Exam 3 Flashcards

1
Q

What are the 3 regions of the brain?

A

Hindbrain
Forebrain
Midbrain

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

What are the components of the hindbrain?

A

Medulla
Pons
Cerebellum

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

What are the components of the midbrain?

A

substantia nigra

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

What are the components of the forebrain?

A

Cerebral cortex

Basal ganglia (striatum, globus pallidus, subthalamic nucleus)

Limbic system (hippocampus, amygdala)

Diencephalon (thalamus, hypothalamus)

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

What structures are associated with the forebrain?

A

“higher” structures

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

What are the functions of the medulla?

A

Autonomic functions (involuntary)

Controls: respiration, cardiac function, vasomotor responses, reflexes (coughing, etc)

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

What are the functions of the pons?

A

Acts as a “bridge” between the cerebellum and the forebrain

-relays signals from the forebrain to the cerebellum

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

What are the functions of the cerebellum?

A

“little brain”

Governs motor coordination for producing smooth movements

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

What is an important point to remember about cerebellum neurodegeneration?

A

Undergoes neurodegeneration in spinocerebellar ataxias

(causes disjointed or jerky movements “ataxias”)

*how we know if this area is damaged

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

What are the two parts of the substantia nigra?

A

SN pars compacta

SN pars reticulata

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

What is the function of the SN pars compacta?

A

Provides INPUT to the basal ganglia

*supplies dopamine to the striatum (goes wrong in Parkinson’s)

-Voluntary motor control and cognitive functions

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

What part of the midbrain is involved in the development of Parkinsons disease?

A

SN pars compacta

-The supply of dopamine by this area to the striatum gets interrupted

-This area undergoes neurodegeneration in Parkinsons

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

What is the function of the SN pars reticulata?

A

Output function

Relays signals from the basal ganglia to the thalamus

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

What is the function of the cortex (cerebrum)?

A

Processing and interpreting information

-decision making, higher level functions

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

What is the function of the basal ganglia?

A

Voluntary motor control

Some cognitive functions

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

What are the two parts of the limbic system?

A

Amygdala

Hippocampus

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

What is the function of the amygdala?

A

Emotions

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

What is the function of the hippocampus?

A

Memory

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

What are the two parts of the diencephalon?

A

Thalamus

Hypothalamus

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

What is the function of the thalamus?

A

“Relay station” to and from the cortex

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

What are the functions of the hypothalamus?

A

Controls involuntary functions

-Internal homeostasis
-Emotions
-Hormonal control
-Direct neural regulation

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

Which structures are associated with controlling involuntary functions?

A

Medulla

Hypothalamus

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

Which structures are associated with controlling voluntary functions?

A

SN pars compacta (movement with intention)

Basal ganglia

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

What is a cortico-thalamic loop?

A

The senses receive information about the environment and pass it through the thalamus to the cortex and then back

-This loop makes decisions about how to interpret and react to the incoming sensory information

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

*What area of the brain is schizophrenia associated with?

A

Frontal cortex

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

Which of the following brain structures is directly involved in controlling involuntary functions?

A. hypothalamus
B. thalamus
C. medulla oblongata
D. A, B, and C
E. A and C

A

E.
hypothalamus and medulla

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

What are the 3 functions of astrocytes?

A

-Provide neurons with growth factors + antioxidants

-Remove excess glutamate

–Support the blood-brain barrier

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

Why is it important for astrocytes to remove glutamate?

A

Too much glutamate leads to excess calcium which overwhelms mitochondria

-mitochondria spit out free radicals which leads to cell death

(too much glutamate is toxic)

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

What is the function of oligodendrocytes?

A

Produce myelin sheath that insulates axons

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

What are the 3 functions of microglia?

A

-Provide growth factors

-Clear debris through phagocytosis

-Neuroinflammation

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

What is an important point to remember about microglia’s function?

A

They are PRO-INFLAMMATORY

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

What is the overarching role of microglia that makes them easier to remember?

A

They are the “immune cells of the brain”

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

Which of the glial cells make up the blood-brain barrier?

A

Astrocytes

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

How does the blood-brain barrier act as a barrier?

A

Cells are held very tight together by tight junctions leaving no spaces between them
-this is what forms the barrier since it is too tight to cross

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

What triggers neurotransmission?

A

Electrical depolarization of the neuron
(influx of Na+)

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

At the beginning of an action potential, is the inside of the cell positive or negative?

A

Negative

-why it is below threshold to start

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

True or False: an action potential leading to neurotransmitter release will only occur when threshold is reached or passed

A

True

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

What causes repolarization?

A

Action potentials open to move K out of the cell
Cl- moves into cell

-this reestablishes the original negative charge

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

What is an afterpotential?

A

The action potential dips below resting potential between repolarization and depolarization

(an overshoot of repolarization)

**part of the refractory period

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

How long do action potentials last?

A

0.2-0.5 msec

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

What is a refractory period?

A

The period after an action potential when a neuron will not fire again

(hyperpolarized phase)

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

True or False: The magnitude of an action potential will always be the same for a single neuron

A

True
*the height of the action potential spike on a graph of them will always be the same

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

What happens when we apply an excitatory neurotransmitter?

A

Action potentials occur more frequently

*depolarizing

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

What happens when we apply an inhibitory neurotransmitter?

A

Action potentials occur less frequently

*polarizing

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

Why is the current carried by a nerve fiber (bundle of axons) greater?

A

Summation

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

What are EPSP’s?

A

Excitatory postsynaptic potentials

*weak signals that do not clear the threshold

-induced by excitatory neurotransmitters

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

What are IPSP’s?

A

Inhibitory postsynaptic potentials

*these lower the polarity far below baseline so that it is harder to clear the threshold and produce an action potential

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

How does an excitatory neuron cause an Excitatory Postsynaptic Potential?

A

Allows Na+ ions to cross the membrane

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

How does an inhibitory neurotransmitter induce hyperpolarization?

A

Allows Cl- ions to cross the membrane

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

What is hyperpolarization?

A

When a cell is so negative that it is below the resting potential

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

True or False: an IPSP can decrease the magnitude of an EPSP

A

True

-because an IPSP makes it harder to reach the threshold

-even though EPSP’s do not reach the threshold, they still will lower and farther away from the threshold than normal

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

NOTE

A

Review lecture 1 slide 20

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

What are the 3 common amino acid neurotransmitters?

A

GABA*

Glycine

Glutamate*

*= focus in CNS

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

What are the 2 types of GABA receptors?

A

GABA (A)

GABA (B,C)

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

What type of receptor is GABA (A)?

A

Ion channel

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

What type of receptor is GABA (B)?

A

GPCR

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

What is GABA?

A

The major inhibitory neurotransmitter in the brain

-depresses neuronal excitability (by increasing Cl- influx)

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

What drugs interact with GABA?

A

CNS depressants

-Sedative hypnotics
-Anticonvulsants
-Anxiolytics

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

What kind of neurotransmitter is GABA?

A

Inhibitory

-hyperpolarizes the neuron

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

What conditions are affected by GABA?

A

Epilepsy

Spasticity

Addiction/Alcohol

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

*What is glycine?

