• 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

1/622

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;

622 Cards in this Set

  • Front
  • Back
  • 3rd side (hint)
what is the Monoamine hypothesis of depression?
too little serotonin and epi lead to depression
what is the Neurotrophic hypothesis of depression?
BDNF regulate neural
plasticity,survival and and neurogenesis.

Depression decreases BDNF and antidepressants
increase BDNF.
What type of depression is this:

Loss (Adverse life events). Physical illness (myocardial infarct, cancer). Drugs (antihypertensives, alcohol, hormones). Other psychiatric disorders (senility).
Reactive
What type of depression is this:

Precipitating life event not adequate for degree of depression. Autonomous (unresponsive to changes in life). May occur at any age (childhood to old age). Biologically determined (family history)
Endogenous

this is where the neurotrophic hypothesis comes into play (that you have neuronal issue)

Will see more PHYSICAL symptoms
What type of depression is this:

Characterized by episodes of mania. Cyclic: mania alone, rare; depression alone, occasional; mania-depression, usual
Manic-depressive
What kind of drug is Fluoxetine? used to treat?
Specific serotonin reuptake inhibitors (SSRIs)

These are a relatively new class of antidepressants and have become drugs of first choice
What kind of drug is Paroxetine? used to treat?
Specific serotonin reuptake inhibitors (SSRIs)

These are a relatively new class of antidepressants and have become drugs of first choice
What effects do SSRIs have on the ANS and CNS?
Nothing significant!

This makes them safe, OD won't likely kill you
what is the half life like for Paroxetine and Sertraline? What is important about this?
Short as compareted to Fluoxetine (1 day vs 3 days)

so you have to make sure to wean them off it slowly or you can cause depression pretty quick

these are all SSRIs
what do SSRIs do to the metabolism of other drugs?
inhibit the metabolism of other drugs
What is the major side effect of SSRIs? why

****TEST
Nausea, headache, diarrhea are most common (stimulation of 5HT3 receptors)

note: another reason people may discontinue the drug is due to sexual dysfunction
pt comes in on an SSRI with anxiety and motor restlessness, what drug(s) could have caused this?
Fluoxetine, and to a lesser extent, sertraline
Which SSRI ) may induce birth defects. Use during 3rd trimester may cause persistent pulmonary hypertension in newborns or withdrawal syndrome.
paroxetine
what do you have to worry about with kids taking SSRIs
high risk of suicide
What must you be careful of when you give an SSRI with SNRIs,TCA, MAOI, meperidine, or triptans?

****TEST
SEROTONIN SYNDROME

mental status changes,(confusion to coma) disorders of motor activity (hyperreflexia, rigidity), and autonomic disturbances ( hyperthermia, hypertens.) All SSRIs)

more dangerous with fluvoxetine (long acting)
PMDD, generalized anxiety disorder, panic, OCD, and bulimia can all be treated with what? (general)
SSRIs
Venlafaxine is what kind of drug?
Serotonin and norephinephrine Reuptake Inhibitors (SNRI)
duloxetine is what kind of drug?
Serotonin and norephinephrine Reuptake Inhibitors (SNRI)
which Serotonin and norephinephrine Reuptake Inhibitors (SNRI) should you avoid in pts with liver disease?
Duloxetine – liver damage (rare) – avoid in pts with chronic liver disease or hepatic insufficiency
Duloxetine does what to P450s?
Duloxetine has greater potential than venlafaxine because it inhibits CYP2D6 and may increase drug levels of TCAs, Type 1C antiarrhythmics (e.g., flecainide) and phenothiazines.

INHIBITS METABOLISM

SNRI
2 drug classes that can treat diabetic peripheral neuropathy?
SNRIs and Tricyclics
milnacipran is what kind of drug? Used to treat?
SNRI

Fibromyalgia

note: all SNRIs can tx fibromyalgia, but milnacipran is used ONLY for fibromyalgia, not depression
Trazodone MOA? Use?
blocking 5HT2A receptors.

Most common use of trazodone is as hypnotic.
Side effects – arrhythmias and priapism
Bupropion MOA? Use? Risk of causing?
causes presynaptic release of DA and NE and blocks their reuptake.

Depression (used after SSRI)

Risk of seizures

note:Less sexual dysfunction, wt gain or sedation than other antidepressants.
Mirtazapine MOA? Use? why it is liked?
blocks H1 receptors, presynaptic a2-adrenergic receptors, increases NE and 5HT release and blocks 5HT2 and 5HT3 receptors

depression

liked: Less sexual dysfunction; more wt gain and sedation.
Doxipen
Maprotiline
Amoxapine

are what kind of drugs
second generation Tricyclic Antidepressants
Imipramine
Desipramine
Amitriptyline

are what kind of drugs?
first generation Tricyclic Antidepressants
MOA of tricyclic antidepressant
block the reuptake of NE and serotonin in the CNS, thereby potentiating transmitter action. This may be an early event in the mechanism of antidepressant action.
which tricyclic has the strongest anticholinergic action?
Amitriptyline
What are the 3 ways you have adverse effects with Tricyclic antidepressants
Antimuscarinic – e.g., dry mouth, constipation, blurred vision, Intensity varies with different drugs (amitriptyline strongest).

CNS – sedation, weakness, fatigue; may switch depressed patient to manic phase; tremor; seizures

Cardiovascular – orthostatic hypotension (a1 block), tachycardia, myocardial infarction, congestive heart failure, arrhythmias. May be less likely or less severe with 2nd generation drugs
what can happen with acute poisoning of tricyclics? tx?
Acute poisoning – life threatening; excitement and restlessness

Convulsions → coma → death. Must support vital functions. Physostigmine can reverse antimuscarinic, cardiotoxic, and neurotoxic effects.
these drugs in addition to treating depression can be used to treat neuropathic pain, nocturnal enuresis, and fibromyalgia
Tricyclics

nocturnal enuresis (imipramine), fibromyalgia (amitriptyline, doxipen).
Tranylcypromine MOA? Use?
Block metabolism of naturally occurring monoamines, e.g., NE, 5HT. Are used in the treatment of depression when tricyclics are not effective and ECT refused.

MAOi
Phenelzine MOA? Use?
Block metabolism of naturally occurring monoamines, e.g., NE, 5HT. Are used in the treatment of depression when tricyclics are not effective and ECT refused.

MAOi
Isocarboxazid MOA? Use?
Block metabolism of naturally occurring monoamines, e.g., NE, 5HT. Are used in the treatment of depression when tricyclics are not effective and ECT refused.

MAOi
2 main reasons we don't use MAOi?

***TEST
Potentiate sympathomimetic amines

*****Dangerous interaction with tyramine (found in food)-->causes release of NE-->Hypertensive crisis

(MAO knocks out Tyramine, without it, it gets into the system and causes the release of NE, leads to hypertensive crisis)
How does St. John's Wort help with depression? What are the problems associated with it?
Contains compounds that inhibit MAO and block 5HT reuptake

Drug interactions are a problem. Hyperforin, a P450 inducer found in St. J. W., increases metabolism of drugs used to treat AIDS and suppress transplant rejection.
MOA of Lithium Carbonate? Use?
interferes with second messenger system, can't remake PIP2, SLOWS DOWN NEURONAL ACTIVITY

stabilize mood in bipolar pts
A bipolar pt comes in complaining that they are constantly thirsty and peeing all the time...what drug is causing this? how?

***
Lithium- Antagonizes ADH

leads to excessive thirst and urination
Primary drug of choice for mood stabilization (bipolar)?
VALPROATE (remember this is also an anti-epileptic!)

others that can be used: carbamazepine (na channel block), lamotrigine (na channel block)
2 drugs that are first choice for depression?
SSRIs or venlafaxine (SNRI)
when is Electroconvulsive therapy (ECT) indicated
when a person is a serious threat to kill themselves when depressed
What are the first line choice drugs for bipolar
Mood stabilizers are mainstays of treatment – Lithium, valproate or lamotrigine are first choice.

Carbamazepine also used.
this is an antidepressant that can be added for severe depression, but don’t use alone. Can be used first line
lamotrigine (another antiepileptic).

An antidepressant can be added for severe depression, but don’t use alone. Certain antidepressants may not be effective.
for a manic episode, what drugs can be added to mood stabilizers?
olanzapine or risperidone (atypical antipsychotics)

added to mood stabilizer (Lithium, valproate or lamotrigine).
what is the Monoamine hypothesis of depression?
too little serotonin and epi lead to depression
what is the Neurotrophic hypothesis of depression?
BDNF regulate neural
plasticity,survival and and neurogenesis.

Depression decreases BDNF and antidepressants
increase BDNF.
What type of depression is this:

Loss (Adverse life events). Physical illness (myocardial infarct, cancer). Drugs (antihypertensives, alcohol, hormones). Other psychiatric disorders (senility).
Reactive
What type of depression is this:

Precipitating life event not adequate for degree of depression. Autonomous (unresponsive to changes in life). May occur at any age (childhood to old age). Biologically determined (family history)
Endogenous

this is where the neurotrophic hypothesis comes into play (that you have neuronal issue)

Will see more PHYSICAL symptoms
What type of depression is this:

Characterized by episodes of mania. Cyclic: mania alone, rare; depression alone, occasional; mania-depression, usual
Manic-depressive
All anti-psychotics share what mechanism in common?
block D2 receptors

(both typical and atypical)

but the importance of this in relation to other receptor actions varies from drug to drug
What kind of drug is Paroxetine? used to treat?
Specific serotonin reuptake inhibitors (SSRIs)

These are a relatively new class of antidepressants and have become drugs of first choice
What kind of drug is Fluoxetine? used to treat?
Specific serotonin reuptake inhibitors (SSRIs)

These are a relatively new class of antidepressants and have become drugs of first choice
what is the half life like for Paroxetine and Sertraline? What is important about this?
Short as compareted to Fluoxetine (1 day vs 3 days)

so you have to make sure to wean them off it slowly or you can cause depression pretty quick

these are all SSRIs
what do SSRIs do to the metabolism of other drugs?
inhibit the metabolism of other drugs
What effects do SSRIs have on the ANS and CNS?
Nothing significant!

This makes them safe, OD won't likely kill you
pt comes in on an SSRI with anxiety and motor restlessness, what drug(s) could have caused this?
Fluoxetine, and to a lesser extent, sertraline
Which SSRI ) may induce birth defects. Use during 3rd trimester may cause persistent pulmonary hypertension in newborns or withdrawal syndrome.
paroxetine
what do you have to worry about with kids taking SSRIs
high risk of suicide
for a manic episode, what drugs can be added to mood stabilizers?
olanzapine or risperidone (atypical antipsychotics)

added to mood stabilizer (Lithium, valproate or lamotrigine).
this is an antidepressant that can be added for severe depression, but don’t use alone. Can be used first line
lamotrigine (another antiepileptic).

An antidepressant can be added for severe depression, but don’t use alone. Certain antidepressants may not be effective.
What are the first line choice drugs for bipolar
Mood stabilizers are mainstays of treatment – Lithium, valproate or lamotrigine are first choice.

Carbamazepine also used.
when is Electroconvulsive therapy (ECT) indicated
when a person is a serious threat to kill themselves when depressed
2 drugs that are first choice for depression?
SSRIs or venlafaxine (SNRI)
Primary drug of choice for mood stabilization (bipolar)?
VALPROATE (remember this is also an anti-epileptic!)

others that can be used: carbamazepine (na channel block), lamotrigine (na channel block)
A bipolar pt comes in complaining that they are constantly thirsty and peeing all the time...what drug is causing this? how?

***
Lithium- Antagonizes ADH

leads to excessive thirst and urination
MOA of Lithium Carbonate? Use?
interferes with second messenger system, can't remake PIP2, SLOWS DOWN NEURONAL ACTIVITY

stabilize mood in bipolar pts
How does St. John's Wort help with depression? What are the problems associated with it?
Contains compounds that inhibit MAO and block 5HT reuptake

Drug interactions are a problem. Hyperforin, a P450 inducer found in St. J. W., increases metabolism of drugs used to treat AIDS and suppress transplant rejection.
2 main reasons we don't use MAOi?

***TEST
Potentiate sympathomimetic amines

*****Dangerous interaction with tyramine (found in food)-->causes release of NE-->Hypertensive crisis

(MAO knocks out Tyramine, without it, it gets into the system and causes the release of NE, leads to hypertensive crisis)
Isocarboxazid MOA? Use?
Block metabolism of naturally occurring monoamines, e.g., NE, 5HT. Are used in the treatment of depression when tricyclics are not effective and ECT refused.

MAOi
Phenelzine MOA? Use?
Block metabolism of naturally occurring monoamines, e.g., NE, 5HT. Are used in the treatment of depression when tricyclics are not effective and ECT refused.

MAOi
Tranylcypromine MOA? Use?
Block metabolism of naturally occurring monoamines, e.g., NE, 5HT. Are used in the treatment of depression when tricyclics are not effective and ECT refused.

MAOi
these drugs in addition to treating depression can be used to treat neuropathic pain, nocturnal enuresis, and fibromyalgia
Tricyclics

nocturnal enuresis (imipramine), fibromyalgia (amitriptyline, doxipen).
what can happen with acute poisoning of tricyclics? tx?
Acute poisoning – life threatening; excitement and restlessness

Convulsions → coma → death. Must support vital functions. Physostigmine can reverse antimuscarinic, cardiotoxic, and neurotoxic effects.
What are the 3 ways you have adverse effects with Tricyclic antidepressants
Antimuscarinic – e.g., dry mouth, constipation, blurred vision, Intensity varies with different drugs (amitriptyline strongest).

CNS – sedation, weakness, fatigue; may switch depressed patient to manic phase; tremor; seizures

Cardiovascular – orthostatic hypotension (a1 block), tachycardia, myocardial infarction, congestive heart failure, arrhythmias. May be less likely or less severe with 2nd generation drugs
which tricyclic has the strongest anticholinergic action?
Amitriptyline
MOA of tricyclic antidepressant
block the reuptake of NE and serotonin in the CNS, thereby potentiating transmitter action. This may be an early event in the mechanism of antidepressant action.
Imipramine
Desipramine
Amitriptyline

are what kind of drugs?
first generation Tricyclic Antidepressants
Doxipen
Maprotiline
Amoxapine

are what kind of drugs
second generation Tricyclic Antidepressants
Mirtazapine MOA? Use? why it is liked?
blocks H1 receptors, presynaptic a2-adrenergic receptors, increases NE and 5HT release and blocks 5HT2 and 5HT3 receptors

depression

liked: Less sexual dysfunction; more wt gain and sedation.
Bupropion MOA? Use? Risk of causing?
causes presynaptic release of DA and NE and blocks their reuptake.

