Cell death (DONE) Flashcards

1
Q

Cell types in the CNS

A

90% glia (can’t transmit AP)
Oligodendrocytes- produce myelin, facilitate transmission
Astrocytes- enable homeostasis, physical barrier/connector
Microglia- immune cells of the brain, phagocytose dead cells and debris
10% neurons
All cell types may be affects- damaging in itself, damage neurons

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

Specific issues in the CNS

A

In the mature CNS, most neurons are post mitotic (not dividing so can’t be replaced)
Cell death in CNS neurons is more severe than in other tissue
May occur during embryonic development (normal), acute e.g. traumatic event, chronic e.g. disease state
May share common mechanisms of cell death

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

What is important about a neuron?

A

Long axon
Metabolic requirements- very energy demanding, cell bodies full of mitochondria, constant use of energy to maintain excitable membrane
Trophic requirements- need energy to move proteins around, growth factors to sustain survival

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

Types of injury to neurons

A

Neuronopathy- death of a neuron, loss of all cytoplasmic processes and the myelin sheath, irreversible
Axonopathy- the toxic effect begins in the axon (associated myelin sheath will also degenerate)
Myelinopathy- the myelinating cell or the myelin sheath is the primary target

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

Axonal transport

A

Transport intracellular material over considerable distances, energy dependent, disturbance of metabolism/ATP may interfere leading to degeneration of the axon, disturbance of cytoskeleton leads to degeneration of the axon
Degeneration of the axon will occur if it is physically cut

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

Axonal transport structures

A

Tubulin exists in alpha and beta heterodimers, polymerise to form micro tubules
MAPs bind to tubulin subunits to regulate their stability/ cross linking
May be stabilizing or destabilizing or guide microtubules
Any disruption in tubulin or MAPs may interfere with axon transport, leading to degeneration

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

Axonapathies

A

PNS axons can regenerate, CNS cannot
Peripherally, if toxin removed, Schwann cells distal to the transection divide and act as a guide for axonal sprouts and synthesise nerve growth factor

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

Axonal transection

A

Axonal transection (chemical or physical)- accumulation of mitochondira, vesicles, filaments and lysosomes (esp. distal) within axonal swellings on either side of the lesion
Distal portion degenerates, macrophages enter the myelin sheath to take up the axonal debris (more rapid peripherally)
Proximal portion may atrophy as the cell body produces fewer cytoskeletal elements

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

Myelinopathies

A

Direct to the myelin or to the myelinating cell
Separation of the myelin lamellae leading to intramyelinic oedema
In the early stages this may be reversible
With severe/prolonged exposure to toxin, segmental demyelination follows

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

Myelinopathies- macrophages

A

Derived from microglia, remove most of the degenerate myelin

Astrocytes, oligodendrocytes and Schwann cells may also take up some components

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

Myelinopathies- remyelination limited in CNS

A

In the periphery, if the axon is intact it may be complete

The distances between nodes of Ranvier in the remyelinated sections are shorter

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

Processes involved in cell death

A

Morphological/physiological characteristics
Biochemical events
Implications of the type of cell death

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

Apoptosis

A

Process of a cell suicide under any condition when carried out by a cascade of executioner proteases; a type of programmed cell death- physiological suicide programme critical for the development and maintenance of healthy tissues

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

Necrosis

A

Accidental death of a group of cells secondary to traumatic injury

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

Mechanism of necrosis

A

Passive disintegration of cell
Dramatic and rapid- every compartment
Environmental insult- excessive injury, normal responses overwhelmed
Neurons have marked dysregulation of ion homeostasis
Breakdown/loss of plasma membrane integrity, swelling, vacuolation of cytoplasm, mitochondrial and ER dilation, gene transcription stops
Take in water and lyse
Contents of cell leaks, lysosomal enzymes escape, inflammatory process to neighbouring cells

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

Apoptosis mechanism

A

Active process
Toxic intracellular material packaged into vesicles- phagocytosed by near cells and metabolised
Damaged cells detach and round up
Cell contents packaged into membrane bound apoptotic bodies, engulfed by neighbouring cells
Reduce any inflammatory processes

17
Q

When do apoptosis and necrosis take place?

A

Apoptosis: physiological process, development, degenerative diseases
Necrosis: pathological, ischaemia, injury, infection, cancer

18
Q

Apoptosis death signals

A

Controlled by a range of cell signals
Intrinsic: response to stress, diverse array of non-receptor mediated stimuli, produce intracellular signals that act directly on targets within the cell, withdrawal of growth factors, hormones and cytokines, DNA damage
Extrinsic: involve transmembrane receptor, mediated interactions

19
Q

Major players are the caspase enzymes

A

Family of proteins > 11 cysteine proteases
Inactive until cleaved
Initiator caspases (regulatory)- 2, 8, 9, 10
Effector caspases (disassembly e.g. activated DNAse, endonucleases)- 3, 6, 7
Cleaved in sequence (initiator caspases activate effector caspases)

