• 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

1/941

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;

941 Cards in this Set

  • Front
  • Back
cholinomimetic drugs that are choline esters
1. acetylcholine 2. methacholine 3. carbachol 4. bethanechol
cholinomimetic drugs that are naturally occurring
1. nitotine 2. muscarine 3. pilocarpine
2 ways that cholinomimetic drugs enhance cholinergic function
1. activate Ach directly through nicotinic and muscarinic receptors and 2. block AchE
function of anticholinesterase agents
indirectly increase cholinergic function
list reversible anticholinesterase agents
1. physostigmine 2. neostigmine 3. edrophonium 4. pyridostigmine 5. donepezil and tacrine
list anticholinesterase irreversible agents
1. parathion and malathion 2. sarin and soman
what is the cholinesterase reactivator?
pralidoxime (2-PAM)
what are the 5 basic steps in neurotransmission
1. synthesis 2. storage 3. release from presynaptic terminal 4. reception and post-syn response 5. termination of signal
examples of muscarinic blocking drugs
1. atropine 2. scopolamine 3. tropicamide 4. ipratropium 5. benzytropine 6. oxybutynin
what are 6 example of places where cholinergic synapses are found?
1. all parasyn neuroeffector junctions 2. all autonomic ganglia 3. skeletal NMJ 4. sympathetic innervation of adrenal medulla 5. sweat glands 6. brain and SC sites
non-innevated ACh receptors are generally of what type?
muscarinic
how do cholinergic drugs decrease colinergic function?
they usually occupy the receptor so the ACh molecules cannot act on them
where does synthesis of ACh occur?
cytoplasm of cholinergic nerves
enzyme that catalyzes ACh synthesis
choline acetylase
rate-limiting step of ACh synthesis
active pumping of choline into the nerve by a transport mechanism
what blocks the transport of choline into the nerve that causes an eventual inhibition of choline function?
hemocholinium (HC-3)
what causes attachment of ACh granules to fuse with nerve terminal membranes during action potential
influx of Ca
what can inhibit exocytosis of ACh into synaptic space?
anything that lowers extracellular Ca i.e. bot toxin and latrotoxin
what prevents release of ACh from nerve terminal
botulinum toxin
what causes excessive fusing of ACh granules w/ nerve mem causing excessive release and depletion of ACh
latrotoxin - spider venom
where are acetylcholinesterases found?
region of all cholinergic synapses
where are pseudocholinesterases found?
plasma and liver
function of pseudocholinesterases
less selective for ACh, acts on many ester linkages, helps degrade ACh that escapes from transmission sites and inactivates ACh injected IV
M2 receptors are found
in heart
M3 receptors are found
peripheral autonomic organs
M1 receptors are found
in GI tract and autonomic ganglia
which muscarinic receptors are located in CNS
M1-M5
what does stimulation of muscarinic receptors in the heart do?
slows heart by increasing K permeability causing prolonged phase 4 depolarizaiton
T/F: muscarinic receps in heart directly stimulate ventricular contraction
F: they have little effect on force of vent contraction
what effect does systemic admin of ACh and muscarinic stimulants have on vascular sm
profound vasodilation and decrease BP
how does ACh cause blood vessel smooth muscle relaxation?
ACh acts on non-innerv muscarinic receptors on endothelial cells that releases EDRF (NO)
what do muscarinic agonists produce in the eye
pupillary constriction (miosis) and spasm of accommodation (cyclotonia)
what do muscarinic agonists do in the GI tract
increase tone and motility
what do muscarinic agonists do in bronchiolar smooth muscle
modest constriction
how must ACh be administered to have an effect
injected
what will be activated if ACh is given in low doses
only muscarinic receptors, much higher amts are needed to stimulate nicotinic
what drug is partially resistant to AChase, is a potent muscarinic stimulant, but has lost almost all nicotinic action of parent molecule
methacholine
why is methacholine not used as much in humans
has too many generalized muscarinic effects
what drug is very refractory to inactivation by AChase, has a potent nicotinic stimulating effects w/ little muscarinic actions, and releases ACh from cholinergic nerve endings
carbachol
what drug is used topically on eye to produce miosis and tx glaucoma
carbachol
which drug is only choline ester that has established clinical use
bethanechol
which drug is refractory to AChase inactivation and has mostly muscarinic stimuating effects, tx post-op urinary retention and atony of GI tract
bethanechol
how is bethanechol used clinically
tx esophageal reflux due to stimulatory action on lower esophageal sphincter
what are responses to injected nicotine
activation of all autonomic ganglia and stim of skeletal muscle endplate nic receptors
antidote for muscarine poisoning
atropine
which drug selectively activates muscarinic receptors, not highly charged so can cross BBB, and used to tx glaucoma
pilocarpine
food, drug and cosmetic act addresses what?
drug safety
what established the FDA?
food, drug and cosmetic act
what does the FDA oversee?
manufacture, labeling and advertisement of drugs
what did durham-humphrey amendment do?
divided drugs into prescription only and OTC
what did the kefauver-harris amendment do?
required that EFFECTIVENESS be shown for all existing and new drugs
what is the Investigational New Drug Application for?
goes from manufacturer to FDA to test a new drug
what are the 3 phases of drug testing?
1- testing in healthy volunteers 2- tx of patients w/ indicated condition 3-private practitioners use drug in less controlled studies
T/F: the New Drug Application can be made by the manufacturer during only phase 1 of clinical testing
F: can be made during any phase
what percentage of drugs that enter human testing reach the market?
12%
how long does drug testing process last?
7-10yrs
what is the patent life for a drug?
17 yrs
which drug class for pregnancy describes when risk cannot be ruled out
C
which drug class for pregnancy describes when no evidence of risk in humans
B
which drug class for pregnancy describes when there is positive evidence of risk
D
which drug class for pregnancy describes when it is contraindicated in pregnancy
X
which drug class for pregnancy describes when controlled studies in both humans and animals failed to show fetal harm
A
what is the only drug labeled as class A for pregnancy
cromalin (for asthma)
which act governs all aspects of controlled substance distribution in US?
comprehensive drug abuse act
what must all controlled substance Rx's contain?
1. date of issue 2. name and address of pt 3. name, address and DEA# of prescriber, 4. manual signature of prescriber on C-II
which drug schedule has no accepted medical use and high abuse potential
C-I
which drug schedule has low abuse potential with low psychological or physical dependence potential?
C-IV
which drug schedule has high abuse potential w/ an accepted medical use with severe psychological and/or physical dependence
C-II
which drug schedule do benzodiazepines, phenobarbital, propoxyphene, and chloral hydrate fall into?
C-IV
what drug schedule do codeine cough syrups, and lomotil fall into?
C-V
how much of a C-II drug can a practitioner CALL in an emergency?
72h
what is the main contributor to life threatening conditions i.e. stroke, heart failure, coronary artery disease and renal failure
hypertensive disease
what is described as mild hypertension
if diastolic is 90-105
what is described as moderate hypertension?
diastolic is 105-120
what is described as severe hypertension
diastolic is above 120
if diastolic BP is sustained above what level that requires immediate medical attention
130
what % of HT patients have HBP secondary to some known, underlying organic prob?
10%
pts w/ HT of unknown etiology are referred to as having what type of HT?
"essential"
the ultimate aim of anti-HT therapy is to produce a sustained lowering of BP by reducing what?
total peripheral resistance (TPR)
renin is secreted by?
juxtaglomerular cells
aldosterone is secreted by?
adrenal cortex
what happens when aldosterone is secreted?
kidney conserves/reabsorbs Na and H20 to increase fluid volume-> increases BP
what are 3 general examples of vasodilators used in HT?
older oral vasodilators, ca blockers, parenteral vasodilators
what are 2 general examples of angiotensin antagonists?
ACE inhibitors and receptor blockers
what are the 3 types of angina/
1. atherosclerotic 2. vasospastic 3. unstable
accumulation of metabolites in striated heart muscle produced in heart when there is inadequate tissue oxygen supply causes what
angina
what is the most frequent cause of angina?
atherosclerotic plaques in large coronary arteries
what condition does the buildup of atherosclerotic plaques in large coronary arteries refer to?
classic/atherosclerotic angina
what causes prinzmetal's or variant angina?
transient spasms of coronary arteries
acute coronary event occurring at rest caused by episodes of increased arterial vasoconstriction in assoc w/ bloot clots occurring in vicinity of arterial occlusion is called
unstable angina
what are the 2 major categories of drugs used in angina pectoris?
vasodilators and cardiac depressants
vasodilators in angina can be broken down into waht 2 groups?
nitrates and calcium blockers
cardiac depressants in angina can be broken down into what 2 groups?
ca blockers and beta blockers
examples of nitrates
nitroglycerin and isosorbide dinitrate
examples of ca channel blockers
nifedipine, verapamil, diltiazem
examples of beta-adrenergic antagonists
propranolol
what drug is taken sublingually, releases NO systemically and dilates all vessels
nitroglycerine
what drug can be taken orally and works the same as nitroglycerine
isosorbide ninitrate
which of the Ca channel blockers is NOT a cardiac depressant?
nifedipine
which of the Ca channel blockers is VERY cardiosuppressant
verapamil
which Ca channel blocker has some cardiac depressant and some vasospastic qualities
diltiazem
T/F: nitrates work against regular and vasospastic angina
T
T/F: all Ca channel blockers are vasorelaxants
T
In CHF, which side of the heart is affected when blood and fluid backs up into lungs producing SOB, being tired and having a cough
left
in CHF, which side of heart is affected when fluid builds up in veins causing peripheral (pedal) edema
right
which cardiac glycoside increases the contractility of the heart
digoxin
how does digoxin work?
