• 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

1/200

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;

200 Cards in this Set

  • Front
  • Back
what are depot formulations?
oily based formations delivered IM that slowly leach into circulation- cannot be recalled after delivery
ion trapping?
drugs trapped in an E due to pH-PCP
problem with levodopa facilitated diffusion?
competition with large neutral amino acids- tryptophan
bioequivalence?
same rate of absorption and same availability
what drug has such a sig. first pass effect that no drug is available for the CNS?
naloxone
what determines effective duration of action of CNS drug more than half-life?
redistribution
what is the therapeutic window?
no relationship between blood levels and CNS effects- best indicated by CSF levels
what receptors are in the CTZ?
serotonergic and dopaminergic
what prevents diffusion within the CNS?
low extracellular space
physical barriers(glia)
enzymatic processes and drug reuptake
what is selegiline biotransformed to?
it is a PD drug transformed to methamphetamine and amphetamine
what CYP's biotransform CNS drugs?
CYP2D6
CYP3A4
features of CYP2D6?
not induced
genetic polymorphism-ability to transform drugs varies across populations
no pharmacokinetic tolerance
features of CYP3A4?
inducible
no genetic polymorphisms
often administered to patients
pharmacokinetic tolerance
high concentration in GI mucosa
INHIBITED by grapefruit juice
INDUCED by St. Johns Wart
what is pharmacokinetic tolerance?
increase in enzyme synthesis due to increased demand
what actively transports drugs out of the brain?
arachnoid granulations
do direct or indirect agents usually have more side effects?
indirect
what type of receptor is the ACh nicotinic receptor?
traditional ionotropic receptor that opens up a ligand-gated Na and K channel
how does a change in receptor number affect drug potency?
up regulation increases agonist potency and decreases antagonist
down regulation is the opposite
how do autoreceptors work?
they reduce NT release and firing rates
so when they are down-regulated it increases release and firing rates(related to anti-depressant efficacy)
what drug treats anxiety of morphine withdrawal?
clonidine- alpha 2 agonist
what is the withdrawal/abstinence syndrome?
upon removal of a drug the system exhibits behavior that is exactly opposite to the originally intended use of the drug
what is physiological tolerance?
reduction in the potency of a drug as a result of a compensatory process to chronic exposure involving secondary actions of the drug
pharmacokinetic tolerance?
reduction in the potency of a drug following chronic exposure due to alterations in entry of drug body- P450 inductions(phenobarbitol)
difference between phenobarbitol and phenytoin?
phenobarbitol induces the P450 that affects itself and other drugs
phenytoin induces enzymes that increase transformation of other drugs only
what is cross-tolerance?
reduction in potency of drug X as a result of chronic drug Y exposure
what is reverse tolerance?
increase in the potency of a drug as a result of chronic exposure- DA agonists
what is disinhibition release?
CNS depressants initially cause a state of excitement due to decrease of CNS inhibition
where are the DA bed nuclei found?
ventral mesencephalon
what is the DA gating function?
regulation of the rate at which neuronal activity passes through the innervated target-more DA, more behavior
DA affect on ACh?
inhibition
features of PD?
hypokinetic- loss of DA
rigidity
loss of postural reflexes
REST tremor
origin and target of nigrostriatal pathway?
O-substantia nigra pars compacta
T- neostriatum(cuadate and putamen)
origin and target of mesolimbic pathway?
O- ventral tegmental area
T- nucleus accumbens
origin and target of mesocortical pathway?
O- ventral tegmental area
T- cortical regions
origin and target of tubero-infandibular pathway?
O- zona incerta, periventricular nucleus
T- pituitary
origin and target of the carotid body pathway?
O- glomus cells
T- nucleus tractus solitarius
origin and target of the SIF cell pathway?
O- paravertebral system
T- post-ganglionic sympathetic fibers
origin and target of the renal system pathway?
O- ?
T- smooth muscle of renal arteries
origin and target of the pyloric sphincter pathway?
O- ?