Depression (used after SSRI)

Risk of seizures

note:Less sexual dysfunction, wt gain or sedation than other antidepressants.
Trazodone MOA? Use?
blocking 5HT2A receptors.

Most common use of trazodone is as hypnotic.
Side effects – arrhythmias and priapism
milnacipran is what kind of drug? Used to treat?
SNRI

Fibromyalgia

note: all SNRIs can tx fibromyalgia, but milnacipran is used ONLY for fibromyalgia, not depression
2 drug classes that can treat diabetic peripheral neuropathy?
SNRIs and Tricyclics
Duloxetine does what to P450s?
Duloxetine has greater potential than venlafaxine because it inhibits CYP2D6 and may increase drug levels of TCAs, Type 1C antiarrhythmics (e.g., flecainide) and phenothiazines.

INHIBITS METABOLISM

SNRI
which Serotonin and norephinephrine Reuptake Inhibitors (SNRI) should you avoid in pts with liver disease?
Duloxetine – liver damage (rare) – avoid in pts with chronic liver disease or hepatic insufficiency
duloxetine is what kind of drug?
Serotonin and norephinephrine Reuptake Inhibitors (SNRI)
Venlafaxine is what kind of drug?
Serotonin and norephinephrine Reuptake Inhibitors (SNRI)
PMDD, generalized anxiety disorder, panic, OCD, and bulimia can all be treated with what? (general)
SSRIs
What must you be careful of when you give an SSRI with SNRIs,TCA, MAOI, meperidine, or triptans?

****TEST
SEROTONIN SYNDROME

mental status changes,(confusion to coma) disorders of motor activity (hyperreflexia, rigidity), and autonomic disturbances ( hyperthermia, hypertens.) All SSRIs)

more dangerous with fluvoxetine (long acting)
What is the major side effect of SSRIs? why

****TEST
Nausea, headache, diarrhea are most common (stimulation of 5HT3 receptors)

note: another reason people may discontinue the drug is due to sexual dysfunction
Please compare the difference in typical and atypical's tendency to bind to different receptors
“Atypical” antipsychotics have different receptor binding profiles, but they all tend to be less potent at D2 receptors than chlorpromazine or halperidol and more potent at 5HT2 and D4 receptors.
how does blocking D2 receptors help with Schizophrenia?
alleviates positive symptoms of schizophrenia (tension, hostility, hyperactivity, combativeness, hallucinations, delusions, insomnia, and anorexia.
Betaseron is what kind of drug?
interferon beta-1b

Mechanism of action – uncertain but beneficial effects may be due to immunomodulatory actions. One thought is that INFs decrease antigen presentation in the CNS, which appears to limit immune attack on myelin
which is more likely to make you want to eat more, typical or atypical antipsychotics?
Atypical
how do anti-psychotic drugs affect the ANS?
peripheral cholinergic block, a-adrenergic block
most important side effects of anti-psychotic drugs
– CV, endocrine, central and autonomic are most important. Other dangerous effects include seizures, agranulocytosis and pigmentary degeneration of retina
Halloperidol is more likely to cause what serious side effect? is this a typical or atypical anti-psychotic?

**
Tardive dyskinesia

Typical
how does blocking D4 and 5HT2 receptors help with SZ?
Blocking D4 and 5HT2 receptors may alleviate negative symptoms (apathy, withdrawal, unresponsiveness).
what 2 antipsychotic drugs cause QT prolongation
thioridazine, ziprasidone
clozapine is likely to cause what side effect?
Seizures
what 2 anti-psychotic drugs are likely to cause weight gain?
clozapine and olanzapine
Most serious adverse affect of Clozapine? When would you use it?
mild leukocytosis, leukopenia, and eosinophilia. Bone marrow suppression or agranulocytosis with clozapine

Last resort drug

anti-psychotic
Phenothiazines is also known as?
Chlorpromazine

or

Fluphenazine
in elderly pts on risperidone what do you have to worry about?

what is another drug that can cause this?
STROKE

Olanzapine

(these are anti-psychotics)
Butyrophenone is what drug?
Haloperidol
2 side effects of Atypicals in general?
weight gain and diabetes
what type of treatment will lead to low number of re-hospitalizations of schizophrenic patients?
Drug therapy + social therapy
Chlorpromazine (Thorazine)
Thioridazine (Mellaril)
Fluphenazine (Prolixin)
Perphenazine (Trilafon)
Thiothixene (Navane)
Haloperidol (Haldol)
Loxapine (Loxitane)
Typical Anti-psychotics
Clozapine (Clozaril)
Risperidone (Risperdal) Paliperidone (Invega)
Olanzapine (Zyprexa)
Quetiapine (Seroquel)
Ziprasidone (Geodon)
Aripiprazole (Abilify)
Asenapine (Safris)
Iloperidone (Fanapt)
Atypical Anti-psychotics
All agents improve positive symptoms, which improve negative sx?
Atypicals
Avonex and Rebif are is what kind of drugs?
interferon beta-1a

Mechanism of action – uncertain but beneficial effects may be due to immunomodulatory actions. One thought is that INFs decrease antigen presentation in the CNS, which appears to limit immune attack on myelin
reteplase (r-PA)
Retavase
fibrinolytic - direct plasminogen activator
is altered t-PA for duration
fibrin specific
inactivated by PAI-1
use of the interferon drugs?
Used to decrease the frequency and severity of exacerbations in patients with relapsing forms of MS. Uncertain whether they actually slow the progression of the disease.
Mitoxantrone is what kind of drug? MOA?

what is important about this drug?
a cancer chemotherapeutic agent recently approved to treat advanced MS

Acts by suppressing immune attack on myelin

important thing to remember: THERE IS A TIME LIMIT TO HOW LONG YOU CAN USE IT (2-3 year)--> will cause problems with cardiac conduction
Natalizumab is what kind of drug? How does it work?
A recombinant humanized MAB

Binds to a specific site on an adhesion molecule on activated lymphocytes and monocytes. This blocks adhesion and prevents leukocyte entry into CNS, therby decreasing immune attack on myelin.
major side effect of Natalizumab?
Progressive multifocal leukoencephalopathy-->FATAL

Binds to a specific site on an adhesion molecule on activated lymphocytes and monocytes. This blocks adhesion and prevents leukocyte entry into CNS, therby decreasing immune attack on myelin.
Fingolimod MOA?

important thing about it?
Mechanism – inhibits migration of T cells out of lymph nodes

The first oral specific therapy for MS!!
MOA of Levodopa?
immediate precursor of dopamine which will cross the blood brain barrier

conversion of l-dopa to dopamine in CNS, thus increasing dopamine in the basal ganglia
adverse effects of Fingolimod
bradycardia and increased infections

Mechanism – inhibits migration of T cells out of lymph nodes
Riluzole major adverse effect?
Hepatotox

MOA: voltage-gated sodium channel blocker which is thought to act by inhibiting glutamate release
What is Riluzole used for? MOA?
is the first (and, so far, only) drug approved for the specific treatment of ALS

voltage-gated sodium channel blocker which is thought to act by inhibiting glutamate release
Galantamine is what kind of drug?

Used to tx?
Acetylcholinesterase inhibitors

Drug treatment Alzheimer’s Disease

– the cognitive deficits of the disease are thought to be related, in part, to degeneration of cholinergic neurons in the cortex and hippocampus resulting in deficient cholinergic neurotransmission.
Donepezil is what kind of drug?

Used to tx?
Acetylcholinesterase inhibitors

Drug treatment Alzheimer’s Disease

– the cognitive deficits of the disease are thought to be related, in part, to degeneration of cholinergic neurons in the cortex and hippocampus resulting in deficient cholinergic neurotransmission.
Alteplase (t-PA)
Cathlo Activase, Activase
Fibrinolytics - direct plasminogen activator
fibrin specific
inactivated by PAI-1
bleed risk only at high doses
Memantine is what kind of drug?

used to tx?
"use-dependent” (only kicks in when there are high lvls of activity) NMDA receptor antagonist. The theory behind its action in Alzheimer’s is that it blocks glutaminergic overstimulation of NMDA receptors, which can be toxic to neurons which are important in learning and memory, but allows low levels of receptor activation.

Drug treatment Alzheimer’s Disease
what effect might gingko have on AZ? Estrogen?
Gingko has been shown to modestly improve memory in some Alzheimer’s patients.

Estrogen may increase risk.
what kind of drug is Glatiramer? used to treat? MOA
a synthetic compound which resembles a component of myelin

Mechanism of action
Since drug resembles a component of myelin, it’s thought that it may protect myelin by acting as a “decoy” attracting immune cells away from myelin

tx: MS
what MS drug has the adverse effects of Flushing, chest tightness or pain, shortness of breath (within 15 min)
Glatiramer
Tacrine is what kind of drug?

Used to tx?
Acetylcholinesterase inhibitors

Drug treatment Alzheimer’s Disease

– the cognitive deficits of the disease are thought to be related, in part, to degeneration of cholinergic neurons in the cortex and hippocampus resulting in deficient cholinergic neurotransmission.
what limits the use of levodopa?
need to have dopamine neurons to convert L dopa to dopamine

over time these neurons are dying
what are the changes you will see in pts who take in levodopa?
Bradykinesia and rigidity are reversed quickly; reversal of tremor requires continued therapy

Changes in mood associated with PD are reversed; patients more alert and interested in environment. Dementia may not reverse
what are the cardiovascular and endocrine effects of L-dopa

(not sure how important this is but here you go anyways)
Cardiovascular
Asymptomatic (usually) orthostatic hypotension
Dopamine stimulates both alpha and beta receptors
Cardiac stimulation

Endocrine
Dopamine important in regulation of anterior pituitary function
Prolactin secretion inhibited
Little change in growth hormone secretion
what are the short and long term side effects of L-Dopa?
Early side effects
Dose dependent; tolerance may develop
GI – nausea, vomiting (80%)
CVS – orthostatic hypotension (30%), cardiac arrhythmias

Long term effects – severity correlates with the degree of clinical improvement, duration of therapy and dose. No tolerance develops.

***Abnormal involuntary movements (dyskinesia)*** (80% after 1 year). Reduction in dosage required

Psychiatric and behavioral disturbances (15%)

***“On-Off” syndrome – oscillations in performance involving rapid changes from akinesia to dyskinesia (different from “end of dose”). **
antipsychotic drugs have what effect on L-dopa?
Typical” antipsychotic drugs are dopaminergic antagonists and thus counteract the effects of dopa
MAOi interact how with L-dopa?
MAO inhibitors increase the effects of dopa, may lead to hypertensive crises

note: Tricyclic antidepressants – may aggravate hypotensive symptoms
Carbidopa MOA?
prevents PERIPHERAL conversion of Dopa to Dopamine

allows more L-dopa to get to the brain to be converted

allows for lower dose of L-dopa to be used

decreases peripheral side effects of L-dopa (less CV, NV)
what are the side effects of Carbidopa?
remember, it decreases L-dopa side effect peripherally BUT

increased central side effects of dopa
Early development of long term side effects possible
Activation of COMT pathway
--so in a way it may progress the disease more quickly
What kind of drug is Tolcapone?
COMT inhibitor

thereby increasing the duration of action of l-dopa and dopamine (COMT inactivates l-dopa and dopamine).

Works in CNS and periphery
MOA of Levodopa?
immediate precursor of dopamine which will cross the blood brain barrier

conversion of l-dopa to dopamine in CNS, thus increasing dopamine in the basal ganglia
major tox of tolcapone? is this a problem in entacapone?
Hepatotox

NO
Ropinirole is what kind of drug?
non-ergot DA agonists

Direct stimulation of DA receptors in striatum
pramipexole is what kind of drug?
non-ergot DA agonists

Direct stimulation of DA receptors in striatum
what is one of the few drugs that may slow the progression of Parkinsons?
pramipexole
impulse control problems is a side effect of what drugs?
Non-ergot DA agonists

pramipexole/Ropinirole
what is a unique side effect of pramipexole?
Pramipexole can cause sudden onset of sleep with no warning

MOA:Non-ergot DA agonist
Use for Apomorphine?
non-ergot DA agonist recently approved for rescue treatment of PD patients with “off” episodes (freezing).

Adverse effects include vomiting, orthostatic hypotension and syncope.
How is Amantadine used in Parkinson's?
An antiviral agent found to be effective against PD
.
Apparently acts by increasing dopamine release from intact dopaminergic neurons. Also blocks NMDA receptors.

Is effective quickly but for short time (6-8 weeks). Also used to control dyskinesias occurring with l-dopa therapy late in progression of disease.
Hallucinations, confusion, and nightmares
Insomnia, dizziness, lethergy, slurred speech
Long term use may result in livido reticularis (net like discoloration of the skin)

are adverse rxns of what drug?
Amantadine

Apparently acts by increasing dopamine release from intact dopaminergic neurons. Also blocks NMDA receptors.
Trihexyphenidyl is what kind of drug? how does it help with PD?
Anticholinergic drugs - used as adjunct to l-dopa therapy

Decrease tremor
Little effect on rigidity and bradykinesia
Generally have little peripheral effect, but may reduce some autonomic symptoms
benztropine is what kind of drug? how does it help with PD?
Anticholinergic drugs - used as adjunct to l-dopa therapy

Decrease tremor
Little effect on rigidity and bradykinesia
Generally have little peripheral effect, but may reduce some autonomic symptoms
adverse effects of anticholinergic drugs used for PD?
CNS – confusion, delerium, somnolence, hallucinations (try to avoid them in old people)

Peripheral – may produce cycloplegia, constipation, and urinary retention in certain patients
MOA of Selegiline?
MAO B Inhibitors

MAO B metabolizes dopamine--prevents this
MOA of Rasagiline?
MAO B Inhibitors

MAO B metabolizes dopamine--prevents this
Pt has Parkinson's, what should you use first? second? third?
Selagilline for neuroprotection

Dopamine Agonists

Levodopa

COMT inhibitor

All others
Tetrabenazine is used to treat what?
approved for treatment of chorea associated with Huntington's Dz
What kind of drug is Entacapone?
COMT inhibitor

thereby increasing the duration of action of l-dopa and dopamine (COMT inactivates l-dopa and dopamine).

only works in the periphery
what limits the use of levodopa?
need to have dopamine neurons to convert L dopa to dopamine

over time these neurons are dying
what are the changes you will see in pts who take in levodopa?
Bradykinesia and rigidity are reversed quickly; reversal of tremor requires continued therapy

Changes in mood associated with PD are reversed; patients more alert and interested in environment. Dementia may not reverse
what are the cardiovascular and endocrine effects of L-dopa

(not sure how important this is but here you go anyways)
Cardiovascular
Asymptomatic (usually) orthostatic hypotension
Dopamine stimulates both alpha and beta receptors
Cardiac stimulation

Endocrine
Dopamine important in regulation of anterior pituitary function
Prolactin secretion inhibited
Little change in growth hormone secretion
what are the short and long term side effects of L-Dopa?
Early side effects
Dose dependent; tolerance may develop
GI – nausea, vomiting (80%)
CVS – orthostatic hypotension (30%), cardiac arrhythmias

Long term effects – severity correlates with the degree of clinical improvement, duration of therapy and dose. No tolerance develops.