A

Similar to GABA

*but acts in the spinal cord

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

What are the 3 Glutamate Receptors?

A

AMPA, NMDA (ion channel)

mGluR (GPCR)

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

What is glutamate?

A

EXCITATORY neurotransmitter in the brain

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

How can excess glutamate cause neuronal damage?

A

By allowing excessive Ca+ influx into the neuron

(overexcites the neuron, leads to cell death)

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

What disease states are associated with glutamate?

A

Epilepsy

Schizophrenia

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

Which neurotransmitter (GABA or Glutamate) is excitatory and which is inhibitory?

A

GABA: inhibitory

Glutamate: excitatory

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

What are the 4 common non-amino acid neurotransmitters?

A

-Acetylcholine

-Dopamine (DA)

-Norepinephrine

-Serotonin (5-HT)

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

What are the two types of acetylcholine receptors?

A

Nicotinic

Muscarinic

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

Where do acetylcholine receptors act?

A

Periphery

-basal forebrain
-pons
-cortex
-basal ganglia

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

*What are examples of drugs that target acetylcholine transmission?

A

Cholinesterase inhibitors

-Aricept (used in Alzheimer’s disease)

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

What disease states are associated with acetylcholine?

A

Cognitive function/decline

Nicotine dependence

Movement disorders

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

What are the two dopamine receptors?

A

D1-like (GS coupled)

D2-like (Gi coupled)

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

What are the functions of the D1-like receptor and the D2-receptor?

A

D1-like: increase cAMP
**Gs coupled

D2-like: decrease cAMP
**Gi coupled

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

Drugs that block dopamine transmission can lead to what?

A

They increase extracellular dopamine concentrations which can produce euphoria and lead to addiction

(ex: cocaine, amphetamine)

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

Excessive dopaminergic signaling can contribute to which disease state?

A

Schizophrenia

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

Loss of dopamine neurons in the SN is responsible for which disease state?

A

Parkinson’s disease

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

What drugs interact with the dopamine pathways?

A

Antipsychotics (D2 receptor antagonists)

D2/D3 and D1 receptor agonists used in Parkinson’s disease

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

What disease states are associated with dopamine transmission?

A

Schizophrenia

Parkinson’s disease

Addiction

Depression

ADHD

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

Where does dopamine transmission occur?

A

Midbrain

-substantia nigra
-pars compacta
-ventral tegmental area (reward + addiction)

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

What are the receptors for norepinephrine?

A

a1, a2 (GPCR)

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

What are the drug targets of norepinephrine?

A

a- and B- adrenergic receptors

norepinephrine transporter (NET)

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

Norepinephrine transporter (NET) inhibitors are used to treat what disease state?

A

Depression

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

Where does norepinephrine transmission occur?

A

Pons

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

What disease states are associated with norepinephrine transmission?

A

-Memory
-Depression
-Addiction
-Pain

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

What are the 2 serotonin receptors?

A

5-HT3

5-HT1,2

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

Where does serotonin transmission occur?

A

Midbrain

Pons

**raphe nuclei in these areas produce serotonin

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

*What is the function of raphe nuclei?

A

Give rise to 5-HT axons

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

What drugs interact with 5-HT receptors?

A

5-HT2A antagonists (atypical antipsychotics)

SERT uptake inhibitors (depression)

5-HT2A agonists (hallucinogenic, ex: LSD)

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

What disease states are associated with serotonin transmission?

A

-Depression
-Mood disorders/anxiety
-Schizophrenia

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

What is multiple sclerosis (MS)?

A

An immune-mediated (inflammatory) disorder involving destruction of the myelin sheath surrounding neuronal axons

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

What are some areas in the body that are affected by MS?

A

-Optic nerve (vision)

-Corticospinal tract (fatigue)

-Cerebellum (walking problems)

-Sensory pathways (pain)

-Vestibular pathways (dizziness)

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

For an environmental insult to cause MS, what age must exposure occur AFTER?

A

15

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

How can viral or bacterial infections increase the risk of MS?

A

By activating autoreactive immune cells

-leads to an autoimmune response in genetically susceptible individuals

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

What specific virus may be involved in the development of MS?

A

Epstein-Barr Virus (EBV)

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

How can Epstein-Barr Virus (EBV) cause MS?

A

Sequence similarities between EBV and self-peptides can result in activation of autoreactive T or B cells

**molecular mimicry

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

Which antigen is responsible for the molecular mimicry of Epstein-Barr Virus in MS?

A

Epstein-Barr nuclear antigen (EBNA)

(resembles myelin basic protein, antigens recognize the host’s myelin basic protein)

**leads to myelin sheath destruction

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

How does MS demonstrate a gene-environment interaction?

A

Patients with a particular HLA phenotype have increased risk of developing MS when they also have anti-EBNA antibodies

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

The first time that an inflammatory demyelinating episode surpasses the clinical threshold is known as what?

A

CIS
Clinically Isolated Syndrome

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

After the CIS, if more inflammatory demyelinating episodes that surpass the clinical threshold occur what are these known as?

A

RRMS
Relapsing Remitting MS

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

When there are no more inflammatory demyelinating episodes but the underlying MS disease continues to progress this is called what?

A

SPMS
Secondary Progressive MS

**inflammatory part is less present but damage and symptoms continue

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

What % of MS cases are Relapsing-Remitting MS (RRMS)?

A

85%

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

What % of MS cases are Primary progressive MS (PPMS)?

A

15%

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

What is Primary Progressive MS (PPMS)?

A

Resembles SPMS

-mean age of onset is later (40)

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

True or False:
If a patient has Relapsing-Remitting MS (RRMS) they will never have Primary Progressive MS (PPMS)

A

True

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

NOTE

A

See lecture 2 sides 7+8

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

How is the overall clinical presentation of MS in a patient determined?

A

A combination of the underlying degeneration (uniform/progressive) AND the host’s immune reaction to it (intermittent/variable)

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

What are the 2 phases of MS?

A

Autoimmune

Degenerative

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

Which phase of MS is the disease trigger?

A

UNCLEAR

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

What is the function of dendritic cells?

A

Present CNS antigens

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

What is the pathophysiology of MS?

A

Dendritic cells present CNS antigens and activate T-cells

B and T cells proliferate and infiltrate the CNS using a4-integrin-mediated binding to penetrate the BBB

B cells mature to plasma cells after encountering an antigen, release antibodies

T cells interact with target ligands on MHC molecules, activate, and results in cytokine release and macrophage stimulation

This causes myelin sheath damage

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

What cells do T cells engage with in the MS autoimmune response?

A

Oligodendrocytes

*destroy these human cells

-antibodies trigger complement activation and cause pore formation and cell damage

-macrophages are recruited and release toxic agents

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

How do macrophages harm the myelin sheath?

A

The release of toxic agents and phagocytosis

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

Do action potentials travel faster or lower in myelinated regions of axons?

A

Faster

-due to the insulating effects of myelin

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

What happens to action potentials in the Node of Ranvier?

A

Action potentials slow down

-current density builds up

(note: this is a zone of demyelination)

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

Voltage-gated Na+ channels in the node of Ranvier are used to do what to action potentials?