T- pyloric sphincter
what is MPTP?
contaminant from opioid synthesis that is converted to MPP+ by MAO-B- high affinity for DA re-uptake pumps and taken up by mit. and uncouples oxidative phosphorylation at complex one
how is DA degraded?
by MAO-B to dihydroxy-phenylacetylaldehyde
by COMT to methoxytyramine
what is the most well-known PD genetic abnormality?
abnormal alpha-synuclein
most genes associated with proteosomal function leading the belief of protein toxicity
what narrows the therapeutic window in PD treatment?
as PD progresses there are less DA receptors so more levodopa is given, but at non-target sites the receptor numbers are normal and increased drug leads to psychosis
big side effect of DA agonists?
nausea and vomiting- DA receptors in CTZ
how does carbidopa change the side effects of levodopa?
without carbidopa the side effects are in the periphery because of AAAD breakdown
with carbidopa the SE's are in the CNS
what is an important co-factor of AAAD?
pyridoxine
what is the on-off phenomenon?
sudden onset of full PD symptoms occurring anytime during dosage of levodopa-due to sensitization of GLU in striatum due to intermittent dosing schedules
pneumonic for anticholinergic tox?
hot as a hare
blind as a stone
mad as a hatter
dry to the bone
problem with antimuscarinic tx. of PD in elderly?
don't tolerate antimuscarinics well-constipation, dry mouth, can't pee
drug regimen of PD?
indirect/direct and antimuscarinics early
MAO-B inhibitor to enhance DA release
hold off on levodopa
what dominates early PD tx?
pramipexole and ropinirole-neuroprotective
what dominates late PD tx?
direct DA agonists
comparison of side effects betwen levodopa and direct DA agonists?
levodopa causes more dyskinesia
direct agonists cause more psychosis
what excitotoxins produce striatal lesions similar to HD?
kainic acid and ibotenic acid
what causes the choreiform movements in HD?
loss of GABA in striatum
what causes the psychosis of HD?
cortical degeneration following loss of striatal trophic support
what is the coprophenomenon?
increased cursing and lewd gestures in tourette's
treatment for increased movement in tourette's?
resperidone and haloperidol- DA antagonists
what disease runs with tourette's?
ADHD
what is the most common neurologic disorder?
essential tremor
what are the positive symptoms of schiz?
bizarre behavior
inability to think coherently
hallucinations or delusions
what are the negative symptoms of schiz?
withdrawn
poverty of speech
blunted affect
inappropriate behavior
what is paradoxical about the treatment of and presentation of schiz?
it is treated with DA antagonists but their brains do not show increased DA or receptor number
what causes the positive symptoms of schiz?
disinhibition of the nucleus accumbens by the prefrontal cortex
what causes the negative symptoms of schiz?
reduced function of the pre-frontal cortex due to underdeveloped parahippocampal gyrus
what defines the typical anti-psychotics(neuroleptics)?
they produce acute extrapyramidal side effects- block of non-target DA receptors that produce parkinsonian side effects, akathisia nd dystonia
time line of typical anti-psychotic side effects
dystonia,akathisias and the parkinsonian
mutually exclusive, usually just parkinsonian
what drugs are used to combat the extrapyramidal side effects of typical anti-psychotics?
trihexyphenidyl and benztropine( antimuscarinics)
effectiveness of DA blockade in nigro-striatal pathway is reduced
what defines the atypical anti-psychotic drugs?
produce less extrapyramidal side effects
what are the main atypical anti-psychotics?
thioridazine and mesoridazine
what are the second-generation atypical anti-psychotics?
risperidone
olanzapine
clozapine
quetiapine
ziprasidone
aripiprazole
side effects of all anti-psychotics?
orthostatic hypotension(alpha)
CV problems(antimuscarinic)
increased prolactin(galactorrhea or gynecomastia)
sedation
antiemetic
weight gain
neuroleptic malignant syndrome(rigidity and hyperthermia)
what side effect is associated with chronic anti-psychotic use?
tardive dyskinesia- facial movements and other dyskinesias
either from withdrawal or breakthrough(worse)
due to DA receptor site up-regulation(permissive) from decreased DA
what treats tardive dyskinesia?
reserpine and tetrabenzine
increased anti-psychotic(may produce iatrogenic(physician induced) movement disorder)
what does efficacy delay of anti-psychotics show?
mechanisms other than DA receptor blockade are involved because the DA antagonists reduce DA activity within an hour
which set of anti-psychotics predominantly reduce positive symptoms?
first generation
what supports the role of NE in depression?
efficacy of NE reuptake inhibitors(desipramine and maprotiline) in treatment
what supports the role of 5HT in depression?
increased receptor numbers
decreased 5HT
decreased 5HIAA-primary metabolite
what causes seasonal affective disorder?
increased melatonin due to prolonged darkness
inhibition of 5HT neurons
melatonin secretion controlled by NE
what breaks down NE?
COMT and MAO-A
how is NE involved in sensory processing?