***Abnormal involuntary movements (dyskinesia)*** (80% after 1 year). Reduction in dosage required

Psychiatric and behavioral disturbances (15%)

***“On-Off” syndrome – oscillations in performance involving rapid changes from akinesia to dyskinesia (different from “end of dose”). **
antipsychotic drugs have what effect on L-dopa?
Typical” antipsychotic drugs are dopaminergic antagonists and thus counteract the effects of dopa
MAOi interact how with L-dopa?
MAO inhibitors increase the effects of dopa, may lead to hypertensive crises

note: Tricyclic antidepressants – may aggravate hypotensive symptoms
Carbidopa MOA?
prevents PERIPHERAL conversion of Dopa to Dopamine

allows more L-dopa to get to the brain to be converted

allows for lower dose of L-dopa to be used

decreases peripheral side effects of L-dopa (less CV, NV)
what are the side effects of Carbidopa?
remember, it decreases L-dopa side effect peripherally BUT

increased central side effects of dopa
Early development of long term side effects possible
Activation of COMT pathway
--so in a way it may progress the disease more quickly
What kind of drug is Tolcapone?
COMT inhibitor

thereby increasing the duration of action of l-dopa and dopamine (COMT inactivates l-dopa and dopamine).

Works in CNS and periphery
What kind of drug is Entacapone?
COMT inhibitor

thereby increasing the duration of action of l-dopa and dopamine (COMT inactivates l-dopa and dopamine).

only works in the periphery
major tox of tolcapone? is this a problem in entacapone?
Hepatotox

NO
Ropinirole is what kind of drug?
non-ergot DA agonists

Direct stimulation of DA receptors in striatum
pramipexole is what kind of drug?
non-ergot DA agonists

Direct stimulation of DA receptors in striatum
what is one of the few drugs that may slow the progression of Parkinsons?
pramipexole
impulse control problems is a side effect of what drugs?
Non-ergot DA agonists

pramipexole/Ropinirole
what is a unique side effect of pramipexole?
Pramipexole can cause sudden onset of sleep with no warning

MOA:Non-ergot DA agonist
Use for Apomorphine?
non-ergot DA agonist recently approved for rescue treatment of PD patients with “off” episodes (freezing).

Adverse effects include vomiting, orthostatic hypotension and syncope.
Tetrabenazine is used to treat what?
approved for treatment of chorea associated with Huntington's Dz
Pt has Parkinson's, what should you use first? second? third?
Selagilline for neuroprotection

Dopamine Agonists

Levodopa

COMT inhibitor

All others
MOA of Rasagiline?
MAO B Inhibitors

MAO B metabolizes dopamine--prevents this
MOA of Selegiline?
MAO B Inhibitors

MAO B metabolizes dopamine--prevents this
adverse effects of anticholinergic drugs used for PD?
CNS – confusion, delerium, somnolence, hallucinations (try to avoid them in old people)

Peripheral – may produce cycloplegia, constipation, and urinary retention in certain patients
benztropine is what kind of drug? how does it help with PD?
Anticholinergic drugs - used as adjunct to l-dopa therapy

Decrease tremor
Little effect on rigidity and bradykinesia
Generally have little peripheral effect, but may reduce some autonomic symptoms
Trihexyphenidyl is what kind of drug? how does it help with PD?
Anticholinergic drugs - used as adjunct to l-dopa therapy

Decrease tremor
Little effect on rigidity and bradykinesia
Generally have little peripheral effect, but may reduce some autonomic symptoms
Hallucinations, confusion, and nightmares
Insomnia, dizziness, lethergy, slurred speech
Long term use may result in livido reticularis (net like discoloration of the skin)

are adverse rxns of what drug?
Amantadine

Apparently acts by increasing dopamine release from intact dopaminergic neurons. Also blocks NMDA receptors.
How is Amantadine used in Parkinson's?
An antiviral agent found to be effective against PD
.
Apparently acts by increasing dopamine release from intact dopaminergic neurons. Also blocks NMDA receptors.

Is effective quickly but for short time (6-8 weeks). Also used to control dyskinesias occurring with l-dopa therapy late in progression of disease.
MOA of Levodopa?
immediate precursor of dopamine which will cross the blood brain barrier

conversion of l-dopa to dopamine in CNS, thus increasing dopamine in the basal ganglia
what limits the use of levodopa?
need to have dopamine neurons to convert L dopa to dopamine

over time these neurons are dying
Tetrabenazine is used to treat what?
approved for treatment of chorea associated with Huntington's Dz
what are the changes you will see in pts who take in levodopa?
Bradykinesia and rigidity are reversed quickly; reversal of tremor requires continued therapy

Changes in mood associated with PD are reversed; patients more alert and interested in environment. Dementia may not reverse
Pt has Parkinson's, what should you use first? second? third?
Selagilline for neuroprotection

Dopamine Agonists

Levodopa

COMT inhibitor

All others
what are the cardiovascular and endocrine effects of L-dopa

(not sure how important this is but here you go anyways)
Cardiovascular
Asymptomatic (usually) orthostatic hypotension
Dopamine stimulates both alpha and beta receptors
Cardiac stimulation

Endocrine
Dopamine important in regulation of anterior pituitary function
Prolactin secretion inhibited
Little change in growth hormone secretion
MOA of Rasagiline?
MAO B Inhibitors

MAO B metabolizes dopamine--prevents this
what are the short and long term side effects of L-Dopa?
Early side effects
Dose dependent; tolerance may develop
GI – nausea, vomiting (80%)
CVS – orthostatic hypotension (30%), cardiac arrhythmias

Long term effects – severity correlates with the degree of clinical improvement, duration of therapy and dose. No tolerance develops.

***Abnormal involuntary movements (dyskinesia)*** (80% after 1 year). Reduction in dosage required

Psychiatric and behavioral disturbances (15%)

***“On-Off” syndrome – oscillations in performance involving rapid changes from akinesia to dyskinesia (different from “end of dose”). **
MOA of Selegiline?
MAO B Inhibitors

MAO B metabolizes dopamine--prevents this
adverse effects of anticholinergic drugs used for PD?
CNS – confusion, delerium, somnolence, hallucinations (try to avoid them in old people)

Peripheral – may produce cycloplegia, constipation, and urinary retention in certain patients
benztropine is what kind of drug? how does it help with PD?
Anticholinergic drugs - used as adjunct to l-dopa therapy

Decrease tremor
Little effect on rigidity and bradykinesia
Generally have little peripheral effect, but may reduce some autonomic symptoms
Trihexyphenidyl is what kind of drug? how does it help with PD?
Anticholinergic drugs - used as adjunct to l-dopa therapy

Decrease tremor
Little effect on rigidity and bradykinesia
Generally have little peripheral effect, but may reduce some autonomic symptoms
Hallucinations, confusion, and nightmares
Insomnia, dizziness, lethergy, slurred speech
Long term use may result in livido reticularis (net like discoloration of the skin)

are adverse rxns of what drug?
Amantadine

Apparently acts by increasing dopamine release from intact dopaminergic neurons. Also blocks NMDA receptors.
How is Amantadine used in Parkinson's?
An antiviral agent found to be effective against PD
.
Apparently acts by increasing dopamine release from intact dopaminergic neurons. Also blocks NMDA receptors.

Is effective quickly but for short time (6-8 weeks). Also used to control dyskinesias occurring with l-dopa therapy late in progression of disease.
Use for Apomorphine?
non-ergot DA agonist recently approved for rescue treatment of PD patients with “off” episodes (freezing).

Adverse effects include vomiting, orthostatic hypotension and syncope.
what is a unique side effect of pramipexole?
Pramipexole can cause sudden onset of sleep with no warning

MOA:Non-ergot DA agonist
what is one of the few drugs that may slow the progression of Parkinsons?
pramipexole
impulse control problems is a side effect of what drugs?
Non-ergot DA agonists

pramipexole/Ropinirole
pramipexole is what kind of drug?
non-ergot DA agonists

Direct stimulation of DA receptors in striatum
[ʒ]
Voiced Palato-Alveolar Fricative
azure [æʒər]
major tox of tolcapone? is this a problem in entacapone?
Hepatotox

NO
What kind of drug is Entacapone?
COMT inhibitor

thereby increasing the duration of action of l-dopa and dopamine (COMT inactivates l-dopa and dopamine).

only works in the periphery
[n]
Voiced Alveolar Nasal
note [nowt]
what are the side effects of Carbidopa?
remember, it decreases L-dopa side effect peripherally BUT

increased central side effects of dopa
Early development of long term side effects possible
Activation of COMT pathway
--so in a way it may progress the disease more quickly
Carbidopa MOA?
prevents PERIPHERAL conversion of Dopa to Dopamine

allows more L-dopa to get to the brain to be converted

allows for lower dose of L-dopa to be used

decreases peripheral side effects of L-dopa (less CV, NV)
MAOi interact how with L-dopa?
MAO inhibitors increase the effects of dopa, may lead to hypertensive crises

note: Tricyclic antidepressants – may aggravate hypotensive symptoms
antipsychotic drugs have what effect on L-dopa?
Typical” antipsychotic drugs are dopaminergic antagonists and thus counteract the effects of dopa
What is a left shift?
Neutrophils are present

(right shift is more lymphocytes than neutrophils)
what is the lymphatic system of the CNS like?
there isn't one!
what will produce a ring enhancement on imaging?
an abscess

has to do with central necrosis and capsule formation
which form of meningitis will have a large number of neutrophils?

what will glucose levels be like in the CSF in this type of infection?
Bacterial

Decrease--using up the glucose
in viral and fungal meningitis what type of cells will be elevated in the CSF
Lymphocytes
which type of meningitis will be self-limited and has a low mortality
Viral
1. Purulent leptomeningeal inflammation, cloudy CSF, vascular engorgement, may extend to cortex

2. Mild leptomeningeal inflammation, brain swelling

which is bacterial and which is viral/
(1-bacterial; 2-viral)
What is a left shift?
Neutrophils are present

(right shift is more lymphocytes than neutrophils)
what are some of the predisposing conditions for focal suppurative infections of the brain (abscess)?
Acute bacterial endocarditis, right to left shunts, chronic pulmonary sepsis
Mastoiditis, paranasal sinusitis, acute otitis, open fracture, previous neurosurgery
what physiologically leads to hydrocephalus? 2
Hydrocephalus can be due to lack of absorption of CSF or due to an obstruction to flow of CSF
what is the therapy of choice for a brain abscess? (reduces mortality to less than 10%)
Surgery and antibiotic therapy can reduce mortality to less than 10%
Hydrocephalus, leptomeningeal fibrosis (may cause cranial nerve impairment) is a result of what type of meningitis?
Bacterial
what are 3 causes of chronic bacterial meningoencephalitis?
Tuberculosis (mycobacterium tuberculosis)

Neurosyphilis (treponema pallidum)

Lyme Disease (borrelia burgdorferi)
pt presents with a headache, maliaise, mental confusion and vomiting. A micro finding is caseating granuloma...
TB
perivascular inflammatory infiltrate of plasma cells and lymphocytes is a micro finding of what?
Neurosyphilis

Chronic Bacterial Meningoencephalitis
what 2 types of pts are likely to get fungal (candida) caused meningitis?

**
HIV and Diabetic pts

immunocompromised
causes of diffuse parenchymal meningoencephalitis? 2

*
Candida, Criptococcus
if you see meningitis associated with vasculitis, what are 2 of your possible causes?

*
Mucor, Aspergillus
if a pt has histoplasmosis in the lung, what is there potential for?

*
abscess in the brain
Chronic memningitis with aids is normally caused by?


**
Toxoplasmosis
necrosis of the temporal lobe is associated with what type of meningoenchepalitis?

how bout Rabies-Negri bodies in hippocampal and Purkinje neurons

and microglial nodules and multinucleated giant cells

*
Herpes-temporal lobe necrosis

Rabies-Negri bodies in hippocampal and Purkinje neurons

HIV-microglial nodules and multinucleated giant cells
what is the lymphatic system of the CNS like?
there isn't one!
what will produce a ring enhancement on imaging?
an abscess

has to do with central necrosis and capsule formation
which form of meningitis will have a large number of neutrophils?

what will glucose levels be like in the CSF in this type of infection?
Bacterial

Decrease--using up the glucose
moa of the -triptan drugs?
5HT1 Receptor Agonists

Mechanism and pharmacological effects – 5HT1D/5HT1B receptor agonists which constrict large cranial blood vessels, decrease inflammation around sensory nerves, and inhibit trigeminal neuronal discharge. These actions relieve pain and other symptoms of migraine
which type of meningitis will be self-limited and has a low mortality
Viral
1. Purulent leptomeningeal inflammation, cloudy CSF, vascular engorgement, may extend to cortex

2. Mild leptomeningeal inflammation, brain swelling

which is bacterial and which is viral/
(1-bacterial; 2-viral)
in viral and fungal meningitis what type of cells will be elevated in the CSF
Lymphocytes
what are some of the predisposing conditions for focal suppurative infections of the brain (abscess)?
Acute bacterial endocarditis, right to left shunts, chronic pulmonary sepsis
Mastoiditis, paranasal sinusitis, acute otitis, open fracture, previous neurosurgery
Hydrocephalus, leptomeningeal fibrosis (may cause cranial nerve impairment) is a result of what type of meningitis?
Bacterial
what physiologically leads to hydrocephalus? 2
Hydrocephalus can be due to lack of absorption of CSF or due to an obstruction to flow of CSF
what are 3 causes of chronic bacterial meningoencephalitis?
Tuberculosis (mycobacterium tuberculosis)

Neurosyphilis (treponema pallidum)

Lyme Disease (borrelia burgdorferi)
pt presents with a headache, maliaise, mental confusion and vomiting. A micro finding is caseating granuloma...
TB
perivascular inflammatory infiltrate of plasma cells and lymphocytes is a micro finding of what?
Neurosyphilis

Chronic Bacterial Meningoencephalitis
causes of diffuse parenchymal meningoencephalitis? 2

*
Candida, Criptococcus
if you see meningitis associated with vasculitis, what are 2 of your possible causes?