A

Essential for replenishing action potentials

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

How does demyelination affect the propagation speed of action potentials?

A

Slows them down

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

How does remyelination occur?

A

Recruits OPCs (oligodendrocyte precursor cells) to the lesion

These turn into myelin-producing oligodendrocytes

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

Why does remyelination typically fail in MS?

A

Lack of OPCs or failure of OPCs to differentiate

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

What is astrogliosis?

A

Invasion and propagation of astrocytes that results in irreversible gliotic plaque formation or scars

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

What cells are activated by demyelination?

A

Microglia

Astrocytes

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

What is the function of microglia and astrocytes in remyelination?

A

Release pro-migratory factors and mitogens

-These recruit OPCs (oligodendrocyte precursors) to the lesion and stimulate their proliferation

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

What is the key step in remyelination that often fails in MS?

A

OPC differentiation

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

What are the 2 pathways that demyelinated axons can take?

A

Remyelination

Degradation

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

What is the function of immunomodulatory therapies in MS?

A

Interfere with T or B cell activation, proliferation, and binding to the BBB

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

What is the function of rescue strategies in MS?

A

Remyelination

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

What contrast agent is used to visualize MS brain lesions?

A

Gadolinium

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

What is Guillain-Barre Syndrome?

A

Acute, inflammatory neuropathy

-Starts as weakness in distal muscles and lower extremities and can progress to total paralysis with death from respiratory failure in days

-Caused by an autoimmune attack on peripheral nerves resulting in demyelination

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

What are the 3 categories of MS treatment?

A
  1. Treatment of Acute Attacks
  2. Disease-Modifying Therapies (DMTs)
  3. Symptomatic Therapies
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
129
Q

What is the goal of disease-modifying therapies with MS?

A

-Reduce relapse rates
-Slow progression of disability

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

Are disease-modifying MS therapies used to treat relapsing or progressive forms of the disease more?

A

Relapsing

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

What are the 4 first-line agents for MS?

A

Interferon B1a (Avonex, Rebif)

Interferon B1b (Betaseron, Extavia)

Glatiramer Acetate (Copaxone)

Fingolimod (Gilenya)

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

What are the 2 second-line agents for MS?

A

Natalizumab (Tysabri)

Mitoxantrone (Novantrone)

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

What are the 3 new drugs for MS?

A

Teriflunomide (Aubagio)

Dimethyl fumarate (Tecfidera)

Cladribine (Mylinax)

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

What are the medications used for treatment of acute MS attacks?

A

Methylprednisolone

Prednisone

Adrenocorticotropic hormone (ACTH)

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

What are the 2 corticosteroids used for acute MS attacks?

A

Methylprednisolone

Prednisone

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

How do corticosteroids work for treatment of acute MS attacks?

A

Up-regulate anti-inflammatory genes

Down-regulate pro-inflammatory genes

Alleviate edema in demyelinated areas

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

Where do Interferon B1a and B1b work?

A

Periphery

BBB

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

What is the function of Interferon B1a and B1b in the periphery?

A

Inhibit autoreactive lymphocytes

(T cells, dendritic cells)

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

What is the function of Interferon B1a and B1b in the BBB?

A

Inhibit BBB penetration by decreasing matrix metalloproteinase (MMP)

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

What reduces the efficacy of Interferon B1a and B1b?

A

neutralizing antibodies

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

What is the MOA of Glatiramer acetate?

A

Synthetic polypeptide

Mimics antigenic properties of myelin basic protein

**Modulates antigen-presenting cells (dendritic cells) which leads to decreased T cell activation

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

What is the MOA of Fingolimod?

A

Sphingosine-1-phosphate (S1P) receptor agonist

-stimulates oligodendrocyte survival (remyelination)
[CNS]

-interferes with lymphocyte movement out of lymphoid organs
[Periphery]

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

What is an important side effect to note with Fingolimod?

A

-Progressive multifocal leukoencephalopathy (PML)

[potentially lethal brain infection]

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

What are considerations to make when deciding which first-line MS treatment to use?

A

Fingolimod is superior to interferon beta

*BUT Fingolimod has more side effects associated with it

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

What is the MOA of Natalizumab?

*2nd line

A

Monoclonal antibody specific for a4 integrin*

a4-integrin pairs with B1-integrin to produce “very late antigen” (VLA-4)

Inhibiting VLA-4 binding to its ligand (VCAM-1 on CNS vascular endothelium) interferes with B and T cell movement across the BBB into the CNS

(interferes with binding of B and T cells to the BBB and makes it difficult to cross)

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

What are some side effects to be aware of with Natalizumab?

A

Progressive multifocal leukoencephalopathy (PML)

Induces neutralizing antibody development which can cause allergic reactions

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

What is the MOA of Mitoxantrone?

*2nd line

A

Anthracenedione with CYTOTOXIC ACTIVITY

-reduces lymphocyte numbers by causing DNA strand breaks via intercalation

-Delays DNA repair via inhibition of topoisomerase II

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

When should Mitoxantrone be considered for use?

A

Secondary Progressive MS (SPMS)

**first cytotoxic drug approved for this

Also can be used for induction therapy and then replaced with 1st-line agent

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

Where does Mitoxantrone act?

A

Periphery

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

What is the MOA of Teriflunomide?

New agent

A

Cytotoxic agent

-Inhibits proliferation of peripheral lymphocytes (B and T)

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

Where does Teriflunomide act?

A

Periphery

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

What is the MOA of Dimethyl fumarate, Diroximel fumarate, and Monomethyl fumarate?

New Agents

A

Metabolized in the GI tract

-Activate Nrf2-mediated cellular antioxidant responses and anti-inflammatory pathways

-Promote remyelination

-Suppress activated T cells and dendritic cells in periphery

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

What is the active form:
Dimethyl fumarate Diroximel fumarate
Monomethyl fumarate

A

Monomethyl ester

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

Where do Dimethyl fumarate, Diroximel fumarate, and Monomethyl fumarate act?

A

Periphery
and
CNS

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
155
Q

What side effect is a concern with Dimethyl fumarate, Diroximel fumarate, and Monomethyl fumarate?

A

Progressive multifocal leukoencephalopathy (PML)

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

Where does the Nrf2 antioxidant response pathway occur?

A

In astrocytes

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

What antioxidant degrades Nrf2?

A

Keap1

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

How does the Nrf2 pathway work?

A

Normally, Nrf2 is continually targeted for destruction by Keap1

When a cell is exposed to toxins or undergoes oxidative stress, Keap1 is covalently modified and cannot degrade Nrf2

Nf2 accumulates and enters the nucleus where it activates gene transcription regulated by the antioxidant response element (ARE)

These genes are involved in glutathione biosynthesis and detoxification

*Anti-oxidant and Anti-inflammatory

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

What is the MOA of Siponimod, Ozanimod, and Ponesimod?

New drugs

A

sphingosine-1-phosphate (S1P) receptor agonists

***Same as fingolimod

-oligodendrocyte survival
-remyelination
-interfere with lymphocyte movement out of lymphoid organs

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

When do we use Siponimod, Ozanimod, and Ponesimod?

