cytokines and chemokines in stroke

A
  • Produced by a range of activated cell types (endothelial cells, microglia, neurones, astrocytes, platelets, leukocytes, fibroblast) within the first few
    hours after ischaemia
  • IL-1 and TNF upregulate adhesion molecules promoting neutrophil migration
  • CSF levels of IL-1, IL-6 and TNF at 24h correlate
    with infarct size
  • Chemokines (e.g. CINC and MCP-1) detected in
    the brain between 6 and 24h attract neutrophils &
    infiltration
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

What are some neuroprotective examples against cytokines?

A
  • IL-1b receptor antagonists
  • TNF-alpha neutralising antibodies and antisense nucleotides
  • TGF b and IL-10 produced by lymphocytes limit
    leukocyte invasion and reduce immune responses
  • Complex protective/harmful effects are seen due
    to multiple sites of action.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

Is neutrophil infiltration correlated with infarct size?

A
  • Preliminary studies supported this concept
  • However, Phase III anti-neutrophil drugs
    failed to improve stroke outcome and antiICAM
    (n=625) increased mortality(Becker et
    al 2002). [Thought to be due to neutrophil activation by
    the mouse antibody (complement)].
  • Neutrophils may have both proinflammatory or anti-inflammatory phenotypes (Easton 2013)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

How does apoptosis occur in stroke?

A
  • Delayed cell death occurring in the penumbra
  • Triggered by free radicals, death receptor, DNA
    damage, protease action, ion imbalance
  • Release of cytochrome c from mitochondria
    activates the formation of an apoptosome complex
    (APAF1 + procaspase 9) and caspase 3 activation
    (detected at ~8h) leading to DNA fragmentation
  • Caspase 3 selective inhibitors (zDEVD.FMK) are
    effective up to 9h after reversible ischaemia.
  • Broad specificity caspase inhibitors (zVAD)/
    caspase 1 deletion protects against ischaemia.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

Describe late stage repair? (Stroke)

A
  • Growth factors are secreted by neurones, astrocytes, microglia, macrophages,vascular and peripheral cells e.g. IGF1, erythropoietin
  • Glutamate-mediated synaptic activity increases BDNF transcription and secretion
  • Neuronal sprouting occurs in an attempt to form contacts
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

What is the limitation of intravenous tPA (IVT)?

A
  • IVT can salvage penumbra if given early but the recanalization rate is low (30% within 4.5h)
  • Poor outcome is linked to the fact that the infarct is already large at the time of recanalization and hence
    the need to slow infarct growth
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

What is the limitation of endovascular thrombectomy (EVT)?

A
  • EVT increases the likelihood of penumbral salvage (60%), however, half the patients who undergo successful canalisation do not achieve functional independence
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

What are the different types of ischaemic stroke?

A

Artery:

  • Acute Ischaemic Stroke (AIS)
  • Lacunar

Venous:
- Cerebral Venous Sinus Thrombosis (CVST)

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

What are the different types of haemorrhagic stroke?

A

Artery:

  • Aneurysm
  • AVM
  • Primary Intracerebral Haemorrhage (PICH)
  • Extradural haemorrhage

Venous:

  • Subdural haemorrhage
  • Cerebral Venous Sinus Thrombosis (CVST)
  • Cavernoma
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

What are specific causes of thrombosis?

A

AADDVISE (Arterial)
OOORR (Venous)
HEPARINISE (Venous/Arterial)

Atherosclerosis
Arteriosclerosis- hypertension
Dissection
Dysplasia, fibromuscular
Vasculitis
- Autoimmune, sarcoid, infection
Injury
- Iatrogenic: radiation, catheter
Spasm: migraine
Extra: embolism, compression

Operation esp. orthopaedic, obstetric
Obesity
Overland flights (e.g. long-haul flights)
Reduced circulating volume (e.g. hypovolaemia)
Right-sided heart failure

Hereditary: factor V Leiden, prothrombin mutations
Endocrine: oestrogen (OCP, HRT), testosterone, diabetic hyperosmolar non-ketotic coma
Polycythaemia and other haemotological disorders
Autoimmune: antiphospholipid syndrome, Behcet’s syndrome
Renal: nephrotic syndrome, volume depletion
Infection: systemic- TB, chlamydia, any other cause of raised CRP, local- otitis media or mastoiditis
Neoplasm: AML, adenocarcinoma
Injury: fracture, sympathetic stress response
Smoking:
Exogenous: chemotherapy, COX-2 inhibitors

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

Underline stroke pathophysiology

A
  • Thromboembolism
  • Hypoperfusion
  • Lacunar infarction
    (Causes include aging, diabetes, hypertension)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

What are the features of lacunar infarction?

A

Acute Syndromes:

  • Pure motor
  • Pure sensory
  • Ataxia-hemiparesis
  • Dysarthria- clumsy hand
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

What are the features of small-vessel ischaemia?

A

Chronic Syndromes:

  • Executive cognitive impairment, bradyphrenia
  • Lower body parkinsonism, gait apraxia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

What are the main clinical syndromes of stroke?

A

MCA, ACA, PCA and brainstem infarction

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

What are different types of MCA stroke?

A
  • Blockage of main branch (horizontal M1 —> wedge infarction)
  • Blockage of Lenticulostriate arteries (end-arteries)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

What does an ACA stroke do?

A
  • Affects medial Brain —> contralateral leg weakness
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

What does a PCA stroke do?

A
  • Affects Occipital cortex —-> Homonymous hemianopia, Neglect
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

What does a brainstem infarction do?

A
  • E.g. PICA infarct —> Contralateral limb symptoms, Ipsilateral Cranial Nerve symptoms
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

How does a stroke lead to respiratory depression?

A
  • Stroke —> Increased ICP —> Midline shift —> Coning —> Respiratory depression
  • Tx: Hemi-craniectomy —> relieve pressure —> 50% survive
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

What are the main investigations requested in stroke?

A
  • CT Scan (Ischaemia: dark, oedema/Haemorrhage: bright, blood)
  • Further: Where is blood clot coming from? —> Carotid doppler
  • MR Angiogram
  • ECG (AF)
  • Echocardiogram (Vegetations)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

What are the causes of death in Stroke patients?

A
  • Herniation —> Respiratory depression
  • Pneumonia (inability to swallow/cough)
  • PE (bedbound/VT)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

What is MS?

A
  • Chronic inflammatory, multi-focal demyelinating condition of the CNS with an unknown cause characterised by loss of myelin and oligodendroglial and axonal pathology
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

What is the epidemiology of MS?

A
  • Affects 2.5 million people worldwide

- Female > Male (2:1)

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

What are the factors affecting MS incidence?

A
  • Latitude Effect
  • Time of exposure
  • Viral hypothesis
  • Genetic factors
  • Role of hormones
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

How does the latitude effect affect MS incidence?

A
  • Higher prevalence in Northern Countries (esp. UK vs World, Scot > Eng)
  • Role of Vit D: Decreased 25(OH)D —> Increased risk of MS (Simpson, 2010)
  • Correlation between low UWB intensity (low sun exposure) and risk of MS —> Prescribe Vit D to MS
  • However Black people are more likely to be Vit D deficient BUT decreased risk of MS
  • Norway North-South gradient risk inverted due to more time spent in outdoor activities during summer
  • Higher fish consumption and use of cod-liver oil supplement
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

How does time of exposure affect MS incidence?

A
  • Migration studies - migration age >15 retain original risk
    Age < 15 have risk of new area - Dean 1971
  • Month of birth effect: Increased risk of MS for May birth (Decreased Vit D in Winter)
  • Season variable of MS activity: ­Increased MS disease activity in spring (Decreased Vit D in winter)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

What is the MS viral hypothesis?

A
  • Hypothesised MS is triggered by a virus (e.g. EBV) - Kurztke
  • EBV sero +ve —> Increased risk of MS
    EBV sero -ve —> 0 risk
    Increased anti-EBNA IgG titre —> Increased MS
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

How do genetic factors influence MS incidence?

A
  • First degree relatives have 10-25 times greater risk of MS
  • 25-30% monozygotic twins
  • 2-3% dizygotic twins
  • 1.9% non-twin siblings
  • HLA-class ΙΙ genes exert the strongest effect, accounting for 20-60% of the genetic risk, with a predominant role played by the HLA-DRB1*15
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

How do the role of hormones influence MS incidence?

A
  • During pregnancy —> Decreased MS relapses
  • Post-partum —> Increased MS relapses

PRIMS study

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

What are relapses?

A
  • Acute neurological deficit lasting more than 24 hours followed by complete or partial recovery (= demyelinating attack)
  • Inflammation —> Demyelination (Seen on MRI T2 as demyelinating plaque = pathological Hallmark of MS)
  • Followed by spontaneous recovery
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

Describe MS progression.

A
  • Insidious onset of irreversible accumulation of neurological deficit >1yr (Retrospective Dx)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

What is RR MS?

A
  • Relapsing-remitting MS (80% of MS)
  • Acute demyelinating attacks followed by partial/complete recovery
  • Asymptomatic between relapses
  • With time, frequency of relapses decreases (Decrease of 17% in Annualised Relapse Rate every 5 years)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

What is SP MS?

A
  • Secondary progressive MS
  • Shift RR MS (inflammation) —> SP MS (degeneration)
  • 10 years from disease onset
  • Increased irreversible disability
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

What is PP MS?

A
  • Primary progressive MS (20% of MS)
  • Disease starts with progression
  • Progressive paraparesis
  • 40% have superimposed paralysis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

What is Clinically isolated syndrome?

A
  • 1st MS-like attack

- No DIT for clinically definite MS

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

What is the disease course of MS?

A
  • Age of Onset ~ 30 years (RRMS)
  • ~ 40 years (SP MS, PP MS)
  • Lasts 40 - 50 years
  • Patient unlikely to die from MS but from decreased QoL
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

What are the presenting symptoms of MS?

A
  • Relapses: Symptoms are highly variable – based on AMOUNT and LOCATION
  • Specific location of inflammation determines specific symptoms
  • E.g. optic neuritis, motor weakness, sensory disturbances
  • In later stage, mainly motor symptoms (Increased disability)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

What is the key diagnostic criteria for MS?

A
  • Exclude DDx + appropriate presentation
  • Dissemination in Time (DIT): demyelination/inflammation on at least 2 separate occasions
  • Dissemination in Space (DIS): demyelination/inflammation in at least 2 different areas of CNS
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

What are the imaging features of MS?

A
  • MRI T2: round demyelinated plaque (white) | 5-10x ­ MRI lesions > Clinical attacks (clinically silent, less important area)
  • Typical locations: Perivascular, Corpus callosum, Cerebellum, Brainstem
  • Over time, they appear (demyelination, inflammation) and disappear (remyelination)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
66
Q

What are the clinical and laboratory tests for MS?

A
  • History: DIS & DIT (H&E sufficient for majority of cases, can use MRI to assess DIS if necessary)
  • MRI: DIS & DIT (new lesion compared to old or presence of GAD-enhancing and non-enhancing lesions)
    GAD enhances NEW lesions <6 weeks and not old lesions >6 weeks
  • CSF: Oligoclonal Band in CSF only (-ve in Serum) – inflammation limited to CNS – suggests MS
  • VEP: DIT asymptomatic + delayed nerve conduction indicates previous attack
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
67
Q

What are potential DDx for MS?

A
  • ADEM (Acute disseminated encephalomyelitis)

- NMO (Neuromyelitis optica

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

What is ADEM?

A
  • Acute inflammatory demyelination of CNS
  • Prodromal infection (respiratory or intestinal)
  • Single attack
  • Very young age < 10
  • Larger inflammation lesions on MRI than MS
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

What is NMO?

A
  • Severe CNS myelination with optic neuritis and acute myelitis
  • Mainly Spinal cord involvement
  • MRI shoes Cord lesion >3 vertebrae
  • Auto-Ab to AQP4 channel
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
70
Q

Describe the variability of disease course severity.