NE inhibits inter-columnar fibers which usually decrease sensory attentiveness in the cortex
in depressed patients there is less NE, so less attentiveness
major side effect of MAOI's?
tyramine effect-accumulation of amines leading to hypertensive crisis
which tests determine the course of depressive treatment?
amphetamine provocative- indicates more NE agent
dexamethasone suppression- indicates use of a serotonergic agent(no reduction of cortisol in 5HT decrease)
what drugs are used for OCD?
SSRI's and heterocyclics(second generation antidepressants)
drugs most used in panic attacks?
SSRI's
what other function to TCA's have?
treatment of night-time enuresis and geriatric incontinence
what does consciousness require?
brain stem
bilateral thalamic
unilateral cortical
which brain areas facilitate dreaming?
locus ceruleus- NE
raphe nuclei- 5HT
what happens in sleep atony(glycine clutch)?
gigantocellular region of lower brainstem activates Renshaw collateral interneuron which depresses activity in lower alpha motor neuron
what is the pedunculo-pontine cholinergic system?
cholinergic fibers project to the thalamus and regulate correspondence between sensory afferent fibers and thalamo-cortical fibers- ACh causes activation of the thalamo-cortical fibers and facilitates sensory traffic to the cortex and arousal
what is adenosine's role in sleep?
it builds up during the day and acts on the purinergic heteroreceptors to reduce ACh and arousal
how does caffeine work?
an antagonist at purinergic heteroreceptors and prevents adenosine action
when do night-time enuresis and night terrors occur?
stage IV sleep
how do depression and REM sleep relate?
depression is associated with an early onset of REM
which CNS depressants do not suppress REM sleep?
chloral hydrate and zolpidem(ambien)
which CNS depressants do not cause respiratory depression?
benzo's
how do benzo's work?
increase the affinity of GABA for the A receptor by increasing the FREQUENCY at which the Cl- gate opens
changes tertiary protein structure
no interference with GABA binding
what is the floor and ceiling effect?
as more drug is given, it has less effect due to decreased NT release
how do benzo's effect adenosine?
inhibit reuptake
how are the benzo's biotransformed?
mostly in liver by N-desalkylation and oxidation followed by conjugation
which benzo's are conjugated outside the liver?
OTL
oxazepam
temazepam
lorazepam
signs of benzo toxicity?
CNS depression
bitter taste
N and V
diarrhea
epigastric distress
what antagonizes benzo toxicity?
flumazenil
how do barbs work?
bind to Cl- ionophore at site independent of GABA site and increases DURATION of time the gate is open
may open the channel without GABA
what herbals contain benzo-like chemicals?
kava,valerian,mandrake
what NT's are in the central nucleus of the amygdala?
GABA
Glu
5HT
NE
what drug is used for stage fright?
propranolol and other beta antagonists
what kind of waves do seizures create?
hypersynchronous low frequency, high amplitude
what is a paroxysmal depolarization shift and what is responsible?
occurs in seizure
NMDA receptor activation with displacement of Mg and influx of Ca leading to excitotoxicity and retrograde release of NO
what cells are responsible for reducing extracellular K?
astrocytes and if they don't it may induce depolarization
how are partial and complex seizures treated differently?
partial are treated by reducing excitation and complex by increasing inhibition
side effects of antiepileptics?
intention tremor
nystagmus
ataxic gait
diplopia
hyperreflexia
seizures at high doses
absence seizures
what does succinic semialdehyde dehydrogenase do?
facilitates GABA entry to the Krebb's cycle, inhibited by valproic acid
what are many of the anti seizure adjunctive agents substrates for?
p glycoprotein, epilepsy induces the production of the protein and may confer resistance
what side effects does the induction of the p450 system by anti seizure meds lead to?
vitamin K deficiency and decrease in steroid efficacy
what leads to fetal hydantoin syndrome?
side effect of phenytoin
usually metabolized to parahydroxyphenytoin by CYP2C8 via arene epoxide intermediate which are detoxified by epoxide hydrolase, during development not all tissues have the hydrolase and the arene attacks DNA leading to transcription errors
problem with phenytoin generics?
not bioequivalent, causes problems when switching
how is phenytoin transported?
mostly bound to albumin, may be displaced by other drugs(aspirin)
when is a seizure considered status epilepticus?
after 20 minutes
management of status epilepticus?
start with benzo(lorazepam,diazepam)
chase with phenytoin
induce barb coma(pentobarbitol)
what is the most common migraine?
without aura
what is a cluster headache
worst pain ever, may be related to circadian rhythm
unilateral and focused around the orbit
what is a non-migraine headache and how do you treat it?
tension in neck and lower occipital region, bilateral and non-throbbing
treat with stress avoidance, NSAIDs and muscle relaxants(benzos)
what is the prostaglandin function in pain?