A

Relapsing Remitting MS (RRMS)

Secondary progressive MS (SPMS)*

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

What is the MOA of Cladribine?

new drug

A

*Cytotoxic prodrug

Taken up into cells by purine nucleoside transporters

Damages DNA and interferes with DNA metabolism
-results in cell death and lymphocyte depletion

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

What was the original use of Cladribine?

A

Chemotherapeutic agent used for leukemia

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

What is an important point to remember about the Cladribine structure?

A

Prodrug

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

What is Rituximab approved to treat?

A

Non-Hodgkin lymphomas

Rheumatoid arthritis

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

What MS patients should we consider using Rituximab in?

A

Primary Progressive MS patients

*effective in some

also for Relapsing Remitting MS (RRMS)

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

What is the MOA of Ocrelizumab?

new drug

A

Monoclonal antibody that targets CD20 (marker on mature B cells)

*only targets mature B cells, good because we still need B cells

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

What is the name of the antisense oligonucleotide in clinical trials for MS?

A

ATL1102

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

What is the MOA of ATL1102?

A

Antisense oligonucleotide that targets VLA-4

*predicted to have the same outcome as natalizumab

(oligonucleotides recognize mRNA and prevent production of protein from them)

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

Which of the following drugs is (are) active in both the periphery and the CNS?

a. dimethyl fumarate
b. natalizumab
c. rituximab
d. teriflunomide
e. none of the above

A

A. Dimethyl fumarate

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

What are 2important diagnostic criteria for MS?

A

Dissemination in Time (DIT)

Dissemination in Space (DIS)

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

What is Dissemination in Time (DIT)?

regarding MS diagnosis criteria

A

Time between evidence of new lesions in subsequent MRIs
(30 days)

*damage that has happened more than once over time is required to diagnose MS

172
Q

What is Dissemination in Space (DIS)?

regarding MS diagnosis criteria

A

Need to have at least 2 lesions in different areas of the brain at 2 different times to be able to diagnose MS

(damage in more than one place)

173
Q

True or False: having 1 lesion is enough to diagnose as MS

A

False, need at least 2

174
Q

What is the most common type of MS?

A

Relapsing Remitting MS

175
Q

How does Secondary Progressive MS present?

A

Normally is a progression of RRMS

-fewer relapses with continuing disability

176
Q

Who is more likely to be diagnosed with Primary Progressive MS?

A

Patients diagnosed later in life

177
Q

What is Progressive Relapsing MS (PRMS)?

A

*Least common form

Steadily worsening disease

Later, clear, acute relapses

May have recovery from acute attacks but no remission between relapses

178
Q

*What is the first choice for MS treatment?

A

High-dose corticosteroid treatment

(oral or IV based on setting)

179
Q

Which corticosteroid is used IV for MS and what is the dose?

A

Methylprednisolone
500mg-1000mg IV daily

3-7 days with or without oral taper over 1-3 weeks

180
Q

Why might we give an IV corticosteroid to an MS patient?

A

Want to blast with anti-inflammatory to bring down lesion and reduce the risk of demyelination

181
Q

What corticosteroid is used outpatient in patients with MS and what is the dose?

A

Prednisone
1250mg

every other day x 5 doses
no need for taper

*note: 50mg is highest tablet strength, taking a large number of tablets

182
Q

What are the oral MS medications?

A

Dimethyl fumarate
Diroximel fumarate
Fingolimod**(only 1st line)
Ozanimod
Ponesimod
Siponimod
Teriflunomide

183
Q

What are the injectable MS medications?

A

Interferon beta-1a
Interferon beta -1b
Peginterferon beta-1a
Glatiramer acetate

184
Q

What are infusion MS medications?

A

Alemtuzumab
Natalizumab
Ocrelizumab

185
Q

What is the only drug for primary progressive MS?

A

Ocrelizumab

186
Q

What must all MS patients do to prevent the development of progressive multifocal leukoencephalopathy?

A

Get tested for JCV antibodies

-disease is caused by reactivation of dormant JCV and causes the myelin sheath to break down

187
Q

What is another important thing all patients with MS should do before starting treatment to prevent their risk of disease?

A

Get up to date on all vaccines before starting MS treatment because they will be at higher risk for diseases that have vaccines to be deadly after starting treatment

*consider varicella vaccine even if they have never had chicken pox

188
Q

What vaccines are preferred for patients with MS?

A

Inactivated vaccines

189
Q

What are 3 important counseling points for Dimethyl Fumarate, Diroximel Fumarate, and Monomethyl Fumarate?

A

-Do not open capsule (do not put on food, do not crush or chew)

-Monitor LFTs (hepatotoxicity)

-Monitor CBC (neutropenia)

190
Q

What are 2 side effects of Dimethyl Fumarate, Diroximel Fumarate, and Monomethyl Fumarate?

A

Progressive multifocal leukoencephalopathy (PML)

Flushing (can take aspirin before to prevent)

191
Q

When are Sphingosine 1-Phosphate Receptor Modulators contraindicated?

A

Past arrhythmia diagnosis

192
Q

What are the Sphingosine 1-Phosphate Receptor Modulators?

A

Fingolimod
Ozanimod
Ponesimod
Siponimod

193
Q

What is an important point to remember when trying to stop Sphingosine 1-Phosphate Receptor Modulators?

A

Discontinuation can worsen MS symptoms

194
Q

MAO inhibitors should be avoided with which drug?

A

Ozanimod

195
Q

What testing is required before starting Siponimod?

A

CYP2C9 genotype testing

196
Q

What side effects may occur with Glatiramer Acetate?

A

-Lipoatrophy
-Chest pain
-Injection side effects

197
Q

What treatment may be preferred with pregnancy?

A

Glatiramer acetate

(teratogenic effects are unknown)

198
Q

What side effects are associated with interferons?

A

*Flu-like symptoms

Psychiatric effects (depression, suicidal thinking)

Elevated LFT

Thyroid dysfunction

199
Q

What assessment should be done before starting interferons?

A

Mental health assessment

200
Q

What are the 3 monoclonal antibody drugs?

A

Alemtuzumab
Natalizumab
Ocrelizumab

201
Q

What monoclonal antibodies require REMS program to be done?

A

Alemtuzumab
Natalizumab

202
Q

What is the only FDA-approved drug for Primary Progressive MS (PPMS)?

A

Ocrelizumab

203
Q

Complete vaccinations are required at least how long before starting monoclonal antibody treatment for MS?

A

6 weeks

204
Q

Which MS drug has an absolute contraindication with pregnancy?

A

Teriflunomide

205
Q

If a patient is taking Teriflunomide and becomes pregnant, what should be done?

A

Take:
-accelerated elimination cholestyramine

or

-activated charcoal for 11 days

206
Q

What is the single MS drug approved to treat gait abnormalities/walking speed?

A

Dalfampridine (Ampyra)

-blocks K channels to prevent cell repolarization
-prolongs action potentials

**may increase walking speed

207
Q

Why do we only use ER medications and not IR?

A

IR dosage form and dose escalation is associated with seizures

*contraindicated in patients with seizure history

208
Q

What are the symptoms of Parkinson’s disease?