*
Mucor, Aspergillus
Chronic memningitis with aids is normally caused by?


**
Toxoplasmosis
what 2 types of pts are likely to get fungal (candida) caused meningitis?

**
HIV and Diabetic pts

immunocompromised
if a pt has histoplasmosis in the lung, what is there potential for?

*
abscess in the brain
necrosis of the temporal lobe is associated with what type of meningoenchepalitis?

how bout Rabies-Negri bodies in hippocampal and Purkinje neurons

and microglial nodules and multinucleated giant cells

*
Herpes-temporal lobe necrosis

Rabies-Negri bodies in hippocampal and Purkinje neurons

HIV-microglial nodules and multinucleated giant cells
what is the therapy of choice for a brain abscess? (reduces mortality to less than 10%)
Surgery and antibiotic therapy can reduce mortality to less than 10%
what are the long acting -triptan drugs?
frovatriptan

naratriptan

-good for outlasting long headaches

note: these also take longer to start working
most common side effect of the triptans? Who are they contraindicated in?
Nausea and vomiting most common after oral administration.

Pain and redness at injection site plus tingling, dizziness, burning sensations,

tightness and pressure in the chest. Contraindicated in patients with coronary artery disease and angina. (will hit 5HT2 receptors on coronary vasculature)
why should triptans be avoided in pts on SSRIs?
triptains affect serotonin

Possibility of serotonin syndrome in patients also taking SSRIs
what is the MOA of Ergotamine and Dihydroergotamine?

how does this help with migranes?

how is it different from the triptans
Ergotamine binds to all subtypes of 5HT1, and 5HT2 receptors as well as adrenergic and dopaminergic receptors. Agonist activity at 5HT1B and/or 5HT1D receptors on cerebral blood vessels probably responsible for anti-migraine activity.

SIDE EFFECTS are really bad-Nausea vomiting, muscle weakness, numbness/tingling in fingers and toes, allergic rxn, chest pain

note: caffeine can help with absorption
What is the major drug interaction of benzodiapepines?
Potentiation of CNS depressant effects of antipsychotics, tricyclic antidepressants, sedative hypnotics, alcohol
which b-blockers can be effective for tx of migraine?
propranolol

timolol
what antiepileptic drug can be good to treat migraine?
Valproate
Amitriptyline can be used to treat?
Migraine
SSRI is a good treatment for what type of headache?
menstrual migraine
Cafergot plus CYP3A4 inhibitor (macrolides, protease inhibitors) may do what?
fatal ischemia

cafergot--Ergotamine+Caffeine
pt has:
Occasional throbbing headaches No major impairment of functioning

stage of migraine?

Therapies?
stage of migraine: Mild

Therapies:
Mild analgesics (NSAID) Combination analgesics Antiemetics, depending on severity
pt has:
Moderate or severe headaches Some impairment of functioning Nausea common


stage of migraine?

Therapies?
stage of migraine: Moderate

Therapies:
Combination analgesics Triptan or Ergot alkaloid Antiemetics
pt has:
More than 3 severe headaches a month Significant functional impairment Marked nausea and/or vomiting


stage of migraine?

Therapies?
stage of migraine: Severe

Therapies:
Triptan or Ergot alkaloid (+/- NSAIDS) Prophylactic medications Antiemetics
first line treatment for acute cluster headache?
Fast acting triptan (nasal or SC) or oxygen are the first choices
how can montelukast be used to tx migraine?
decrease frequency by decreasing inflammation
prevention tx for verapamil?
Verapamil is drug of choice; melatonin may be effective
Felbamate (Felbatol) – was approved for use in....? What is a stipulation of its use?
adults with partial seizures and in children with seizures associated with Lennox-Gastaut Syndrome

can only be gotten for pts who this is the ONLY DRUG THAT WILL WORK...people have to sign a waiver to use it because it causes high incidence of aplastic anemia, and liver failure
MOA of Vigabatrin?

side effects?

use?
Inhibits GABA degradation (GABA transaminase) and reuptake

Adverse rxns – **vision loss in 30% of pts**. Also headache, somnolence, fatigue, dizziness.
Used as last resort for infantile spasms.

Also for refractory complex partial seizures
MOA of Rufinamide (Benzel)?

tox?

use?
Action on sodium channels

Adverse rxns – somnolence, vomiting

Used to treat Lennox-Gastaut syndrome
lamotrigene use?

MOA?

**
Adjunct drugs for partial seizures
approved add-on drug

acts by slowing recovery in Na+ channels, inhibiting voltage gated Ca channels, and perhaps by inhibiting glutamate release

also:probably block L-type calcium channels to some extent
gabapentin use?
Adjunct drugs for partial seizures
approved add-on drug

act at a subunit of voltage-gated calcium channels to decrease depolarization-induced calcium influx, which decreases release of excitatory neurotransmitters (glutamate).

also:probably block L-type calcium channels to some extent
topiramate use?
Adjunct drugs for partial seizures; approved add-on drug; treats TREMORS and prevents migraines

mechanism may involve block of sodium channels and AMPA/Kainate receptors. Other possible uses include treatment of tremors and prevention of migraine.
Gabapentin use?
effective in treatment of neuropathic pain and fibromyalgia
pregabalin use?
effective in treatment of neuropathic pain and fibromyalgia
Side effects of Lamotrigene? what was the use of this again?
Most common – weakness, diplopia, ataxia, nausea, irritability, life-threatening rash, Stevens-Johnson syndrome. Also inhibits carbarmazepine metabolism. Increased risk of cleft lip/palate(?)

Adjunct drugs for partial seizures
approved add-on drug

act at a subunit of voltage-gated calcium channels to decrease depolarization-induced calcium influx, which decreases release of excitatory neurotransmitters (glutamate).
Common side effects of Topiramate?
. Slight increase in incidence of kidney stones
What is the major drug interaction of benzodiapepines?
Potentiation of CNS depressant effects of antipsychotics, tricyclic antidepressants, sedative hypnotics, alcohol
Zonisamide should be avoided in what kind of pts?
Avoid in patients allergic to sulfonamides
What is status epilepticus?

some causes?
A state of continuous seizure activity without recovery of consciousness. Generalized tonic-clonic type is life-threatening (10% fatal)

Some causes – sudden withdrawal of CNS depressants or antiepileptic agents, or drug poisoning
drug therapy tx for status epilepticus?
IV diazepam or lorazepam followed by maintenance therapy of slow IV infusion of phenytoin (fosphenytoin) or phenobarbital
What are the Drugs of Choice for a Primary Generalized Tonic-Clonic (Grand Mal)

**
Valproate
Carbamazepine
Phenytoin
What are the Drugs of Choice for Partial Including Secondarily Generalized seizures?
Carbamazepine
Penytoin
Valproate
What are the Drugs of Choice for Absence (Petit Mal) seizures?
Ethosuximide
Valproate
What are the Drugs of Choice for Atypical Absence, Myoclonic, Atonic seizures?
Valproate
important side effect of Tiagabine?

***
Off-label use in patients without epilepsy may cause seizures.
what is always found as a symptom of epilepsy? if epilepsy is untreated what 2 things might this lead to?
Abnormal and excessive EEG discharges (always) – if untreated, may lead to anoxic brain damage. Also danger of “excitotoxicity”. (will just keep happening over and over)
Spread of epileptic activity to affect both hemispheres
Loss of consciousness
Motor pattern similar to “generalized seizures”

type of seizure?
Partial seizures secondarily generalized

will start as a localized partial seizure, then it will become a generalized tonic clonic
Consciousness preserved
Localized EEG spikes
Various manifestations of motor or sensory disturbances related to specific cortical area affected

type of seizure?
simple partial
Disturbed consciousness
Focal EEG spikes originate from temporal lobe
Confused behavior, automatisms, sensory, emotional distortions

type of seizure?
Complex partial
Major convulsion typified by tonic contraction, conic jerking, and a prolonged post-seizure (postictal) depression of CNS activity
Loss of consciousness
Generalized high voltage EEG spikes

type of seizure?
Generalized seizures
Convulsive type:

Tonic-clonic (Grand Mal)
this is a disease in which you have mental retardation, and a mirage of seizure types (tonic clonic, absence, etc)
Lennox-Gastaut Syndrome
What is status epilepticus?
repeated seizures

one seizure leads to another

medical emergency
3 mechanisms of antieplieptic drugs?
Limit sustained repetitive firing of neurons by slowing rate of recovery in Na+ - channels

Enhancing GABA-mediated synaptic inhibition

Limit activation of CA++ - channels
this drug Prevents seizures without producing general CNS depression, by reducing the rate at which Na channels recover from inactivation, thus slowing firing
Phenytoin
Brief, abrupt loss of consciousness
With or without clonic motor activity
Generalized 3 per second spike and wave EEG activity

type of seizure?
Generalized
Nonconvulsive type

Absence
MOA of phenytoin?
reduce rate at which Na+ channels recover from inactivation, thereby slowing firing.

note: Prevent seizures without producing general CNS depression
describe how phenytoin is metabolized.

Also, how is 90% of it found in the body? what does this imply?
98% metabolized by liver microsomal enzymes; zero order (no matter the dose it can only be cleared at a certain rate) elimination kinetic at therapeutic concentration (1st order at low doses)

90% plasma protein bound--if displaced you will have loads of side effects
what is an important tox of Phenytoin assoc with administration of the drug?
Acute toxicity after rapid IV administration – cardiovascular collapse and/or CNS depression

note: Fosphenytoin (a soluble pro-drug of phenytoin) decreases, but does not eliminate, this problem.
chronic side effect of Phenytoin? (2 key)
behavioral changes,
***gingival hyperplasia, peripheral neuropathy,
osteomalacia***
GI-disturbances,
megaloblastic anemia (decreased vitamin B12), hirsutism, depression of serum folate and vitamin K level.
What drug class is Phenytoin?
D

It can be used if you have no choice, but it is teratogenic
what happens with sudden withdrawal of Phenytoin
STATUS EPILEPTICUS

causes tons of seizures!!

make sure if you need to change the drug that you SLOWLY take them off phenytoin
Chloramphenicol, dicumerol, cimetidine, and sulfonamides have what effect on Phenytoin?
Inhibit its metabolism

What were those drugs again? There were 4 of them..yeah slack ass, flip it over for the answer, and if you didn't remember, mark that shit wrong.
what effect will Phenytoin have on the CYP system?
Induces hepatic drug metabolizing enzymes (CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP3A4)
Dicumerol, salicylates, BZDs have what effect on phenytoin?
displace from protein binding sites to increase plasma level
Acutely, what effect do phenobarbital and ethanol have on phenytoin?

Chronically?
compete for the same metabolic sites leading to an INCREASE in plasma phenytoin (inhibit metabolism)

Chronically, phenobarbital is a classic inducer of drug metabolizing enzymes. So chronically, both phenobarbital and ethanol increase metabolism of Phenytoin
what effect does phenytoin have on OCP? why is this especially important
it will decrease their effectiveness

this is important because it is also a teratogen, so if you get pregnant while on it, you are harming the baby
therapeutic use of Phenytoin?
Effective for all types of seizures except absence
Tonic-clonic-seizures in adults or older children
Used in status epilepticus
Wound healing
what effect does Carbamazepine have on phenytoin
enhances phenytoin metabolism, decreasing plasma levels
what effect does phenobarbital have on drug metabolizing enzymes?
induces drug metabolizing enzymes
what 2 pts (think demographics) would you want to avoid phenobarbital in? why?
hyperactivity (children)

confusion in elderly patients, decreased cognitive ability
what drug can Precipitate acute intermittent porphyria
Phenobarbital

penetrate the brain and cause death of neurons
what category drug is phenobarbital?
Potentially teratogenic (Pregnancy Category D)
MOA of Phenobarbital?
Potentiates GABA inhibitory transmission and decreases glutamate excitation

note: can cause sedation, but can be used at low doses so its not a big deal
MOA of Primidone
Structurally similar to phenobarbital
Also has similar pharmacology but less potent-prevents Na channel recovery-->inhibition
aplastic anemia, agranulocytosis can be a side effect of what anti-seizure drug?
Carbamazepine
if you are Asian, and you need to go on Carbamazepine, what should you have done? why?
FDA recommends genetic testing before starting therapy in patients of Asian descent.

Hypersensitivity reactions (SJS) in Asians with HLA-B* 1502 gene
Oxcarbazepine works like Carbamazepine (how was this agian?)...what is different about it?
slows recovery of voltage-sensitive Na+ channels (also like phenytoin)

less likely to cause SJS and agranulocytosis
Carbamazepine MOA?
Anticonvulsant effects resemble phenytoin (slows recovery of voltage-sensitive Na+ channels).

note: Structurally related to tricyclic antidepressants--so it takes a while to start working
what anti-seizure drug can cause Parkinsonism?
Ethosuximide
MOA of Valproic acid?
acts via several mechanisms. Blocks recovery in Na+ channels, hyperpolarizes cell via an action on K+ channels and perhaps an action on GABA systems (inhibition)
Valproic acid is used for what kind of seizures?
approved for absence and complex partial seizures in U.S. May also be useful in treatment of grand mal, myoclonic and febrile seizures. Also used in patients who don’t respond to any drug, regardless of seizure type

pretty much all of them!
It is the Amiodarone of seizure drugs
biggest potential tox assoc with Valproic Acid?

which population is this risk highest in?
Hepatotoxicity

especially in kids under 2
Ethosuximide MOA?
Increases neuronal refractory period by decreasing conductance in Ca++ channels (T current).