sensitizes pain fibers to the pain signal by altering phosphorylation status of the primary afferent terminal
what is caffeine's effect on drug kinetics?
enhances absorption of NSAIDS
what is the prophylactic treatment of choice for migraines?
propranolol
what is the vascular hypothesis of Wolff?
theory of migraine headache
release of serotonin from platelets causes vasoconstriction and ischemia
subsequent loss of serotonin leads to atonic arteries that stretch conn. tissue in response to pulse pressure producing throbbing
what is the sterile inflammatory hypothesis?
theory of migraines
release of substance P,CGRP and neurokinin from trigeminal nerve, induce vasodilation and histamine release producing inflammation and neural irritation
what is the biobehavioral hypothesis?
theory of migraine
spreading depression of Leao creates a wave of depolarization across cortex activating the ophthalmic limb of trigeminal nerve
stress precipitation and explains menstrual cycle involvement
how are anti-migraine meds administered?
suppository
what are the side effects of ergot alkaloids?
parethesias-tingling
leg cramps
confusion
what NT is most responsible for migraines?
CGRP
problem with newer triptans?
enter CNS and associated with serotonin syndrome in patients taking SSRIs
treatment of plan for migraines?
NSAIDs
sumatriptan
prokinetic with chronic treatment
what are the most dangerous drugs in frequent use?
anesthetics, very low TI
what is the minimal alveolar concentration?
measure of anesthetic potency and describes the gas concentration in blood needed to produce no reflex response to a knife cut in 50% of the population at room temp at sea level
relationship of solubility and anesthetic effect?
higher solubility, slower anesthetic induction
how are anesthetics administered?
in a closed system and not biotransformed
what is diffusion hypoxia?
when NO leaves the blood to enter the lung so quickly it displaces too much partial pressure leaving it insufficient for oxygenation
which anesthetic is biotransformed?
halothane
what is the Overton-Meyer hypothesis?
suggests anesthetic drugs disrupt membrane fluidity and Na channel function, found to be only at toxic levels
what is the MOA of anesthetics?
stereoselective-receptor based
drugs bind at interface between the protein and surrounding lipid to activate GABA-A receptors and depression of ion channels, bind in regions most associated with consciousness(thalamus,cortex,brain stem)
effects of anesthetics?
respiratory depressant(except NO)
bronchodilators
cardiac depressant(except NO)
vascular smooth mm relaxer and causes hypoperfusion(NO helps by vasocx.)
skeletal mm relaxers at high tensions,
malignant hyperthermia-genetic predisposition, abnormal Ca regulation in sarcoplasmic reticulum due to splice variants
what is a bad side effect of halothane?
halothane hepatitis- invokes immunological response that attacks the liver
biotransformed by CYP2E1 and 2A6 to trifluoroacetyl chloride then trifluoroacetic acid, may bind covalently to hepatocytes or react with 2E1, delayed response to 2E1
what adjuvants are used in anesthesia?
diazepam-facilitates drowsiness, anterograde amnesia
atropine reduces bronchial secretions and counteracts cardiac depression
muscle relaxants to reduce amount required to paralyze skeletal mm
opioids- pain
what is the effect of inducing agents in anesthetic treatment?
make patient more comfortable
reduce duration of disinhibition release motor excitement
NO,thiopental,high dose opioids
what is neurolept-anesthesia?
droperidol and NO
what is neurolept-analgesia?
droperidol and opioid fentanyl
may produce chest wall rigidity
what are the IV anesthetic agents?
etomidate
propofol
thiopental
ketamine
how do all local anesthetics work?
Na channel antagonists, enter channel from inside neuron- prevented during inflammation due to low pH
what are local anesthetics formulated with?
Ep or other vasocx. to keep drug local
CO2 to keep solution acidic
buffer to overcome effects on pH
problems with local anesthetics?
cardiovascular toxicity due to Na channel block
CNS depressant which decreases respiration
how are local anesthetics broken down?
esters converted to PABA which is a common allergen and has risk of delayed type hypersensitivity reaction
amides biotransformed in liver
what two systems are involved in pain?
sensory and limbic
what system is associated with the suffering aspect of pain?
limbic system
what do C fibers transmit?
slow pain
where do pain fibers synapse?
substantia gelatinosa
what forms the pain gate?
serotonin fibers from the raphe nuclei
NE fibers from the locus ceruleus
enkephalinergic heteroreceptors
what is neuropathic pain?
