• Shuffle
    Toggle On
    Toggle Off
  • Alphabetize
    Toggle On
    Toggle Off
  • Front First
    Toggle On
    Toggle Off
  • Both Sides
    Toggle On
    Toggle Off
  • Read
    Toggle On
    Toggle Off
Reading...
Front

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/93

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

93 Cards in this Set

  • Front
  • Back
Define Neurodegenerative disorders
Progressive and irreversible loss of neurons from specific regions of the brain
Neurodegenerative disorders are primarily disorders associated with what?
aging
What are the % of patients over age of 65 that have Parkinson's and Alzheimer's?
1%
10%
What % of each generation in families carrying gene have Huntington's?
50%
Define dementia?
taken from Latin, from de- "without" + ment, the root of mens "mind") is a serious loss of cognitive function in a previously unimpaired person.
How can dementia be induced?
Drug induced
Depression induced
Stress induced
Head injury
Illnesses (kidney, heart failure)
What are the striking feature of PD, HD, AZ & ALS
selective vulnerability
Pd is extensive destruction of what? But what regions are not effected?
extensive destruction of Dopa neurons in substantia nigra but not cortex and other regions
AZ is what kind of injury?
neuronal injury most severe in hippocampus and neocortex (specific functional regions)
HD is what kind of gene? How is it expressed? Where is it pronounced?
mutant gene is expressed through out brain and other organs yet pathology pronounced in neostriatum
ALS is the loss of what?
loss of spinal motor neurons and cortical neurons
PD creates mutations in what genes?
mutations in alpha-synuclein, parkin, UCHL1 and DJ-1 genes lead to specific forms of the disease
AZ creates mutations in genes for what two things?
mutations in genes for amyloid precursor protein (APP) and presenilins (involved in APP processing)
ALS creates mutations in what?
mutations for SOD1 account for  2% of cases
What are environmental triggers that play a role in neurodegenerative disorders?
Infectious agents, environmental toxins and acquired brain injury play role in neurodegenerative disorders.
What is Excitotoxicity?
neural injury that results from the presence of excess glutamate in the brain.
What is a major “excitatory” neurotransmitter
Glutamate
What is impaired during Excitotoxicity? What does this increase with?
The brain’s ability to respond to excess synaptic glutamate levels is impaired


This increases with age, exposure to environmental toxins, stroke, head trauma
During neuronal injury in neurodegenerative disease what happens to proteins?
Proteins misfold, etc, causing dysfunction of normal cellular processes
What is oxidative metabolism required for? What is a consequence of it?
Oxidative metabolism required for survival but a consequence is the generation of reactive oxygen species (ROS) such as hydrogen peroxide and oxyradicals.
What is Excitotoxicity?
neural injury that results from the presence of excess glutamate in the brain.
What is a major “excitatory” neurotransmitter
Glutamate
What is impaired during Excitotoxicity? What does this increase with?
The brain’s ability to respond to excess synaptic glutamate levels is impaired