1. inhibits Na/K-ATPase pump 2. causes elevated [Na] intracellular 3. heart shifts to Na/Ca pump causing increased [Ca] intracellular 4. increased cardiac contraction
examples of diuretics for CHF
chlorotiazide and furosemide
example of beta-adrenoceptor agonists for CHF
dobutamine
examples of angiotensin antagonists
ACE inhibitor - lisinopril and angiotensin receptor agonist - losartan
examples of beta-adrenoceptor antagonists
metoprolol and carvedilol
which CHF drugs are used to treat both CHF and HBP
chlorothiazide, lisinopril, and losartan
what are the 2 types of arrhthmias
1. atrial tachycardia 2. ventricular arrhthmias
what types of drugs are used to treat atrial tachycardia
beta-adrenergic antagonists
examples of beta-adrenergic antagonists
propranolol and esmolol
what type of drugs used to tx ventricular arrhthmias
local anesthetics (lidocaine)
which drug blocks beta adrenergic receptors for the long run: propranolol or esmolol?
propranolol
why is esmolol short acting?
b/c broken down by enzymes in RBC's
what is the cause of about 1 in 6 strokes
atrial fibrillation
what limits cardiac output and exercise with atrial flutter/fibrillation?
rapid ventricular response rate and increase in HR w/ exercise
what is the principal goal of antiarrhythmic drug therapy in atrial flutter/fibrillation
reduction in ventricular rate and improvement in exercise tolerance
how do you reduce ventricular rate and improve exercise tolerance?
1. conversion of atrial flutter to atrial fibrillation 2. slowing conduciton and prolongation of refractoriness w/in AV node 3. conversion of atrial fibrillation to sinue rhythm
T/F: atrial flutter is more common than atrial fib
F
how can you convert atrial fib to sinus rhythm safely
w/ DC cardioversion or IV ibutilide
T/F: the maintenance of sinus rhythm is possible in pts w/ organic heart disease
F
what is most appropriate long-term means of slowing ventricular HR and supressing increase in AV nodal conduction accompanying exercise
depression of AV nodal function
exogenous chemical agent used to modify physiology
drug
use of a chemical agent for therapeutic purposes
pharmacology
adverse affect of a drug
toxicology
therapeutic goal
mimic a normal physiology or inhibit an abnormal physiology
4 descriptors of drug action
symptomatic, psysiological, cellular, molecular
what body does to drug is called
pharmacokinetics
effect of drug on body (drug action)
pharmacodynamics
what are the order of drug receptor bonds in order from strongest to weakest
covalent - ionic - hydrophobic
distribution of hydrophilic drugs through large aqueous compartments - driven by the concentration gradient of permeating drug
aqueous diffusion
distribution of hydrophobic drugs across membranes
lipid diffusion
what is the most important limiting factor in drug distribution
lipid diffusion
proteins in the membrane that help transport compounds that are too insoluble in lipids or too large to go through membranes
special carriers
only what types of drugs can pass through membranes?
uncharged
whether a drug is charged or uncharged is determined by:
1. whether drug is a weak acid or a weak base 2. pH of compartment
the percentage of drug protonated can be quantified by what?
henderson-hasselbach equation
which type of receptor mediates the actions of endogenous chemical agents
regulatory proteins (opiod receptors - morphine)
what are the 4 types of receptors
1. regulatory proteins 2. enzymes 3. transport proteins 4. structural proteins
example of enzyme receptor
dihydropolate reductase - methotrexate
example of transport proteins receptors
Na/K ATPase - digitalis glycosides
example of structural protein receptor
tubulin - colchicine
concentration of drug needed to achieve 50% of max effect
EC50
max drug response
Emax
max num of binding sites/receptors
Bmax
concentration of drug required for 50% of binding sites to be occupied
Kd
which type of antagonist reversibly binds to a receptor
competitive antagonist
which type of antagonist binds to receptor and changes maximal effect
irreversible antagoist
with a competitive antagonist, what do you need to do to produce same physiologic response
increase concentration of agonist
what happens if you increase concentration of agonist w/ an irreversible antagonist
doesn't change physiological response
agonists that do not produce max effect produced by other agonists
partial agonists
what happens to Emax and EC w/ irreversible antagonists
Emax decreases and Ec doesn't change
what happens if you give increasing concentrations of partial agonist?
attenuate response of full agonist
which drug is more potent: the one that produces a greater response or the one that has a lower EC50?
lower EC50 (less of this drug is required to produce the 50% max response of a larger dose of the other drug
which drug has a greater efficacy: the drug that produces a greater response or the one with a lower EC50
produces a greater response
concentration at which 50% of the patients achieve he desired response
median effective dose (ED50)
concentration that is toxic to 50% of the patients
median toxic dose - TD50
concentration that is lethal to 50% of pts
median lethal dose - LD50
relationship b/w the amount of drug required to produce the desired effect to amt that produces an undesired effect
therapeutic index/therapeutic window (ratio of ED50 to TD50)
individual, unusual response to a drug
idiosyncratic
diminished/increased drug effect than what is seen in most indivduals
hyperreactive/hyperresponsive
allergic or immunologic responses to drugs
hypersensitive
decreased responsiveness to a drug w/ repeated administration
tolerance
rapid dimishment of responsiveness to a drug
tachyphylaxis
which route of administration has highest bioavailability (F%)
IV
fraction of unchanged drug reaching systemic circulation following administration
bioavailability
amt of drug metabolized by liver
first-pass effect
apparent volume into which a drug homogeneously distributes in the body
volume of distribution (Vd)
T/F: rate of clearance increases w/ drug concentration
T
the rate of elimination in first order elimination is dependent on what
drug concentration
most drugs follow what order of elimination
first order elimination
T/F: zero order elimination is dependent on drug concentration
F
examples of zero order
ethanol, phenytoin, aspirin
how do Vd and Cl affect half life
increase Vd=increase half life
immediate effects of drugs are related to what
plasma concentrations
delayed effects of drugs are due to what
turnover of physiologic substrate or slow distribution of drug (ex warfarin)
since drug binding is irreversible in cumulative effects, what is required for effect?
dosing over time
conversion of compounds from lipophilic substances to polar
biotransformation
biotransformation has what 2 actions
1. terminates biological activity of a drug 2. facilitates elimination of drug from body
over 90% of drugs have what 2 organs as their sites of biotransformation?
liver and kidneys
phase I metabolism
introduce/unmask a functional group
phase II metabolism
conjugation of a polar group
what types of reactions take place in Phase I cytochrome P450-dependent rxns
oxidations - hydroxylations, oxidative dealkylation, oxidation, deamination
what types of reactions are part of cytochrome P450-independent rxns
dehydrogenation, reductions, hydrolysis
which cytochrome P450-dependent drugs undergo hydroxlations
propanolol, ibuprofen
which cytochrome P450-dependent drugs undergo oxidative dealkylation
morphine, caffeine
which cytochrome P450-dependent drugs undergo oxidation
thioridazine
which cytochrome P450-dependent drugs undergo deamination
amphetamine, diazepam
which cytochrome P450-independent drugs undergo dehydrogenation
ethanol
which cytochrome P450-independent drugs undergo reductions
chloramphenicol
which cytochrome P450-independent drugs undergo hydrolysis
aspirin
which phase II metabolism drugs undergo glucoronidation
morphine, acetominophen
which phase II metabolism drugs undergo acetylation
mescaline
which phase II metabolism drugs undergo glutathione conjugation
ethacrynic acid
which phase II metabolism drugs undergo glycine conjugation
nicotinic acid
which phase II metabolism drugs undergo sulfate conjugation
acetaminophen
which phase II metabolism drugs undergo methylation
epinephrine
which phase II metabolism drugs undergo water conjugation
leukotriene A4
endogenous compounds that may affect drug biotransformation
glutathione, glucuronic acid, sulfate
parasym pregang neurons project where?
to gang at or near target (usually long)
parasym postgang neurons project where?
from gang to targets (usually short)
CN3 projects to what parasym ganglion
ciliary
CN7 projects to what parasym ganglions
pterygopalatine and submandibular
CN9 projects to what parasym ganglion
otic
sympathetic system originates where
thoracolumbar spinal cord
where do sympathetic pregang project to? long or short?
project to sympathetic chain or preaortic ganglia - short projection
where do sympathetic postgang project to? long or short?
from ganglia to targets - long projections
what are the cholinergic nicotinic receptors
ganglionic (Nn) and skeletal muscle (Nm)
what is the agonist/antagonist for ganglionic receptors
agonist-nicotine, antag-hexamethonium
what is the agonist/antag for Nm receptors
ag-nicotine, antag-curare
what is the ag/antag for muscarinic receptors
ag-muscarie, antag-atropine
what is the ag/antag for alpha adrenergic receptors?
ag-ephedrine, antag-phenoxybenzamine
what is the ag/antag for beta adrenergic receptors
ag-ephedrine, antag-propanolol
when the parasym pregang synapses, what NT is released
Ach
when the parasym postgang synapses, what NT is released
Ach
when the sym pregang synapses, what NT is released
Ach
when the sym postgang synapses, what NT is released
NE
what does the activation of the adrenal medulla cause?
release of Epi or NE, general sympathetic activation
where do parasyms come from to activate everything from belt down
sacral spinal nerves
outflow from CNS in sym vs parasym
sym-thoracolumbar (T1-L2)
location of ganglia in sym vs para
sym-close to CNS
where are B receptors located
B1-heart, B2-bv's, lungs, bronchi, B3-adipose
what happens to HR, force of ventricular contraction, and conduction velocity when innerv by sym vs para
sym- all 3 are increased, para-HR and conduction velocity are decreased and no effect on force of vent contraction
what happens to iris when innervated by sym vs para
sym-pupils dilate, para-constrict
what happens to bronchi when innerv. by sym vs para
sym-dilated (B2), para-constricted
does sym or para stimulate GI tract
para
what happens to salivary glands when innerv by sym vs para
sym-stimulated (viscous), para-stimulated (watery)
what does receptor "m" mean
cholinergic inhibitory, stimulated by ACh: vasodilation
what receptors are found in skeletal muscle, innerv or non-innerv?
a-innerv, B2-non-innerv, m-both
what receptors are found in gut, liver, spleen- innerv or non-innerv?
a-innerv, B2-non-innerv, m-non-innerv
what receptors are found in skin-innerv or non?
a-innerv, B2 - a few innerv, m-a few innerv
what receptors are found in coronaries - innerv or non?