A
  • High variability: Spectrum from Benign MS (slow deterioration) to Malignant MS (rapid deterioration)
  • Expanded Disability Status Scale (EDSS) rates disability in MS patients (EDSS 6 = walking assistance)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

What are the clinical and radiological factors affecting MS prognosis?

A

Poor prognostic factors:

  • Older age of onset
  • PP MS
  • Early relapses
  • MRI lesion load
  • Male
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
72
Q

What are the basic pathophysiological mechanisms that lead to brain injury following trauma?

A
  • Focal: Fractures | Contusions | Haemorrhage —> CT/MRI

- Diffuse: Diffuse Axonal Injury | Diffuse Vascular Injury —> “Advanced MRI”

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

What is CT?

A
  • 3D reconstruction based on differential attenuation of X-ray beams passed through an object from multiple directions
  • Tailor CT to specific degree of attenuation (alter the window)
  • Water = 0, Bone > 1000
  • Hounsfield units
  • Nobel 1979
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
74
Q

What are the advantages of CT?

A
  • Fast
  • Cheap
  • Better than MRI for bony abnormalities
  • Useful for imaging Acute bleeds and fractures
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
75
Q

What are the disadvantages of CT?

A
  • Poor resolution

- Can’t see subtle changes in brain structure

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

What is an MRI?

A
  • Large magnet aligns all protons, 2nd RF misaligns protons which relax to original position releasing energy which is detected
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
77
Q

Describe T1-weighted MRI scan

A
  • Good tissue discrimination
  • Dark CSF
  • Bright fat dark lesions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
78
Q

Describe T2-weighted MRI scan

A
  • Sensitive to water (oedema)
  • Bright CSF
  • Dark fat bright lesions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
79
Q

Describe FLAIR

A
  • Sensitive to water (oedema)
  • Dark CSF
  • Dark fat bright lesions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
80
Q

Describe echo imaging / MRI SWI (Susceptibility Weigh Imaging)

A
  • Useful for subtle injuries/microbleeds —> Any bleed will leave some haemosiderin (containing Iron) leftover
  • Microbleeds have a Parafalcine distribution (suggests axonal injury)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
81
Q

Describe Diffusion Tensor Imaging

A
  • Useful to look at specific tracts (white matter tracts)
  • Diffusion of water molecules is constrained by property of the tissue
  • In axons, normally diffuse along length
  • Diffusion Tensor Imaging measures how anisotrophic (e.g. the direction, scale 0-1) the water diffusion is
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
82
Q

What are some features of Prion diseases?

A
  • Transmissible Spongiform Encephalopathies
  • A series of diseases with a common molecular pathway
  • Spongiform = vacuoles
  • Transmissable factor
  • No DNA or RNA involved
  • Prion (PRoteinacious Infectious ONly)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

What are the human forms of Prion disease?

A
  • Creutzfeldt-Jacob Disease
  • Gerstmann-Straussler-Sheinker Syndrome
  • Fatal familial insomnia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
84
Q

What are the animal forms of Prion disease?

A
  • Scrapie
  • Bovine Spongiform Encephalopathy
  • Feline Spongiform Encephalopathy
  • Chronic Wasting Disease
  • Transmissible Mink Encephalopathy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
85
Q

What is the epidemiology of Prion disease?

A
  • 1-2 cases per million population
  • M = F
  • 10 - 15% familial
  • Age onset average 55-75
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
86
Q

What is the neuropathology of prion disease characterised by?

A
  • Spongiform change: microscopic vacuoles, found in cerebral cortex and cerebellum —> ataxia
  • Neuronal loss + Synapatic loss: general atrophy (widening sulci, shrinking gyri, enlarged ventricles)
  • Astrogliosis: Activated astrocytes (GFAP marker) react to vacuolar change and prion desposition
  • Accumulation of PrP
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
87
Q

What are some features of sporadic CJD?

A
  • Progressive dementia
  • Ataxia
  • EEG changes
  • Death within one year
  • Biopsy/Autopsy for Dx
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
88
Q

What are some features of iatrogenic CJD?

A

Can be caused by

  • GH (extracted from cadaveric pituitatries)
  • Dural transplant
  • Neurosurgery
  • Slow incubation period
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
89
Q

What are some features of GSS?

A

(Genetic version of CJD)

  • Autosomal dominant
  • Progressive dementia
  • Mild phenotype (4-5 years)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
90
Q

What are some features of fatal familial insomnia?

A
  • Autosomal dominant
  • Sleep disturbances
  • Neuropsychiatric presentation (tiredness, psychoses)
  • Late dementia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
91
Q

What are some features of a prion protein?

A
  • Normal cellular protein , PrPc
  • Expressed in neurons and glia
  • Chromosome 20
  • Membrane associated
  • Unknown function
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
92
Q

What are some genetic features of prion proteins?

A
  • Many different types of mutations
    Valine or Methionine
  • Varying level of penetrance
  • Codon 129 (VV, MM, VM)
  • VV/MM are at a higher risk than VM
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
93
Q

What is PrPp?

A
  • Abnormal protein
  • Accumulates within cells and in amyloid deposits
  • Resistant to degradation by proteinase K
  • Detectable by ICC
  • No amino acid difference betwen PrPc and PrPp
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
94
Q

How does PrPc become PrPp?

A
  • Energy unfolds PrPc from alpha-helical structure to beta-pleated sheet (=Amyloid) - Equilibrium influenced by genetic susceptibility
  • Beta-pleated sheet structure converts other host proteins into beta-pleated sheet structure (Autocatalytical conversion)
  • Irreversible propagation —> Amyloid fibrils deposited in the brain

Nat Rev Neurosci, 6, 23-34 (2005)

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

What are the mechanisms that convert PrPc into PrPp?

A

1) Post-translational modification alters conformation
2) Mutation in Prion gene predisposes to PrPp conformation

PrP knockout mice are immune to PrP infection (from other sources)

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

Describe disease strains

A
  • Same protein can lead to different strains
  • Multiple passages of infection of Scrape in mice lines
  • –> different disease strains
  • Number of polysaccharide chains, either 1 (=A) or 2 (=B), attached to Prion protein can modulate clinical phenotype
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
97
Q

Described glycosylated patterns

A
  • Type 1 = 20kDa band predominant
  • Type 2 = 19kDa band predominant
  • A = A band (monoglycosylated 25kDa) predominant
  • B = B band (19kDa band predominant)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
98
Q

What is the species barrier with regards to the emergence of new variant CJD (vCJD)?

A
  • Scott, 1989: Hamster PrPp (hPrPp) can only lead to Scrapie in Hamsters and transgenic mice expressing hPrPp
  • Species-specific infection
  • Inoculation of wild-type mice with hPrPp does not cause Scrapie
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
99
Q

What are features of vCJD?

A
  • Sporadic neuropsychiatric disorder
  • Patients <45 years old
  • Cerebellar ataxia
  • Dementia
  • Longer duration than CJD
  • Linked to BSE
  • Diagnosed at autopsy since 1990
  • All 129 MM homozygotes

Will RG et al (1996):

  • Widespread vacoulation
  • Florid plaques (PrPp deposits)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
100
Q

What are the mechanisms of spread literature?

A

1) & 2) Beekes et al., FEBS, 2007, 274; 588-605

3) Frost B & Diamond MI (2010) Nat Rev Neurosci 11 155-159

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

Describe CJD diagnosis

A
  • Genetic CJD: genetic sig/PRNP seq
  • vCJD: PrP deposits in peripheral lymphoid tissue
  • Sporadic CJD (Brain biopsy)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
102
Q

What is the treatment for vCJD?

A
  • Symptomatic relief
  • Pentosan polysulphate
    • Post-exposure prophylaxis to prevent peripheral replication and neuroinvasion
  • Ablation of FDCs
  • Beta-sheet breaker peptides
  • Vaccination
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
103
Q

What is the epidemiology of CNS Trauma?

A
  • Single largest cause of death in people under 45
  • 9 deaths from head injury per 100,000
  • Account for 25% of all trauma deaths
  • High morbidity:
    19% vegetative or severely disabled
    31% good recovery
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
104
Q

What are the different types of head trauma?

A
  • Non-missile
  • Missile
  • Focal or Diffuse
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
105
Q

What are features of non-missile head trauma?

A
  • Acceleration/deceleration
  • Rotation force (midline structures vulnerable)
  • RTA, falls, assaults
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
106
Q

What are features of non-missile traumatic brain injury?

A
  • Acceleration/deceleration
  • Rotation forces (midline structures vulnerable)
  • RTA, Falls, Assaults
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
107
Q

What is a feature of missile TBI?

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

What are features of focal TBI?

A
  • Fractures
  • Contusions
  • Haemorrhage —> CT/MRI
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
109
Q

What are features of diffuse TBI?

A
  • Diffuse Axonal Injury

- Diffuse Vascular Injury —> Advanced MRI

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

What is primary trauma damage?

A
  • Damage that has occurred, cannot be changed

- Depends on cause, type, location, age, drugs, pre-existing disease, genetics

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

What are the different types of primary trauma damage?

A
  • Scalp lactation
  • Skull fractures
  • Cerebral contusion
  • Intracranial haemorrhage
  • Diffuse axonal injury
  • All fatal non-missile head injury cases will have surface contusion and diffuse axonal injury
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
112
Q

What are features of skull fractures?

A
  • Base of skull fracture —> CSF leakage —> Otorrhea/Rhinorrhoea, Battle’s sign/Racoon’s eyes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
113
Q

What are features of a cerebral contusion?

A

High risk areas:

  • Orbitofrontal cortex
  • Temporal lobe
  • Occipital lobe
  • Inferior surface of the brain
  • Coup-contrecoup damage associated with acceleration/deceleration injury
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
114
Q

What are features of intracranial haemorrhage?

A
  • Extradural
  • Subdural/Burst lobe
  • Subarachnoid
  • Intracerebral
  • Tx: Surgical evacuation (drill 3 bore holes, remove bone flap, tie off vessel and reseal)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
115
Q

What are some features of diffuse axonal injury?

A
  • Due to shear & tensile forces on axons —> Retraction balls (marker of axonal damage)
  • Grade 1 (Parasagittal Frontal, Internal Capsule), Grade 2 (+ Corpus Callosum), Grade 3 (+ Dorsal Brainstem)
  • DAI is present in nearly all head injuries and is always present in fatal head injuries
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
116
Q

Describe primary axotomy

A
  • Tear of axolemma —> Ca2+ influx —> activate proteases —> cytoskeletal dysfunction —> disconnect
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
117
Q

Describe secondary axotomy

A
  • Rupture —> membrane sealing (imperfect stabilisation) —> highly susceptible to secondary insult (rugby)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
118
Q

What are the general mechanisms of diffuse axonal injury?

A
  • Shearing forces —> Axotomy —> Stops normal axonal transport —> Build-up of toxic proteins within the Axon
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
119
Q

How can DAI be detected?

A
  • Immunostaining for APP (undergoes axonal transporter, marker of axonal damage) or Silver staining
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
120
Q

What are different types of secondary trauma damage?

A
  • Ischaemia/Hypoxia
  • Cerebral swelling
  • Infection (if open head injury)
  • Seizure (Glutamate excitotoxicity)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
121
Q

How does cerebral swelling cause secondary damage?

A
  • Increased ICP —> Midline shift + herniation (most common secondary cause of death)
  • ICP measured using a Bolt monitor
  • MOA unknown: ?Vasodilation (to aid perfusion), ?BBB breakdown —> vasogenic oedema
  • Tx: Diuretics, Craniotomy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
122
Q

What are the different types of herniation?

A
  • Subfalcine herniation (under falx cerebri)
  • Tentorial herniation (under tentorium cerebelli)
  • Tonsillar herniation (cerebella tonsils under foramen magnum)
  • Coning (brainstem into foramen magnum)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
123
Q

What are the molecular and cellular pathways that have been implicated in TBI?