This increases with age, exposure to environmental toxins, stroke, head trauma
During neuronal injury in neurodegenerative disease what happens to proteins?
Proteins misfold, etc, causing dysfunction of normal cellular processes
What is oxidative metabolism required for? What is a consequence of it?
Oxidative metabolism required for survival but a consequence is the generation of reactive oxygen species (ROS) such as hydrogen peroxide and oxyradicals.
Alterations in neuronal mitochondrial energy metabolism increases with what?
age
what increase the production of ROS?
Environmental toxins
What can ROs cause to break and what can it cause to reorganize?
ROS cause DNA strand breaks, nucleotide reorganization
What are 4 clinical features of Parkinson's disease?
Bradykinesia (slow movement)
Muscle rigidity
Resting tremor
Impaired postural balance, altered gait, falling
What are the 2 pathological hallmarks of PD?
loss of the pigmented dopaminergic neurons of the substantia nigra (70-80% dopaminergic neurons)
↓ dopamine action in corpus striatum (Regions involved in motor control)
Untreated PD progresses how long and to what state?
progresses over 5-10 yr to akinetic state
In PD, what does metabolism of Dopa via MAO yield? Hydrogen peroxide + Fe2+ (abundant in basal ganglia) yields what? Degeneration of dopa neurons could occur due to what?
1. Metabolism of Dopa via MAO yields hydrogen peroxide
2. Hydrogen peroxide + Fe2+ (abundant in basal ganglia) yields free radicals
3. Degeneration of dopa neurons could occur due to deficiencies in protective mechanisms
Levodopa is what kind of precursor and what is it a precursor for?
metabolic precursor of dopa.
What is the single most effective treatment of PD?
Levodopa
Levodopa is converted how?
Levodopa is inert; conversion by decarboxylation to dopamine in presynaptic terminals of dopaminergic neurons of striatum
Levodopa restores what signaling? How?
Restores dopa signaling in corpus striatum by ↑dopa synthesis in surviving neurons of substantia nigra.
What is Levodopa's half life? What can it result in>
Short t1/2 (1-2 hr): can result in fluctuation in plasma [ drug] and response
What is Carbidopa? What is used in combo with it?
Dopa decarboxylase inhibitor that does not cross BBB. Used in combo w/levodopa
When carbidopa is used with levodopa what is decreased and what is increased?
↓ metabolism of levodopa in GI tract and periphery →↑ availability of levodopa to CNS (<1% levodopa reaches CNS if dosed alone)
What kind of receptor is used to treat parkinson's? What are 4 drug examples of it?
Dopamine Receptor Agonists: (bromocriptine, ropinirole, pramipexole, rotigotine)
What are advantages of Dopamine Receptor Agonists compared to levodopa?
Enzymatic conversion not required →don’t depend on functional capacity of nigrostriatal neurons.
Longer acting that levodopa; avoid fluctuations in motor activity.
MAY have neuroprotective effect via ↓ free radical formation from dopa metabolism (by reducing release of endogenous dopa and reducing need for levodopa)
Dopa agonists may delay levodopa treatment in early PD and may ↓ dose of levodopa in advanced PD
Ineffective in patients who have shown no therapeutic response to levodopa.
What 2 components are involved with metabolism of dopamina and? levodopa
Catechol-O-Methyltransferase (COMT) Inhibitors and MOA
COMT inhibitors (selective, reversible) block what? What does this increase?
the peripheral conversion of levodopa to 3-O-methylDOPA → increasing the plasma t½ of levodopa as well as the fraction of each dose that reaches the CNS.
Catechol-O-Methyltransferase (COMT) Inhibitors are absorbed how? Bound how? what kind of vd?
Readily absorbed, highly protein bound, limited Vd
what drug crosses the BBB and inhibits COMT in the CNS (therapeutic effect in PERIPHERY)? What drug does not?
Tolcapone, but not entacapone,
What can tolcapone cause? When is it used? What is monitored?
Tolcapone can cause fulminating hepatic necrosis→ only use when other Rx has failed and monitor liver function
What are Selective MAO-B Inhibitors? Where are they expressed and what are they involved in?
MAO-B is predominant isoform expressed in striatum and involved in dopa metabolism.
What drug inhibitits MAO-B? At what dose levels? What does it increase?
Selegiline (deprenyl) selective inhibits MAO-B (but not MAO-A) at low to moderate doses →↑ [dopa].
Rasagline is what kind of inhibitor? What is its potenency compared to selegiline?
Rasagline: irreversible, selective MAO-B inhibitor; 5X > potency than selegiline.
What are 3 drugs that are Muscarinic Receptor Antagonists?
Trihexyphenidyl, benztropine, biperiden
What is amantadine?
antiviral agent for influenza A. Modest efficacy for PD.
amantadine increases release of what? What kind of blocker is it?What kind of weak receptor does it have? How is it compared to levodopa?
Complex pharmacology; ↑ dopa release in the striatum, anticholinergic, weak NMDA glutamate receptor antagonist. Less efficacious than levodopa but fewer side effects.
How many cases of Alzheimer’s Disease in the us is there?
there are as many as 5.3 million Americans living with AZ
What kind onset and progression is there for AZ?
Gradual onset but “relentless progression”
What is the first sign of AZ? What signs are there later?
First sign: impaired short term memory
Later, other cognitive deficits appear:
Visual-spatial processing deficits
“Ideomotor apraxia” (loss of ability to use common tools)
What 2 components remain normal until very late in the AZ disease?
Arousal and motor function remain normal until very late in the disease
AZ characterized by what two components? What neurons appear to be lost?
AZ characterized by neuronal atrophy and a loss of cortical and subcortical neurons
In particular, cholinergic neurons in appear to be lost
what kind of projections to the parietal lobe are less active during AZ?
Serotonergic
What plaques are common in az?
Amyloid and senile plaques are common