a-innerv, B2-non, m-non
where do parasyms project to in the eye?
ciliary muscle and sphincter muscle or iris
where do syms project to in the eye?
dilator muscle of iris
sympathetic innervation of bv's are usually mediated by which receptor
b2
which wins out? vascular resistance or CO?
vascular resistance
T/F: all somatic nerves release ACh
T
what happens to BP if ACh is given in small doses
decrease in BP
what happens to BP if ACh is given in a small dose after atropine is given
no effect on BP b/c atropine blocked musc receptors
what happens to BP if ACh is given in a large dose after atropine is given
big increase in BP b/c ACh is now hitting nicotinic receptors
when nicotine is given repeatedly or in large doses, why does it block nicotinic receptors?
sustained depolarization
agents that indirectly enhance cholinergic function
anticholinesterases
anticholinesterases only have an effect on which type of receptors
only at innervated ACh receptors
main clinical use for AChase inhibitors
tx myasthenina gravis and glaucoma
what is used to tx atropine poisoning
physostigmine
what drugs are antidotes to curare
AChase inhibitors
main clinical use for physostigmine
topical for glaucoma
what actions does physostigmine have in eye
miosis and contraction of ciliary muscle
how is it imp that physostigmine is not highly charged
can enter CNS
what drug is the best choice for atropine poisoning
physostigmine
neostigmine directly stimulates nicotinic sites where?
on skeletal muscle endplates
the dual action of neostigmine makes it a drug of choice for what
myasthenia gravis
pyridostigmine is used to tx what
myasthenia gravis
which AChase inhibitors are CNS acting and used to tx cognitive dysfunction in Alzeimer's dx
donepezil and tacrine
which cholinomimetic can stimulate salivary flow
pilocarpine
which drug is used to distinguish myasthenia gravis from AChE blockade (cholinergic crisis)
edrophonium
Can be used as topical treatments for glaucoma,but most are insecticides and military nerve agents
irreversible AChases
what insecticide is converted in the liver to the active form
parathion
sarin and soman
Gaseous nerve agents used in chemical warfare. NOT reversible by cholinesterase reactivators
Used to treat muscarinic intoxication and intoxication with some irreversible organophosphate anti-cholinesterases
pralidoxime (2-PAM)
paralysis of the NMJ's due to excessive ACHASE inhibition
cholinergic crisis
paralysis of the NMJ's due to insufficient ACHASE inhibition
myasthenic crisis
what would muscarinic receptor blockers do to parasym innervated organ
lack of motility and secretions
what is the diff b/w atropine and scopolamine
atropine produces CNS excitation, scopolamine causes CNS depression
low doses of atropine cause what
slight paradoxical bradycardia due to CNS stim
higher doses of atropine cause what
they block peripheral muscarinic receptors at heart to produce tachycardia
atropine effect on heart
low doses- bradycardia, higher doses-tachycardia
atropine effect on skin
inhibition of sweating, hot dry skin
atropine effect on eye
mild to marked mydriasis, mild to marked cycloplegia
atropine effect on viscera
dry mouth, decreased saliv, reduced GI and urinary tone
atropine effect on CNS
confusion, restlessness, excitement --- hallucinations, delirium, coma
characteristics of atropine poisoning
hot dry skin, dilated pupils, maniacal behavior
what is a less potent and much shorter acting muscarinic antag
tropicamide
use for tropicamide
opthalmological exams
ipratropium is mainly used in what type of pts
COPD (asthma secondarily)
how does ipratropium work in COPD pts
bronchodilation w/o affecting bronchial secretions
musc blocker that crosses BBB and tx parkinson's
benztropine
musc blocker that tx bladder spasms postop that decreases bladder tone and improves continence
oxybutynin
will innervated or non-innerv receptors be affected by ganglionic transmission
innerv
ganglionic stim will excite which organs
all autonomic organs
which neurons/cells does nicotine stimulate
postgang neurons and adrenal chromaffin cells
prototype ganglionic blocking drug
hexamethonium
nicotine blocks autonomic ganglia when given in high doses due to what
sustained depolarization of postgang cells
T/F: hexamethonium blocks NMJ's
F - does not
predominant tone and effect of ganglionic blockade at arterioles
sypathetic - vasodilation
predominant tone and effect of ganglionic blockade at veins
sym - dilation
predominant tone and effect of ganglionic blockade at heart
parasym - tachycardia
predominant tone and effect of ganglionic blockade at iris
parasym - mydriasis
predominant tone and effect of ganglionic blockade at ciliary muscle
parasym - cycloplegia
predominant tone and effect of ganglionic blockade at GI tract
parasym - reduced tone and motility
predominant tone and effect of ganglionic blockade at urinary bladder
parasym - urinary retention
predominant tone and effect of ganglionic blockade at salivary glands
parasym - xerostomia
predominant tone and effect of ganglionic blockade at sweat glands
sym - anhidrosis
drugs can cause neuromuscular blockade by one of what 2 ways
1. compete w/ ACh for nicotinic receptors 2. sustained depolarization
prototype NMJ blocking drug
curare
how do local anes and tetrodotoxin inhibit nerve action potential
interfere w/ Na transport
what inhibits ACh release by blocking reuptake of choline into nerve ending
hemicholinium
what inhibits ACh release by blocking ACh release mech
botulinum toxin
what enhances ACh release
spider venom (latrotoxin) and catecholamines
what inhibits binding of ACh to receptor
NMJ blockers and snake alpha-bungarotoxins
T/F: competitive NMJ blocking drugs enter BBB
F: highly charged
T/F: competitive NMJ blocking drugs produce reduction in pain sensation
FALSE
gold standard of competitive NMJ blockers
tubocurarine
main side effect for tubocurarine - eliminated by?
hypotension - liver
how is doxacurium different from tubocurarine?
doesn't block autonomic ganglia or release histamine
organ of excretion for doxacurium
kidney
NMJ blocker w/ steroid nucleus
pancuronium
how is pancuronium same as doxacurium
no autonomic blockade no histamine release
more potent, shorter acting analog of pancuronium
vecuronium
only drug used clinically as depolarizing NMJ blocker
succinylcholine
how is succinylcholine inactivated
rapidly metabolized by plasma and liver pesudocholinesterases
drug of choice to relax laryngeal muscles before intubation
succinylcholine
what syndrome can appear due to succinylcholien
malignant hyperthermia
benzodiazeepine that enhances GABAergic inhibition in CNS
diazepam
side effect for diazepam
sedation at effective dose
function of diazepam
reduces all muscle spasms
drug that causes decrease in release of Glu and decrease in skeletal muscle tone while experiencing less sedation than diazepam
baclofen
drug used for relief of spasticity caused by MS and spinal cord injury
baclofen
drug that is CNS alpha2 stim, reduces skeletal muscle spasticity, causes less hypotension, but causes sedation and dry mouth
tizanidine
related to tricyclic antidepressants, tx muscular spasms assoc w/ musculoskeletal conditions
cyclobenzaprine
spasmolytic w/ side effects that are atropine-like and not effective in tx for spasms from spinal cord injury or cerebral palsy
cyclobenzaprine
Acts directly on skeletal muscle abd disrupts excitation-contraction coupling by blocking release of Ca needed for contraction
dantrolene
drug of choice for treating malignant hyperthermia
dantrolene
main adverse effects of dantrolene
generalized msucle weakness and sedation
muscle relaxant to tx generalized spastic disorders (cerebral palsy), effective longterm blockade of trans to muscle
botulinum toxin
drugs to tx paralytic ileus
physostigmine and bethanechol
drug to tx atony of bladder
bethanechol
drug to tx glaucoma
physostigmine, pilocarpine
drug to tx myasthenia gravis
neostigmine
drug to reverse NMJ blockade
neostigmine + atropine
preanesthetic medication
scopolamine
drug used in opthalmological exam
tropicamide
drug to tx motion sickness
scopolamine
4 sympathomimetics: catecholamines
1. Epi 2. NE 3. isoproterenol 4. dopa
6 non-catecholamine sympathomimetics - alpha agonists
1. tyramine 2. amphetamine 3. methamphetamine 4. methylphenidate 5. ephedrine 6. phenylephrine
5 non-catecholamine sympathomimetics - beta adrenoceptor agonists
1. terbutaline 2. albuterol 3. salmeterol 4. ritodrine 5. dobutamine
sympathomimetics: inhibitors of reuptake
1. cocaine 2. tricyclic antidepressants
CNS acting sympatho-inhibitory agents
1. clonidine 2. a-methyl DOPA
alpha-adrenoceptor blockers
1. phentolamine 2. phenoxybenzamine 3. prazosin 4. doxazosin 5. terazosin
beta-adrenoceptor blockers
1. propanolol 2. metoprolol 3. atenolol 4. pindolol 5. timolol 6. esmolol
alpha and beta-adrenoceptor antagonist
1. labetalol 2. carvedilol
most imp regulation of NE and Epi synthesis
end product inhibition
those neurons that synthesize, store, and release catecholamines
adrenergic neurons
what do adrenergic nerves regulate the function of
heart and blood vessels, visceral smooth muscle and glandular tissue
catecholamine molecule
amine with benzene ring with OH sub on 3 and 4
catecholamine released from post ganglionic sympathetic nerve fibers in peripheral nervous system
NE
what is released from chromaffin cells of the adrenal medulla
epi
is epi released from peripher nerves
no
NT released from neurons in CNS
epi and dopamine
aa that cat are derived from
tyrosine
tyrosine is converted to what
DOPA
DOPA is converted to..
dopamine
dopamine is converted to ? where?
NE in the storage vesicle
in the granule, NE is associated with ? and ?