A

(Similar to ischaemic damage in Stroke Lectures)

  • Neurodegeneration: acute head injury may initiate protective response, if chronic, may cause neurodegeneration
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
124
Q

What characterises Alzheimer’s disease pathology?

A
  • A-beta plaques and Tau neurofibrillary tangles (APP is a marker of traumatic axonal injury)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
125
Q

What does the literature on CTE say about boxing?

A
  • Boxing is associated with cognitive issues and molecular changes similar to AD (but boxers much earlier onset)
  • Key features in Boxers: Tear in midline structures - Earlier cognitive changes - Tau tangles (Corsellis) - A beta plaques (Roberts)
  • 50% of Boxers with Dementia were found to have CTE (Astrocytic Tau Pathology) - the other 50% did not have Tau pathology
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
126
Q

What does the Glasgow group say about CTE?

A
  • 152 patients who died with 30 days of head injuries
  • 30% have A beta pathology - De novo AD pathology is linked to acute trauma? Is this age related (e.g. present regardless)?
  • If they survived, would they get AD pathology? or would A beta be cleared?
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
127
Q

What is the proposed mechanism of CTE?

A
  • Inflammation is thought to be initially protective and persistence (cytokine cycle - IL-1, ApoE) —> neurodegeneration
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
128
Q

What does McKee AC, 2009 say about CTE?

A
  • Newly described Tau pathology
  • Cause and effect is not known - Don’t know if clinical picture is same as pathology
  • Tau pathology in CTE is within Astrocytes (Astrocyte Tau Tangles) & found at base of sulci (where impact force concentrate)
    • AD Tau in Neurons - Astrocytic neurofibrillary Tau tangles found at bases of sulci, sub-pial, peri-vascular + cortex
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
129
Q

What does McKee AC, 2012 say?

A
  • n = 85 athletes/military personnel with repetitive mild TBI (McKee AC, 2012)
  • 80% of repetitive mild TBI had CTE pathology
  • However some had concomitant AD/PD/FTLD
  • Therefore difficult to assess cause and effect of Tau and clinical phenotype
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
130
Q

What does Tau PET ligand (11C-PBB3) do?

A
  • Visualises Tau pathology in vivo (useful for CTE and AD)

- A beta is less relevant for CTE

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

What does Goldstein, 2012 show?

A
  • Blast neurotrauma mouse model (blast head injury) shows Tau-related changes
  • But head injury in rodents if different in humans
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
132
Q

What is ARTAG?

A
  • Age-related Tau-Astrogliopathy
  • ARTAG has no clinical phenotype
  • Age-related Tau pathology in Astrocytes
  • ARTAG confounds CTE
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
133
Q

What are the future implications of CTE?

A
  • Need longitudinal studies to follow progression of retired sportsman to assess cognitive capacity and imaging changes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
134
Q

What are treatments for CTE?

A
  • Neuroprotection
  • Anti-inflammatories (but thought to initially be protective)
  • Protease inhibitors (decreased Ca2+ activated proteases)
  • Hypothermia/Hyperbaric treatment
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
135
Q

What are the main categories of MS treatment?

A
  • Education and counselling
  • Management of acute attacks
  • Prevention of disease activity (disease modifying treatments)
  • Symptomatic therapy
  • Physical therapy
  • Treatment of complications
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
136
Q

What is the management of acute MS attacks?

A
  • High-dose IV methylprednisolone

- Decide on necessity for treatment as Sx tend to be self-resolving (treat disabiling Sx e.g. double vision)

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

What is symptomatic therapy for MS?

A
  • Spasticity: stretching, physical therapy, Baclofen, Botulinum toxin
  • Paroxysmal pain: Gabapentin, Carbamazepine - no treatment for neuropathic pain
  • Chronic dysaesthetic pain: Amitroptyline
  • Fatigue: “Energy-savings”, Amantadeine
  • Depression: Antidepressants
  • Immobility (most concerning to pt): Encourage activity from start, Physiotherapy, Aids
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
138
Q

What is the aim of disease modifying treatments?

A
  • Decrease inflammation/demyelination —> Decrease axonal damage —> Prevent disability
  • Many DMTs for RRMS (hard to test if they delay progression)
  • Limited progression for Progressive MS
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
139
Q

What are different types of Injectable DMTs?

A
  • IFN-beta

- Glatiramer acetate

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

What’s the MOA of IFN-beta?

A
  • Decreased T-cell activation
    (Decreased MHC Class II, Decreased co-stimulation)
  • Decreased pro-inflammatory cytokines release
  • Decreased transmigration
    (Decreased VLA4, Decreased MMP, VCAM decoys)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
141
Q

What are the results of IFN-beta on MS?

A

ADVANCE trial

  • RRMS: 30% Decrease in ARR
    ? delay progression
  • CIS: delay 2nd episode
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
142
Q

Is IFN-beta safe?

A
  • yes
  • Injection site infection
  • Flu-like Sx
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
143
Q

What are further details about IFN-beta?

A
  • 1st line active MS

- 1st DMT available for RRMS

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

What is the MOA of Glatiramer acetate?

A
  • Decoy effect

Resembles MBP

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

What are the results of Glatiramer acetate on MS?

A
  • RRMS: 30% Decrease in ARR
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
146
Q

Is Glatiramer acetate safe?

A

Yes

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

Further details on Glatiramer acetate?

A
  • 1st line active MS

- Only DMT for emergency

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

What are the different types of oral DMT?

A
  • Fingolimod

- Dimethyl fumerate

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

What is the MOA of Fingolimod?

A
  • Prevents T cells leaving lymph nodes
    (Decreased T cell sensitivity to chemotactic cues)

(S1P1 receptor agonist —> S1P1 receptor internalisation)

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

What are the results with Fingolimod?

A

TRANSFORM trial

  • 50% decrease in ARR vs IFN-beta

FREEDOM trial

  • 50% decrease in ARR vs placebo
  • Decreased disability progression
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
151
Q

What are the side effects of Fingolimod?

A
  • Increased infections
  • Lymphopaenia
  • Bradycardia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
152
Q

What are further details of Fingolimod?

A
  • For highly active MS

- 1st oral DMT for RRMS

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

What is the MOA of dimethyl fumarate?

A
  • Decreasd pro-inflammatory cytokine production
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
154
Q

What are the results of dimethyl fumarate?

A

DEFINE

  • 50% decrease in ARR vs placebo
  • Decreased disability progression
  • Decreased MRI lesions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
155
Q

Is dimethyl fumarate safe?

A
  • Safer than Fingolimod

S/E

  • Flushing
  • GI S/E
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
156
Q

Further details on dimethyl fumarate?

A
  • 1st line active MS

- 1st oral drug offered as 1st line (choice)

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

What are the different types of Monoclonal Ab DMT?

A
  • Natalizumab
  • Alemtuzumab
  • Ocrelizumab
158
Q

What is the MOA for Natalizumab?

A
  • Decreased transmigration

block VLA-4 & VCAM-1 interaction

159
Q

What are results for Natalizumab?

A

AFFIRM

  • 70% decrease in ARR vs placebo
  • Decreased disease progression
  • Decreased MRI lesions (90%)
160
Q

What are the side effects of Natalizumab?

A

Rare S/E

  • Herpes
  • PML (4 in 1000)
    • 20% mortality
    • rapid decreased cognition
161
Q

What are further details on Natalizumab?

A
  • For highly active MS
162
Q

What is the MOA of Alemutzumab?

A
  • Depletes T and B cells

- Immune population rebuilds

163
Q

What are the results for Alemutzumab?

A

CARE MS I

  • 55% Decrease in ARR vs IFN-beta (similar to Natalizumab)
    no diff in disease progression
  • Decreased MRI lesions
  • No evidence of disease activity (NEDA)
164
Q

What are the side effects of Alemutzumab?

A
  • Opportunistic infections

- Autoimmunity (50% Graves’ in 5 years)

165
Q

What are further details on Alemutzumab?

A
  • 1st line active MS

- 2x injections

166
Q

What is the MOA of Ocrelizumab?

A
  • Targets CD20+ B cells
167
Q

What are the results of Ocrelizumab?

A

OPERA

  • 50% decrease in ARR vs IFN-beta
  • PPMS: 25% decrease in disability progression vs Placebo
168
Q

What are further details on Ocrelizumab?

A
  • 1st line active MS

- PPMS: 1st DMT for PPMS

169
Q

Describe autologous haematopoietic stem cell transplant (AHSCT)

A
  • Harvest stem cells
  • Immunosuppression
  • Repopulate with stem cells
  • Atkins, 2006: n = 24 had no further disease
170
Q

What is relapse-remitting MS?

A
  • Unpredictable attacks which may or may not leave permanent deficits followed by remission
171
Q

What is primary-progressing MS?

A
  • Steady increase in disability without attacks
172
Q

What is secondary progressive MS?

A
  • Initial relapse-remitting MS that suddenly begins to decline without periods of remission
173
Q

What is progressive-relapsing mutliple sclerosis?

A
  • Steady decline since onset with super-imposed attacks
174
Q

What are the clinico-pathological correlations of MS?

A
  • Inflammatory foci without demyelination = acute relapses
  • Primary demyelination = acute & chronic
  • Grey matter demyelination = progressive
  • Axonal loss in lesions = progressive symptoms
  • Grey matter neuronal axonal loss = Progressive motor, sensory and cognitive
  • Diffuse white matter changes = Fatigue?
  • Diffuse grey matter changes = motor, sensory and cognitive symptoms
  • Fatigue?
175
Q

What are some histological features seen in MS?

A
  • Brain atrophy
  • Vesicular enlargement
  • White Matter Lesions
    • Periventricular
    • Perivascular
    • Disseminated
  • Grey Matter Lesions
    • Sub-meningeal
  • Remyelinating areas
  • Lesions anywhere on the CNS
176
Q

What cells are involved in the perivascular immune cell infiltration of MS?

A
  • CD4 and CD8 T-Cells

- CD20 B-Cells

177
Q

What are the different lesion stages of MS?

A
  • Acute active: Macrophages throughout the lesions with synchronous myelin destruction
  • Chronic active: Numerous macrophages at expanding plaque edge; centre contains few cells
  • Chronic inactive: Hypocellular plaques with no macrophages and no ongoing demyelination
  • Shadow plaque: Represent remyelinated axons with thin myelin sheaths
  • Destructive plaques: Destruction of axons, oligodendrocytes, astrocytes and myelin loss - Marburg type MS and Devic’s disease
178
Q

What are the inflammatory mechanisms of neuronal damage in MS?

A
  • By-stander effect

- Autoimmune-mediated degradation of myelin sheath and oligodendrocytes

179
Q

What is the Bystander effect? (Neuronal damage)

A
  • Release of free radicals by peripheral immune cells and activated microglia
    • Nitric Oxide (NO), peroxynitrite (OON-), hydroxyl radicals (OH-)
  • Glutamate release by activated microglia results in excitotoxicity to oligodendrocytes (which express NMDA and AMPA receptors)
  • Hypoxia-like events
  • Mitochondrial dysfunction
  • Cytotoxic cytokine release by immune cells and microglia
    • TNF, lymphotoxin, IL-1beta, interferon-Y
180
Q

Describe autoimmune-mediated degradation of myelin sheath and oligodendrocytes

A
  • Macrophage-mediated
  • Presence of anti-myelin antibodies against multiple antigens (MOG, MBP…)
  • Epitope spreading —> oligoclonal bands
  • Complement activatio
181
Q

How does demyelination cause chronic symptoms?

A
  • Bystander effect: Inflammatory immune cells release antibodies and cytotoxic mediators that damage myelin
  • Without myelin, electrical signals leak out and fade away, making axons more vulnerable to damage
  • Leads to axonal loss, neuronal damage and neurodegeneration
182
Q

How does Na+/Ca2+ imbalance cause degeneration of chronically demyelinated axons?