T currents in thalamic neurons important in generation of absence seizures
why are Benzodiazepines typically not used for tx of seizures?

MOA
tolerance builds up quick

Suppression of seizure spread in the cortex, thalamus, and limbic structures
May facilitate GABA mediated pre- and post-synaptic inhibition
what is the difference btw a bizarre and non-bizarre delusion?
Delusion-erroneous beliefs that usually involve a misinterpretation of perceptions or experiences

Bizarre-clearly implausible, not understandable, and not derived from ordinary life experiences (guy thinks someone took all his organs while he slept even though he has no scars)

Non bizarre- involve situations that can conceivably occur in real life
Felbamate (Felbatol) – was approved for use in....? What is a stipulation of its use?
adults with partial seizures and in children with seizures associated with Lennox-Gastaut Syndrome

can only be gotten for pts who this is the ONLY DRUG THAT WILL WORK...people have to sign a waiver to use it because it causes high incidence of aplastic anemia, and liver failure
MOA of Vigabatrin?

side effects?

use?
Inhibits GABA degradation (GABA transaminase) and reuptake

Adverse rxns – **vision loss in 30% of pts**. Also headache, somnolence, fatigue, dizziness.
Used as last resort for infantile spasms.

Also for refractory complex partial seizures
MOA of Rufinamide (Benzel)?

tox?

use?
Action on sodium channels

Adverse rxns – somnolence, vomiting

Used to treat Lennox-Gastaut syndrome
lamotrigene use?

MOA?

**
Adjunct drugs for partial seizures
approved add-on drug

acts by slowing recovery in Na+ channels, inhibiting voltage gated Ca channels, and perhaps by inhibiting glutamate release

also:probably block L-type calcium channels to some extent
gabapentin use?
Adjunct drugs for partial seizures
approved add-on drug

act at a subunit of voltage-gated calcium channels to decrease depolarization-induced calcium influx, which decreases release of excitatory neurotransmitters (glutamate).

also:probably block L-type calcium channels to some extent
topiramate use?
Adjunct drugs for partial seizures; approved add-on drug; treats TREMORS and prevents migraines

mechanism may involve block of sodium channels and AMPA/Kainate receptors. Other possible uses include treatment of tremors and prevention of migraine.
Gabapentin use?
effective in treatment of neuropathic pain and fibromyalgia
pregabalin use?
effective in treatment of neuropathic pain and fibromyalgia
Side effects of Lamotrigene? what was the use of this again?
Most common – weakness, diplopia, ataxia, nausea, irritability, life-threatening rash, Stevens-Johnson syndrome. Also inhibits carbarmazepine metabolism. Increased risk of cleft lip/palate(?)

Adjunct drugs for partial seizures
approved add-on drug

act at a subunit of voltage-gated calcium channels to decrease depolarization-induced calcium influx, which decreases release of excitatory neurotransmitters (glutamate).
Common side effects of Topiramate?
. Slight increase in incidence of kidney stones
Zonisamide should be avoided in what kind of pts?
Avoid in patients allergic to sulfonamides
What is status epilepticus?

some causes?
A state of continuous seizure activity without recovery of consciousness. Generalized tonic-clonic type is life-threatening (10% fatal)

Some causes – sudden withdrawal of CNS depressants or antiepileptic agents, or drug poisoning
important side effect of Tiagabine?

***
Off-label use in patients without epilepsy may cause seizures.
drug therapy tx for status epilepticus?
IV diazepam or lorazepam followed by maintenance therapy of slow IV infusion of phenytoin (fosphenytoin) or phenobarbital
What are the Drugs of Choice for Partial Including Secondarily Generalized seizures?
Carbamazepine
Penytoin
Valproate
What are the Drugs of Choice for Atypical Absence, Myoclonic, Atonic seizures?
Valproate
What are the Drugs of Choice for Absence (Petit Mal) seizures?
Ethosuximide
Valproate
What are the Drugs of Choice for a Primary Generalized Tonic-Clonic (Grand Mal)

**
Valproate
Carbamazepine
Phenytoin
what is always found as a symptom of epilepsy? if epilepsy is untreated what 2 things might this lead to?
Abnormal and excessive EEG discharges (always) – if untreated, may lead to anoxic brain damage. Also danger of “excitotoxicity”. (will just keep happening over and over)
Disturbed consciousness
Focal EEG spikes originate from temporal lobe
Confused behavior, automatisms, sensory, emotional distortions

type of seizure?
Complex partial
Consciousness preserved
Localized EEG spikes
Various manifestations of motor or sensory disturbances related to specific cortical area affected

type of seizure?
simple partial
Spread of epileptic activity to affect both hemispheres
Loss of consciousness
Motor pattern similar to “generalized seizures”

type of seizure?
Partial seizures secondarily generalized

will start as a localized partial seizure, then it will become a generalized tonic clonic
Brief, abrupt loss of consciousness
With or without clonic motor activity
Generalized 3 per second spike and wave EEG activity

type of seizure?
Generalized
Nonconvulsive type

Absence
Major convulsion typified by tonic contraction, conic jerking, and a prolonged post-seizure (postictal) depression of CNS activity
Loss of consciousness
Generalized high voltage EEG spikes

type of seizure?
Generalized seizures
Convulsive type:

Tonic-clonic (Grand Mal)
this is a disease in which you have mental retardation, and a mirage of seizure types (tonic clonic, absence, etc)
Lennox-Gastaut Syndrome
What is status epilepticus?
repeated seizures

one seizure leads to another

medical emergency
3 mechanisms of antieplieptic drugs?
Limit sustained repetitive firing of neurons by slowing rate of recovery in Na+ - channels

Enhancing GABA-mediated synaptic inhibition

Limit activation of CA++ - channels
MOA of phenytoin?
reduce rate at which Na+ channels recover from inactivation, thereby slowing firing.

note: Prevent seizures without producing general CNS depression
describe how phenytoin is metabolized.

Also, how is 90% of it found in the body? what does this imply?
98% metabolized by liver microsomal enzymes; zero order (no matter the dose it can only be cleared at a certain rate) elimination kinetic at therapeutic concentration (1st order at low doses)

90% plasma protein bound--if displaced you will have loads of side effects
this drug Prevents seizures without producing general CNS depression, by reducing the rate at which Na channels recover from inactivation, thus slowing firing
Phenytoin
what effect will Phenytoin have on the CYP system?
Induces hepatic drug metabolizing enzymes (CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP3A4)
what is an important tox of Phenytoin assoc with administration of the drug?
Acute toxicity after rapid IV administration – cardiovascular collapse and/or CNS depression

note: Fosphenytoin (a soluble pro-drug of phenytoin) decreases, but does not eliminate, this problem.
chronic side effect of Phenytoin? (2 key)
behavioral changes,
***gingival hyperplasia, peripheral neuropathy,
osteomalacia***
GI-disturbances,
megaloblastic anemia (decreased vitamin B12), hirsutism, depression of serum folate and vitamin K level.
what happens with sudden withdrawal of Phenytoin
STATUS EPILEPTICUS

causes tons of seizures!!

make sure if you need to change the drug that you SLOWLY take them off phenytoin
What drug class is Phenytoin?
D

It can be used if you have no choice, but it is teratogenic
Acutely, what effect do phenobarbital and ethanol have on phenytoin?

Chronically?
compete for the same metabolic sites leading to an INCREASE in plasma phenytoin (inhibit metabolism)

Chronically, phenobarbital is a classic inducer of drug metabolizing enzymes. So chronically, both phenobarbital and ethanol increase metabolism of Phenytoin
Dicumerol, salicylates, BZDs have what effect on phenytoin?
displace from protein binding sites to increase plasma level
Chloramphenicol, dicumerol, cimetidine, and sulfonamides have what effect on Phenytoin?
Inhibit its metabolism

What were those drugs again? There were 4 of them..yeah slack ass, flip it over for the answer, and if you didn't remember, mark that shit wrong.
what effect does Carbamazepine have on phenytoin
enhances phenytoin metabolism, decreasing plasma levels
what effect does phenytoin have on OCP? why is this especially important
it will decrease their effectiveness

this is important because it is also a teratogen, so if you get pregnant while on it, you are harming the baby
why are Benzodiazepines typically not used for tx of seizures?

MOA
tolerance builds up quick

Suppression of seizure spread in the cortex, thalamus, and limbic structures
May facilitate GABA mediated pre- and post-synaptic inhibition
biggest potential tox assoc with Valproic Acid?

which population is this risk highest in?
Hepatotoxicity

especially in kids under 2
Valproic acid is used for what kind of seizures?
approved for absence and complex partial seizures in U.S. May also be useful in treatment of grand mal, myoclonic and febrile seizures. Also used in patients who don’t respond to any drug, regardless of seizure type

pretty much all of them!
It is the Amiodarone of seizure drugs
MOA of Valproic acid?
acts via several mechanisms. Blocks recovery in Na+ channels, hyperpolarizes cell via an action on K+ channels and perhaps an action on GABA systems (inhibition)
what anti-seizure drug can cause Parkinsonism?
Ethosuximide
Ethosuximide MOA?
Increases neuronal refractory period by decreasing conductance in Ca++ channels (T current).

T currents in thalamic neurons important in generation of absence seizures
Oxcarbazepine works like Carbamazepine (how was this agian?)...what is different about it?
slows recovery of voltage-sensitive Na+ channels (also like phenytoin)

less likely to cause SJS and agranulocytosis
if you are Asian, and you need to go on Carbamazepine, what should you have done? why?
FDA recommends genetic testing before starting therapy in patients of Asian descent.

Hypersensitivity reactions (SJS) in Asians with HLA-B* 1502 gene
aplastic anemia, agranulocytosis can be a side effect of what anti-seizure drug?
Carbamazepine
Carbamazepine MOA?
Anticonvulsant effects resemble phenytoin (slows recovery of voltage-sensitive Na+ channels).

note: Structurally related to tricyclic antidepressants--so it takes a while to start working
MOA of Primidone
Structurally similar to phenobarbital
Also has similar pharmacology but less potent-prevents Na channel recovery-->inhibition
what drug can Precipitate acute intermittent porphyria
Phenobarbital

penetrate the brain and cause death of neurons
what category drug is phenobarbital?
Potentially teratogenic (Pregnancy Category D)
what 2 pts (think demographics) would you want to avoid phenobarbital in? why?
hyperactivity (children)

confusion in elderly patients, decreased cognitive ability
what effect does phenobarbital have on drug metabolizing enzymes?
induces drug metabolizing enzymes
MOA of Phenobarbital?
Potentiates GABA inhibitory transmission and decreases glutamate excitation

note: can cause sedation, but can be used at low doses so its not a big deal
therapeutic use of Phenytoin?
Effective for all types of seizures except absence
Tonic-clonic-seizures in adults or older children
Used in status epilepticus
Wound healing
PT comes in with headache or other acute neuro findings in the ER

What is the first imaging study you want to use?

******
NON CONTRAST CT
in Schizophrenia, you need to have 2 or more of the following symptoms for at least 1 month...what are these sx? (5)
Delusions
Hallucinations
Disorganized speech
Disorganized or catatonic behavior
Negative symptoms (e.g., flat affect, avolition)
what is the difference btw positive and negative symptoms?

examples?
Positive=Present and shouldn’t be
Delusions
Hallucinations
Disorganized Speech
Disorganized Behavior

Negative=
Missing and should be there
Flattened Affect
Alogia-impoverished speech
Avolition- can’t initiate or persist in goal-directed activities
Anhedonia- loss of interest or pleasure
Delusions
Hallucinations
Disorganized speech
Disorganized or catatonic behavior
Negative symptoms (e.g., flat affect, avolition)

describes what kind of Schizophrenia?
Paranoid
Disorganized Speech
Disorganized Behavior
Flat/Inappropriate Affect
Active, but aimless
Little contact w/reality
Does not meet criteria for Catatonic type.

describes what kind of Schizophrenia?
Disorganized
Motoric immobility
Excessive motor activity
Extreme negativism or mutism
Peculiarities of voluntary movement as evidenced by posturing
Echolalia or Echopraxia

describes what kind of Schizophrenia?
Catatonic
What is one of the worst prognostic factors in Schizophrenia?
Assaultiveness
Acute onset, later age of onset, mood symptoms present, and no psychiatric history are what type of prognostic factors for Schizophrenia?
Good prognostic factor
How is Schizophreniform different from Schizophrenia?

***TEST
Schizophreniform:

Essential features are identical to those of Schizophrenia except for two differences:
Total duration of illness is at least 1 month but less than 6 months.
Impaired social or occupational functioning is not required
What is a delusional disorder?
Nonbizarre delusions-1 month
Criterion A for Schizophrenia never been met
Functioning is not markedly impaired
Behavior is not obviously odd or bizarre
If mood episodes have occurred concurrently with delusions, their total duration has been brief relative to the duration of the delusional periods.

example: Erotomanic- think someone of high stature in love with you

Grandiose: you have a connection with god
What is Shared Psychotic Disorder (Folie à Deux)
A delusion develops in an individual in the context of a close relationship with another person, who has an already-established delusion.
The delusion is similar in content to that of the person who already has the established delusion.

seen in dependent child parent relationship
Prominent hallucinations or delusions
Evidence (hx, physical exam, lab findings) that the disturbance is the direct physiological consequence of a GMC.
Not better accounted for by another mental disorder.
Does not occur exclusively during a delirium.
Psychotic Disorder Due to a General Medical Condition
Substance-Induced Psychotic Disorder

define it (pretty simple)
Prominent hallucinations or delusions. Note: Do not include hallucinations if the person has insight that they are substance induced.
There is evidence (hx, physical exam, lab findings) that
The symptoms developed during, or within a month of, Substance Intoxication or Withdrawal
Medication use is etiologically related to the disturbance
Not better accounted for by a psychotic disorder that is not substance induced.
Does not occur exclusively during a delirium.
What is the key to Schizoaffective disorder
An uninterrupted period of illness during which, at some time, there is either a Major Depressive Episode, a Manic Episode, or a Mixed Episode concurrent with symptoms that meet criteria A for Schizophrenia

**During the same period of illness there have been delusions or hallucinations for at least 2 weeks in the absence of prominent mood symptoms.**
What is one key reason why CT is better than MRI?