ATP and a protein called chromoganin
what stimulates NE systhesis
increased neuronal activity by increasing tyrosine hydroxlase
which receptors can NE activate when released
a1, a2, or b1 on postsyn cell
rate limiting step in syn of NE in nerve terminals and epi in adrenal gland
Tyrosine hydroxylase (conversion of tyrosine to dopa)
what happens when action potential invades nerve terminal
influx of calcium
what enzymatically inactivates NE that enters postsyn cell
COMT (uptake II)
T or F Synaptic NE can also stimulate presynaptic alpha 2 receptors that act as a neg feedback to inhi further release of NT
TRUE
which enzymes in liver can inactivate NE that diffuses into systemic circ
MAO and COMT
what is the most important mech for the term of NE action, and what is it called?
re uptake back into the nerve terminal 60-90% - uptake I
if NE is not sequestered by the storage vesicle in the presyn term, what and where will it be inactivated?
MAO
MAO is assoc w/ what organelle
mitochondria
COMT is in high concen where
cytosol of liver cells
MAO is in high concentration in what organ
liver
what metabolite is sometimes screened for in patients suspected of harboring a tumor of the adrenal gland called pheochromocytoma
VMA
alpha 1 receptors are located where
post junctionally at most symp neuroeffector synapses except the heart and JG cells of the kidney
activation of alpha 1 receptors is excitatory or inhibitory
excitatory ( increased bp, pupil dilation via contraction of the radial muscle of iris, piloerection, viscous salivation, contraction of GI an durinary bladder
what systems are inhibited with alpha 1 receptors
Gi and urinary smooth muscle
what is the function of alpha 2 receptors
same as a1 + prejunc mem to provide a neg feedback mechanism to prevent excessive release of NE
where are the 2 places that beta 1 receptors are found
heart and JG apparatus of the kidney
what are the only receptors potently stimulated by NE
beta 1 receptors
what are beta receptors responsible for?
increasing heart rate, conduction, ad contractile force as well as the neural release of renin from the JG cells
what are the only innervated beta adrenoreceptors
beta 1
do beta 2 adrencoreceptors prefer epi or NE
epi
what is the result of beta 1 receptor stimulation
increased cardiac output
what will beta 2 receptors activation cause?
vascular vasodilation and reduction of bp, relaxation of smooth muscle of bronchial tree, inhibition of GI and urinary tract smooth muscle, and relaxation of gravid uterus
do b2 act directly on innerv or noninnerv
non-innerv
why are catechols not effective by oral route
b/c substrates for MAO and COMT
sympathomimetic drugs
mimic sympathetic function
2 ways sympathomimetics mimic sym function
directly by stim the adrenergic rec or indirectly allowing more NE to be present in the synaptic cleft
isoproterenol is a synthetic catecholamine iwth preference for which receptors?
all b adrenoreceptors
epinephrine works on what receptors
all alpha and beta
alpha adrenoreceptor activation is...
vasoconstriction
beta adrenoreceptor activation is...
vasodilation
are there more alpha or beta receptors
alpha (but less sensitive to epi)
which receptors are more sensitive to epi
beta (even though fewer in number)
which receptor is fewer in number, more sensitive to epi and has longer lasting effects
beta receptors
which receptor is greater in number, less sensitive to epi and has shorter duration of effects
alpha
what would the effect of a small bolus of epi be?
vasodilation due to beta2 response
what would the effect of a large bolus of epi be?
vasoconstriction and a pressor response -- alpha response
reversal of pressor response to depressor response after a-adrenoceptor blockade is called
epi reversal
when epinephrime stimulates beta 1 receptors, what is the effect on the heart?
increase heart rate and contractility
epi effect on b2 receptors
relax bronchial smooth muscle in the treatment of asthma and for treating bronchospasm during anaplylactic shock
epi acts via beta adrenoreceptors mechanisms to increase adenylate cyclase activity to form cAMP that has what metabolic effects?
1) increase plasma glucose by breakdown of liver glycogen 2) inhibits synthesis of glycogen 3) stimulates gluconeogensis 4) breaks down fats to fatty acids
NE acts on which receptors?
all alpha, and beta 1
a bolus injection of NE will have what outcome?
increase blood pressure due to both vasoconstriction and increased cardiac output
which has a longer 1/2 life: epi or NE?
epi
drug that potently acts on all b receptors to relieve bronchoconstrictive states
isoproterenol (ISO)
untoward side effects from isoproterenol
cardiac
A CNS defect in dopaminergic mechanisms results in what disease
parkinsons
dopamine has what effect on the kidney
vasodilation
at higher doses, what is the effect of dopamine
stimulate alpha and beta receptors to increase bp, and cardiac output
what drug, found in certain foods like beer, cheese, and wine, can cause a hypertensive crisis in pts taking MAO inhibitor to treat depression?
tyramine
tyramine causes massive release of what from sym nerve endings
NE
why is injestion of tyramine not harmful?
readily inactivated by MAO in gut and liver
what must happen for tyramine effect to manifest
nerve endings must contain NE
amphetamine and meth are what kinds of drugs
indirect sympathomimetics
why are amphetamines so toxic and lethal?
bc the ED50 increases but the LD50 remains the same
chronic toxicity to amphetamines can mimic what?
paranoid schizophrenia
what is the major finding at autopsy of amphetamine overdose>
cerebral hemorrhage that may be secondary to intense vasoconstriction of blood vessels
what are the 2 valid reasons for amphetamine use
1) hyperkinetic children to increase attention span 2) narcolepsy
methylphenidate is a mild CNS stimulant that causes what?
gerenalized CNS excitation that may cause convulsions
primary use of methylphenidate (Ritalin)
ADHD
medical uses for ephedrine
bronichial asthma (b effect) ear and nasal congestion OTC, opthalmology for short lasting mydriasis without cycloplegia
medical uses for phenylephrine
opthal for mydriasis, nasal spray
terbutaline is a modification of what drug
metaproterenol
which is longer acting: metaproterenol or terbutaline
terbutaline
which drug, terbutaline or metaproterenol, has a higher incidence of cardiac side effect?
terbulatine
drug used to prevent premature child birth
terbutaline
drug for astham, similar to terbutaline
albuterol
long acting beta 2 agonist that is used to treat bronchial asthma
salmeterol
beta 2 agonist that is approved for use to relax smooth muscle of the uterus and to delay premature child labor
ritodrine
what is the preferred method of delivery for ritrodrine
orally, even though only 30% effective
synthetic catecholamine analog of dopamine that is a highly selective beta 1 adrenoreceptor
dobutamine
does dobutamine stimulate dopamine receptors?
no
what is the effect of dobutamine on the heart?
increased cardiac output without the vasoconstriction associated with NE that would result in increased cardiac work
what drug is used for short term treatment of cardiac insufficiency
dobutamine
what is the effect of sympatho-inhibitory agents on the peripheral nervous system
vasoconstriction by alpha 2's, short lived
what is the effect of sympatho - inhibitory agents on the CNS?
hypotensive action that lasts for hours
those neurons that synthesize, store, and release catecholamines
adrenergic neurons
what do adrenergic nerves regulate the function of
heart and blood vessels, visceral smooth muscle and glandular tissue
catecholamine molecule
amine with benzene ring with OH sub on 3 and 4
catecholamine released from post ganglionic sympathetic nerve fibers in peripheral nervous system
NE
what is released from chromaffin cells of the adrenal medulla
epi
is epi released from peripher nerves
no
NT released from neurons in CNS
epi and dopamine
aa that cat are derived from
tyrosine
tyrosine is converted to what
DOPA
DOPA is converted to..
dopamine
dopamine is converted to ? where?
NE in the storage vesicle
in the granule, NE is associated with ? and ?
ATP and a protein called chromoganin
what stimulates NE systhesis
increased neuronal activity by increasing tyrosine hydroxlase
what is the rate limiting step in epi synthesis
Tyrosine hydroxylase
what happens when action potential invades nerve terminal
influx of calcium
how else can NE be released
sympathomimetic amines such as tyramine and amphetamine
T or F Synaptic NE can also stimulate presynaptic alpha 2 receptors that act as a neg feedback to inhi further release of NT
TRUE
What does COMT do
catechol - o - methyltransferase enzymatically inactivates NE that enters the post synaptic cell
what is this referred to as?
uptake II
what is NT degraded by if it diffuses into the systemic circulation
MAO and COMT in liver
what is the most important mech for the term of NE action
re uptake back into the nerve terminal 60-90%
if NE is not sequestered by the storage vesicle in the presyn term, what and where will it be inactivated?
MAO in the mitochondria
MAO is in high concentration in what organ
liver
what metabolite is sometimes screened for in patients suspected of harboring a tumor of the adrenal gland called pheochromocytoma
VMA
alpha 1 receptors are located where
post junctionally at most symp neuroeffector synapses except the heart and JF cell so fthe kidney
activation of alpha 1 receptors is excitatory or inhibitory
excitatory ( increased bp, pupil dilation via contraction of the radial muscle of iris, piloerection, viscous salivation, contraction of GI an durinary bladder
what systems are inhibited with alpha 1 receptors
Gi and urinary smooth muscle
what is the function of alpha 2 receptors
provide a neg feedback mechanism to prevent excessive release of NE
where are the 2 places that beta 1 receptors are found
heart and JG apparatus of the kidney
what are the only receptors potently stimulated by NE
beta 1 receptors
what are beta receptors responsible for?
increasing heart rate, conduction, ad contractile force as well as the neural release of renin from the JG cells
what are the only innervated beta adrenoreceptors
beta 1
do beta 1 adrencoreceptors prefer epi or NE
epi
what is the result of beta 1 receptor stimulation
increased cardiac output
what will beta 2 receptors activation cause?
vascular vasodilation and reduction of bp, relaxation of smooth muscle of bronchial tree, inhibition of GI and urinary tract smooth muscle, and relaxation of gravid uterus
sympathomimetic drugs
mimic sympathetic function
2 ways they can do this
directly by stim the adrenergic rec or indirectly allowing more NE to be present in the synaptic cleft
isoproterenol is a synthetic catecholamine iwth preference for which receptors?
all b adrenoreceptors
epinephrine works on what receptors
all alpha and beta
is epi effective when given orally
no
alpha adrenoreceptor activation is...
vasoconstriction
beta adrenoreceptor activation is...
vasodilation
are there more alpha or beta receptors
alpha (but less sensitive to epi)
which receptors are more sensitive to epi
beta (even though fewer in number)
which receptor is fewer in number, more sensitive to epi and has longer lasting effects
beta receptors
which receptor is greater in number, less sensitive to epi and has shorter duration of effects
alpha
what would the effect of a small bolus of epi be?
vasodilation due to beta response
what would the effect of a large bolus of epi be?
vasoconstriction and a pressor response -- alpha response
when epinephrime stimulates beta 1 receptors, what is the effect on the heart?
increase heart rate and contractility
epi works on beta 2 receptors to do what?
relax bronchial smooth muscle in the treatment of asthma and for treating bronchospasm during anaplylactic shock
epi acts via beta adrenoreceptors mechanisms to increase adenylate cyclase activity to form cAMP that has what metabolic effects?