A
  • In addition to normal Na+ channels at the nodes of myelinated axons, there are Na+/Ca2+ exchangers which allows sodium in and removes calcium
  • Na+ entry is also balanced by internodal Na+/K+ ATPase which uses ATP to pump K+ in and Na+ out
  • Na+ channels are diffusely distributed along demyelinated axons —> increased Na+ influx during AP transmission and increased ATP demand for operating Na+/K+ ATPase
  • Alterations in ATP production reduce Na+/K+ exchange capacity and increase axonal accumulation of Na+
  • Increased axonal Na+ reverses Na+/Ca2+ exchanger resulting in increased axonal Ca2+
  • Increased Ca2+ activates proteolytic enzymes, leading to damage of axoplasmic contents and axonal death
183
Q

Is axon loss responsible for clinical progression of MS?

A
  • Roughly 40% loss of corticospinal tract axons in cervical cord of SPMS
  • Axon loss is greatest during early inflammatory attacks and continues but decreases throughout course of MS
  • Identified by accumulation of APP and end bulbs
  • Only axon loss correlates with increasing chronic clinical disability in relapsing-remitting MOG-EAE
184
Q

How does neurodegeneration lead to chronic progression?

A
  • Inflammation can be resolved, remyelination can be repaired but axon and neuronal loss is irreversible
  • Irreversible but there is spare capacity
185
Q

What is the correlation between disease progression and WM/GM lesions?

A
  • Poor correlation between disease progression and inflammation/demyelination
    • Inflammation and demyelination in the white matter are key features of MS visible on MRI
    • MRI lesion load and rate of appearance of new lesions correlates poorly with clinical progression
    • Immunomodulatory therapies reduce relapse rate but fail to prevent long term progression of disability
  • Good correlation between disease progression and grey matter atrophy
186
Q

What are features of the lymphoid-like structures in MS?

A
  • A feature of meningeal inflammation
  • CD20+ cells
  • Ki67+ B-cells
  • Ig plasma cells (parafollicular)
  • CD35+ FDCs (reticular network)
  • CXCL13+ FDCs
  • CD4+ and CD8+ T-cells
  • Majority of B-cells appear to be CD27+ antigens
187
Q

What’s the relationship between meningeal inflammation and GM demyelination?

A
  • Positive correlation
188
Q

Does remyelination occur in MS?

A
  • Yes
  • Indicated by presence of thinly remyelinated sheaths
  • Remyelination is a frequent finding at the edge zones of inactive plaques
  • Suggested to be extensive in early stages but fails later on —> death of oligodendrocytes/increased damage
  • Complete remyelination of lesions can occur
  • Schwann cell remyelination is found in spinal cord
189
Q

What are glial progenitor cells’ role in remyelination?

A
  • Glial progenitors in the adult CNS are cycling
  • When isolated into culture, they differentiate into oligodendrocytes
  • Glial progenitors are thought to be responsible for oligodendrocyte replacement following demyelination
190
Q

What are possible mechanisms of repair failure?

A
  • Hostile environment/Inflammation/Axonal damage
  • No migration to lesion
  • No differentiation into oligodendrocytes
  • Differentiation but not maturation/effector function (no myelin formation)
  • Axons not reactive to myelin (no sheath formation)
191
Q

What is the definition of epilepsy?

A
  • The tendency to have recurrent (>1), unprovoked seizures
  • Need to have at least 2 seizures
  • Seizure must be unprovoked - anyone can have a seizure under appropriate stimulus
192
Q

What is the definition of a seizure?

A
  • Abnormal synchronised discharge of neurones
193
Q

What is the 10/20 convention?

A
  • Distance between electrodes within 10-20% of AP and lateral dimensions of skull
194
Q

How does an EEG work?

A
  • Synchronised discharge of set of cerebral neurons —> Na+ influx —> Sudden depletion in Na+ ions —> EEG -ve deflection
195
Q

What are the issues with EEGs?

A
  • Many layers of scalp diminish signal
  • Bell’s reflex (blinking artefact)
  • Scalp muscles give artefact
196
Q

What does epilepsy show on an EEG?

A
  • Spike and slow wave

- Normal variation: sharp transient (commonly misdiagnosed as epilepsy)

197
Q

What is the epidemiology of epilepsy?

A
  • Bimodal peak in age-specific incidence: Childhood (genetics) and later life (neurodegenerative disease or acquired CNS injury)
  • Increased risk —> increased risk of developing epilepsy
  • Prevalence = 0.5%
198
Q

How do epilepsy-related deaths occur?

A

1000 deaths per year in UK due to
- SUDEP

  • Status epilepticus (seizures fail to self-resolve —> can’t breathe —> hypoxia)
  • LOC —> Drowning
199
Q

What are the two main aetiologies in epilepsy?

A
  • Genetic disposition (normal brain with no obvious cause e.g. idiopathic)
  • Secondary to Brain Injury
200
Q

Describe the genetic disposition of epilepsy

A

Mendelian inheritance: 2% risk

Complex inheritance: 47% risk
- GWAS found 3 loci (2014): SCN1a (voltage-gated Na+ channel), PCDH7, FANCL

  • Trio studies look for de novo protein-truncating new mutations in affected children with healthy parents
    • Everyone has 1-4 de novo mutations - Healthy people: fall in tolerance genes or no change in aa sequence
    • Disease: fall in intolerant gene (synaptogenesis, synaptic transmission)
201
Q

Describe the “secondary to brain injury” mechanism

A

Environmentally acquired: Head injury, Tumour, Stroke, Infection

  • Innate immunity: Maroso (2010) induced status epilepticus using Pilocarpine in mouse model
    • Found increased HMGB1 —> activate TLR-4 on Neurons, Microglia, Astrocyte
    • –> release of pro-inflammatory cytokines —> modification of NMDA receptors —> Hyperexcitability —> Chronic seizures/Epilepsy is pro-inflammatory —> Vicious cycle
  • Adaptive immunity: Auto-antibodies against voltage-gated K+ channel, NMDA, AMPA, GABA-A R, GAD..etc

Inherited brain injury: Malformation of Cortical Development (MCD)

202
Q

What are the different types of seizures in generalised epilepsy?

A
  • Symptoms depend on location

Generalised epilepsy = Extensive, synchronised discharge in both cerebral hemispheres + LOC, EEG: global ictal discharge

  • Absence (brief LOC)
  • Tonic (contract)
  • Tonic-clonic (tense, jerk, repeat)
  • Myoclonic (fasciculation)
  • Atonic (no tone)
203
Q

What are the different types of seizures in focal epilepsy?

A

Focal epilepsy = Localised, synchronised discharge on part of the brain

  • Simple Partial (no LOC)
  • Complex Partial (with LOC)
  • Secondary Generalised (Partial —> Generalised)
204
Q

What are the decisions to treat epilepsy?

A
  • Benefits (seizure suppression) vs Harms (stigma)
  • Number of seizures at presentation (single seizure with normal MRI/EEG has 50% risk of 2nd seizure - Increased seizures —> Increased risk)
  • Seizure type, seizure severity, cause of seizure
205
Q

What are the MOA of AEDs?

A
  • Increased GABA inhibition
  • Decreased Glutamate mediation
  • Decreased voltage-gated Na+ channel activity
  • Decreased voltage-gated Ca2+ channel activity
206
Q

How does increased GABA inhibition work?

A
  • Benzodiazepine: allosteric modulation of GABA receptor —> Increased GABA influx
  • Barbiturates: allosteraic modulation of GABA receptor —> Increased GABA influx
  • Vitabatrin: inhibits GABA transaminase —> Decreased GABA degradation —> Increased synaptic GABA (not used, S/E 1 in 3 causes blindness
  • Tiagabine: inhibits GATI —> Decreased re-uptake —> Increased synaptic GABA
207
Q

How does decreased glutamate mediation work?

A
  • Presynaptic: Levetiracetam binds to presynaptic vesicle protein SV2A —> Decreased exocytosis
  • Postsynaptic: Parampanel: AMPA receptor antagonist, Felbamate: NMDA receptor antagonist
208
Q

What drugs block the voltage-gated Na+ channel?

A
  • Phenytoin, Carbamazepine, Valproate

- Use-dependent blockage (only blocks during seizures, minimises S/E)

209
Q

How does Gabapentin decrease Ca2+ channel activity?

A
  • Inhibits alpha2-beta subunit of Ca2+ channel —> decreased Ca2+ influx —> Decreased exocytosis
210
Q

What is classical Parkinsonism?

A
  • Bradykinesia
  • Rigidity
  • Rest tremor
  • Response to L-dopa
211
Q

What are the causes of Parkinsonism?

A
  • Idiopathic PD
  • Drug-induced PD
  • MSA
  • PSP
  • CBD
  • FTLD
212
Q

What are the different types of MSA?

A
  • Parkinsonism-type (MSA-P)

- Cerebellar type (MSA-C)

213
Q

What is the macroscopic pathology of MSA?

A
  • Cortical atrophy
  • Cerebellar atrophy
  • Pallor of locus coerulus
  • Pallor of SN
214
Q

What is the microscopic pathology of MSA?

A
  • Mixed neuronal and glia alpha-synuclein pathology

- Papp-Lantos bodies (Olig inclusions of alpha-syn)

215
Q

What are the symptoms of MSA?

A
  • Cerebellar signs, PD Sx
216
Q

What are features of CBD?

A
  • Alien limb phenomenon
  • Taupathy
  • PD Sx
217
Q

What is the microscopic pathology of CBD?

A
  • Fronto-parietal atrophy
  • Deep cerebellar nuclei affected
  • SN affected
218
Q

What is the microscopic pathology of CBD?

A
  • Mixed neuronal and glial pathology
  • Astrocytic plaques (tau in astrocytes)
  • Balloon Neurons (enlarged neurons with tau)
219
Q

What are features of PSP?

A
  • Tauopathy

- Supranuclear gaze palsy (lack of vertical eye movement)

220
Q

What is the macroscopic pathology of PSP?

A
  • Atrophy of basal ganglia, subthalamic nucleus and brainstem
  • Neuronal loss
  • Reactive gliosis
221
Q

What are microscopic features of PSP?

A
  • Neuronal and glial Tau +ve inclusion
  • Tufted astrocytes (many processes)
  • Coiled bodies (Oligoinclusions of Tau)
222
Q

What are some features of Tau?

A
  • Coded by MAPT gene on Chromosome 17
  • Tau stabilises microtubules
  • MAPT KO has no phenotype (other MAPs compensate)
  • Alternative splicing gives 6 isoforms: 3R-/4R- (MT-binding domain, depends on Exon 10) , 0N/1N/2N (Depends on additional inserts)
223
Q

Describe Tau phosphorylation

A
  • Tau phosphorylation —> Decreased MT Binding
  • Tau kinases: GSK-3beta and CDK5 (potential therapeutic targets)

Hyperphosphorylated Tau neurofibrillary tangles have a good clinicopathological correlation with Alzheimer’s disease

224
Q

Western Blot of Tau

A

Do card

225
Q

How can Parkinson’s plus disorder be distinguished clinicallly while patient is alive?

A
  • Biomarkers

- Imaging

226
Q

What biomarkers are seen in Parkinson’s plus disorders?

A
  • MSA has increased alpha-synclein in CSF IN PM (distinguishes it from other alpha-syncleinopathies)
  • CSF neurofilament levels may show signs of CNS damage
  • However a signal protein will not define a disease —> need an algorithm to assess many different proteins
227
Q

What does imaging do for Parkinson’s plus disorders?

A

PET imaging can distinguish PSP from CBD, MSA-P and PD

228
Q

What is it widely believed that MS is initiated by?

A
  • Autoreactive T and B cells reacting to an unknown antigen
229
Q

What happens during the early stages of MS?

A
  • Perivascular infiltrates
  • Axonal damage
  • Neurodegeneration
230
Q

Where do the early stages of disease occur?