A

TRAP

Tremor
Rigidity
Akinesia (slow movement)
Postural Instability (balance, coordination)

-mask-like appearance
-speech and cognitive deficit, depression

209
Q

What causes Parkinson’s Disease?

A

Loss of dopaminergic neurons in the substantia nigra

210
Q

What % of nigral dopamine neurons are lost in Parkinson’s disease before motor symptoms present?

A

50% of nigral dopamine neurons

Or: 70-80% of nerve terminals in striatum

211
Q

Besides loss of dopaminergic neurons, what else is a common characteristic of Parkinson’s disease?

A

Presence of Lewy Bodies

212
Q

What are Lewy Bodies?

A

Dense, spherical protein deposits

-seen in the brains of Parkinson’s patients

-Enriched with fibrillar forms of the protein a-synuclein

213
Q

Where is it suggested that Parkinson’s disease may begin?

A

Brainstem

(because this is the first place Lewy bodies have found to show up in)

214
Q

What area of the brain needs to be affected for classic Parkinson’s symptoms to appear?

A

Substantia nigra

**undergoes neurodegeneration

215
Q

What are the 2 pathways that dopamine neurons signal through?

A

Direct pathway

Indirect pathway

216
Q

What receptors are involved in the direct pathway of dopamine neuron signaling?

A

D1 receptors (in the striatum)

217
Q

What receptors are involved in the indirect pathway of dopamine neuron signaling?

A

D2 receptors (in the striatum)

218
Q

What signaling is favored in signaling from the SNpc to D1 and D2 receptors in the striatum?

A

Thalamocortical signaling

(this is disrupted in PD)

219
Q

What happens when dopamine is released to the direct and indirect pathways?

A

Increased signaling from the thalamus to the cortex occurs

-this causes increased motor signaling

220
Q

When the direct or indirect dopamine pathways are interrupted, what is observed?

A

Interruption of motor functions

221
Q

Antimuscarinics are used in Parkinson’s for what purpose?

A

Adjunct therapy to treat tremor

222
Q

What is the antimuscarinic used in Parkinson’s?

A

Benztropine

223
Q

How do antimuscarinics work in Parkinson’s?

A

Acetylcholine is excitatory and Dopamine is inhibitory

A loss of dopamine results in excess energy in cholinergic pathways

Cholinergic antagonists help compensate for this

*most effective treatments increase dopaminergic transmission

224
Q

What drug is considered the “Gold Standard” of Parkinson’s treatment?

A

L-DOPA

225
Q

What is L-DOPA?

A

Precursor of dopamine

226
Q

How does L-DOPA differ from dopamine?

A

It is orally active and can enter the CNS

*dopamine cannot because it has a positive charge at neutral pH

227
Q

What is a common side effect of L-DOPA?

A

Nausea

228
Q

What drug can be added to L-DOPA in the combination product “Sinemet”?

A

Carbidopa

229
Q

When we co-administer carbidopa, how much can we decrease the dose of L-DOPA by?

A

4x

230
Q

Where does carbidopa work?

A

*Peripherally acting DOPA decarboxylase inhibitor

231
Q

Where in the body do we want L-DOPA converted to dopamine?

A

Substantia nigra

*but not the periphery because dopamine cannot cross the BBB

232
Q

Will carbidopa cross the BBB?

A

NO

-has a charge, only works in periphery

233
Q

What are the challenges of L-DOPA treatment?

A

*On/off oscillations (after years)

*Dyskinesia (right after dose, produces exaggerated motor effects)

Off state (when plasma levels decline, drug may not have an effect)

234
Q

How can we avoid the challenges of L-DOPA treatment?

A

Administer in a continuous manner instead of pulsatile

(Subq infusion in trials currently)

235
Q

How can we avoid limitations associated with the prodrug conversion of L-DOPA?

A

L-DOPA must be converted to dopamine by DOPA decarboxylase

-the progressive disease and death of dopamine neurons causes patients to become unresponsive to L-DOPA

**We can use dopamine receptor agonists instead
(because postsynaptic dopamine receptors are still present in the striatum)

236
Q

What are the dopamine receptor agonists?

A

Apomorphine
Ropinirole
Pramipexole
Rotigotine

237
Q

What kind of receptor agonist is Apomorphine?

A

Mixed D1/D2 agonist

238
Q

When is Apomorphine used?

A

Late stages of Parkinson’s Disease to provide rapid relief of the off state

239
Q

Why is the usefulness of Apomorphine limited?

A

It has potent emetic (vomiting) effects

240
Q

What kind of receptor agonists are Ropinirole, Pramipexole, and Rotigotine?

A

D2/D3 agonists

241
Q

Which Parkinson’s drug is a patch?

A

Rotigotine

242
Q

What are the three MAO-B inhibitors?

A

Selegiline
Rasagiline

Safinamide (reversible)

243
Q

What is the function of the MAO-B inhibitors?

A

Inhibit dopamine metabolism

244
Q

Are MAO-B inhibitors reversible or irreversible?

A

Irreversible

(propargylamines)

245
Q

When are MAO-B inhibitors used?

A

Can be used as initial monotherapy

Can be used as an adjunct to L-DOPA

246
Q

What is the reversible MAO-B inhibitor?

A

Safinamide

247
Q

When is Safinamide used?

A

Adjunct to L-DOPA/Carbidopa

248
Q

What are the COMT inhibitors?

A

Entacapone

Tolcapone

249
Q

What is the MOA of COMT inhibitors?

A

Decrease metabolism of L-DOPA in the periphery

Tolcapone: Increases CNS dopamine levels

250
Q

What drugs are included in the combination product Stalevo?

A

L-DOPA
Carbidopa
Entacapone

251
Q

What is bradykinesia?

A

Slowness of movement

252
Q

What is the Parkinsonian gait?

A

Dragging of feet (difficult to get them off the ground)

Width of step shortened

**fall risk

253
Q

What is an important non-motor symptom of Parkinson’s disease to remember?

A

Constipation

**dopaminergic interruption has a variety of roles including gut movement

254
Q

What are some non-pharmacologic therapies for Parkinson’s disease?

A

-Exercise/physical therapy

-Nutritional counseling

-Occupational therapy

-Psychotherapy/support groups

-Speech therapy

255
Q

What is the first step that needs to be done in Parkinson’s treatment?

A

Rule out drug-induced Parkinson’s

256
Q

What are the 1st line agents for Parkinson’s?

A

-Dopamine precursor
-Dopamine agonists
-MAO-B inhibitor

257
Q

What are the 2nd line agents for Parkinson’s disease?

A

COMT inhibitors

Amantadine

258
Q

When are dopamine agonists considered first-line?

A

age < 60 years

Higher risk for dyskinesia

259
Q

Rank the following drugs in order of the MOST efficacy with motor symptoms to LEAST efficacy with motor symptoms:

Dopamine Agonists
Levodopa/Carbidopa
MAOB-I

A

Most:
Levodopa/Carbidopa
Dopamine Agonists
MAOB-I

260
Q

What are the two types of dopamine agonists?

A

Non-ergot

Ergot

261
Q

What are the 3 first-line treatment agents for Parkinson’s disease?