1) increase plasma glucose by breakdown of liver glycogen 2) inhibits synthesis of glycogen 3) stimulates gluconeogensis 4) breaks down fats to fatty acids
NE acts on which receptors?
all alpha, and beta 1
a bolus injection of NE will have what outcome?
increase blood pressure due to both vasoconstriction and increased cardiac output
which has a longer 1/2 life: epi or NE?
epi
isoproterenol has what effect
potently acts on all B receptors to relieve bronchoconstrictive states
A CNS defect in dopaminergic mechanisms results in what disease
parkinsons
dopamine has what effect on the kidney
vasodilation
at higher doses, what is the effect of dopamine
stimulate alpha and beta receptors to increase bp, and cardiac output
what drug, found in certain foods like beer, cheese, and wine, can cause a hypertensive crisis in pts taking MAO inhibitor to treat depression?
tyramine
amphetamine and meth are what kinds of drugs
indirect sympathomimetics
why are amphetamines so toxic and lethal?
bc the ED50 increases but the LD50 remains the same
chronic toxicity to amphetamines can mimic what?
paranoid schizophrenia
what is the major finding at autopsy of amphetamine overdose>
cerebral hemorrhage that may be secondary to intense vasoconstriction of blood vessels
what are the 2 valid reasons for amphetamine use
1) hyperkinetic children to increase attention span 2) narcolepsy
methylphenidate is a mild CNS stimulant that causes what?
gerenalized CNS excitation that may cause convulsions
primary use of methylphenidate (Ritalin)
ADHD
ephedrine
agonist on all alpha and beta receptors in the body
medical uses for ephedrine
bronichial asthma (b effect) ear and nasal congestion OTC, opthalmology for short lasting mydriasis without cycloplegia
phenylephrine has what primary effect
alpha adrenergic effects
medical uses for phenylephrine
opthal for mydriasis, nasal spray
terbutaline is a modification of what drug
metaproterenol
which is longer acting
terbutaline
what drug is used to prevent premature child birth
terbutaline
which drug, terbutaline or metaproterenol, has a higher incidence of cardiac side effect?
terbulatine
drug for astham, similar to terbutaline
albuterol
long acting beta 2 agonist that is used to treat bronchial asthma
salmeterol
beta 2 agonist that is approved for use to relax smooth muscle of the uterus and to delay premature child labor
ritodrine
what is the preferred method of delivery for ritrodrine
orally, even though only 30% effective
synthetic catecholamine analog of dopamine that is a highly selective beta 1 adrenoreceptor
dobutamine
does dobutamine stimulate dopamine receptors?
no
what is the effect of dobutamine on the heart?
increased cardiac output without the vasoconstriction associated with NE that would result in increased cardiac work
what drug is used for short term treatment of cardiac insufficiency
dobutamine
what is the effect of sympatho-inhibitory agents on the peripheral nervous system
vasoconstriction by alpha 2's, short lived
what is the effect of sympatho - inhibitory agents on the CNS?
hypotensive action that lasts for hours
potent alpha 2 stimulant that readily penetrates into the CNS where it acts to decreases sympathetic tone to blood vessels and the heart
also may enhance vagal outflow
drug that has been used in treating the excessive symp activity exp during withdrawal from opoid and ethanol addiction
clonidine
what is a problem exp with withdrawal from clonidine itself?
dramatic rebound hypertension following cessation of clonidine therapy
what type of drug is alpha methyl dopa
antihypertensive - alpha 2
why does a methylnorepinephrine have a longer half life than NE?
not metabolized by MAO
problems exp with alpha blockade
postural hypotension, reflex tachycardia
what will alpha blockade produce?
reduction in bp
what are teh 4 most common side effects seen with alpha blockade?
hypotension, reflex tachycardia, failure to ejaculate, nasal congestion
what are the 5 alpha antagonists?
phentolamine, phenyoxybenzamine, prazosin, terazosin, doxazosin
what is phentolamine used for?
screening test for pheochromocytoma
what does phentolamine cause many false positive reactions
bc it releases histamine
which drug has a higher preference for alpha 1 receptors? phentolamine or phenxoybezamine
phenoxybenzamine
why does phenoxybenzamine have a longer duration?
because it binds comp at first, then after 30-60 min binds irreversibly by non competitive inhibition
why is prazosin useful in treating some hypertensive patients?
does not produce the large tachycardia seen with other alpha antagonists
what drugs are commonly used to treat the symptoms of benign prostatic hypertrophy?
terzosin and doxazosin
what is unique about terazosin and doxazosin?
longer half life permitting once per day dosing
beta 1 blockage will produce what outcomes
reduction in heart rate, myocardial contractility, conduction velocity and renin release
beta 2 blockade will produce what effect
prevention of relaxation of bronchial smooth muscle
beta blockers are used to treat what 5 conditions?
hypertension, angina, certain cardiac arrythmias, and CHF
what drugs are used to decrease the risk of myocardial reinfarction in patients at risk
beta blockers
what are 2 other uses of beta blockers?
glaucoma and prevention of migraines
when should beta blockers be used with caution? what 3 conditions?
bronchial astham, COPD, diabetes mellitus
what should beta blockers be used with caution in pts with diabetes?
they wont experience the usual expected tachycardia warning them of a hypoglycemic state
which beta blockers are non selective?
nadolol, timolol, propanolol, pindolol (never trick pink panthers)
which beta blockers are cardioselective?
betaxolol, esmolol, atenolol, metaprolol (BEAM)
which beta blocker is almost as potent of a LA as lidocaine?
propanolol
what is the effect of metoprolol?
selective for B1, so reduces plasma renin, but not as much effect on bronchial smooth muscle
what are the side effects of metoprolol?
fatigue, insomnia, headache, dizziness
what about atenolol is different from metoprolol?
much longer half life and less side effects
should atenolol be administered to pts with severe renal disease?
NO
what drug has Intrinsic sympathomimetic activity?
pindolol
which beta blocker is best tolerated during exercise?
pindolol, bc it is intrinsic sympathetic activity
what is the clinical use of timolol?
topical to relieve intraocular pressure
which drug is the treatment of choice for open angle glaucoma?
timolol
what beta blocker is suitable for patients where compliance is a particular problem?
nadolol- bc of long duration
why does esmolol have a very short half life?
rapid hydrolysis by cytosolic red blood cell esterases
what is the clinical use of esmolol?
acute emergency control of ventricular heart rate in pts with atrial fibrillation
why is esmolol used in acute emergency control of ventricular heart rate?
bc of its short half life of 10-15 min
which drug has both alpha and beta blocking properties?
labetalol
which blocking effect predominates?
alpha blocking effect predominates
which drug acts as a "free radical scavenger" in addition to being used for CHF?
carvedilol
cholinomimetic drugs that are choline esters
1. acetylcholine 2. methacholine 3. carbachol 4. bethanechol
cholinomimetic drugs that are naturally occurring
1. nitotine 2. muscarine 3. pilocarpine
2 ways that cholinomimetic drugs enhance cholinergic function
1. activate Ach directly through nicotinic and muscarinic receptors and 2. block AchE
function of anticholinesterase agents
indirectly increase cholinergic function
list reversible anticholinesterase agents
1. physostigmine 2. neostigmine 3. edrophonium 4. pyridostigmine 5. donepezil and tacrine
list anticholinesterase irreversible agents
1. parathion and malathion 2. sarin and soman
what is the cholinesterase reactivator?
pralidoxime (2-PAM)
what are the 5 basic steps in neurotransmission
1. synthesis 2. storage 3. release from presynaptic terminal 4. reception and post-syn response 5. termination of signal
examples of muscarinic blocking drugs
1. atropine 2. scopolamine 3. tropicamide 4. ipratropium 5. benzytropine 6. oxybutynin
what are 6 example of places where cholinergic synapses are found?
1. all parasyn neuroeffector junctions 2. all autonomic ganglia 3. skeletal NMJ 4. sympathetic innervation of adrenal medulla 5. sweat glands 6. brain and SC sites
non-innevated ACh receptors are generally of what type?
muscarinic
how do cholinergic drugs decrease colinergic function?
they usually occupy the receptor so the ACh molecules cannot act on them
where does synthesis of ACh occur?
cytoplasm of cholinergic nerves
enzyme that catalyzes ACh synthesis
choline acetylase
rate-limiting step of ACh synthesis
active pumping of choline into the nerve by a transport mechanism
what blocks the transport of choline into the nerve that causes an eventual inhibition of choline function?
hemocholinium (HC-3)
what causes attachment of ACh granules to fuse with nerve terminal membranes during action potential
influx of Ca
what can inhibit exocytosis of ACh into synaptic space?
anything that lowers extracellular Ca i.e. bot toxin and latrotoxin
what prevents release of ACh from nerve terminal
botulinum toxin
what causes excessive fusing of ACh granules w/ nerve mem causing excessive release and depletion of ACh
latrotoxin - spider venom
where are pseudocholinesterases found?
plasma and liver
M2 receptors are found
in heart
M3 receptors are found
peripheral autonomic organs
M1 receptors are found
in GI tract and autonomic ganglia
which muscarinic receptors are located in CNS
M1-M5
what does stimulation of muscarinic receptors in the heart do?
slows heart by increasing K permeability causing prolonged phase 4 depolarizaiton
T/F: muscarinic receps in heart directly stimulate ventricular contraction
F: they have little effect on force of vent contraction
what effect does systemic admin of ACh and muscarinic stimulants have on vascular sm
profound vasodilation and decrease BP
how does ACh cause blood vessel smooth muscle relaxation?