arises from trauma to the CNS or toxic insult
central activation of the pathway by non-chemical or mechanical means
what is the treatment for shingles?
carbamazepine
gabapentin
valproic acid
amitriptyline
what is a vaccine for shingles?
zostavax
what do opioid analgesics mimic?
enkephalins and endorphins
where do enkephalins and endorphins work?
spinal cord
periaqueductal gray
limbic system
what are the two enkephalins?
met-Enk
leu-Enk
how do enkephalins work?
reduce substance P release from C fibers in the periphery
effects the PAG
how do endorphins work?
paracrine effects
mediates ACTH
what are the opioid receptors?
enkephalin receptors
1 mu- brain and spinal cord, G proteins close Ca channels,decrease NT release and open K channels for hyperpolorization, produce the side effects of euphoria and resp. dep.
2 kappa- spinal cord
3 delta- limbic system, emotional and behavioral effects
4 epsilon-unknown action
how do direct acting opioid agonists work?
affect pain threshold without affecting consciousness
inhibit substance P release in substantia gelatinosa and activate descending pain suppression pathways from the PAG
what is the triad of opioid OD?
coma
respiratory depression
miosis
what drugs are associated with the most severe withdrawal symptoms?
barbs-seizures
what type of pain do opioids affect?
slow pain, suffering
how do opioids and elderly interact?
effects are increased due to decreased blood flow to the liver
what is the deal with loperamide and quinidine?
loperamide is an anti-diarrheal agent but only acts peripherally due to P-glycoprotein action which pumps it out of the brain, taken with quinidine which inhibits P-gp it can produce euphoria and respiratory depression
define drug abuse
consumption of a drug by self-administration that is outside the societal norm
define recreational drug use
self-administration of drugs to experience the euphoriant properties, leads to a pattern of dependence, higher doses that normally prescribed
define drug dependence
a pattern of use where the individual perceives the need to continue the use, in chronic pain or abuser
define addiction
a pattern of compulsive abuse, extended in time, where the individual is preoccupied with getting the drug
define narcotic
all drugs of abuse not including alcohol or tobacco
why are drugs abused?
provide incentive salience- an act leading to a reinforcement that further perpetuates incentive to produce that behavior again, not due to tolerance, withdrawal symptoms
common action of all drugs of abuse?
increase DA in the nucleus accumbens
what is the effect of continued drug abuse?
produces reverse tolerance in the mesolimbic pathway while tolerance in other brain regions-continue to seek out euphoric behavior which has tolerance(frontal cortex) while the reward center has become sensitized
what drugs do not increase DA in the N. accumbens?
pot
benzo's
inhalants
what are the anorexiants?
phenteramine
fenfluramine
phenylpropanolamine
phendimetrazine
benzphetamine
more NE and 5HT action, no DA action
actions of alcohol dehydrogenase?
readily saturated
genetic variants
not inducible
how do alcoholics develop tolerance?
greater percentage of alcohol is biotransformed by the p450 system following induction
what enzyme in the alcohol pathway shows genetic variance in the pacific rim?
aldehyde dehydrogenase
what is a Mickey Finn?
ethanol and chloral hydrate(trichloroethanol),they share alcohol dehydrogenase which favors chloral hydrate and leads to sedation
how do you treat methanol and ethylene glycol toxicity?
ethanol followed by dialysis
what is used in alcohol OD?
diazepam
what is used to treat phencyclidine OD?
diazepam
why are anticholinergics abused?
produce delirium with delusions
difference between hallucination and delusion?
hallucination is a misperception of reality, a delusion is an image that is not there
what is another name for pseudocholinesterase?
butyrylcholinesterase, widely distributed in blood
how do the nootropics inhibit ACh metabolism?
1 reversible binding to choline site
2 binding to anionic site
3 act as AChE substrate but are transformed slower
what insecticides inhibit AChE?
parathione,diazonon and malathione
what are the nootropic toxic effects?
Defecation
Urination
Miosis
Bowl disturbance
Emesis
Lacrimation
Salivation
why do people use inhalants?
euphoria from oxidative phosphorylation uncoupling
little dependence liability
toxicity due to contaminants
what is pyramiding?
cyclic steroid regimen
starts at low doses and increases for 6-12 weeks and then decreases back to zero
what are the toxic effects of steroids?
paranoia,agitation,aggressiveness, increased libido,hallucinations,psychomotor retardation or acceleration,fatal liver cysts and cancer,blood clots,hypertension,acne
withdrawal symptoms of steroids?
depressed mood, fatigue, restlessness,loss of appetite,insomnia,reduced sex drive,headache and joint pain