What are amyloid plaques caused by? What is it accompanied by?
Amyloid plaques are caused by accumulation of the protein b-amyloid
Accompanied by neuronal degeneration
What is b-amyloid? What is it the product of? Where is APP expressed? It maybe what kind of receptor? How is B-amloid hypothesized to be produced?
1. b-amyloid is a short polypeptide (42-43 AA)
2.Product of the amyloid precursor protein (APP)
3.( APP is normally expressed in many neurons)
4. may be a membrane-bound receptor
5.b-amyloid is hypothesized to be produced via abnormal proteolytic cleavage of APP
The probability of developing b-amyloid plaques is what? What may this be due to?
1. Heritable
2.May be due to inheritance of abnormal genes involved in APP itself
May also be due to the inheritance of abnormal genes involved in processing APP
What activity is altered in Alzheimer’s disease?
Tau Protein
What is tau protein associated with? What 2 places is it found in? What are microtubules important for?
Tau protein is associated with microtubule stability
Found in both neuronal and non-neuronal tissue
Microtubules are important to neuronal architecture integrity, neurotransmitter transport
Tau protein is though to by ___ in alzheimers?
hyper-phosphorylated
what does the hyper-phosphorylation of Tau protein leads to the development of? What 3 things does it lead to the loss of?
1.The development of tangles
Loss of structural integrity of neurons
2.Loss of efficient neurotransmitter transport from the nucleus to the pre-synaptic area
Loss of efficient recycling of neurotransmitter metabolites
In later stages of the disease, Neurofibrillary tangles are numerous in what?
The hippocampus
Areas of cortex involved in abstract thinking, language, our general (not tactile) perception of the world and interaction in it
In AZ, what happens to the neurotransmitter content of cerebral cortex?
Neurotransmitter content of cerebral cortex ↓↓ consistent with neuronal loss
In AZ there is Striking and disproportionate deficiency of what?
acetylcholine (Ach)
IN AZ acetylcholine neurons are though to degenerate in both what areas? What is thought to happen to Ach? What activity is also decreased?
1.ACh neurons are thought to degenerate in both subcortical and cortical areas
2.ACh receptor levels are also thought to decrease
3.Choline acetyl transferase (CAT) activity is decreased
CAT is the final rate-limiting enzyme involved in ACh production
What does reduced Choline acetyl transferase (CAT) activity = ?
= reduced ACh in secretory vesicles
What is the “Cholinergic hypothesis”?
What can Central cholinergic antagonists (atropine) induce?
1.deficiency in Ach is critical in genesis of AZ
2.induce a state similar to dementia
AZ pathology includes defects in what 3 things?
includes defects in serotonin, glutamate and several neuropeptides
IN AZ there are destruction of what 3 things?
Destruction of not only cholinergic neurons but also cortical and hippocampal targets that receive cholinergic input
Alzheimer’s patients exhibit what increased levels? What can these levels be measured in?
1.Glutamate
2.Increased glutamate can be measured in the CSF
In AZ patients, what does excessive glutamate result in? What does glutamate play major rules in? What can high levels of glutamate lead to?
1. Excessive glutamate results in disrupted cognition
2.Glutamate plays a major role in learning and memory
3.High levels of glutamate ultimately can lead to excitotoxicity
excitotoxicity in AZ patients lead to what?
Toxicity is associated with the over-activation of NMDA (glutamate) receptors
Influx of Ca2+
Cell death
What is the primary approach in tx of AZ?
to attempt to augment cholinergic function
ACh neurotransmission in AZ patients is intricately involved in multiple what?
involved in multiple hippocampus, cortex functions
What are 4 drugs Acetylcholinesterase (AChE) inhibitors (AChE)?
Tacrine (Cognex
Donepezil(Aricept)
Rivastigmine (Excelon)
Galantamine (Razadyne)
Tacrine is what kind of AChE inhibitor? What does it show effectiveness in improving?
Centrally acting AChE inhibitor
Shows some effectiveness for improving memory
Donepezil is what kind of inhibitor? What does it show moderate improvement in? What does its long t1/2 allow?
Reversible, non-competitive central AChE inhibitor, Modest improvement in cognitive scores
Long t1/2 allows qd dosing
Rivastigmine is what kind of inhibitor?
Reversible, non-competitive AChE inhibitor
Galantamine is what kind of inhibitor?
Competitive inhibitor of AChE
AZ rx memantin is hypothesized to be what? What does it block the flow of? What does it slow the rate of?
1. hypothesized to be a non-competitive NMDA receptor antagonist
2.Blocks the flow of Na+ and Ca++ through the NMDA receptor channel
3.slow rate of memory los?
What kind of disorder is Huntinton's Disease?
HD is an autosomal dominant disorder with nearly complete penetrance.
What gene does the HD mutation lie within? Equally heritable from both parents, however 80% of patients developing symptoms before age 20 inherit from which parent?
1.IT15 gene
2. Father
What is anticipation?
tendency for the age of disease onset to ↓ w/each succeeding generation.
What does tx of HD focus on?
focuses on reducing symptoms, preventing complications, and providing support and assistance to the patient and family/caregivers.
What is the only FDA-approved Rx for ALS?
Riluzole
Riluzole delays what? Has what kind of effects? Inhibits the release of?What does it block? Inhibits what channels?
1.Delays onset of ventilator dependence or tracheotomy in some patients
2.Has both pre- and post-synaptic effects
3.Inhibits glutamine release
4. Blocks post-synaptic NMDA and kinate-type glutamate receptors
5. Inhibits voltage-dependent sodium channels
What is spasticity?
increase in muscle tone w/initial resistance to passive displacement of a limb at a joint, followed by sudden relaxation
What are drug tx of spasticity?
baclofen (GABA- B receptor agonist) and tizanidine (2 adrenergic receptor agonist in CNS)