***
it is good for quick evaluation of acute bleeding: Excellent for acute intracranial hemorrhage


(also, MRI might require pre-approval for access)
where do epidural bleeds most commonly occur
fronto-temporal area
What is this? Where is it occuring
Epidural bleed

frontal lobe
what are the Hounsfield units for:
water
bone
air
blood
Fat
0 = water density
+ 1000 = bone
-1000 = air
+ 60 – +80 = blood
- 5 to –12 = fat
T1 vs T2 on MRI?
T1 = the “anatomic” sequence
T2 = the “pathology” sequence


On T1 water is dark black
On T2 water is bright white
T1 or T2?
What 4 things are black on this type of image?
T1

Cortical bone
Bone
Moving blood
Fluid
T1 or T2
T2
Which study, MRI or CT can detect ischemic injusry 24-36 hours earlier than CT?
MRI
This imaging study has no bone artifact (especially in the posterior fossa), and is better at assessment of tumors, white matter, and disease/early edema?
MRI
Which imaging study (CT or MRI) has unchanged multi-planar capability?

**
MRI
A 26 y/o patient presents to the ED after being struck by a softball bat. You want to rule our intracranial hemorrhage. You order:
A. CT
B. MRI
CT (no contrast)

Cranial bone will be white
Which modality is best for evaluating white matter disease?
A. CT
B. MRI
MRI
CT or MRI?
MRI
type of image? Path?
what normally causes an epidural hematoma?

Subdural?
epi: skull fracture

sub: direct blow to the head w change in lvl of conciousness
What happens in shear injuries?
diffuse axonal injury

can be due to rotational injury (not necessarily blow to a head)
path?

will it cross sutural lines?

what blood vessel causes it?
epidural hematoma

DOESN'T CROSS SUTURAL LINES

assoc with skull fracture

due to arterial tear (middle meningeal artery)
epidural hematoma vs subdural hematoma

which crosses suture lines?
epidural hematoma :DOESN'T CROSS SUTURAL LINES

Sub: does
for an epidural hematoma, at what amount of bleeding do you have to consider operative tx?
less than 5mm: NON OPERATIVE

greater than 5mm: drain
epidural hematoma in kid
describe some of the characteristics of subdural hematomas (shape, blood source, who it happens to)
crecentic,
bridging veins,
old people who fall
SDH more common with ____ while

EDH more common with _____

(cause of injury)
SDH more common with falls
EDH more common with MVA
Subdural Hematoma
this type of bleed is Spread over a larger area, limited by the falx and the tentorium
Subdural Hematoma
there are 3 densities seen on CT with a SDH..what are they and what are the time frames assoc with them
Acute 0 to 3-4 days hyperdense

Subacute 3 to 20 days isodense

Chronic > 20 days hypodense
SDH
epidural hematoma
this type of injury Occur with rapid acceleration/deceleration
Usually involves large WM tracts: corpus callosum, brainstem and deep white matter
Occurs at gray-white matter interface due to slight differences in mass
Minor differences in tissue inertia
Shear Injuries
Intraparenchymal hemorrhage from shear injuries
MRI showing
intra parenchymal
hemorrrhage
Left Middle Cerebral Artery Infarction with midline shift:

what is the most useful landmark for measuring midline shift?
Most useful landmark for measuring midline shift: Septum Pellucidum

note:This type of edema is called cytotoxic edema


cytotoxic edema visible on CT and MRI
what is key to remember about cytotoxic edema?
Affects both gray and white matter
History: stroke symptoms
cytotoxic edema
44 y/o female
New onset seizures
Fell, hit head

hemorrhage or tumor?
This is an enhancing
tumor with edema
confined to the
white matter (vasogenic
edema)
vasogenic edema is found where?

cytogenic?

causes?
***
vasogenic edema: CONFINED TO WHITE MATTER (tumor or infection)

where cytogenic could be in white or gray (infarct or stroke)
SUMMARY SLIDE

CT is 1st in the emergent setting to rule out hemorrhage.
MRI is more sensitive for edema, tumors, infections and white matter disease.
Be able to distinguish SDH from EDH.

Be able to distinguish cytotoxic (stroke) from vasogenic (tumor or infection)
CT is 1st in the emergent setting to rule out hemorrhage.--FAST

MRI is more sensitive for edema, tumors, infections and white matter disease.--CONTRAST MAKES IT EVEN MORE SENSITIVE

Be able to distinguish SDH from EDH.--SDH more cresentic, covers further distance, not confined by sutures

Be able to distinguish cytotoxic (stroke-effects both grey and white matter) from vasogenic (tumor or infection--just in white matter)
Define the purpose of the Mental Status Exam

(o)
Examination of neuropsychiatric functioning

Comprehensive description of a patient’s appearance, behavior, thinking, feeling, etc.

Meaningful only in the context of other baseline data (e.g., history, physical / neurologic exam)

Ex. Tearful patient may be reacting to stress or pain, be depressed, have neurological disease, or other cause

The MSE ≠ MMSE
Explain how the Mental Status Exam is performed

(o)
By observation
i.e., many areas assessed while obtaining the history
e.g., speech, behavior, affect

By asking relevant questions to elicit symptoms that usually cannot be observed
e.g., hallucinations, paranoid ideation, mood

By performing cognitive screening tests
e.g., MMSE, other bedside tests
Identify the major components of the Mental Status Exam

(o)
General appearance, attitude (toward the examiner), and behavior

Motor activity (overactivity, underactivity, abnormal movements, catatonia)

Speech

Mood and affect

Thought process (form) and content

Perception

Cognitive function

Insight and judgment
What is the difference btw catatonia, catalepsy, and cataplexy?

***
Catatonia: motor symptom: Dx by any two of the following: a. motor immobility, b. motor excitement, c. negativism or mutism, d. posturing, stereotypies or mannerism, e. echolalia (parroting) or echopraxia (miming)

Catalepsy: feature of catatonia, waxy flexibility (muscular rigidity and fixity of posture regardless of external stimuli, as well as decreased sensitivity to pain.)

Cataplexy: sudden loss of muscle tone leading to collapse, related to narcolepsy
describe the following mood/affects

Constricted/Restricted (flat)
Labile
Expansive
define the following disturbances in thought process:

Circumstantiality
Tangentiality
Looseness of associations
Verbigeration
“Word Salad”
Neologisms
Clang associations
Circumstantiality (excessive detail but gets to point)
Tangentiality (never gets to point of message)
Looseness of associations (ideas loosely connected)
Verbigeration (meaningless repetition of words/phrases)
“Word Salad” (incoherent collection of words/phrases)
Neologisms (creation of new words)
Clang associations (rhyming/punning; no logical connection)
describe circumstantial thought

*board fodder
Describe Tangential thought

*
Describe flight of ideas

*****
what is the difference between hypnagogic and hypnopompic hallucinations? are these pathological or non pathological?

***
Nonpathological: hypnagogic (when you go to sleep), hypnopompic (when you wake up)

Pathological: Auditory, visual, tactile, olfactory, gustatory, somatic
what are
Perceptual disturbances? (4)

****
include:
hallucinations,
derealization (where I am at doesn't feel real, i am floating in the space I am in),
depresonalization (feel like you are floating outside of yourself),
déjà vu
What are illusions (with respect to perceptual disturbances?)

**
misinterpretation of real stimuli
on the MMSE what score points to cognitive problems?
less than 20
Recognize the difference between the Folstein MMSE and the mental status examination

(o)

note: answer extrapolated from Wiki
The mini–mental state examination (MMSE) or Folstein test is a brief 30-point questionnaire test that is used to screen for cognitive impairment. It is commonly used in medicine to screen for dementia. It is also used to estimate the severity of cognitive impairment at a given point in time and to follow the course of cognitive changes in an individual over time, thus making it an effective way to document an individual's response to treatment.

Mental status is way more in depth
One-half of all lifetime cases of mental illness begin by age...
14

three-quarters by age 24.
Define psychiatric illness

(o)
Any of various conditions characterized by impairment of an individual's normal cognitive, emotional, or behavioral functioning, and caused by social, psychological, biochemical, genetic, or other factors, such as infection or head trauma.

Also called emotional illness, mental illness, mental disorder.
what is the difference btw primary and secondary psychiatric disorders?
Primary psychiatric disorders
No underlying medical disorder to account for the diagnosis
Not caused by medication
Not caused by substance abuse or withdrawal

Secondary psychiatric disorders
Due to a medical condition
Due to a medication(s)
Due to substance abuse or withdrawal
Identify the domains of psychiatric illness (6)

(o)
Some People Can Eat Bad Eggs
Identify the classification systems used to diagnose psychiatric illness

(o)
Diagnostic and Statistical Manual of Mental Disorders, 4th Edition, Text Revision (DSM-IV-TR)
Published by the American Psychiatric Association (2000)
Most recent update of criteria: DSM-IV (1994)
Current DSM bases diagnostic codes on ICD-9-CM

International Classification of Diseases, 9th Revision, Clinical Modification (ICD-9-CM)
Published by the US government in 1979
Official coding system in the US for all diseases (Ch.5 Mental Dis)
Revises codes annually but has not kept up with diagnostic changes in the DSM
What is an important thing to remember about the DSM IV and diagnosis it gives

****
Contains a listing of psychiatric disorders, diagnostic codes, and criteria for each disorder

Also contains text with information about the disorder (e.g., associated features, prevalence, course, familial patterns, age-,culture- and gender-specific features, differential diagnosis)

***Does NOT contain information about presumed etiology or treatment****
Describe the DSM-IV-TR multiaxial diagnostic system

(o)
Axis I: Clinical Disorders (psychiatric)

Axis II: Personality disorders and traits, mental retardation, prominent defense mechanisms

Axis III: General Medical Conditions

Axis IV: Psychosocial and environmental problems (stressors)

Axis V: Global Assessment of Functioning (1-100 scale)


EXAMPLE
Axis I 296.22 Major Depressive Disorder, single episode, moderate
305.00 Alcohol Abuse

Axis II 301.6 Dependent Personality Disorder

Axis III Hepatitis B

Axis IV Threat of job loss

Axis V GAF = 45 (current)
Almost all psychiatric diagnoses are made...
CLINICALLY
Identify the components of a psychiatric evaluation

(o)
History (Identifying Data/Reason for admission or evaluation, Past Psychiatric History, Substance History, Mental Status Exam)




Exam (neuro/mental)

Radiological and lab
SUMMARY SLIDE FOR LECTURE 2 OF PSYCH
Psychiatric symptoms can be seen in many conditions

The etiology of most primary psychiatric disorders is not known

Diagnosis is based primarily on history and mental status exam

Physical exam, neurologic exam, lab, and other studies are performed to rule out possible non-psychiatric causes of psychiatric symptoms

The DSM-IV-TR increases reliability of diagnosis
explain why most mechanisms of action given the neuropharmacologic drugs are approximations.

(o)
Entire mechanism is generally unknown. Approximations of

CNS mechanism made from information regarding effects of drugs in isolated systems.
CNS as a “Black Box”
Extrapolation from biochemical events to behavior is difficult.
define what is meant by specific mechanism of action.

(o)
A specific drug action affects a recognized protein target, i.e., a receptor, an ion channel, an enzyme, or a transporter.
describe the various events in synaptic transmission which might be altered by drugs

(o)
describe the various events in synaptic transmission which might be altered by drugs

(o)
describe the various events in synaptic transmission which might be altered by drugs

(o)
what is the difference between a neurotransmitter and a neuromodulator?

(o)
Neurotransmitter – a substance which is released locally and causes a change in post-synaptic potential.

Neuromodulator – a substance which acts to modify the response of the synapse to a neurotransmitter. (make a stimulus stronger or weaker)
describe the various events in synaptic transmission which might be altered by drugs

(o)
recognize the substances which have been proposed to act as CNS neurotransmitters

(o)
Acetylcholine
Amino acids
Biogenic amines
Peptides
Purines (ATP, adenosine)
Nitric acid
Endocannabinoids
differentiate between ionotropic and metabotrophic receptors.

(o)
Ionotropic receptors (also known as ligand-gated ion channels) are associated with ion channels, and change ionic conductance.
Mediate fast, short effects.

Metabotropic receptors are coupled with enzymes via G-proteins and other intermediates.
Mediate slow, long effects.
describe the blood brain barrier and its implications in drug therapy (what are the characteristics that regulate diffusion of substance through capillaries)

(o)
A term used to describe diffusional barriers retarding the passage of substances from the central circulation to nerve cells.