20
Q

How are caspases activated? Role of BCL proteins

A

Bcl-2 (B cell lymphoma 2) family- control integrity and response of mitochondria
Anti and pro apoptotic members:
Anti-apoptotic- Bcl2- stabilises mitochondrial function
Pro-apoptotic- Bad, Bid, Bax and Bak- pore formation in mitochondrial membrane
Other proteins may interact with Bcl2- mitochondrial dysfunction

21
Q

Activation of caspases by BCL

A

When cell death triggers occur:
Pro-apoptotic BCL members increase in relation to anti-apoptotic members and oligomerise
Opening of the mitochondrial pore, permeabilising the outer membrane
Permits release of pro-death proteins- cytochrome C from mitochondrial inter membrane space

22
Q

Apaf-1 in the activation of caspases

A

Apaf-1 apoptotic protease activating factor exists normally in cytosol as inactive monomer
Interacts with cytochrome C release from mitochondria and caspase 9- forms the apatosome
Transmits apoptotic signals- activate caspase 3 (effector caspase)

23
Q

Apoptosis growth factor

A

Growth factors activate PI3K or Akt
These phosphorylate proapoptotic BAD, inhibiting its ability to build up or oligomerise and activate its pro-apoptotic activity
No mitochondrial pore formation

24
Q

What triggers apoptosis or necrosis in CNS?

A

Triggers of cell death: withdrawal of growth factors, DNA damage, increased calcium, excitotoxicity, oxidative stress, mitochondrial dysfunction
Each may contribute to apoptosis or necrosis depending on severity and duration of the trigger as well as the type and age of tissue involved
Necrosis- sustained duration triggers- severe intensity
Apoptosis- transient duration triggers- less severe intensity

25
Q

Calcium functions

A

Calcium may originate extracellularly or from intracellular stores
Timing and duration of signalling events important

26
Q

The control of calcium

A

Plasma membrane channels: voltage operated ca channels, receptor operated ca channels, store operated ca channels, non-selective channels
Intracellular Ca channels- InsP3 channels, ryanodine receptors

27
Q

How does calcium kill cells?

A

Activate proteases
Mitochondrial dysfunction
Oxidative stress
Excitotoxicity

28
Q

Calcium activated proteases

A

Calpain- cleaves cytoskeletal proteins including microtubule subunits and MAPs
Phospholipases e.g. PLA2- hydrolyses membrane phospholipids- may disrupt neuronal membrane

29
Q

Mitochondrial dysfunction

A

Excess calcium in the cytosol- opening of the mitochondrial permeability transition pore
Mitochondria swell and release proteins that can lead to apoptosis
Can also cause mitochondria to release more calcium
Production of ATP may be stopped

30
Q

Oxidative stress

A

The steady state level of damage caused to cells or tissue by the generation of reactive oxygen species (ROS)
ROS are formed as a product of biological reactions and metabolism
Such as free radicals and peroxides
Normally controlled by the cell, in excess can cause damage to DNA leading to release of cytochrome C, high levels lead to necrosis

31
Q

Membrane associated oxidative stress

A

Lipid peroxidation by hydroxyl radicals and peroxynitrite
Other receptor activation
Disruption of ion movement across the membrane
Promotes depolarisation and Ca influx through NMDA receptors
Ca concentrations continue to rise as NMDA channels over activate beyond tolerable levels

32
Q

Combatting cell death- therapeutic implications

A

Caspase inhibitors prevent the apoptotic cascade
Growth factors prevent apoptosis, stimulate survival, outgrowth
Anti-oxidants- reduce oxidative stress
Anti-glutamatergics- reduce excitotoxicity

33
Q

What type of damage/disease can lead to the generation of these triggers in the CNS?

A
Traumatic brain injury
Change in/damage to the BBB
Toxins- common mechanisms: reduced axonal transport, interference in metabolism- oxidative stress
Ischaemic event
Neurodegenerative diseases
34
Q

Cell death in neurodegenerative diseases

A

Huntington’s disease: striatum and related cortical structures
Parkinson’s disease: substantia nigra and related cortical structures
Alzheimer’s disease: hippocampus and related cortical and limbic structures
Amyotrophic lateral sclerosis: spinal cord and precentral gyrus

35
Q

Ageing

A

Major risk factor for neurodegenerative diseases
Causes: increased oxidative stress, protein accumulation, DNA damage, mitochondrial instability
Additional factors contribute to disease onset: lack of trophic support, excitotoxicity, oxidative stress

36
Q

Common features of neurodegenerative diseases

A

All progressive disease states
All have specific patterns of cell death in the brain with predominantly affected regions
All have silent period before disease onset whilst degeneration is occurring
Protein deposits
Genetic vs environment: most have genetic components, genetic risk factors and environmental causes
No predictive testing for any, except genetic testing

37
Q

Apoptosis in neurodegeneration

A

Most ND associated with specific proteins
Accumulation in cells
Gene mutations

38
Q

Features of neurodegenerative diseases

A

All have protein deposits- end stage
Before deposited soluble monomers build up into oligomers
Currently believe that these oligomers are the toxic form not the insoluble protein deposits

39
Q

Can the brain repair itself?

A

Areas of cell proliferation in the adult brain
New neurons can be formed
But it is not enough