ACh acts on non-innerv muscarinic receptors on endothelial cells that releases EDRF (NO)
what do muscarinic agonists produce in the eye
pupillary constriction (miosis) and spasm of accommodation (cyclotonia)
what do muscarinic agonists do in the GI tract
increase tone and motility
what do muscarinic agonists do in bronchiolar smooth muscle
modest constriction
how must ACh be administered to have an effect
injected
what will be activated if ACh is given in low doses
only muscarinic receptors, much higher amts are needed to stimulate nicotinic
what drug is partially resistant to AChase, is a potent muscarinic stimulant, but has lost almost all nicotinic action of parent molecule
methacholine
why is methacholine not used as much in humans
has too many generalized muscarinic effects
what drug is very refractory to inactivation by AChase, has a potent nicotinic stimulating effects w/ little muscarinic actions, and releases ACh from cholinergic nerve endings
carbachol
what drug is used topically on eye to produce miosis and tx glaucoma
carbachol
which drug is only choline ester that has established clinical use
bethanechol
which drug is refractory to AChase inactivation and has mostly muscarinic stimuating effects, tx post-op urinary retention and atony of GI tract
bethanechol
how is bethanechol used clinically
tx esophageal reflux due to stimulatory action on lower esophageal sphincter
what are responses to injected nicotine
activation of all autonomic ganglia and stim of skeletal muscle endplate nic receptors
antidote for muscarine poisoning
atropine
which drug selectively activates muscarinic receptors, not highly charged so can cross BBB, and used to tx glaucoma
pilocarpine
T/F: all somatic nerves release ACh
T
what happens to BP if ACh is given in small doses
decrease in BP
what happens to BP if ACh is given in a small dose after atropine is given
no effect on BP b/c atropine blocked musc receptors
what happens to BP if ACh is given in a large dose after atropine is given
big increase in BP b/c ACh is now hitting nicotinic receptors
when nicotine is given repeatedly or in large doses, why does it block nicotinic receptors?
sustained depolarization
agents that indirectly enhance cholinergic function
anticholinesterases
anticholinesterases only have an effect on which type of receptors
only at innervated ACh receptors
main clinical use for AChase inhibitors
tx myasthenina gravis and glaucoma
what is used to tx atropine poisoning
physostigmine
what drugs are antidotes to curare
AChase inhibitors
main clinical use for physostigmine
topical for glaucoma
what actions does physostigmine have in eye
miosis and contraction of ciliary muscle
how is it imp that physostigmine is not highly charged
can enter CNS
what drug is the best choice for atropine poisoning
physostigmine
neostigmine directly stimulates nicotinic sites where?
on skeletal muscle endplates
the dual action of neostigmine makes it a drug of choice for what
myasthenia gravis
pyridostigmine is used to tx what
myasthenia gravis
which AChase inhibitors are CNS acting and used to tx cognitive dysfunction in Alzeimer's dx
donepezil and tacrine
which cholinomimetic can stimulate salivary flow
pilocarpine
which drug is used to distinguish myasthenia gravis from AChE blockade (cholinergic crisis)
edrophonium
Can be used as topical treatments for glaucoma,but most are insecticides and military nerve agents
irreversible AChases
what insecticide is converted in the liver to the active form
parathion
sarin and soman
Gaseous nerve agents used in chemical warfare. NOT reversible by cholinesterase reactivators
Used to treat muscarinic intoxication and intoxication with some irreversible organophosphate anti-cholinesterases
pralidoxime (2-PAM)
paralysis of the NMJ's due to excessive ACHASE inhibition
cholinergic crisis
paralysis of the NMJ's due to insufficient ACHASE inhibition
myasthenic crisis
what would muscarinic receptor blockers do to parasym innervated organ
lack of motility and secretions
what is the diff b/w atropine and scopolamine
atropine produces CNS excitation, scopolamine causes CNS depression
low doses of atropine cause what
slight paradoxical bradycardia due to CNS stim
higher doses of atropine cause what
they block peripheral muscarinic receptors at heart to produce tachycardia
atropine effect on heart
low doses- bradycardia, higher doses-tachycardia
atropine effect on skin
inhibition of sweating, hot dry skin
atropine effect on eye
mild to marked mydriasis, mild to marked cycloplegia
atropine effect on viscera
dry mouth, decreased saliv, reduced GI and urinary tone
atropine effect on CNS
confusion, restlessness, excitement --- hallucinations, delirium, coma
characteristics of atropine poisoning
hot dry skin, dilated pupils, maniacal behavior
what is a less potent and much shorter acting muscarinic antag
tropicamide
use for tropicamide
opthalmological exams
ipratropium is mainly used in what type of pts
COPD (asthma secondarily)
how does ipratropium work in COPD pts
bronchodilation w/o affecting bronchial secretions
musc blocker that crosses BBB and tx parkinson's
benztropine
musc blocker that tx bladder spasms postop that decreases bladder tone and improves continence
oxybutynin
will innervated or non-innerv receptors be affected by ganglionic transmission
innerv
ganglionic stim will excite which organs
all autonomic organs
which neurons/cells does nicotine stimulate
postgang neurons and adrenal chromaffin cells
prototype ganglionic blocking drug
hexamethonium
nicotine blocks autonomic ganglia when given in high doses due to what
sustained depolarization of postgang cells
T/F: hexamethonium blocks NMJ's
F - does not
predominant tone and effect of ganglionic blockade at arterioles
sypathetic - vasodilation
predominant tone and effect of ganglionic blockade at veins
sym - dilation
predominant tone and effect of ganglionic blockade at heart
parasym - tachycardia
predominant tone and effect of ganglionic blockade at iris
parasym - mydriasis
predominant tone and effect of ganglionic blockade at ciliary muscle
parasym - cycloplegia
predominant tone and effect of ganglionic blockade at GI tract
parasym - reduced tone and motility
predominant tone and effect of ganglionic blockade at urinary bladder
parasym - urinary retention
predominant tone and effect of ganglionic blockade at salivary glands
parasym - xerostomia
predominant tone and effect of ganglionic blockade at sweat glands
sym - anhidrosis
drugs can cause neuromuscular blockade by one of what 2 ways
1. compete w/ ACh for nicotinic receptors 2. sustained depolarization
how do local anes and tetrodotoxin inhibit nerve action potential
interfere w/ Na transport
what inhibits ACh release by blocking reuptake of choline into nerve ending
hemicholinium
what inhibits ACh release by blocking ACh release mech
botulinum toxin
what enhances ACh release
spider venom (latrotoxin) and catecholamines
what inhibits binding of ACh to receptor
NMJ blockers and snake alpha-bungarotoxins
T/F: competitive NMJ blocking drugs enter BBB
F: highly charged
T/F: competitive NMJ blocking drugs produce reduction in pain sensation
FALSE
gold standard of competitive NMJ blockers
tubocurarine
main side effect for tubocurarine - eliminated by?
hypotension - liver
how is doxacurium different from tubocurarine?
doesn't block autonomic ganglia or release histamine
organ of excretion for doxacurium
kidney
NMJ blocker w/ steroid nucleus
pancuronium
how is pancuronium same as doxacurium
no autonomic blockade no histamine release
more potent, shorter acting analog of pancuronium
vecuronium
only drug used clinically as depolarizing NMJ blocker
succinylcholine
how is succinylcholine inactivated
rapidly metabolized by plasma and liver pesudocholinesterases
drug of choice to relax laryngeal muscles before intubation
succinylcholine
what syndrome can appear due to succinylcholien
malignant hyperthermia
benzodiazeepine that enhances GABAergic inhibition in CNS
diazepam
side effect for diazepam
sedation at effective dose
function of diazepam
reduces all muscle spasms
drug that causes decrease in release of Glu and decrease in skeletal muscle tone while experiencing less sedation than diazepam
baclofen
drug used for relief of spasticity caused by MS and spinal cord injury
baclofen
drug that is CNS alpha2 stim, reduces skeletal muscle spasticity, causes less hypotension, but causes sedation and dry mouth
tizanidine
related to tricyclic antidepressants, tx muscular spasms assoc w/ musculoskeletal conditions
cyclobenzaprine
spasmolytic w/ side effects that are atropine-like and not effective in tx for spasms from spinal cord injury or cerebral palsy
cyclobenzaprine
Acts directly on skeletal muscle abd disrupts excitation-contraction coupling by blocking release of Ca needed for contraction
dantrolene
drug of choice for treating malignant hyperthermia
dantrolene
main adverse effects of dantrolene
generalized msucle weakness and sedation
muscle relaxant to tx generalized spastic disorders (cerebral palsy), effective longterm blockade of trans to muscle
botulinum toxin
drugs to tx paralytic ileus
physostigmine and bethanechol
cholinomimetic drugs that are choline esters
1. acetylcholine 2. methacholine 3. carbachol 4. bethanechol
cholinomimetic drugs that are naturally occurring
1. nitotine 2. muscarine 3. pilocarpine
2 ways that cholinomimetic drugs enhance cholinergic function
1. activate Ach directly through nicotinic and muscarinic receptors and 2. block AchE
function of anticholinesterase agents
indirectly increase cholinergic function
list reversible anticholinesterase agents
1. physostigmine 2. neostigmine 3. edrophonium 4. pyridostigmine 5. donepezil and tacrine
list anticholinesterase irreversible agents
1. parathion and malathion 2. sarin and soman
what is the cholinesterase reactivator?
pralidoxime (2-PAM)
what are the 5 basic steps in neurotransmission
1. synthesis 2. storage 3. release from presynaptic terminal 4. reception and post-syn response 5. termination of signal
examples of muscarinic blocking drugs
1. atropine 2. scopolamine 3. tropicamide 4. ipratropium 5. benzytropine 6. oxybutynin
what are 6 example of places where cholinergic synapses are found?