A
  • Perivascular cuffs
  • Parenchyma (Intra-parenchyma infiltrates)
  • Meninges (Meningeal inflammation- B cells)
  • Lesions (Macrophages)
231
Q

Where are the B cells seen in MS?

A
  • Perivascular and meningeal locations

- Isolate B cells in parenchyma (active) or perivascular (chronic)

232
Q

What are the possible events leading to immune inactivation in MS?

A
  • Bystander mechanism (TCR dependent mechanism)
  • Role of EBV - infects B cells (persistent latent infection)
    • Sero +ve —> Increased risk of MS
    • Increased anti-EBNA —-> Increased risk of MS
233
Q

What is the rough immunological outline in MS?

A

1) Lymph node: APC (DC) presents autoreactive antigen to activate naive CD4 T cell —> T cell differentiates
2) Activated T cell extravasates into parenchyma
3) B cells migrate into the parenchyma and differentiate into plasma cells —> Immunoglobins —> Inflammatory

Macrophages produce ROS
Macrophages primed to M1 (pro-inflammaory) or M2 (anti-inflammatory)

4) Neuronal death —> neurodegeneration

Overall: Believed MS initiated by CD4 cells, amplified by CD8 cells infiltrating CNS, propagated by T/B cells, Macrophages

234
Q

What are possible antigens in MS?

A
  • Myelin basic protein (MBP)
  • Proteolipid protein (PLP)
  • MOG
235
Q

What are the pro-inflammatory T cells in MS?

A
  • CD4 Th1

- CD4 Th17

236
Q

How does CD4 Th1 exhibit its effects?

A

BOTH PRO-INFLAMMATORY CELLS:
Extravasation of autoreactive cells —> induces M1 microglia —> Neurotoxic mediators —> Neuronal death —> Protein debris —> DC captures antigens and go to lymph nodes —> vicious cycle

  • Th1 produces IFN-lambda, require IL-12 for differentiation
  • Increased MS activity correlates with IFN-lambda and IL-12 expression
  • IFN-lambda administration exacerbates MS
  • Transfer of Th1 —> EAE (mouse model of MS)
237
Q

How does CD4 Th12 exhibit its effects

A

BOTH PRO-INFLAMMATORY CELLS:
Extravasation of autoreactive cells —> induces M1 microglia —> Neurotoxic mediators —> Neuronal death —> Protein debris —> DC captures antigens and go to lymph nodes —> vicious cycle

  • Th17 produces IL-17. require IL-23 for differentiation
  • Increased IL-17 producing cells found in MS lesions
  • IL-23 deficiency —> resistance of EAE (greater role in initiating MS than Th1)
238
Q

What are the anti-inflammatory cells involved in MS?

A
  • CD4 Th2

- CD4 Treg

239
Q

How does CD4 Th2 exhibit its effects?

A

BOTH ANTI-INFLAMMATORY CELLS:
Induces M2 microglia —> Maintain tolerance in the CNS

  • Th2 secretes anti-inflammatory cytokines (IL-4, IL-5, IL-13)
240
Q

How does CD4 Treg exhibit its effects?

A

BOTH ANTI-INFLAMMATORY CELLS:
Induces M2 microglia —> Maintain tolerance in the CNS

  • Treg produces anti-inflammatory cytokines
  • MS: Decreased Treg activity and decreased removal of autoreactive T cells
  • CD25 is a MS susceptibility gene (GWAS) - essential for Treg development
241
Q

What other T cells are involved in MS?

A
  • CD8 cytotoxic T cells: Found at edge of inflammatory lesions and perivascular areas (clonal expression)
  • CD8 MAIT cells: Gut lymphocyte found in post-mortem MS brain tissue
  • Reg CD8 T cell subsets: AHSCT —> Increased CD8+ CD57+ cells (maintain tolerance once repopulated post-transplant
242
Q

What happens once activated T-cells extravasate into blood parenchyma?

A
  • Local DC activates more pro-inflammatory T cells —> Increased damage
  • Entry into CNS via BBB or Blood-CSF barrier (Choroid plexus —> SAD —> Pia —> Brain) - believed initiating mechanism
  • Lymphocyte require activation to enter CNS tissue
243
Q

Describe B Cell migration in MS

A
  • B cells migrate into the parenchyma and differentiate into plasma cells —> Immunoglobins —> Inflammatory
  • B cells locations: Intrameningeal + within White Matter | Demyelination located near Follicles – relationship?
  • IgG oligoclonal bands| APC |Clonal expansion in lesion | Ectopic B cell follicles in meninges & brain parenchyma
244
Q

What are macrophages and microglia’s role in MS?

A
  • Macrophages produce ROS —> axonal damage

- Microglia primedto M1 (pro-inflammatory) or M2 (anti-inflammatory)

245
Q

What are features of M1 microglia?

A
  • Pro-inflammatory phenotype
  • Injured neurons release inflammatory mediators —> M1 phenotype (and expanded by pro-inflammatory CD4
  • M1 Microglia —-> Phagocytosis + Cytokine release —> Neuronal death (MS lesion)
246
Q

What are features of M2 microglia?

A
  • Anti-inflammatory phenotype
  • Sustained by Th2/Treg
  • Th2 releases IL-4 —> M2 microglia release neurotrophic factors —> Increased neuronal survival
247
Q

How does neuronal death lead to neurodegeneration?

A
  • Inflammation —> Immune cells infiltrate —> Pro-inflammatory/Cytokine damage —> Axonal/Neuronal damage
  • Inflammation-dependent mechanisms: Loss of synapses | Retrograde and Anterograde Wallerian degeneration —> Axonal + Neuronal damage/loss
248
Q

What genes are strongly associated with MS?

A
  • HLA Class II genes - strongest, IL-7R, IL-2Ralpha, CD58
  • Many T helper cell differentiation pathway associated with MS risk
  • Different HLAs combinations modulate risk

No significant difference found between genome and epigenome of MS and non-MS twin–Baranzini, 2010

249
Q

How does the gut influence autoimmunity/MS?

A
  • Transfer of gut microbiome from MS —> mice model

- Dietary fatty acids influence GI T cell differentiation (long-chain FA —> Th1 and Th17 | short-chain FA —> Treg

250
Q

What are the CSF immunological abnormalities in MS?

A
  • CSF oligoclonal bands in >90% MS cases | Increased leukocytes, ­Increased CSF protein
251
Q

What is the overall alteration in T cell function in MS?

A
  • Increased freq of T cells responding to myelin antigens | Decreased Treg activity (to autoreactive T cells)
252
Q

Describe MS Comorbidity

A
  • MS is associated with increased ­incidence of other autoimmune conditions (esp Thyroiditis) and asymptomatic auto-Ab
253
Q

What is treatment for MS?

A
  • Only immune-modulatory | No regenerative or neuroprotective therapies | Limited Tx for Progressive MS
  • Acute relapse —> High-dose corticosteroids
  • Immunomodulatory treatment (1st line) - Injectables —> IFN-b, GA | Orals —> Dimethyl Fumarate, Teriflunomide
  • Block immune cell entry: to CNS - Natalizumab (remain in circ) | in periphery —> Gylenia (remain in LN)
    Modulating / Neutralising immune cells —> Daclizumab
  • Immunosuppressing / Depleting —> Alemtuzumab
  • Only for RRMS —> Autologous haematopoietic cell transplant (AHSCT) —> Decreased relapses, stabilises disease
254
Q

What are some symptoms of Parkinson’s disease?

A
  • Bradykinesia
  • Tremor
  • Postural instability
  • Rigidity
255
Q

What is Parkinson’s disease?

A
  • Progressive neurological disorder charactersied by bradykinesia, tremor, rigidity and postural instability (at least two)
  • Commonly associated features: autonomic dysfunction, cognitive disturbance, depression, dysphagia
256
Q

What is parkinsonism?

A
  • Clinical syndrome with some or all of these clinical features:
  • Bradykinesia
  • Tremor
  • Postural instability
  • Rigidity
257
Q

What are Parkinsonian disorders?

A
  • Disorders in which Parkinsonism is a prominent feature = akinetic-rigid disorders
258
Q

What did Ehringer & Hornkiewicz find about PD patients in 1960?

A
  • In the caudate/putamen of PD patients dopamine concentrations were only 10% of those found in controls
259
Q

How does Parkinson’s cause impaired mobility?

A
  • Loss of dopamine release in the striatum causes the acetycholine producers there to overstimulate their target neurons, thereby triggering a chain reaction of abnormal signalling leading to impaired mobility
  • Necessary to lose 80-85% of dopaminergic neurons and deplete dopamine levels by 70% before clinical symptoms of PD appear
260
Q

Why is MPTP used to model PD in animals?

A
  • MPTP=1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine
  • Prodrug to the neurotoxin MPP+
  • May be accidentally produced during the manufacture of the opioid drug desmethylprodine (MPPP)
  • Intoxication causes parkinsonism associated with degeneration of dopaminergic neurons
261
Q

What is the main site of PD and its clinical correlate?

A
  • SN

- Akinetic-rigid syndrome

262
Q

What is the main site(s) of Parkinson’s with dementia (PDD) and its clinical correlate?

A
  • SN, cerebral cortex

- Dementia more than one year after PD Dx

263
Q

What is the main site(s) of DLB and its clinical correlate?

A
  • SN, cerebral cortex

- Dementia less than one year after akinetic-rigid syndrome

264
Q

What is the main site(s) of autonomic failure and its clinical correlate?

A
  • Sympathetic neurons in the spinal cord

- Autonomic failure

265
Q

What is the main site(s) of LB dysphagia and its clinical correlate?

A
  • Dorsal vagal nucleus

- Dysphagia

266
Q

What is the pathological correlate of PD?

A
  • Neuronal inclusions composed of alpha-synuclein (Lewy bodies)
  • Neuronal loss in the substantia nigra (ventrolateral) with dopaminergic denervation of the striatum is the pathology correlate of main symptoms.
267
Q

Describe the differential regional and cellular vulnerability concept in PD?

A
  • Distinctive physiological phenotype of adult SNc DA neurons
  • Pacemaker-like properties of these cells, leading to frequent intracellular calcium transients
  • Pacemaking is necessary to maintain a basal DA tone in target structures, like the striatum; without it, movement ceases
  • Ventral tegmental area (VTA) DA neurons (also slow
    pacemakers)
    • No Calcium transients
    • Less Ca2+ channel density
    • Express high levels of the Ca2+-buffering protein calbindin
268
Q

What is α-synuclein?

A
  • α-synuclein is a member of the human synuclein family along with β-synuclein and γ-synuclein
  • It is abundant in the brain
  • It has an unknown physiological function but may be involved in the regulation of synaptic plasticity and neurotransmitter release (due to its presynaptic
    location)
  • Natively unfolded protein enriched in presynaptic terminals
  • “Amyloid-like” aggregation, pathologic post-translational modifications (including phosphorylation), truncation and oxidative damage
269
Q

What is the diagnostic gold standard for PD?

A
  • Now α-synuclein immunostaining is considered as the diagnostic gold standard
  • •Spillantini reported that Lewy bodies and Lewy neurites are immunoreactive for α-synuclein
270
Q

Where does Parkinson’s arise from the brain?

A
  • Begins in dorsal motor nucleus of the vagus nerve and anterior olfactory nucleus
  • Proceeds in rostral direction toward neocortex
  • Many cases also have Alzheimer-type plaques and tangles.
  • Conversely, a substantial number of individuals with Alzheimer disease develop Lewy pathology, especially in the amygdala
  • There may be involvement in the substantia nigra without obvious involvement
    of dorsal motor nucleus of the vagus
271
Q

Where outside the brain is Lewy Body pathology also observed?

A
  • Peripheral ganglia
    • Epicardial nerve vesicles
    • Paravertebral sympathetic ganglia
  • GI tract
    • Intermediolateral nucleus with affected preganglionic sympathetic neurons
    • Coeliac ganglion (postganglionic sympathetic neurons)
    • Auerbach’s plexus in the stomach
    • Enteric nervous system
272
Q

What are special types of PD?