A

Dopamine Precursors (L-DOPA, Carbidopa)

Dopamine Agonists

MAO-B inhibitors

262
Q

What are the non-ergot drugs?

A

Pramipexole
Ropinirole
Rotigotine
Apomorphine

263
Q

What are dopamine agonists used for?

A

Minimize LD motor fluctuations

264
Q

Which dopamine agonists are rarely used?

A

Ergots

*have toxicity

265
Q

Which drugs can have Impulse Control Disorder (ICD)?

A

Dopamine Agonists

266
Q
A
267
Q

Which drug can cause insomnia because it is metabolized to a stimulant?

A

Selegiline

268
Q

Which drug group in Parkinsons has a risk of serotonin syndrome?

A

MAO-B inhibitors

*drug-drug interaction
(these minimize dopamine breakdown but can also inhibit breakdown of other neurotransmitters like serotonin)

269
Q

What are the COMT inhibitors?

A

Entacapone
Opicapone
Tolcapone

“capone”

270
Q

When are COMT inhibitors used?

A

As a combination product to manage symptom fluctuation

271
Q

Which drug can cause brown/orange urine discoloration as a side effect?

A

Entacapone

272
Q

When is Amantadine used?

A

To manage motor fluctuations in Parkinsons disease

*rarely used monotherapy

273
Q

What is a common side effect of Amantadine?

A

Confusion/hallucinations

274
Q

What are the anticholinergics used in Parkinsons Disease?

A

Benztropine

Trihexyphenidyl

275
Q

When are anticholinergics used in Parkinsons disease?

A

Management of tremor-dominant symptoms in patients <65 years old

276
Q

What medications can worsen Parkinson’s disease?

A

Dopamine Antagonists

-Antipsychotics
-Metoclopramide
-Prochlorperazine
-Promethazine

277
Q

NOTE

A

Review lecture 6 slide 35

278
Q

What gender is more likely to get Alzheimer’s disease?

A

Females

2:1 ratio female:male

279
Q

What is the neuropathology of Alzheimer’s disease?

A

Loss of brain volume

Amyloid plaque formation

Neurofibrillary tangle formation

Synapse loss

280
Q

Where are amyloid plaques located?

A

Extracellular

281
Q

Where are neurofibrillary tangles located?

A

Intracellular

282
Q

What are Amyloid Plaques made of?

A

Amyloid-B peptide (AB)

283
Q

What are neurofibrillary tangles made of?

A

Hyper-phosphorylated tau

284
Q

Neuropathology associated with Alzheimer’s disease typically affects what areas of the brain?

A

Areas of higher cognitive function:
-Entorhinal cortex
-Hippocampus
-Basal forebrain cholinergic systems
-Neocortex

(these have to do with memory, learning, consolidation, and cognition)

285
Q

Neurons with neurofibrillary tangles and amyloid plaques often develop what?

A

Destruction of synapses

286
Q

Destruction of synapses causes what?

A

Reduced levels of neurotransmitters

*Acetylcholine
-Serotonin
-Norepinephrine
-Dopamine

*Also dysregulated glutamate (toxicity)

287
Q

What is the key pathogenic molecule in Alzheimer’s disease?
AB or tau?

A

AB

288
Q

What gene encodes the AB precursor protein?

A

APP (amyloid precursor protein)

**mutations in this are linked to early onset Alzheimer’s

289
Q

What syndrome is associated with Alzheimer’s-like symptoms late in life and why?

A

Down’s syndrome

*because the APP gene is located on chromosome 21 and patients with Down’s syndrome have an extra copy of this

290
Q

What is the function of B-secretase and y-secretase?

A

Release AB peptide from APP (amyloid precursor protein)

291
Q

What is the function of a-secretase?

A

Cleaves APP (amyloid precursor protein) in the middle of the AB segment

-releases non-toxic fragment

***this prevents the release of AB which prevents amyloid plaque formation!

292
Q

What is the difference between AB42 and AB40?

A

*differ in the number of amino acids in their structure

*AB42 more readily forms amyloid plaques

293
Q

How can mutations in the APP gene lead to greater amyloid plaque formation?

A

Mutations favor more cleavage by B- or y-secretase

*this leads to more AB42 production over AP40

294
Q

What do the presenilin proteins do?

A

Cleave AB from APP

*mutations in the gene encoding these lead to early onset Alzheimer’s

*mutations lead to production of more AB42 by altering y-secretase cleavage of APP

295
Q

What are the presenilin proteins components of?

A

y-secretase complex

296
Q

What are the 2 presenilin proteins?

A

PSEN1 and PSEN2

297
Q

How does AB aggregation effect tau pathology?

A

Kinase activation leads to tau hyper-phosphorylation

This creates neurofibrillary tangles

Ultimately results in disruption of cytoskeleton and axonal trafficking (synapse transport)

298
Q

Neurofibrillary tangle formation results in what?

A

Cytoskeletal defects

The cytoskeletal tracks are disrupted leading to defects in axonal transport and synaptic dysfunction

299
Q

What is the function of normal tau?

A

Binds and stabilizes microtubules

*hyperphosphorylation causes tau to fall off and form tangles

300
Q

How does AB aggregation affect microglial activation?

A

Increases microglial activation

-microglia release pro-inflammatory cytokines that cause neuroinflammation

-microglia also release reactive nitrogen species and reactive oxygen species that cause oxidative stress

301
Q

What two things are caused by microglial activation?

A

Neuroinflammation

Oxidative stress

302
Q

Individuals with what gene/allele are more likely to develop Alzheimer’s disease?

A

ApoE4

*note allele 4

303
Q

Inheritance of what Apo allele decreases the risk of developing Alzheimer’s?

A

ApoE2

304
Q

What are the cholinesterase inhibitors used to treat Alzheimer’s disease?

A

Donepezil

Rivastigmine

Galantamine

305
Q

What is the function of Donepezil?

A

Specific, reversible inhibitor of acetylcholinesterase

306
Q

What is the function of Rivastigmine?

A

Inhibits acetylcholinesterase AND butyrylcholinesterase

307
Q

What is the function of Galantamine?

A

Selective, reversible inhibitor of acetylcholinesterase

AND enhances the action of acetylcholine on nicotinic receptors

308
Q

What is the anti-glutamatergic therapy used in Alzheimer’s disease?

A

Memantine

309
Q

How does Memantine work?

A

NMDA (glutamate receptor) antagonist

-blocks glutamatergic neurotransmission through noncompetitive mechanism

-reduces excitotoxicity (caused by excess glutamate signaling)

310
Q

What are the possible targets for disease-modifying therapy of Alzheimer’s disease?

A

AB generation
AB aggregation
AB clearance

Tau kinase inhibitors
Glutamate-mediated excitotoxicity
Inflammation and Oxidative stress

311
Q

What do the AB antibodies: Aducanumab, Lecanemab, and Donanemab target?

A

AB clearance

312
Q

What radiolabeled agent is used to visualize Alzheimer’s disease?

A

Florbetapir (18F)

*binds B-amyloid
*visualized by PET scanning

313
Q

What is the radiolabeled agent specific for tau?