1. all parasyn neuroeffector junctions 2. all autonomic ganglia 3. skeletal NMJ 4. sympathetic innervation of adrenal medulla 5. sweat glands 6. brain and SC sites
non-innevated ACh receptors are generally of what type?
muscarinic
how do cholinergic drugs decrease colinergic function?
they usually occupy the receptor so the ACh molecules cannot act on them
where does synthesis of ACh occur?
cytoplasm of cholinergic nerves
enzyme that catalyzes ACh synthesis
choline acetylase
rate-limiting step of ACh synthesis
active pumping of choline into the nerve by a transport mechanism
what blocks the transport of choline into the nerve that causes an eventual inhibition of choline function?
hemocholinium (HC-3)
what causes attachment of ACh granules to fuse with nerve terminal membranes during action potential
influx of Ca
what can inhibit exocytosis of ACh into synaptic space?
anything that lowers extracellular Ca i.e. bot toxin and latrotoxin
what prevents release of ACh from nerve terminal
botulinum toxin
what causes excessive fusing of ACh granules w/ nerve mem causing excessive release and depletion of ACh
latrotoxin - spider venom
where are pseudocholinesterases found?
plasma and liver
M2 receptors are found
in heart
M3 receptors are found
peripheral autonomic organs
M1 receptors are found
in GI tract and autonomic ganglia
which muscarinic receptors are located in CNS
M1-M5
what does stimulation of muscarinic receptors in the heart do?
slows heart by increasing K permeability causing prolonged phase 4 depolarizaiton
T/F: muscarinic receps in heart directly stimulate ventricular contraction
F: they have little effect on force of vent contraction
what effect does systemic admin of ACh and muscarinic stimulants have on vascular sm
profound vasodilation and decrease BP
how does ACh cause blood vessel smooth muscle relaxation?
ACh acts on non-innerv muscarinic receptors on endothelial cells that releases EDRF (NO)
what do muscarinic agonists produce in the eye
pupillary constriction (miosis) and spasm of accommodation (cyclotonia)
what do muscarinic agonists do in the GI tract
increase tone and motility
what do muscarinic agonists do in bronchiolar smooth muscle
modest constriction
how must ACh be administered to have an effect
injected
what will be activated if ACh is given in low doses
only muscarinic receptors, much higher amts are needed to stimulate nicotinic
what drug is partially resistant to AChase, is a potent muscarinic stimulant, but has lost almost all nicotinic action of parent molecule
methacholine
why is methacholine not used as much in humans
has too many generalized muscarinic effects
what drug is very refractory to inactivation by AChase, has a potent nicotinic stimulating effects w/ little muscarinic actions, and releases ACh from cholinergic nerve endings
carbachol
what drug is used topically on eye to produce miosis and tx glaucoma
carbachol
which drug is only choline ester that has established clinical use
bethanechol
which drug is refractory to AChase inactivation and has mostly muscarinic stimuating effects, tx post-op urinary retention and atony of GI tract
bethanechol
how is bethanechol used clinically
tx esophageal reflux due to stimulatory action on lower esophageal sphincter
what are responses to injected nicotine
activation of all autonomic ganglia and stim of skeletal muscle endplate nic receptors
antidote for muscarine poisoning
atropine
which drug selectively activates muscarinic receptors, not highly charged so can cross BBB, and used to tx glaucoma
pilocarpine
T/F: all somatic nerves release ACh
T
what happens to BP if ACh is given in small doses
decrease in BP
what happens to BP if ACh is given in a small dose after atropine is given
no effect on BP b/c atropine blocked musc receptors
what happens to BP if ACh is given in a large dose after atropine is given
big increase in BP b/c ACh is now hitting nicotinic receptors
when nicotine is given repeatedly or in large doses, why does it block nicotinic receptors?
sustained depolarization
agents that indirectly enhance cholinergic function
anticholinesterases
anticholinesterases only have an effect on which type of receptors
only at innervated ACh receptors
main clinical use for AChase inhibitors
tx myasthenina gravis and glaucoma
what is used to tx atropine poisoning
physostigmine
what drugs are antidotes to curare
AChase inhibitors
main clinical use for physostigmine
topical for glaucoma
what actions does physostigmine have in eye
miosis and contraction of ciliary muscle
how is it imp that physostigmine is not highly charged
can enter CNS
what drug is the best choice for atropine poisoning
physostigmine
neostigmine directly stimulates nicotinic sites where?
on skeletal muscle endplates
the dual action of neostigmine makes it a drug of choice for what
myasthenia gravis
pyridostigmine is used to tx what
myasthenia gravis
which AChase inhibitors are CNS acting and used to tx cognitive dysfunction in Alzeimer's dx
donepezil and tacrine
which cholinomimetic can stimulate salivary flow
pilocarpine
which drug is used to distinguish myasthenia gravis from AChE blockade (cholinergic crisis)
edrophonium
Can be used as topical treatments for glaucoma,but most are insecticides and military nerve agents
irreversible AChases
what insecticide is converted in the liver to the active form
parathion
sarin and soman
Gaseous nerve agents used in chemical warfare. NOT reversible by cholinesterase reactivators
Used to treat muscarinic intoxication and intoxication with some irreversible organophosphate anti-cholinesterases
pralidoxime (2-PAM)
paralysis of the NMJ's due to excessive ACHASE inhibition
cholinergic crisis
paralysis of the NMJ's due to insufficient ACHASE inhibition
myasthenic crisis
what would muscarinic receptor blockers do to parasym innervated organ
lack of motility and secretions
what is the diff b/w atropine and scopolamine
atropine produces CNS excitation, scopolamine causes CNS depression
low doses of atropine cause what
slight paradoxical bradycardia due to CNS stim
higher doses of atropine cause what
they block peripheral muscarinic receptors at heart to produce tachycardia
atropine effect on heart
low doses- bradycardia, higher doses-tachycardia
atropine effect on skin
inhibition of sweating, hot dry skin
atropine effect on eye
mild to marked mydriasis, mild to marked cycloplegia
atropine effect on viscera
dry mouth, decreased saliv, reduced GI and urinary tone
atropine effect on CNS
confusion, restlessness, excitement --- hallucinations, delirium, coma
characteristics of atropine poisoning
hot dry skin, dilated pupils, maniacal behavior
what is a less potent and much shorter acting muscarinic antag
tropicamide
use for tropicamide
opthalmological exams
ipratropium is mainly used in what type of pts
COPD (asthma secondarily)
how does ipratropium work in COPD pts
bronchodilation w/o affecting bronchial secretions
musc blocker that crosses BBB and tx parkinson's
benztropine
musc blocker that tx bladder spasms postop that decreases bladder tone and improves continence
oxybutynin
will innervated or non-innerv receptors be affected by ganglionic transmission
innerv
ganglionic stim will excite which organs
all autonomic organs
which neurons/cells does nicotine stimulate
postgang neurons and adrenal chromaffin cells
prototype ganglionic blocking drug
hexamethonium
nicotine blocks autonomic ganglia when given in high doses due to what
sustained depolarization of postgang cells
T/F: hexamethonium blocks NMJ's
F - does not
predominant tone and effect of ganglionic blockade at arterioles
sypathetic - vasodilation
predominant tone and effect of ganglionic blockade at veins
sym - dilation
predominant tone and effect of ganglionic blockade at heart
parasym - tachycardia
predominant tone and effect of ganglionic blockade at iris
parasym - mydriasis
predominant tone and effect of ganglionic blockade at ciliary muscle
parasym - cycloplegia
predominant tone and effect of ganglionic blockade at GI tract
parasym - reduced tone and motility
predominant tone and effect of ganglionic blockade at urinary bladder
parasym - urinary retention
predominant tone and effect of ganglionic blockade at salivary glands
parasym - xerostomia
predominant tone and effect of ganglionic blockade at sweat glands
sym - anhidrosis
drugs can cause neuromuscular blockade by one of what 2 ways
1. compete w/ ACh for nicotinic receptors 2. sustained depolarization
how do local anes and tetrodotoxin inhibit nerve action potential
interfere w/ Na transport
what inhibits ACh release by blocking reuptake of choline into nerve ending
hemicholinium
what inhibits ACh release by blocking ACh release mech
botulinum toxin
what enhances ACh release
spider venom (latrotoxin) and catecholamines
what inhibits binding of ACh to receptor
NMJ blockers and snake alpha-bungarotoxins
T/F: competitive NMJ blocking drugs enter BBB
F: highly charged
cholinomimetic drugs that are choline esters
1. acetylcholine 2. methacholine 3. carbachol 4. bethanechol
cholinomimetic drugs that are naturally occurring
1. nitotine 2. muscarine 3. pilocarpine
2 ways that cholinomimetic drugs enhance cholinergic function
1. activate Ach directly through nicotinic and muscarinic receptors and 2. block AchE
function of anticholinesterase agents
indirectly increase cholinergic function
list reversible anticholinesterase agents
1. physostigmine 2. neostigmine 3. edrophonium 4. pyridostigmine 5. donepezil and tacrine
list anticholinesterase irreversible agents
1. parathion and malathion 2. sarin and soman
what is the cholinesterase reactivator?
pralidoxime (2-PAM)
what are the 5 basic steps in neurotransmission
1. synthesis 2. storage 3. release from presynaptic terminal 4. reception and post-syn response 5. termination of signal
examples of muscarinic blocking drugs
1. atropine 2. scopolamine 3. tropicamide 4. ipratropium 5. benzytropine 6. oxybutynin
what are 6 example of places where cholinergic synapses are found?