A
  • Heterozygous GBA mutation carriers
    • 9–12% risk of PD, compared with 2.6% in the general population
    • Earlier age of onset and more-severe nonmotor symptoms
    • Lewy bodies and Lewy neurites contain glucocerebrosidase
    • Link between lysosomal dysfunction, α-synuclein aggregation and
      PD.
  • Heterozygous mutations in leucine-rich repeat kinase 2 (LRRK2)
    • 1% of idiopathic PD cases and 4% of familial PD cases
    • Typical LB pathology (majority)
    • Tau inclusions
    • No inclusions
  • Recessive forms of juvenile-onset parkinsonism
    • Parkin (an E3 ubiquitin ligase): majority no LBs
    • PINK1 (a mitochondrial protein kinase): LBs
273
Q

What can be used to visual early PD pathological lesions?

A
  • alpha-synuclein proximity ligation assay
274
Q

What is ALS?

A
  • chronic neurodegenerative condition causing
    muscle wasting, paralysis and death usually
    within 3-5 years due to respiratory failure
275
Q

What is ALS’ incidence?

A
  • New cases per annum: roughly 2/100,00 per year

- M > F

276
Q

What is ALS’ prevalence?

A
  • 5/8 per 100,000
277
Q

What is the lifetime risk of people with ALS in the UK?

A
  • M: 1 in 350

- F: 1 in 472

278
Q

What components are affected in ALS? (Neurons)

A
  • Upper motor neurons
  • Lower motor neurons
  • Two distinct and connected systems that are essential components in ALS
  • Both have long axons (length/cell diameter roughly 10,000)
  • Very active
  • High energy demands
279
Q

What are clinical signs of ALS?

A
  • Muscle atrophy and spasticity
  • Progressive denervation and secondary muscle weakness of limbs, trunk, tongue and respiratory (intercostal) muscles. Onset usually occurs in distal muscles of a single limb or may be bulbar, followed by widespread progression
  • Impaired swallowing and speech (‘bulbar signs’)
  • Spastic weakness and paralysis affect all skeletal muscle
  • Respiratory failure
  • No impairment of bladder, bowel or sexual function
  • Occulomotor, sensory and autonomic function spared
  • Cognitive function may be affected in a minority of cases
280
Q

What is the onset of ALS?

A
  • Starts focally and spreads
  • First signs usually occur in limb extremities or tongue
  • Wasting of thenar hand muscles
  • Wasting of the tongue muscle (‘bulbar onset’)
281
Q

What are early diagnostic tests of ALS?

A
  • Muscle biopsy

- Electromyogram (nerve conduction test)

282
Q

What are LMN signs in ALS?

A
  • muscle weakness
  • wasting
  • fasciculations
  • cramps
283
Q

What are UMN signs in ALS?

A
  • stiffness and slowness of movement
  • slow and clumsy speech
  • Babinski signs are often present
284
Q

What are features of primary lateral sclerosis?

A
  • Effects on UMNs predominate, spasticity, hyperreflexia, surivial is 20 years
285
Q

Describe treatment for ALS

A
  • Speech and swallowing: speech therapist
  • Mobility around home: occupational therapist
  • Swallowing and feeding: NG or PEG tube
  • Breathing: Oxygen, assisted ventilation (face mask)
  • Symptomatic treatments: cramps/muscle spasm
  • Slowing disease progression: Riluzole extends survial by roughly 3 months
286
Q

What is ALS characterised by?

A
  • Motor neuron loss in spinal cord, brain stem, motor cortex
  • Corticospinal tract degeneration
  • Ubiquitinated inclusions in neuronal cell bodies and proximal axons
287
Q

What is an ubiquitinated protein?

A
  • Post-translational modification by a small 76 aa regulatory protein, ubiquitin
  • Ubiquitinated inclusions are characteristic of ALS (sporadic ALS and familial ALS) and a subset of cases of frontotemporal dementia
  • TDP-43 identified as a major component of ubiquitinated inclusions in SALS, FALS and FTLD*
288
Q

What % of ALS and FTD cases are characterised by TDP-43+ve ubiquitinated inclusions

A
  • ~97% ALS cases (Familial ALS and Sporadic ALS)

- ~50% FTD (familial and sporadic, FTLD-TDP)

289
Q

What are the mutations that cause Familial ALS (FALS)?

A
  • SOD1
  • TARDBP
  • FUS
  • c9ORF72
  • Abnormalities in RNA binding proteins, proteostasis, cytoskeletal proteins
290
Q

What is TARDBP?

A
  • TAR DNA binding protein
  • TARDBP encodes TDP-43, binds TAR DNA sequences in DNA/RNA acting as a transcriptional repressor, inhibits splicing and regulates mRNA transport/ local translation
291
Q

What is the effect of TDP-43 mutations?

A
  • TDP-43 is cleaved, locates to cytoplasm
  • Hyperphosphorylated, ubiquitinated aggregates in cytoplasm, MNs, glia, neurites
  • Animal models: Neurodegeneration (cortical and spinal
    neurons) BUT without consistent formation of inclusions
  • Both wild-type (8) and mutant (4) TDP-43 rodent
    transgenics produce neurodegeneration and paralysis
    (threshold for toxicity may vary).
292
Q

How does TARDBP regulated RNA processing?

A
  • Repressor
  • Exon skipping
    Binds to CFTR pre mRNA
    UG intronic tract
293
Q

How does FUS regulate RNA processing?

A
  • ACTIVATOR nuclear hormone receptors, NFkB. (RNA polymerase complex)
  • Part of spliceosome machinery
294
Q

What are further actions of TDP-43?

A
  • 30% TDP-43 is cytoplasmic
  • Activity and cell stress stimulate nuclear efflux of both TDP-43 and FUS to the cytoplasm where they are found in RNA transport granules (stress granules and processing bodies).
  • TDP-43 also regulates local protein synthesis in dendrites (as occurs in LTP) and loss of TDP-43 reduces dendritic branching/ synapse formation and FUS knockout reduces spine formation
295
Q

What causes TDP-43 mislocalisation?

A
  • Inhibition of FUS nuclear transport protein
    (transportin) causes FUS accumulation in cytoplasm.
  • Similar inhibition of the TDP-43 nuclear transport protein (Importin) causes accumulation of soluble TDP-43 in cytoplasm.
  • Transgenic mice with a defective Nuclear Localisation Signal (NLS) show accumulation of insoluble, phosphorylated TDP-43 in brain and spinal cord, loss of endogenous nuclear
    mouse TDP-43, brain atrophy, muscle denervation, dramatic motor neuron loss and progressive motor impairments leading to death. (Mihevc et al 2016)
296
Q

How do stress granules form in the cytoplasm in ALS?

A
  • Effects on nuclear transport proteins are relevant to
    SALS, as these proteins decrease in abundance with
    age and could lead to increased cytoplasmic TDP-43.

—>

  • Cell stress, oxidative stress, heat shock, ER stress

—>

  • Formation of stress granules in the cytoplasm
    containing housekeeping mRNAs that do not require
    translation during stress also contain FUS and TDP-43
297
Q

What is the common pathway for ALS neurodegeneration?

A
  • Nuclear proteins (TDP-43 and FUS) that mislocalise to the cytosol
    TARDBP, FUS, ageing

—>

  • TDP-43+ve/FUS+ve - Ubiquitinated protein aggregates

—>

  • Neurodegeneration
298
Q

What chromosome locus is ALS and FTD linked to?

A
  • Chromosome 9p locus
299
Q

What genes are in the 9p21 ALS/FTD locus?

A
  • Chromosome 9 open reading frame 72 (C9ORF72)
  • Expanded repeats in intron 1 of C9ORF72 in FTD and ALS
  • TDP-43 +ve inclusions in cytoplasm
  • C9ORF72 containing 3 GGGGCC repeats arose in primate evolution: present in human, chimpanzee
    and gorilla but no other species
300
Q

What are some features of C9ORF72?

A
  • Unknown function likely to be a member of the DENN protein group involved in membrane fusion
  • Reduced mRNA levels
  • RNA foci accumulate in ALS (spinal cord, animal models and iPSCs) BUT overexpression of the repeat not associated with neurodegeneration
  • RAN peptides: Bi-direction transcription and translation of expanded repeat containing sequences
  • C9ORF72-specific pathology includes p62+ve cytoplasmic and nuclear inclusions in hippocampus and cerebellum that are TDP-43-ve
  • DENN = Differentially expressed in normal and neoplastic cells (Guanine nucleotide
    exchange factors for small GTPases like Rab)
301
Q

What is the putative mechanism of C9orf 72 pathogenesis?

A
  • Bi-direction transcription of expanded repeat containing sequences followed by translation of repeat associated non-AUG initiated (RAN) translation of aggregation-prone dipeptide repeat peptides (DPRs) (Cruts et al 2013).
  • Generates 5 DPRs: Poly GA, GP, GR, PR, PA
  • Poly GA is the most highly aggregatable and interferes most with proteostasis (Yamakawa et al 2014)
302
Q

Is the anatomical distribution of DPR species
related to neurodegeneration or clinical
phenotype (e.g. motor neurons and ALS)?

A
  • Inconsistent results from biological studies and
    low sample number/ restricted number of
    anatomical regions/ limited antibody use
303
Q

What did Mackenzie et al (2015) and Davidson et al (2016) find out?

A
  • Confirmed the presence of 5 DPR species (sense being most abundant) in unaffected regions: granule cells of hippocampus and cerebellum
  • BUT showed a lack of association between DPR and
    neurodegeneration and clinical features e.g. VERY
    RARE in ALS lower motor neurons
  • Degeneration and loss of anterior horn cells occurs
    in the absence DPR
304
Q

What is clinical characteristics of C9ORF72 FALS

cases compared to other FALS cases?

A
  • Age of onset
    C9ORF72+ve: 56
    All others: 61
  • Co-morbidity with FTD (%)
    C9ORF72+ve: 50
    All others: 12
  • Survival (months)
    C9ORF72+ve: 20
    All others: 26
305
Q

What factors can affect TDP-43+ve and FUS+ve ubiquitinated protein aggregates?

A
  • Activation of cell stress responses and protein degradation pathways impaired/overloaded
  • Trigger factors, cell stress, excitotoxicity, risk factors, ageing, variable penetrance
306
Q

How are FALS genes important in protein regulation?

A
  • Unfolded protein response (UPR): protective pathway increases levels of protein chaperones to facilitate folding
  • Protein degradation via the proteasome and autophagy via the lysosome (aggrephagy)
307
Q

What is VAP B?

A
  • Vesicle associated protein B
  • A VAPB mutation was first described in a Brazilian
    family linked to 20q13 (Nishimura et al 2004)
  • A second UK FALS-associated mutation was found (Chen et al 2010)
  • VAPB is localised in motor neurons and is significantly decreased in sporadic ALS spinal cord
308
Q

What is P4HB?

A
  • an ER foldase, that is induced in ER stress
    and SALS, that is a disease modifier (“risk factor”)
  • Disease onset or duration may vary substantially even
    within a family harbouring the same mutation (e.g. 0.5 to
    20 years duration). Risk factors are thought to contribute to this variable penetrance and modify disease onset/progression
309
Q

How does the waste disposal system work in ALS?

A
  • SOD1: ALS1, binds to Derlin 1
  • VCP: ALS14 and IBMPFD, ER protein export to the proteasome
  • Ubiquilin 2: X-linked FALS and SALS, binds to poly-ubi chains and components of the proteasome
310
Q

What is autophagy?

A
  • Aggrephagy of misfolded or aggregated proteins
    is activated by the failure of proteasomal degradation and molecular chaperones to resolve aggregate build-up.
  • Involves P62 (Ubiquitin binding protein Sequestosome and 1 SQSTM1) and OPTN ALS12 slow
    progressing AR/AD
311
Q

What is a novel ALS gene by Cirulli et al 2015?