Characteristics which regulate diffusion of substances through capillaries are:

Molecular weight
Lipid solubility
Ionization at physiological pH (ionized drugs are not lipid soluble)
define epilepsy...

incidence?
2 or more unprovoked seizures

incidence is highest in kids and old folk
in-between epileptic seizures, what is a common finding on EEG?
Spikes
What happens in an absence seizure? clinically and EEG

(o)
Clinic: pt suddenly stops responding/no eye contact for about 7 seconds and then just resumes back to normal (full recovery)

EEG: sudden spiking in all leads, quick onset, quick ending
What happens in a tonic-clonic seizure? clinically and EEG

(o)
Clinic: abrupt onset with no warning, pt starts arching back (tonic) and then clonic (shaking all over), NO IMMEDIATE RECOVERY, pt might have pissed themselves, bit their tongue, want to go to bed--this is what you classically think of as a seizure

EEG: sudden rapid hashy spiking (tonic) in all leads, followed by spike then pause, spike then pause (in all leads) leading to the jerkiness (clonic). When the seizure ends, the EEG is not completely normal (shows the disorientation when it ends)
What happens in a Partial Simple Seizure? clinically and EEG

(o)
specific to what brain area is affected

for example, a pt could have a left motor seizure causing a right arm tremor. Pt is wide awake, aware of the symptoms, but can't stop them

EEG: Will only see irregular activity in certain leads (that correspond to one side of the brain, thus focal)
What happens in a Partial Complex Seizure? clinically and EEG

(o)
occurs in the Temporal lobe

usually clouding of consciousness

EEG: Will only see irregular activity in certain leads (that correspond to one side of the brain, thus focal)
Can occur in 4 places:
anterior-deja vu/psychic symptoms
Sylvian- Epigastric sensation, movement of mouth and face
Mid-temporal - auditory hallucinations, depersonalization
posterior temporal-Complex visual hallucinations
a pt says every now and then I keep seeing a pack of wild dogs that nobody else can see...what is this?
Partial-Complex Seizure
what is common and similar in Absence and Partial Complex Seizures
Consider:
brain involvement
Age of pt
Duration
Sx recalled by pt
Movements or behaviors
After-effects
EEG pattern
Underlying Cause
Effective Tx

TEST!!!!!!!!!!!!!!!!!!!!!!!!!
ONLY THING SHARED: STARING AND DECREASED RESPONSIVENESS
The student will be able to correlate the age of onset of first seizure with likely underlying causes.
Consider:
Idiopathic
Febrile
Birth Injury
Metabolic
Infection
Trauma
Tumor
Stroke

(o)
The student will be able to describe the means and rationale for investigation of a new seizure disorder by history, examination and ancillary testing.-JUST READ

(o)
CBC—is there an elevated white count to imply infection?
Chemistries—especially glucose, sodium, calcium, renal function tests and liver function tests.
Brain imaging to look for macroscopic focal disease, such as tumor, stroke, arteriovenous malformation or developmental anomaly.
An MRI scan including coronal sections through the medial temporal lobes is ideal, but there are circumstances in which a CT is substituted, e.g. when the patient is restless, claustrophobic or has a pacemaker.
An EEG, preferably sleep-deprived to increase sensitivity. Does the EEG show abnormalities, especially spikes, highly associated with seizures? If so, are they focal or generalized?
The student will be able to explain and account for differences in brain-injury patterns caused by high- and low-velocity missiles.

(o)
k.e.= ½ m v2

so the faster the traveling bullet (military weapon) will be more likely to kill you than a slow moving bullet (civilian weapon)
please define a concussion

(o)
Diffuse, physiological and microscopic insult, with or without diffuse axonal injury

Usually defined as involving a period of confusion or unconsciousness, even if just brief

Numerous common associated symptoms, e.g. headache, dizziness, irritability, emotionality, poor concentration
highschool football player gets hit on the head in the second half of the game and dies. Why did this happen? this is an example of?

*
You look back and see that the kid got hit on the head in the 1st half but it was dismissed or not realized

the SECOND IMPACT SYNDROME says that the brain is waaayyyy more vulnerable to a 2nd injury

this is more common in younger ppl
Little Johnny gets hit in the head during a game and gets knocked out. Mom and Dad bring him to you and ask if he can play the next game in 2 days. What do you tell them
have to be at least a week of no symptoms before they can resume play
pleas define a brain contusion..where do they tend to occur?

(o)
Focal hemorrhages within brain tissue
Big enough to see with naked eye in a pathology specimen or as an area of intensity (whiteness) on a CT scan
Tend to occur around edges of brain, particularly at base of cerebrum and in anterior temporal lobes
describe a subdural hematoma including the location, and appearance on CT initially and chronically

(o)
Traumatic hemorrhage from veins inside dura but outside brain

This behaves like an actual space, so, acutely, blood runs from anterior to posterior pole

The hematomas appear initially white (X-ray dense) on CT

Over time, the hematomas get diluted and resorbed, and become less X-ray dense
describe an epidural hematoma including the location, and appearance on CT initially and chronically...what is the cause?

(o)
Traumatic hemorrhage from meningeal arteries inside skull but outside dura

This is a potential space through which the fresh blood dissects from edge to edge, creating a lens-shaped hematoma that does not extend from pole to pole (the subdural does)

The hematomas appear white (X-ray dense) on CTs acutely, but become less dense as they dilute and resorb
please describe the Glasgow Coma scale, including the 3 aspects, and scoring ranges
pt presents with multiple cognitive deficits including memory impairment, aphasia, apraxia (arm won't do what you tell it to), agnosia (trouble remembering what objects are), or trouble in executive functioning (have trouble cooking). What do they have?

(o)
Alzheimer's Dz

note: the shrinking effect on the brain is bigger due to age than Alzheimers (like an 80 year old with AD would have a larger brain than a normal 90 year old)
on a PET scan, what will be a difference seen in a pt with Alzheimer's dz?
less activity posteriorly
amyloid plaques are commonly seen on silver stain in what? what is another pathological finding of this problem?
Alzheimer's Dz

Neurofibrillary Tangles (used to be neurotubules, will be in the cells)
What are 2 pharmacological tx for AD?
Acetylcholinesterase Inhibitors [Examples: Donepezil (Aricept) Galantamine (Reminyl, Razadyne)
Rivastigmine (Exelon)] --used in mild to moderate disease

NMDA (Glutamate) Receptor Blocker (Memantine (Namenda))-- Moderate level of disease
Progressive dementia clinically similar to Alzheimer’s disease

Early, prominent involvement of frontal and temporal lobes (“frontotemporal dementia”)

(o)
Pick's Disease

will find Pick Bodies intracellularly

note: PET scan will show less activity in the frontal and temporal lobes (who'da thought)
Hypokinesia/akinesia
Bradykinesia
Rigidity (not to be confused with spasticity or paratonia)
Stooped posture
Small-stepped gait
“Rest” tremor, typically unilateral or asymmetric

area all seen in what?
Parkinsonism
associated with degeneration of substantia nigra cells containing intracellular Lewy bodies

(o)
Parkinson's Disease
Usual fluctuations in cognitive function, involving alertness and attention
Visual hallucinations are common
Will also have movement problems
Pathologically, intracellular Lewy bodies appear diffusely in the cerebral cortex

(o)
Dementia with Lewy Bodies

Clinically resembles a blend between Alzheimer’s and Parkinson’s diseases

note: in Parkinson's, the Lewy Bodies are mostly in the Substantia Nigra
flip for a comparison of dementia with Lewy Bodies, Alzheimer's Disease, Parkinson's
flip for a comparison of dementia with Lewy Bodies, Alzheimer's Disease, Parkinson's
pt presents to your office due to changes in mood. Upon walking you notice that they seem to flick their hand, or kind of dance around. What is this? Genetics?

(o)
Huntington’s Disease

Autosomal dominant with high penetrance
Abnormal gene on chromosome 4

note: the dancing/flicking movement is known as CHOREA
What change in the brain would you see in a pt with Huntington's disease?
Absence of the Caudate Nucleus
A rapidly progressive, fatal dementia involving prominent motor symptoms, such as rigidity, clumsiness, and myoclonic jerks
A transmissible “spongiform” encephalopathy involving abnormal proteins called prions
Diagnosed with certainty only by brain biopsy

(o)
Creutzfeldt-Jakob Disease
What are the 3 categories of Creutzfeldt-Jakob Disease? Which are we most concerned about?
Sporadic CJD appearing without known risk factors accounts for 85% of cases.

Hereditary CJD involves positive family history or positive testing for a genetic mutation. 5-10% of U.S cases.

Acquired CJD involves transmission by exposure to brain or nervous system tissue. 1% of cases. (****transmission from open wounds, retina transplant, etc)
Increased levels of CSF protein 14-3-3 would give you the diagnosis of?
Creutzfeldt-Jakob Disease
inflammatory demyelinating plaques in the central nervous system is the hallmark of what?

(o)
Multiple Sclerosis (MS)

has to do with OLIGODENDROCYTES (not Schwann--peripheral)
T1 “Black Holes” and brain atrophy can be seen in what?
Multiple Sclerosis

note: Gilenya ™ - Fingolimod is the first oral pill tx for MS
(o)
2. The student will be able to correlate brain tumors and their usual anatomic compartments.

3. The student will be able to provide examples of brain tumors that are more or less common in younger and older patients.

(o)
midline tumors and tumors of cerebellar lobes are more common in adults or kids?
kids
this is the Fastest-growing and most lethal brain tumor in astrocytoma series
Infiltrates and spreads widely within brain, so surgery cannot entirely remove
Surgery still warranted for biopsy, with or without de-bulking and insertion of Gliadel wafer
Radiation therapy usually applied

*
Glioblastoma Multiforme
this brain tumor is chiefly in adults 30-50, has Intermediate growth rates depending on grade, can sit dormant for years with no effect. Radiation therapy can be applied, but is usually reserved for when the tumor is perceived as growing because there is a lifetime limit to how much can be given.
Cerebral Astrocytomas
Usually the slowest growing brain tumors, but can eventually crowd the brain.
Often discovered as an incidental finding on a scan done for another reason
Are usually calcified and are therefore very recognizable on CT scans.
Meningiomas
please give an example that fits each of the following:
A fast-growing intracranial tumor displaces brain.

A slow-growing intracranial tumor replaces brain
A fast-growing intracranial tumor displaces brain. (Examples: glioblastoma multiforme, metastases.)

A slow-growing intracranial tumor replaces brain. (Example: typical meningioma.)
Most frequent incidence between ages of 30 and 50
Also calcify and usually show this on CT scans.
Intermediate growth rate, and is usually "managed" (surgery, RT) rather than cured
Glial cell in origin
Oligodendrogliomas
Can be hormonally active or disruptive
Can compress optic chiasm from below and produce visual field losses
Often removed surgically through the nose and sphenoid sinus
Pituitary Adenomas
Usually arise from the 8th cranial nerve as Schwannomas
Auditory symptoms (unilateral hearing loss and tinnitus) are prominent
Meningiomas and other tumors can also appear in this location
Cerebellopontine Angle Tumors
Comprise about 1/3 of childhood posterior fossa tumors
Predilection for cerebellar vermis
Though malignant, they are highly radio-sensitive
Medulloblastomas
posterior fossa tumor of childhood
Often encapsulated
Complete surgical extirpation often possible
Cerebellar Astrocytoma
2 of the most common primary cancers in existence are what? so what can this lead to?

(o)
breast and Lung

can lead to brain (intermediate tenancy to go to the brain)
The student will be able to describe more and less common sources of metastatic brain tumors.

aka what are the most and least likely to go to the brain***

(o)
Prostate: DOES NOT go to brain

Melanoma: GOES TO BRAIN (very high propensity)
The student will be able to describe treatments for tumors of the nervous system as well as their optimum applications and limitations. What are the options (4)

(o)
High-dose corticosteroids
Surgery
Radiation therapy
Chemotherapy
How do high dose corticosteroids help treat brain tumors?

(o)
Buys time by shrinking the edema around the tumor deposit, rather than affecting the tumor itself.
Effective against extracellular edema, but not intracellular edema, such as that seen in ischemia.
A typical starting dose is dexamethasone 10 mg, followed by 4 mg q.i.d
how does radiation therapy help treat CNS tumors?

(o)
RT is usually palliative--to shrink but not eliminate tumor deposits
For brain metastases, RT is sufficiently effective that the patient usually dies of non-CNS complications before CNS complications.
The treatment itself can damage the CNS.
When do you often see chemotherapy for CNS tumor treatment?

(o)
Usually reserved for glioblastoma multiforme (Fastest-growing and most lethal brain tumor in astrocytoma series
Infiltrates and spreads widely within brain, so surgery cannot entirely remove)


IV administration is complicated by intact blood-brain barrier blocking entry into CNS.
BCNU (carmustine) given IV probably crosses BBB to some extent.
Gliadel® wafers (also carmustine) can be implanted within the tumor bed.
what are some of the important areas that can be damaged by occluding the middle cerebral arteries?
Broca/Wiernicke's

Motor/Sensory cortex

Frontal eyelid

Optic radiation... others
anterior cerebral artery blockage can lead to problems where
leg and foot of motor and sensory
Posterior cerebral artery occlusion will give you what major problem?
visual deficits
non-traumatic Subarachnoid Hemorrhage is normally due to what?
aneurysm
where do aneurysms typically occur? (berry)
close to the circle of willis

most in Anterior communicating (like in marfans)
besides trauma, what are the 2 major causes of intraparenchymal hemorrhage?

(o)
Hypertension-associated

Arterio-venous malformation (AVM)

note: this is literally bleeding into brain tissue
where do Charcot-Bouchard microaneursyms occur? how are these different from Berry aneurysms

*
on penetrating arteries (deep in the brain, leading to areas like the basal ganglia)--assoc with intraparenchymal hemorrhage

different from saccular (Berry) aneurysms occurring near the circle of Willis. assoc with subarachnoid hemorrhage
large wedge shaped damage vs small pockets of damage...

please list what type of vessels were damaged

(o)
Wedges: large vessels

small pockets/lacunae: small (will be in penetrating arteries
what is the difference between an embolism and thrombosis

(o)
embolism: some garbage from upstream got dislodged, then it gets stuck in the brain

thrombosis: clot...can lead to breaking off and causing an embolism

so: embolism arises from somewhere else, thrombus occurs from the site
what is most commonly going to cause an embolism?
Atrial fibrillation

(irregularly irregular)
what is the concept of ischemic penumbra?

*
There is a central core of forever-lost brain cells that no treatment can revive surrounded by a larger zone of sick brain cells that may or may not recover – depending on acute management.
in stroke, what effect can blood sugar have on acute stoke management?
Too high can be damaging!!
what are some of the modifiable atherosclerotic risk factors? 4 (+1 non-modifiable)

(o)
Blood pressure
Blood sugar
Lipids (statin drug)
Smoking


note: Family History
shortly after stroke it is important to monitor BP. If it is really high what should you do? Should you let a pt walk right away?
we lost autoregulation, so be careful not to drop BP too aggressively-->can extend the stoke

so if their BP is high, don't worry about it for the immediate time being

also, don't let them walk, they may have an unsafe drop in BP
if you are going to give a drug for acute treatment of thrombotic stroke, which of the following would you use?

t-PA
heparin
aspirin
ASPIRIN has small benefit acutely

note: heparin would be good to prevent DVT from bed rest
what commonly causes stroke for old and young folks?

(o)
old folks: atherosclerosis or emboli from heart

young folks: congenital heart/great vessel defects; hypercoaguable states, migraine with aura + smoking + contraceptives
Giant Cell Arteritis
Tumor
Subarachnoid Hemorrhage
Subdural Hematoma
Epidural Hematoma
Medication Overuse Headaches

are examples of what type of headache?