A

Flortaucipir (18F)

314
Q

True or False: Not every dementia patient has Alzheimer’s

A

True

315
Q

What are the characteristics of Vascular Dementia?

A

-Impaired judgment or executive function

-Result from brain injury associated with vascular disease or stroke

316
Q

What are the characteristics of Dementia with Lewy Bodies (DLB)?

A

-Combination of cognitive decline and Parkinsonian symptoms

-Visual hallucinations

-Cortical Lewy Bodies

317
Q

What are the characteristics of Frontotemporal Dementia (FTD)?

AKA Pick’s Disease

A

-Disinhibited behavior
-Tau accumulation (Pick’s bodies)

318
Q

What percent of patients with epilepsy will experience their first seizure by age 20?

A

> 80%

319
Q

What is the percent chance of having epilepsy after having a single seizure?

A

20%

320
Q

What is the percent chance of having epilepsy after having 2 seizures?

A

60%

321
Q

What is the percent chance of having epilepsy after having 3 seizures?

A

75%

322
Q

What are the types of generalized seizures (non-absence types)?

A

-Myoclonic
-Tonic
-Clonic
-Atonic
-Tonic-Clonic

323
Q

What is a myoclonic seizure?

A

Shock-like muscle contraction

(ex: isolated jerking of head, trunk, body)

*isolated jerks

324
Q

What is a tonic seizure?

A

Rigidity

-results from increased extensor muscle tone

**occur in children

325
Q

What is a clonic seizure?

A

Rapid, repetitive motor activity

*occur in babies and young children

326
Q

What is an atonic seizure?

A

Sudden loss of muscle tone

-patient falls if standing
-“drop attacks”

327
Q

What is a tonic-clonic seizure?

A

Tonic (rigid) phase immediately followed by Clonic (repetitive activity) stage

“Grand Mal” seizure

328
Q

What is the most severe seizure type?

A

Tonic-Clonic

329
Q

What is a “seizure”?

A

Paroxysmal disorder of the CNS

-characterized by abnormal cerebral neuronal discharge

**with or without loss of consciousness

330
Q

What is “epilepsy”?

A

Repeated seizures

*due to damage, irritation, or chemical imbalance in brain
-leads to sudden, excessive, synchronous electrical discharge

331
Q

Where does a seizure originate from?

A

Gray matter

332
Q

What causes seizures?

A

Disordered, Synchronous, and Rhythmic firing of brain neurons

“Synchronized Hyperexcitability”

333
Q

What can prolonged seizures cause?

A

Ischemia

*because the brain uses more energy and oxygen than it can manufacture

334
Q

What are the 3 seizure classifications?

A

Focal onset

Generalized Onset

Unknown Onset

335
Q

What is the most common classification of seizures?

A

Focal

336
Q

What is a Focal seizure?

A

Seizure activity starts in one area of the brain

“focal point”

337
Q

Where do focal seizures commonly start?

A

Temporal lobe

338
Q

What typically causes Focal seizures?

A

Some kind of damage to the brain

(disease, trauma, lesion, etc)

339
Q

What is a Focal to Bilateral seizure?

A

A focal seizure that progresses to a generalized seizure

-typically by projections to the thalamus

340
Q

What is a Generalized seizure?

A

Involves both brain hemispheres and multiple locations in the brain

341
Q

What is the main cause of a Generalized seizure?

A

Genetic

342
Q

What is an important point to remember about Generalized seizures?

A

Patients are unconscious

343
Q

What genes indicate that a patient has epilepsy?

A

GABRA1
GABRB2+3
GABRG2

SCN1A+B
SCN2A
SCN8A

*GABA receptors and Sodium Channels

344
Q

What does an EEG of a focal seizure look like?

A

Sharp waves shown only in the electrodes placed where the seizure is coming from on the brain

(less pronounced than generalized, only appears in some rows of the EEG)

345
Q

What does the EEG of a generalized seizure look like?

A

Shows involvement of both hemispheres

-crazy peaks along the entire EEG in every row

346
Q

What are the two types of focal seizures?

A

Aware

Impaired Awareness

347
Q

What is the defining factor of Aware type seizures?

A

No loss of consciousness

348
Q

What seizure types may involve the subject experiencing auras?

A

Aware Type Focal seizures
+
Impaired Awareness Focal seizures

349
Q

What are the symptoms of an Aura associated with a seizure?

A

Abdominal discomfort
Sense of fear
Unpleasant smell
Abnormal electrical activity

350
Q

What is a simple partial seizure?

A

Old term for Aware type focal seizure

351
Q

What is a complex partial seizure?

A

Old term for Impaired awareness seizure

352
Q

What is a postictal state?

A

Altered state of consciousness after a seizure

*occurs with impaired awareness focal seizures

353
Q

What type of seizure is an absence seizure?

A

Generalized

354
Q

What are the two types of Absence seizures?

A

Typical

Atypical

355
Q

What is a Typical Generalized seizure?

A

Brief loss of consciousness (10-45 seconds)

-staring or eye flickering, often repetitive

**No convulsions, aura, or postictal period
*Begin abruptly

356
Q

What is an Atypical Generalized seizure?

A

*Slower onset than typical

**Difficult to control pharmacologically

357
Q

Do tonic-clonic seizures have an aura?

A

No aura in the tonic phase!

***can have a brief aura if they are focal-to-bilateral tonic-clonic seizures

358
Q

What is Status Epilepticus?

A

Repetitive seizure activity in which the patient does not regain consciousness between seizures

OR

A single seizure lasting > or = 30mins (after 5 minutes it is considered this)

*note: this is a medical emergency
*convulsions occur

359
Q

What is the therapeutic goal with Status Epilepticus?

A

Bring seizures under control within 60 mins

360
Q

True or False:
One seizure is considered epilepsy

A

False

361
Q

When can we gradually withdraw epilepsy drugs?

A

Patients clinically free of seizures for 2-5 years

362
Q

Which of the following types of convulsions can be preceded by an aura phase?

A. typical absence (petit-mal)
B. primary generalized tonic-clonic
C. focal to bilateral (secondary generalized) tonic-clonic
D. all of the above

A

C. focal to bilateral (secondary generalized) tonic-clonic

363
Q

What is a “secondarily generalized” seizure?

A

old term for Focal-to-Bilateral seizure

364
Q

What is a paroxysmal depolarizing shift (PDS)?

A

A large depolarization that triggers a burst of action potentials

365
Q

What occurs during depolarization?

A

Influx of cations (ex: Ca+)

-through activation of AMPA and NMDA by glutamate

**Becomes more positive
**Excites the neuron

366
Q

What occurs during hyperpolarization?

A

Influx of anions (ex: Cl-)

Efflux of K+

*follows depolarization
*Involves activation of GABA receptors

**Becomes more negative
**Inhibits the neuron

367
Q

What are the two ways we can reduce excitation?

A

Reduce glutamate (excitatory neuron leads to excess movements)

Increase GABA (inhibitory interneuron)

368
Q

Activation of GABA receptors cause what?

A

Hyperpolarization

**want this with seizures to prevent them

369
Q

Activation of Glutamate causes what?