A
  • TANK-binding Kinase 1 (TBK1)
312
Q

What does TBK1 do?

A
  • Phosphorylates optineurin (OPTN) and p62 (SQSTM1)
    and proteins involved innnate immunity
  • Enhances the binding of OPTN to the autophagosome
    protein LC3, facilitating the autophagic turnover of
    infectious bacteria, coated with ubiquitylated proteins, a
    specific cargo of the OPTN adaptor
  • Strengthens the importance of autophagy in ALS
    pathogenesis
  • TBK1 variants found in 1.097% of cases and 0.194% of controls
313
Q

How does Riluzole work?

A
  • an anti-epileptic drug,
  • targets voltage-dependent Na+ channels and
    reduces glutamate release
314
Q

What triggers ALS that is specific for motor neurones?

A
  • Constant excitatory activity (NMDA receptors) activated by Glutamate and D-serine
  • D-serine is significantly elevated in SALS
  • High levels of D-amino acid oxidase (DAO) are vital to metabolise and regulate D-serine levels
315
Q

What is the effect of R199WDAO?

A
  • A pathogenic mutation in DAO, R199WDAO, is an enzyme that metabolises D-serine was identified in FALS that is transmitted with disease
  • R199WDAO increases ubiquitinated protein aggregates
  • Autophagy: increase in autophagosomes and LC3-II in
    motor neuron cell line (NSC-34)
  • Significant loss of spinal cord motor neurons occurs in transgenic mice expressing R199WDAO and in sporadic cases
316
Q

What is the latest FDA approved drug for ALS?

A
  • Edavarone

- A free radical scavenger in stroke

317
Q

What is ENCALS statement on Edavarone?

A
  • FDA approval based on a single positive study showing that the drug may slow the progression of disease ONLY in a subgroup of patients.
  • An initial study of 100 patients with drug and 100 with placebo: no significant effect but a hint of an effect
  • Second smaller study for 6 months using only patients with mild to moderate swallowing and motor dysfunction (ALS FRS-R) within two years of diagnosis with normal respiratory function.
  • RESULT: Treated patients declined 5.1 points on the ALS-FRS compared to 7.5 points on placebo.
  • BUT previous studies show a decline of 5.6 in most ALS patients.
  • Recommendation: Longer studies (2 years) and testing the effect on survival
  • Cons: no effect on severely affected cases
318
Q

What is used in ALS treatment? (Drugs & Gene Therapy)

A
  • No effective cure but treatment of symptoms
  • Riluzole (since 1995): mean increased survival up to 3 months
  • Edavarone (2017): an anti-oxidant

Where the causal gene is known:
- Targeted gene delivery or gene deletion using direct
delivery of antisense oligonucleotides (ASOs) by
adeno-associated viral (AAV) vectors to “neutralise”
the mutation”. Phase 1 Clinical trial SOD1 Mutation
Intrathecal injection with adenovirus gene delivery.

  • CRISPR/Cas9 technology to target mutant genes and
    reintroduce wild type copies.
319
Q

What are possible treatment targets for ALS?

A
  • Motor neuron susceptibility and trigger factors?
  • Prevention of nuclear RNA foci (C9orf72 specific)?
  • Prevention of cytosolic inclusion formation?
  • Up-regulate UPR, UPS, autophagy?
  • Upregulate molecular chaperones (HSPs)?
  • Reduce D-serine levels?
  • Designer Drug treatment: 15,000 patients and
    7,500 controls Project MinE (US) to develop basis
    for treatment from Whole Genome Sequencing,
    metabolomics and proteomic analysis.
320
Q

What are symptoms of Parkinson’s Disease Dementia?

A
  • Mainly frontal synptoms
  • Visuospatial symptoms
  • Hallucinations
321
Q

What imaging is used for parkinsonism?

A
  • Fluorodopa (18F) —> PD: Bilateral loss of dopamine signal
  • Need to lose 80% dopamine or 50% dopaminergic neurons for symptoms
322
Q

What neurotransmitters are involved in PD?

A
  • Dopamine
  • Acetycholine
  • Excitatory amino acids (Serotonin, NA, Adenosine, Opioids)
323
Q

What motor symptoms appear in PD?

A
  • Responsive to medication: Tremor, Rigidity, Bradykinesia
  • Loss of Postural reflexes
  • Shuffling gait, Freezing episodes, Maskes facies
  • Symptoms worsen with time: Early stages (motor) —> Later stages (non-motor, severe loss of mobility and independence)
324
Q

What are non-motor symptoms of PD?

A
  • Neuropsychiatric: Hallucinations, confusion, depression (50%)
  • Autonomic : Bladder, bowel, hypotension
  • Sleep: restless legs, REM Sleep Behaviour Disorders
    • Symptoms worsen with time: Early stages (motor) —> Later stages (non-motor, severe loss of mobility and independence)
325
Q

What are NICE guidelines (2006) for PD treatment?

A
  • Physiotherapy
  • Occupational Therapy
  • Speech and Language Therapy
  • Exercise/Movement: Yoga, Tai-Chi, Le Trepidant (the shaking chair)
  • Pharmacological
326
Q

How is dopamine used in PD treatment?

A
  • Indirect agonists
  • Direct dopamine agonists
  • Enzyme blockers
327
Q

What are examples of indirect dopamine agonists?

A
  • L-DOPA (precursor, GOLD STANDARD, given with enzyme blockers
  • Amantadine (Increased DA release, MOA unknown)
  • N.B. protein load (food) may interfere with L-DOPA absorption as L-DOPA also relies on Amino Acid Transporters
  • L-DOPA benefits wears off - Therapeutic window: Low = no response, high = dyskinesia - requires surviving neurons
  • For Young-onset PD, give DA agonist (keep L-DOPA for future use), as diseases progresses –> decreased L-DOPA effectiveness
328
Q

What are examples of direct dopamine agonists?

A
  • Apomorphine
  • Ropinorole
  • More side effects (N&V, Gambling)
  • When effect wanes —> add L-DOPA
  • Early (delay L-DOPA use), late (Lower L-DOPA use)
329
Q

What are examples of enzyme blockers used in PD?

A
  • DOPA decarboxylase inhibitor: Sinemet
  • MAO inhibitor: Selegiline
  • COMT inhibitor: Entacapone
330
Q

What are some other examples of pharmacological therapy (apart from dopamine)?

A
  • Anticholinergics
  • Cell therapy
    Fetal Cell Transplant: extracting dopaminergic neurons from aborted foetuses and transplant into striatum
331
Q

How do you treat the secondary symptoms in PD?

A
  • Autonomic features: Decreased BP (Fluid, Salts, Fludrocortisone), Bladder freq/urgency (Desmopressin), Drooling (Anticholinergics)
  • Cognition: depression, anxiety, dementia, hallucinations (identify triggers)
  • Sleep (avoid drugs, treat depression, sleep hygiene), Pain
332
Q

Why is it difficult to stop progression of PD?

A
  • Widespread pathological CNS involvement
  • Infections may precipitate features, PD patients have a high risk of falls, Only adjust/add/remove one drug at a time
  • Not all PD patients have worsening symptoms due to illness or the treatment - PD patients can have other conditions
333
Q

Who do dopaminergic nenrons degenerate?

A
  • Oxidative stress
  • Ubiquitin-Proteasome dysfunction
  • Neuroinflammation
  • Mit. dysfunction
334
Q

What is the Ubiquitin-Proteosome pathway?

A
  • Polyubiquitin chain conjugated to substrate to tag it for degradation by Protease
  • Enzymes involved in Ubiquitination: E1, E2, E3 (Parkin gene associated with E3 ligase)
  • PD causing mutation (Parkin) disrupts Ubiquitination —> decreased degradation of damaged proteins —> protein aggregation —> cell death
335
Q

How do ROS make midbrain dopaminergic neurons vulnerable to neurodegeneration?

A
  • Dopamine and its metabolites have intrinsic tendency to form ROS
  • SNc is rich in Iron: Redox cycles of Iron generates ROS
  • Mitochondrial abnormalities (e.g. complex 1 defect) —> uncouples redox reactions and regenerates ROS
  • PD: deficient in Antioxidant molecules (Glutathione)
336
Q

How does mitochondrial dysfunction contribute to neuronal vulnerability?

A

Release of Cytochrome C —> Apoptosis

  • ROS affect mitochondria - PD: Complex 1 dysfunction —> mitochondria become inefficient
  • PD: alpha-synuclein aggregates inhibit Complex 1 of mitochondria
  • Damaged mitochondria —> disrupted mitochondrial potential - PD genes (DJ1) affect integrity of mit. Membrane
337
Q

How does neuroinflammation cause neuronal vulnerability?

A
  • Microglial inactivation from direct (MPTP, 6-OHDA- neuron death activate microglia) & indirect (LPS) neurotoxins
338
Q

Describe the environmental toxin theory of SNc dopaminergic neuron susceptibility?

A
  • Environmental toxins lead to specific degeneration of SNc neurons
  • MPTP-induced Parkinsonism accidentally discovered from Heroin users; MPTP is a Mitochondrial Complex 1 inhibitor
  • Uptake mechanisms of MPP+(MPTP metabolite) are specific to SNc DA neurons; MPP+ concentrates in mitochondria
  • This isn’t true PD- this is artificially killing DA neurons to produce PD-like state, Langston 1984
339
Q

Describe the transcriptional profile theory of SNc dopaminergic neuron susceptibility?

A
  • Transcriptional profile may confer inherent vulnerability of SNc dopaminergic neurons
  • Neurodevelopment defines the transcriptional profile of a cell which defines its properties
  • Neurodevelopment —> Neural stem progenitor cell —> mDA progenitor —> mDA mature neuron
  • (1) Defined as Midbrain neurons (2) Defined by Dopaminergic neurons (3) In adult, become survival/maintenance factors
  • (1) Regional identity: Otx2 (SHH, Engrailed, Wnt1) (2) Specficiation: LMX1a (FGF8, SHH) (3) Survival: Engrailed, Nurr1
340
Q

What does Nurr1 do?

A
  • Nurr1 regulates NT identify of neuron

- Nurr1 deficiency —> DA neurons cannot produce dopamine (Le WD)

341
Q

What does FoxA2 do?

A
  • Involved in late-stage DA neuron development

- Heterozygote FoxA2 (+/-) mice —> unilateral Da neuron degeneration (Ferri ALM)

342
Q

What are Engrailed genes?

A
  • Engrailed 1/2
  • Engrailed KO —> total loss of DA neurons (dose-dep)
  • Heterozygote: cell lost by P90 (Sgado)
343
Q

How are VTA neurons more resilient than SNc neurons?

A
  • Otx2: upregulated in VTA neurons > SNc
  • Otx2 overexpressionin SNc in neuroprotective (animal/cellular models) (Chung CY)
  • Overexpressionof pro-survival genes (found upregulated in VTA but downregulated in SNc) is neuroprotective
344
Q

What are features of SNCA?

A
  • 1st PD gene found
  • unknown role (possibly related to pre-synaptic vesicle trafficking)
  • Found as aggregates in Lewy Bodies
  • Pathological roles of alpha-synuclein: Proteasome inhibition, Complex 1 inhibition, Autophagy inhibition, form LBs
  • SNCA KO Mice show resistance to MPTP-induced dopaminergic toxicity
345
Q

What are features of Parkin?

A
  • Most common genetic cause
  • Autosomal recessive PD
  • Parkin = E3 ligase adds Ub + aids degradation
  • Parkin KO —> inability to remove/break down damaged proteins and organelles
346
Q

What are features of PINK-1?

A
  • Autosomal recessive
  • PINK-1 marks mitochondria (to be removed by Parkin)
  • Parkin overexpression rescues PINK-1 KO
347
Q

What are features of DJ1?

A
  • Inhibits aggregation of alpha-synuclein (via its chaperone activity)
  • Antioxidant
  • Modulates mit. membrane potential
  • DJ1 KO —> disrupted mit. membrane potential —> marked by PINK-1 to be removed by Parkin for degeneration
348
Q

What are causes of PD?