1. all parasyn neuroeffector junctions 2. all autonomic ganglia 3. skeletal NMJ 4. sympathetic innervation of adrenal medulla 5. sweat glands 6. brain and SC sites
non-innevated ACh receptors are generally of what type?
muscarinic
how do cholinergic drugs decrease colinergic function?
they usually occupy the receptor so the ACh molecules cannot act on them
enzyme that catalyzes ACh synthesis
choline acetylase
rate-limiting step of ACh synthesis
active pumping of choline into the nerve by a transport mechanism
what blocks the transport of choline into the nerve that causes an eventual inhibition of choline function?
hemocholinium (HC-3)
what causes attachment of ACh granules to fuse with nerve terminal membranes during action potential
influx of Ca
what can inhibit exocytosis of ACh into synaptic space?
anything that lowers extracellular Ca i.e. bot toxin and latrotoxin
what prevents release of ACh from nerve terminal
botulinum toxin
what causes excessive fusing of ACh granules w/ nerve mem causing excessive release and depletion of ACh
latrotoxin - spider venom
where are pseudocholinesterases found?
plasma and liver
M2 receptors are found
in heart
M3 receptors are found
peripheral autonomic organs
M1 receptors are found
in GI tract and autonomic ganglia
which muscarinic receptors are located in CNS
M1-M5
what does stimulation of muscarinic receptors in the heart do?
slows heart by increasing K permeability causing prolonged phase 4 depolarizaiton
T/F: muscarinic receps in heart directly stimulate ventricular contraction
F: they have little effect on force of vent contraction
what effect does systemic admin of ACh and muscarinic stimulants have on vascular sm
profound vasodilation and decrease BP
how does ACh cause blood vessel smooth muscle relaxation?
ACh acts on non-innerv muscarinic receptors on endothelial cells that releases EDRF (NO)
what do muscarinic agonists produce in the eye
pupillary constriction (miosis) and spasm of accommodation (cyclotonia)
what do muscarinic agonists do in the GI tract
increase tone and motility
what do muscarinic agonists do in bronchiolar smooth muscle
modest constriction
how must ACh be administered to have an effect
injected
what will be activated if ACh is given in low doses
only muscarinic receptors, much higher amts are needed to stimulate nicotinic
what drug is partially resistant to AChase, is a potent muscarinic stimulant, but has lost almost all nicotinic action of parent molecule
methacholine
why is methacholine not used as much in humans
has too many generalized muscarinic effects
what drug is very refractory to inactivation by AChase, has a potent nicotinic stimulating effects w/ little muscarinic actions, and releases ACh from cholinergic nerve endings
carbachol
what drug is used topically on eye to produce miosis and tx glaucoma
carbachol
which drug is only choline ester that has established clinical use
bethanechol
which drug is refractory to AChase inactivation and has mostly muscarinic stimuating effects, tx post-op urinary retention and atony of GI tract
bethanechol
how is bethanechol used clinically
tx esophageal reflux due to stimulatory action on lower esophageal sphincter
what are responses to injected nicotine
activation of all autonomic ganglia and stim of skeletal muscle endplate nic receptors
antidote for muscarine poisoning
atropine
which drug selectively activates muscarinic receptors, not highly charged so can cross BBB, and used to tx glaucoma
pilocarpine
T/F: all somatic nerves release ACh
T
what happens to BP if ACh is given in small doses
decrease in BP
what happens to BP if ACh is given in a small dose after atropine is given
no effect on BP b/c atropine blocked musc receptors
what happens to BP if ACh is given in a large dose after atropine is given
big increase in BP b/c ACh is now hitting nicotinic receptors
when nicotine is given repeatedly or in large doses, why does it block nicotinic receptors?
sustained depolarization
agents that indirectly enhance cholinergic function
anticholinesterases
anticholinesterases only have an effect on which type of receptors
only at innervated ACh receptors
main clinical use for AChase inhibitors
tx myasthenina gravis and glaucoma
what is used to tx atropine poisoning
physostigmine
what drugs are antidotes to curare
AChase inhibitors
main clinical use for physostigmine
topical for glaucoma
what actions does physostigmine have in eye
miosis and contraction of ciliary muscle
how is it imp that physostigmine is not highly charged
can enter CNS
what drug is the best choice for atropine poisoning
physostigmine
neostigmine directly stimulates nicotinic sites where?
on skeletal muscle endplates
the dual action of neostigmine makes it a drug of choice for what
myasthenia gravis
pyridostigmine is used to tx what
myasthenia gravis
which AChase inhibitors are CNS acting and used to tx cognitive dysfunction in Alzeimer's dx
donepezil and tacrine
which cholinomimetic can stimulate salivary flow
pilocarpine
which drug is used to distinguish myasthenia gravis from AChE blockade (cholinergic crisis)
edrophonium
Can be used as topical treatments for glaucoma,but most are insecticides and military nerve agents
irreversible AChases
what insecticide is converted in the liver to the active form
parathion
sarin and soman
Gaseous nerve agents used in chemical warfare. NOT reversible by cholinesterase reactivators
Used to treat muscarinic intoxication and intoxication with some irreversible organophosphate anti-cholinesterases
pralidoxime (2-PAM)
paralysis of the NMJ's due to excessive ACHASE inhibition
cholinergic crisis
paralysis of the NMJ's due to insufficient ACHASE inhibition
myasthenic crisis
what would muscarinic receptor blockers do to parasym innervated organ
lack of motility and secretions
what is the diff b/w atropine and scopolamine
atropine produces CNS excitation, scopolamine causes CNS depression
low doses of atropine cause what
slight paradoxical bradycardia due to CNS stim
higher doses of atropine cause what
they block peripheral muscarinic receptors at heart to produce tachycardia
atropine effect on heart
low doses- bradycardia, higher doses-tachycardia
atropine effect on skin
inhibition of sweating, hot dry skin
atropine effect on eye
mild to marked mydriasis, mild to marked cycloplegia
atropine effect on viscera
dry mouth, decreased saliv, reduced GI and urinary tone
atropine effect on CNS
confusion, restlessness, excitement --- hallucinations, delirium, coma
characteristics of atropine poisoning
hot dry skin, dilated pupils, maniacal behavior
what is a less potent and much shorter acting muscarinic antag
tropicamide
use for tropicamide
opthalmological exams
ipratropium is mainly used in what type of pts
COPD (asthma secondarily)
how does ipratropium work in COPD pts
bronchodilation w/o affecting bronchial secretions
musc blocker that crosses BBB and tx parkinson's
benztropine
musc blocker that tx bladder spasms postop that decreases bladder tone and improves continence
oxybutynin
will innervated or non-innerv receptors be affected by ganglionic transmission
innerv
ganglionic stim will excite which organs
all autonomic organs
which neurons/cells does nicotine stimulate
postgang neurons and adrenal chromaffin cells
prototype ganglionic blocking drug
hexamethonium
nicotine blocks autonomic ganglia when given in high doses due to what
sustained depolarization of postgang cells
T/F: hexamethonium blocks NMJ's
F - does not
predominant tone and effect of ganglionic blockade at arterioles
sypathetic - vasodilation
predominant tone and effect of ganglionic blockade at veins
sym - dilation
predominant tone and effect of ganglionic blockade at heart
parasym - tachycardia
predominant tone and effect of ganglionic blockade at iris
parasym - mydriasis
predominant tone and effect of ganglionic blockade at ciliary muscle
parasym - cycloplegia
predominant tone and effect of ganglionic blockade at GI tract
parasym - reduced tone and motility
predominant tone and effect of ganglionic blockade at urinary bladder
parasym - urinary retention
predominant tone and effect of ganglionic blockade at salivary glands
parasym - xerostomia
predominant tone and effect of ganglionic blockade at sweat glands
sym - anhidrosis
drugs can cause neuromuscular blockade by one of what 2 ways
1. compete w/ ACh for nicotinic receptors 2. sustained depolarization
how do local anes and tetrodotoxin inhibit nerve action potential
interfere w/ Na transport
what inhibits ACh release by blocking reuptake of choline into nerve ending
hemicholinium
what inhibits ACh release by blocking ACh release mech
botulinum toxin
what enhances ACh release
spider venom (latrotoxin) and catecholamines
what inhibits binding of ACh to receptor
NMJ blockers and snake alpha-bungarotoxins
T/F: competitive NMJ blocking drugs enter BBB
F: highly charged
T/F: competitive NMJ blocking drugs produce reduction in pain sensation
FALSE
gold standard of competitive NMJ blockers
tubocurarine
main side effect for tubocurarine - eliminated by?
hypotension - liver
how is doxacurium different from tubocurarine?
doesn't block autonomic ganglia or release histamine
organ of excretion for doxacurium
kidney
NMJ blocker w/ steroid nucleus
pancuronium
how is pancuronium same as doxacurium
no autonomic blockade no histamine release
more potent, shorter acting analog of pancuronium
vecuronium
only drug used clinically as depolarizing NMJ blocker
succinylcholine
how is succinylcholine inactivated
rapidly metabolized by plasma and liver pesudocholinesterases
drug of choice to relax laryngeal muscles before intubation
succinylcholine
what syndrome can appear due to succinylcholien
malignant hyperthermia
benzodiazeepine that enhances GABAergic inhibition in CNS
diazepam
side effect for diazepam
sedation at effective dose
function of diazepam
reduces all muscle spasms
drug that causes decrease in release of Glu and decrease in skeletal muscle tone while experiencing less sedation than diazepam
baclofen
drug used for relief of spasticity caused by MS and spinal cord injury
baclofen
drug that is CNS alpha2 stim, reduces skeletal muscle spasticity, causes less hypotension, but causes sedation and dry mouth
tizanidine
related to tricyclic antidepressants, tx muscular spasms assoc w/ musculoskeletal conditions
cyclobenzaprine
spasmolytic w/ side effects that are atropine-like and not effective in tx for spasms from spinal cord injury or cerebral palsy
cyclobenzaprine
Acts directly on skeletal muscle abd disrupts excitation-contraction coupling by blocking release of Ca needed for contraction
dantrolene
drug of choice for treating malignant hyperthermia
dantrolene
main adverse effects of dantrolene
generalized msucle weakness and sedation
muscle relaxant to tx generalized spastic disorders (cerebral palsy), effective longterm blockade of trans to muscle
botulinum toxin
drugs to tx paralytic ileus
physostigmine and bethanechol
drug to tx atony of bladder
bethanechol
drug to tx glaucoma
physostigmine, pilocarpine
drug to tx myasthenia gravis
neostigmine
drug to reverse NMJ blockade
neostigmine + atropine
preanesthetic medication
scopolamine
drug used in opthalmological exam
tropicamide
drug to tx motion sickness
scopolamine