A

Depolarization

370
Q

What are AMPA and NMDA?

A

Glutamate-mediated excitatory receptors

*activation is important in depolarization

371
Q

Which one causes seizures?
Break down of Glutamate
Breakdown of GABA

A

The breakdown of GABA-mediated inhibition allows action potentials to propagate to distant neurons which causes a spread of seizure activity

-this makes seizures change from focal to secondary generalized

372
Q

What is a potential trigger of status epilepticus?

A

Sudden discontinuation of AEDs (anti-epileptic drugs)

373
Q

What drugs increase the risk for seizures?

A

**Alcohol
-Theophylline
-CNS stimulants (coffee)
*Bupropion
*Oral contraceptives
-Withdrawal from depressants
*Clozapine

374
Q

Which of the following occurs during the hyperpolarization phase of a PDS?

A. Influx of Cl- ions resulting from GABA receptor activation
B. Influx of K+ through voltage- and calcium-dependent K+ channels
C. Activation of NMDA receptors
D. All of the above

A

A. influx of Cl- ions resulting from GABA receptor activation

375
Q

What is the MOA of anticonvulsant drugs?

A

Stabilize and reduce neuronal excitability

376
Q

What are the 5 ways that anticonvulsant drugs reduce excitability?

A
  1. Decrease sodium influx and prolong inactivation of their channels
  2. Reduce calcium influx
  3. Enhance GABA-mediated neuronal inhibition
  4. Antagonize excitatory transmitters (glutamate!)
  5. Other targets (levetiracetam!)
377
Q

What method of reducing excitability by anticonvulsant drugs is most critical for absence seizures?

A

Reduction of calcium influx

378
Q

What are the excitatory presynaptic targets of anticonvulsant drugs?

A

Na+ channels

Ca+ channels

379
Q

What are the excitatory post-synaptic targets of anticonvulsant drugs?

A

NMDA receptors

AMPA receptors

380
Q

What are the inhibitory presynaptic targets of anticonvulsant drugs?

A

GABA transporter (GAT-1)

GABA transaminase (GABA-T)

381
Q

What are the inhibitory post-synaptic targets of anticonvulsant drugs?

A

GABA A receptors

GABA B receptors

382
Q

What common structural feature do anticonvulsant drugs share?

A

Heterocyclic ring structure

383
Q

What drugs are used to treat Focal Seizures and Generalized Tonic-Clonic seizures?

A

Hydantoins

384
Q

What Hydantoins are used to treat focal and tonic-clonic seizures?

A

Phenytoin*

Fosphenytoin **injectable prodrug

385
Q

What is the MOA of phenytoin?

A

Binds and stabilizes the inactive state of Na+ channels

**not isoform selective, works in brain and other body parts

386
Q

What are two important points about phenytoin elimination kinetics?

A

Dose-dependent

Has non-linear pharmacokinetics

387
Q

What do phenytoin, carbamazepine, and phenobarbital do that can cause drug interactions?

A

Induce liver cytochrome P450

**increases metabolism of other drugs

388
Q

What are two side effects to remember for phenytoin?

A

Gingival hyperplasia
Hirsutism

389
Q

What is the MOA of carbamazepine and oxcarbazepine?

A

Stabilize inactivated state of Na+ channels

390
Q

Which iminostilbene has reduced toxicity?

A

Oxcarbazepine

(less toxic than carbamazepine)

391
Q

What is the MOA of Lacosamide?

A

Enhances inactivation of voltage-gated Na+ channels

392
Q

What drugs target Excitatory Presynaptic Na+ Channels?

A

Phenytoin
Carbamazepine
Lacosamide

393
Q

Where do barbiturates and Benzodiazepines bind?

A

Allosteric regulatory site on GABA A receptor

(directly bind GABA receptor)

394
Q

What is the MOA of phenobarbital?

A

Binds an allosteric regulatory site on GABA A receptor

-Increases duration of Cl- channel opening

(enhances GABA signaling)

395
Q

True or False: primidone is more similar to phenytoin than phenobarbital

A

True

396
Q

What are the benzodiazepines?

A

Diazepam

Clonazepam

397
Q

What is the MOA of Diazepam and Clonazepam?

A

Binds an allosteric regulatory site on GABA A receptor

-Increases FREQUENCY of Cl- channel opening

(enhances GABA signaling)

398
Q

What type of treatment are Diazepam and Clonazepam not useful for?

A

Chronic treatment
(because of tolerance)

399
Q

What is clonazepam useful for?

A

Acute epilepsy treatment

**Absence seizures

400
Q

What drugs target the inhibitory post-synaptic GABA A receptors?

A

Phenobarbital

Benzodiazepines

401
Q

What is ataxia?

A

Impaired coordination

402
Q

What is the MOA of Gabapentin and Pregabalin?

A

Increase GABA release

Decrease Ca+ influx

Reduce glutamate release

**mimic GABA

403
Q

When is Vigabatrin used?

A

Adjunct therapy for refractory patients

404
Q

What is the MOA of Vigabatrin?

A

Irreversible inhibitor of GABA transaminase (GABA-T)

405
Q

What is GABA transaminase (GABA-T)?

A

Enzyme responsible for degrading GABA

*bad

406
Q

When is Tiagabine used?

A

Adjunct therapy

407
Q

What is the MOA of Tiagabine?

A

Inhibits GABA transporter (GAT-1)

408
Q

What is the GABA transporter (GAT-1)?

A

Transporter responsible for transporting GABA out of the system

*bad

409
Q

What is the function of the NMDA receptor?

A

Glutamate binding triggers Na+ and Ca+ influx and K+ efflux

410
Q

What is the function of the AMPA receptor?

A

Glutamate binding triggers Na+ influx and K+ efflux

411
Q

What is the MOA of Felbamate?

A

NMDA receptor antagonist

412
Q

When is Felbamate used?

A

3rd line agent for refractory cases

413
Q

What is an important side effect of Felbamate?

A

Severe hepatitis

414
Q

What is the MOA of Topiramate?

A

AMPA and Kainate receptor antagonist

415
Q

When is Topiramate used?

A

Monotherapy or Adjunct

416
Q

What are the succinimides?

A

Ethosuximide

417
Q

What is the MOA of ethosuximide?

A

Blocks T-type Ca+ channels

418
Q

What do T-type Ca+ channels do?

A

Generate the rhythmic discharge of an absence attack

419
Q

Which of the following statements is TRUE?

A. Tiagabine inhibits GABA transaminase
B. Gabapentin increases Cl- influx in postsynaptic neurons
C. Topiramate is an NMDA receptor antagonist
D. Phenytoin is stabilized by the co-administration of carbamazepine

A

B

420
Q

What two drugs have a risk for seizure recurrence with withdrawal?

A

Phenytoin

Valproate

421
Q

What is considered “drug-resistant” epilepsy?

A

Failure of at least two trials of antiseizure medication

422
Q

What type of drug therapy is used with status epilepticus?

A

IV

423
Q

How many attacks are needed to classify Migraine without Aura?

A

5 attacks

424
Q

How many attacks are needed to classify Migraine with Aura?

A

2 attacks

425
Q
A