A
  • Sporadic PD
    Genetic risk factors: Tau, LRRK2, alpha-synuclein
    Environmental risk factors: Ageing, Toxins (MPTP)
  • Familial PD
    Autosomal dominant: LRRK2, alpha-synuclein
    Autosomal recessive: Parkin, PINK-1, DJ1
349
Q

What percentage of PD cases are familial?

A
  • 10%
350
Q

What are the principal gene mutations associated with familial PD?

A
  • SNCA
  • LRRK2
  • Parkin
  • PINK-1
  • DJ-1
351
Q

What are the features of SNCA?

A
  • SNCA is 1st genetic mutation identified (Polymeropoulous, 1997)
  • Found in the Brain and Heart
  • ?role in pre-syanptic vesicle trafficking
  • High penetrance, all 3 mutations cause disease, SNCA mutation —> alpha-synuclein more likely to misfold + aggregate
  • Pathological alpha-synuclein aggregates —> Proteasome inhibition, Complex 1 inhibition, Autophagy inhibition
  • When alpha-synuclein reaches certain level, cell may efflux them —> ?prion-like spread
352
Q

What are the features of LRRK2?

A
  • cytoplasmic kinase (many functions), low penetrance, (many mutations- same family, same mutation, different phenotypes)
  • LRRK2 interacts with Parkin, LRRK2 mutations results in abnormal shapes of alpha-synuclein pathology (pleomorphic pathology)
  • LRRK2 mutation results in variety of pathologies (Clinical Dx of PD may not be true PD at PM), Most common mutation of fPD
353
Q

What are the features of Parkin?

A
  • Ubiquitin E3 ligase

- Role in Mitophagy

354
Q

What are the features of PINK-1?

A
  • Mitochondrial kinase

- PINK-1 marks Mitochondria for Parkin to bind and induce mitophagy

355
Q

What are the features of DJ-1?

A
  • Protein chaperone
  • Protects cells from oxidative stress
  • Stabilises mit. membrane potential
  • DJ-1 mutation —> PINK1 marks mit.
356
Q

What are the risk genes associated with the development of idiopathic PD?

A
  • MAPT (Tau)
  • SNCA (alpha-synuclein)
  • LRRK2 (interacts with Parkin)
  • HLA-DRB5 (MHC Class II)
  • LAMP3
  • Identified by GWAS, suggests Tau in disease pathology rather than just age-related changes in Tau
357
Q

Which pathways do each SNP affect?

A
  • Ubiqutin-Proteasome system: Parkin
  • Protein aggregation: SNCA, MAPT
  • Mitochondrial clearance: Parkin, PINK-1, DJ-1
  • Protein/Membrane trafficking: LRRK2, MAPT
  • Neuroinflammation & Complement: HLA
  • Synaptic function: SCNA, LRRK2
358
Q

What are the relative risks of fPD and idio PD?

A
  • Familial PD —> Very rare, high risk (SNCA)

- idio PD —> Rare, medium risk (LRRK2) or Common, low risk (SNPs)

359
Q

How is neuroinflammation involved in PD?

A
  • PD tissues show neuroinflammation
  • Activated microglia may damage DA neurons
  • Anti-inflammatories may reduce cell loss
360
Q

What is the epidemiological evidence regarding PD neuroinflammation?

A
  • Regularly taking Ibuprofen has a lower incidence fo PD

- Glitazone (anti-inflammatory) for Diabetes given 2 days after 6-OHDA lesion in animal models: almost total protection

361
Q

How is Pioglitazone involved in PD?

A
  • PPAR-gamma agonist
  • Modules inflammation and induces neuroprotection in Parkinsonian monkeys
  • Pioglitazone Phase II Clinical Trial (multicentre, double-blind RCT): No benefit - too little too late? Few SNc neurons to rescue?
362
Q

Why may potential PD drugs be given too late?

A
  • Symptoms only arise after 60-70% loss of dopaminergic neurons in SNc BUT pathology occurs years before diagnosis of PD
363
Q

What is the premotor phase of PD?

A
  • Onset of neurodegeneration 5 - 10 years before diagnosis
  • Precursor symptoms: Anosmia, constipation, REM Sleep BD
  • Ideally target neuroprotection in premotor phase
364
Q

What are progressive symptoms of PD?

A
  • Motor symptoms

- Cognitive impairment

365
Q

What are the challenges in PD treatment?

A
  • PD pathogenesis is not well understood - what is the trigger?
  • Limitations in Clinical Trial Design (stratify rapid/slow progressers)
  • No validated biomarkers
  • 10% early stage patient - no PD
366
Q

What is epigenetics?

A
  • DNA Methylation
  • Physical blockage of DNA
  • Long-term repression
367
Q

What is histone modification?

A
  • Short-term repression, methylation/acetylation —> Increased electrostatic repulsion of histones —> Increased transcription factor accessibility —> gene expression
368
Q

What do histone acetyltransferases (HATs) and histone deacetylases (HDACs)?

A
  • HATs acetylate histones (Increased gene expression)
  • HDACs deacetylate histones (Decreased gene expression)
  • Neuronal death due to repression of essential proteins or expression of detrimental factors
369
Q

What are epigenetic changes in PD?

A
  • Misfolded alpha-synuclein enters nucleus —> binds to histone H3 –> supresses gene expression
370
Q

What is a potential epigenetic target for PD?

A
  • HDAC inhibition
371
Q

How does Valproate work?

A
  • General inhibition of HDACs (Class I & II)
372
Q

What is the unilateral Lactacystin lesion model of PD?

A
  • Lactacystin (proteasome inhibitor) injected directly into nigrostriatal pathway
  • Lactacystin —> Epigenetics (hypoacetylation —> Decreased gene expression of BDNF, hsp70 and Bcl-2 - factors for neuronal survival)
  • Valproate: reverses Histone acetylation —> Hyper-expression of BDNF, hsp70, Bcl-2, Dose-dependent neurorestoration
  • However when clinically translated, high dose Valproate induces S/E, Proof-of-principle of using epigenetics to treat PD
  • Future: potent selective HDAC inhibitor
373
Q

What are toxin-based models of PD?

A
  • Toxins injected locally or systematically (if can cross BBB) to kill DA neruons
  • Rodents bred in standardised conditions
  • Uniform brain at certain age
  • Can predict location of brain structures
374
Q

What is the 6-OHDA toxin-based model?

A
  • 6-OHDA: oxidative product of Dopamine, usually in tiny amounts that is cleared
  • Unilateral stereotactic injection: into SNc (rapid cell loss, 3d), into Medial forebrain bundle (7d), into Striatum (14d)
  • Dose-responsive loss of nigrostriatal DA neurons - ideally develop for diff. stages of cell loss (e.g. 60-70% cell loss)
  • PD Features: Mitochondrial inhibition, selective neuronal loss, oxidative stress, NO altered proteins
375
Q

What is the MPTP toxin-based model?

A
  • MPTP crosses BBB converted by Glia by MOA-B to MPP+ —> enters DA neuron (complex 1 mit. inhibitor) —> bilateral damage
  • Systemic administration - Only in susceptible species: Humans, Primates, Mice (C57Black) - Only loss in SNc not VTA/NAc
  • Behaviour changes reversed by standard PD treatment - However: high mortality, ethically difficult to use Primates
  • PD Features: Mitochondrial inhibiton, selective neuronal loss, oxidative stress, NO altered proteins - yes if +Probenicid
376
Q

What is the Lactacystine toxin-based model?

A
  • Lactacystine: Proteasome inhibitor, more stable model, but does not cross BBB
  • Unilateral stereotactic injection: into SNc/MFB, chronic progressive model (c.w. rapid cell loss in 6-OHDA and MPTP) + spread
  • PD Features: NO Mitochondrial inhibition, selective neuronal loss, oxidative stress, altered proteins
377
Q

What is the PSI toxin-based model?

A
  • Proteasome inhibitor —> Increased build up of abnormal/altered proteins —> Protein inclusions —> cell death
  • Systemic administration however not very reproducible model
378
Q

What are other toxin-based models are used?

A
  • Manganese (mining ore)
  • Carbon disulphide (rubber processing)
  • Cycad seeds (toxic seeds from Guam)
379
Q

What is the alpha-synuclein Drosophiliae model?

A
  • Either Increased alpha-syn expression or mutated alpha-syn —> selective dopaminergic neuronal death
  • Lewy Body-like structures
  • Movement disorder - change in climbing behaviour in Drosophiliae
380
Q

What is the alpha-synuclein transgenic mice model?

A
  • Need to wait 18 months for pathology
  • Transgenic animals are expensive
  • Increased WT 1x human alpha-synuclein expression —> neuronal inclusions (hLB sunflower structure, rLB is disorganised - no neuronal loss
  • Mice require 2-3 mutations in alpha-synuclein —> Age-dependent decreased in SNc TH cell number, decreased motor function, no inclusion formation
  • But no human has more than 1 alpha-syn mutation (as it has high penetrance)
  • Is double mutant model valid?
  • Expensive
381
Q

What is the rAAV alpha-synuclein overpexpression model?

A
  • Viral vector delivers alpha-synuclein (WT or mutated) to DA neurons —> Increased alpha-synuclein expression
  • Causes deficit in motor behaviour, decreased dopaminergic striatal terminals and cell bodies, progressive model (wait 6 months)
382
Q

What is the injection of pre-formed fibrils of alpha-synuclein model?

A
  • Stereotactic injection —> taken up by striatal injections —> Lewy Bodies
  • Oligomeric alpha-syn is toxic, inclusions spread over time, 30% nigrostriatal DA cell loss BUT no behavioural deficit
  • Injections of Protofibrils in SNc or gut mirrors spread of altered proteins in similar pattern to PD - Braak hypothesis
383
Q

What is the LRRK2 Transgenic Mice?

A
  • Cell death in Neuroblastoma cells and mouse cortical neurons
  • Movement dysfunction, abnormal proteins, no neuronal loss in SNc —> +MPTP to induce stress/neurodegeneration - is this PD?
  • Animal models can cope with slight genetic change due to lack of stress compared with humans
384
Q

What is the MitoPark Mouse model?

A
  • Increase in age leads to increase in mtDNA mutations, mitochondrial transcription factor A regulated mtDNA transcription
  • Tfam KO mice —> mitochondrial respiratory chain deficiency in DA neurons —> Decreased ATP —> neuronal loss
  • PD Features: Adult-onset progressive decrease in motor function - loss of DA neurons - loss of Str DA - Intraneuronal inclusions
385
Q

How are induced Pluripotent Stem cells used to model PD?

A
  • Dopaminergic neurons (‘Disease in a dish’) - carry the same mutations
386
Q

How can Gait be tested in PD animal models?

A
  • Forepaw reaching behaviour
  • Amphetamine-induced rotational assessment
  • Walking on a beam (number of falls recorded)
387
Q

Describe forepaw reaching behaviour

A
  • Measure R and L paw use in Perspex cylinder for Push off, Exploration, Landing
  • Unilateral lesion: asymmetry in paw use
388
Q

Describe the Amphetamine-induced rotational assessment

A
  • unilateral DA release (inhibition on intact side) - turn TOWARDS
  • Asymmetry causes animals to walk in circles
  • Number of turns correlates with size of lesions
389
Q

How can cognitive dysfunction be tested in PD animal models?

A
  • Using learning paradigms
390
Q

Critically evaluate the use of animals in PD modelling

A
  • PD can results in many motor and non-motor symptoms - cannot mirror all these symptoms, can only mirror certain processes
391
Q

What is an example of a drug which has been developed for PD?

A
  • Deferiprone (iron chelator)
  • PD have increased iron in SNc (iron redox —> ROS)
  • Animal model (Ferrocene deposits iron globally, not just SNc like PD)
  • Animal models show iron chelators to be neuroprotective