(o)
Secondary Headaches
a pt shows up with really bad headaches and insists they have a tumor that is causing it. You respond..
they are extremely rare, and not likely the cause of your headache
What is a medication overuse headache?

(o)
person needed to take medicine for a headache

they took it too often for a long enough time

you transform it into a 2ndary headache that is more difficult to treat

headaches will occur everyday, present throughout the whole day, and is mild to moderate. It worsens when the medication is discontinued
an elderly female presents to your office with a unilateral headache, jaw claudication and impaired vision. What does this pt have? is this a primary or secondary headache? FIRST AID REVIEW: What is the tx??

(o)
Giant cell arteritis

secondary

High dose steroids
Migraine without Aura
Migraine with Aura
Tension-Type Headaches
Cluster Headaches

are what kind of headaches?

(o)
Primary
pt presents with a unilateral headache that feels like it is "pulsing". They say it is aggravated with activity and they have some nausea. They say they only have light sensitivity some of the times when this occurs...what is it?
Migraine without aura
what are some of the common triggers for migraines?
Hormonal – menstruation, ovulation, OCP, HRT
Dietary – alcohol, nitrite-laden meat, MSG, aspartame, chocolate, aged cheese, missing a meal
Psychological – stress, post-stress let-down, anxiety, worry, depression
Physical/Environmental – glare, flashing lights, visual stimulation, fluorescent lighting, odors, weather changes, high altitudes
Sleep-related – lack of sleep, excessive sleep
Miscellaneous – head trauma, physical exertion, fatigue
Drugs – NTG, histamine, reserpine, hydralazine, ranitidine, estrogen
what is the most common aura in migraines with aura? some examples?
VISUAL

scotomas (blind spots), photopsia (unformed flashes of light), fortification spectra (scintillating zig-zag lines), distortions in shape and size. In “retinal migraine,” these occur unilaterally instead of bilaterally.
what are the 5 types of migraine auras?
Visual: scotomas (blind spots), photopsia (unformed flashes of light), fortification spectra (scintillating zig-zag lines), distortions in shape and size. In “retinal migraine,” these occur unilaterally instead of bilaterally.

Somatosensory: unilateral paresthesias (spontaneous sensations) or hypesthesia (decreased perception of applied stimuli).

Motor: hemiparesis.

Language: aphasia (impairment of language comprehension or expression).

Brainstem: loss or change in level of consciousness, diplopia, tinnitus or hearing loss, vertigo, dysarthria, ataxia, bilateral sensory or motor symptoms.
pt comes in with a mild/moderate pressing headache that is bilateral. It does not get worse with activity and there is no nausea associated with it...what is this?

(o)
Tension type headache

THE ANTI MIGRAINE (pretty much the opposite: no light sensitivity, no nausea, doesn't get worse with activity)
what does the "cluster" refer to in cluster headache?
The "cluster" means clustering in time (not location)
pt presents with recurrent headaches occurring around the the eye lasting for about 15 minutes to 2 hours. He says the pain is intense but then goes away. What is this?

(o)
Cluster headache
Clumsiness in greater proportion than loss of power
Babinski sign present (immediate)
Spasticity (delayed)
Increased tendon reflexes (delayed)
Little if any atrophy

what type of lesion?

(o)
Upper motor neuron lesion
what is Spasticity...what is it associated with (upper or lower mn)

(o)
A unidirectional increase in muscular tone

Involves increased tone in “anti-gravity” muscles

In the upper extremities, resistance to stretching out biceps, but not triceps
In the lower extremities, resistance to stretching out quadriceps but not hamstrings

seen in upper motor neuron
What is Rigidity? what is it seen in?

(o)
Seen in Parkinson’s disease and other extrapyramidal disorders

A bi-directional increase in tone to passive range of motion

The resistance to passive range of motion is relatively independent of degree of force or rate of speed
What is Paratonia? commonly seen in?

(o)
feels like the pt is trying to stop you from moving the limb

Commonly seen in dementia

Also known as gegenhalten (to hold against)

A bidirectional resistance to passive range of motion that increases with force and rate of movement, as if voluntary
Loss of power in greater proportion than clumsiness
Hypotonia
Muscle atrophy
Muscle fasciculations

what type of lesion?

(o)
Lower Motoneuron
Compare Duchenne, Facioscapulohumeral, and Limb girdle Muscular dystrophies for the following characteristics:
Age of onset
Sex
Pseudohypertorphy
Initial distribution
Involvement of the face
Rate of progression
Contractures and deformity
Inheritance

(o)
what is the genetic defect in duchenne muscular dystrophy?
Dystrophin gene on short arm of X chromosome codes for dystrophin protein, which is a cytoskeletal structural protein.

this is damaged
a boy presents to your office and on physical exam you note that their calves are pretty toned. Mom says that when the boy is on the ground he does this strange thing where he arches his back and climbs his way off the ground...what is this called? sign of?
Gower's sign

in Duchenne
you are in your neuro office and examining a young lady patient who has issues with smiling. You ask her to smile and she physically can't. Her forehead is unwrinkeld and the corners of her mouth do not curl. On physical exam, you note winging of the scapula. What does this pt have?
Facioscapulohumeral Muscular Dystrophy

One key gene is on the long arm of chromosome 4. The function of the gene product is as yet unknown.
a man presents to you with a seemingly triangular face. On a hunch, you hit their hypothenar eminence with your reflex hammer and it causes their thumb to move towards their pinky. To confirm what you think, you have the pt squeeze your hand and tell them to open it as soon as you tell them. When you say go, their hand stays in a grip...what do they have? What is the genetics going on?
Myotonic Dystrophy
A gene near the centromere of chromosome 19 codes for myotonic dystrophy protein kinase. In M.D. dozens to hundreds of “CTG” repeats occur in the gene. The transcribed mRNA gets trapped within the cell’s nucleus.
what happens in Myasthenia gravis physiologically?

(o)
antibodies to the AChR
pt presents to your office with muscle weakness. They say they are fine in the morning, but by the end of the day they have drooping eyelids, jaw drop, and drooling...what do they likely have?
MG
what are some of the associated dz with MG? what is the most important one?
THYMOMA (malignant mediastinal tumor)

thyroid disease

widespread immune complex disease
what is a polyneuropathy

(o)
Bilaterally symmetric
Distinct from mononeuropathy and multiple mononeuropathies

so the key is that you have to have the deficit on the same spot on both sides

mulitple mononeuorpathies would be like radial on one side, ulnar on the other

poly is like whole left and right hand (stocking and glove is a common characteristic of polyneuopathy)
Distal-greater-than-proximal weakness
Distal-greater-than-proximal numbness
A “stocking-glove” pattern of weakness, numbness or both
“Length-dependent” neuropathy
Disrupts DTRs more than other forms of lower-motoneuron weakness

this describes?

(o)
Polyneuropathy

the longer the axon the worse it is affected
Werdnig-Hoffman is a motoneuron disease affecting what population?

*
NEONATES

you will see characteristic FROG LEG position and floppy baby
(other thing that causes floppy baby? Botulin toxin)

BABIES WILL NOT HAVE REFLEXES
a 60 year old man presents to your office and says that he cannot do routine tasks such as going up steps, getting out of a chair, or swallowing. You note widespread atrophy. What do you suspect...
Amyotrophic Lateral Sclerosis

ALS
of the following, which will have a MARKEDLY increase CK level?

Myopathy
MG
Polyneuropathy
Motorneuron disease
Myopathy

(think duchenne's muscular dystrophy)
what is a pathologic finding of babinski's sign?
dorsiflexion of the big toe
. The student will be able to measure and record visual acuity according to generally agreed upon conventions
Eye chart for visual acuity testing. Standard distances are 20 feet for a wall chart and 14 inches for a hand-held card.

Read down to the smallest characters possible or until reaching the 20/20 line, whichever comes first. The acuity is the last line correctly read.

say they read like all of the letters but miss 1 or 2 you say 20/20 minus (assuming they read at the 20 20 line)
a cut at the optic nerve would cause what visual problem?
a cut at the optic chiasm would cause what visual problem?
a cut at the optic tract would cause what visual problem?
left homonomous hemianopia
right temporal damage could cause what visual damage?
4
what system is responsible for constricting the pupil?
PNS
please diagram the light reflex pathway...is this PNS or SNS?
CN II

pretectal nucleus

Edinger Westphal nucleus

ciliary ganglion

contract pupil

(This is PNS)
please diagram out the sympathetic pathway for pupilary dilation
ipsilateral hypothalamus

synapse at T1 (IML)

travel up the sympathetic chain and to the apex of the lung (look out for horners)

goes to carotid bifurcation but then sends to the pupil for dilation
how can a tumor of the apex of the lung lead to pupil issues?
this is horner's syndrome due to a pancoast tumor

you will see a constricted pupil because the sympathetics (which would normally dilate the pupil) stop at the apex of the lung and are disrupted by the tumor
pt presents with a droopy eyelid and constricted pupil...
ipsilateral sympathetic chain issue--HORNERS

miosis-small pupil

ptosis- droppy eyelid
please list the primary and secondary action of the extraocular muscles. consider what would happen with a lesion
What do CN 6 and 4 do?
LR6SO4

LR (lateral rectus) connected to CN 6
SO (superior oblique) connected to CN 4
Remaining EOMs and levator palpebrae connected to CN 3
right lower face is weak, upper face is fine

is this an upper or lower motor neuron problem

(o)
UPPER
right lower face is weak, upper face is also weak

is this an upper or lower motor neuron problem

(o)
LOWER
The student will be able to grade muscular strength according to the MRC (Medical Research Council) scale.

(o)
afferent vs efferent
afferent: carry nerve impulses from receptors or sense organs towards the central nervous system

efferent: from CNS to target
what makes up the brainstem?
the midbrain (sup/inf colliculus)

pons

medulla
4 lobes of the brain? major functions?
frontal: high order processing, planning, etc

parietal: somatosensory

temporal: sound

Occipital: vision
where is broca's area located? (what lobe)

where is Weirnikes area?
B: Frontal lobe

W: Temporal
post central gyrus=?

pre=?
post central=somatosensory

pre= motor
cerebellum function?
compare planned motor behavior with actual movement

checks/balances
functions of the insular lobe
emotions, drives, drug addiction etc
function of the calcarine fissure
primary visual processing area
difference btw gray and white matter?
gray: cell bodies (and some axons)

white: axons
what makes up the basal ganglia?
Caudate Nucleus

Putamen

Globus Pallidus
just list the cranial nerves and their functions
* I-Olfactory nerve,
* II-Optic nerve,
* III-Oculomotor nerve,
* IV-Trochlear nerve/pathic nerve,
* V-Trigeminal nerve/dentist nerve
* VI-Abducens nerve,
* VII-Facial nerve,
* VIII-Vestibulocochlear nerve/Auditory nerve,
* IX-Glossopharyngeal nerve,
* X-Vagus nerve,
* XI-Accessory nerve/Spinal accessory nerve and
* XII-Hypoglossal nerve.
Dorsal horn is associated with what?

Ventral?
Dorsal: Sensory

Ventral Motor
consider the locations of white and gray matter in the spinal cord and brain (which is deep and which is superficial in each)
spinal cord: gray matter deep

brain: gray matter superficial
what divides the gracile and cuneate fasciculi?
posterior intermediate sulcus
what is the difference btw the gracile and cuneate fasiculate
Gracile: carries axons from lower extremities

cuneate: upper (t6 and up)

REMEMBER: Feet in the grass

Part of the DCML pathway for sensory info (pressure, vibration, fine touch)
what type of information is carried by the Dorsal column/ medial lemniscus pathway
fine touch
proprioception,
vibration.
lateral corticospinal tract does what?
somatomotor
does a 1st, 2nd, or 3rd order neuron cross?
2nd will cross

such as the second order neurons from the nucleus cuneatus crossing as internal arcuate fibers in the medulla
what does the anterolateral system (ALS) do?
pain
temperature
touch (crude)
please diagram out the DCML pathway...what info does this carry?
please diagram out the Anterior Lateral System, and list what it carries
Please diagram out fine touch and vibration of the face
please diagram orofacial pain and temp
The left optic tract carries info from what visual field?
right visual field
What is the job of the Lateral Geniculate Nucleus (LGN)?
visual processing

receives synapse from eyes
where will superior visual field info end up?
inferior calcarine fissure

(remember they also travel inferior in the meyers loop)
a pituitary tumor causes pressure on the optic chiasm...what will this lead to?


****TEST
Bitemporal hemianopia

Will only be able to see info from medial visual fields (peripheral vision lost-->tunnel vision)
pt falls off their bike and was not wearing a helmet. They now say that they have lost vision just in the very center of each eye...what is going on?
Central scotomas
diagram out the corticospinal and corticobulbar tracts
pt has flaccid paralysis...what is the problem
lower motor neuron
pt has spastic paralysis...what is the problem?
upper motor neuron
middle cerebral artery damage will lead to issues in what structure
face

comes out of sylvian fissure and branches like a tree
anterior cerebral artery damage will lead to issues in what structure
legs and feet
describe the role of the Direct pathway and the indirect pathway in the basal ganglia

**TEST
Direct pathway: starting movement

indirect pathway: stopping movement
which pathway (direct or indirect) encourages GPm activity...what does this do?
INDIRECT

GPm has tonic inhibition of the thalamus (which is always wanting to activate movement...it's like GO GO GO)

thus the indirect by activating GPm will stop movement (this is the key thing to remember)

note: in the direct pathway, you inhibit GPm so inhibiting the inhibition leads to activity
FIRST AID REVIEW QUESTION

abdominal pain radiating to the back, weight loss, migratory thrombophlebitis and obstructive jaundice should make you think what?
Pancreatic adenocarcinoma

more common in pancreatic head

aggressive (prognosis is 6 months or less)

CEA and CA19-9 tumor markers

assoc with Cigarettes and chronic pancreatitis but not EtOH
FIRST AID REVIEW QUESTION

What are some of the causes of aplastic anemia?
Radiatoin and drugs (Benzene, chloramphenicol, alkylating agents, antimetabolites)

Viral agents (B19 parvo, EBV, HIV, HCV)

Fanconi's anemia

Idiopathic