final exam Flashcards
What is Pharmacokinetics?
What the body does to the drug
Ex) Half life after metabolizing
What is Phamacodynamics?
What a drug does to the body
Ex) Increases heart rate
What is a poison?
a nonbiological substance that has negative effects on the body.
Ex) lead
What is a toxin?
biological substance that has negative effects on the body (created from living substances)
Ex) poison mushrooms
What is a Stereoisomerism?
Isomers that differ in spatial arrangement of atoms, rather than order of atomic connectivity
(Optical Isomers or mirror images)
Where does orthosteric bonding take place?
Binding at the primary active site on the receptor
Where does allosteric bonding take place?
Drug binds to something other than the primary active site (Non-competitive)
What is the relationship between strength and bond specificity?
It is inversely proportional.
The stronger the bond, the less specific the bond. The weaker the bond, the more specific the bond.
What is the pharmacokinetics acronym?
ADME
Absorption
Distribution
Metabolism
Excretion
What is EC(50) and E(max) in the Drug Concentration Response Curve?
EC(50) is a point on the horizontal axis (Drug Concentration) where you see 50% of drug effects.
E(max) is a point on the curve where you max out the drug effects right before the curve plateaus.
What is K(d) and B(max) on the Drug Concentration Response Curve?
K(d) is a point on the horizontal axis (Drug Concentration) where 50% of the receptors are bound.
B(max) = max receptors bound
What is the difference between K(d) and EC(50)?
K(d) is referring to 50% of receptors bound vs EC(50) refers to 50% max effect of drug.
Describe the relationship between an agonist and an allosteric antagonist?
You cannot outcompete an allosteric antagonist (non-competitive antagonist) because it’s not competing for the same receptor site.
If you keep increasing dose of agonist, you will only see a toxic effect - insurmountable
Describe the relationship between an agonist and an allosteric activator?
An agonist and and allosteric activator (allosteric agonist) work together to get an increased result. An allosteric activator binds outside out of the active site.
What is an inverse agonist?
Acts as an antagonist; has a greater affinity to R(i) and stabilizes R(i) form.
Can shut down the downstream response.
Drops BELOW the constitutive activity
In practice, we will refer to as antagonist.
What is the relationship between K(d) and receptor affinity?
The relationship is inverse.
Low K(d) = high drug/receptor affinity; the drug binds well to the receptor.
High K(d) = low drug/receptor affinity; the drug doesn’t bind well to the receptor.
What is physiological antagonism?
Acts at a different receptor but produces an opposite physiological effect to that of the agonist
Effect: Does not compete with the agonist at the same receptor, but rather opposes its action through a different mechanism.
Example: Epinephrine (which increases heart rate) can act as a physiological antagonist to histamine (which decreases heart rate), even though they act on different receptors.
Describe a graph in which an agonist is alone, an agonist + competitive antagonist, agonist + allosteric agonist, and an agonist + allosteric inhibitor
Describe a graph showing constitutive activity, full agonist, partial agonist, antagonist, and an inverse agonist
How does therapeutic index correlate with drug safety?
The larger the therapeutic index the safer the drug is; est. margin of safety
If pH>pKa
favors unprotonated form
If pH<pKa
favors protonated form
If a weak acid is protonated it is…
Not charged
If a weak base is protonated it is…
charged
In order to cross barriers, a compound will want to be ____
uncharged; more lipid soluble
If aspirin (weak acid) has pKa = 3.5, what form does it take in the stomach (pH = 1.5)?
Protonated, uncharged
If aspirin (weak acid) has pKa = 3.5, what form does it take in the intestine (pH = 6.5)?
Unprotonated, charged
What medication for morning sickness was rejected by the FDA due to it causing Phocomelia?
Thalidomide
What are the 4 mechanisms of transmembrane signaling?
- Direct crossing to intercellular receptor (lipid soluble)
- Enzymatic action mediated by ligand binding
a. Tyrosine kinase activated receptors - Ligand gated ion channel
- G protein receptor
What receptors are intracellular?
Lipid soluble - uncharged
Gasses - easily diffuses through membrane
What are the steps to the GPCR cascade?
- Drug binds to receptor - activates alpha subunit of G protein (conformation change)
- G protein releases GDP and binds GTP
- G protein activates effector protein
- Secondary messenger cascade or ion conductance
Differentiate between desensitization and resensitization
- When first give agonist, response jumps up, peaks, then declines (muted response with same amount of agonist) - desensitization
- Stop giving agonist, then wait, then give agonist again → see same response - resensitization
Describe the steps to the desensitization process
- Drug binds to receptor → promotes receptor interaction with G proteins in cytoplasm (closed → open conformation)
- Agonist-activated receptors phosphorylated by G protein-coupled receptor kinase (GPK), preventing receptor interaction with G protein → attracts beta-arestin (B-Arr)
- B-Arr receptor complex binds to coated pits, promoting receptor internalization
- 2 possibilities:
Possibility 1 - resensitization (Drug falls off receptor→ phosphatase dephosphorylates receptor and receptor recyles back to cell membrane)
Possibility 2 → degradation (Can’t get drug off of receptor → lysosome merges with drug/receptor complex → enzymes from lysosome degrade receptor)
What are the 2 signal types for ion channels?
Ligand and voltage gated
Differentiate ionotropic from metabotropic ligand-gated ion channels
Ionotropic - Ligand binding site and channel on same protein
Metabotropic - Ligand activates GPCR, second messenger activity opens channel
First-order elimination
Rate of elimination varies with concentration - clearance constant
Zero-order elimination
Rate of elimination is constant, clearance varies with concentration
What is meant by a high Vd?
The higher the Vd, the less amount of drug is in the blood; drug distributed in other areas
What is the difference between elimination and clearance?
Elimination changes based on clearance (doesn’t change)
What is clearance?
Ability of body to eliminate drug in relation to drug concentration in body
What is volume of distribution?
Apparent volume in blood
- How much did we give them vs how much stayed in blood (how much stayed in blood vs how much went to other areas)
What are the main two components of standard drug dosing?
Volume of distribution and clearance
What is rational dosing?
Goal – to achieve desired beneficial effect with minimal adverse effects
Describe what occurs in zero-order elimination
Occurs when the body’s ability to eliminate a drug has reached its maximum capability (i.e., all transporters are being used). As the dose and drug concentration increase, the amount of drug eliminated per hour does not increase, and the fraction of drug removed declines.
What order of elimination do most drugs follow?
First-order
What are the 4 parameters affective passive diffusion?
Molecular weight
pKa
lipid solubility
plasma protein binding
How many half-lives are usually required for drug to achieve full effects?
4
What are the 4 phase I reactions?
Oxidation, reduction, dehydrogenation, and hydrolysis
Cytochrome 450 enzymes are _________
oxidases
Cytochrome p450 oxidation occurs in the ________ of the hepatocyte
Sarcoplasmic endoplasmic reticulum
Drugs that increase cytochrome P450 activity
induction
Drugs that decrease or irreversibly inhibit cytochrome P450 activity
inhibition
Cytochrome P450 induction causes a decreased drug effect – IF metabolism ________ drug
deactivates
Cytochrome P450 induction causes an increased drug effect – IF metabolism ________ drug
activates
Cytochrome P450 inhibition causes an increased drug effect – IF metabolism ________ drug
inactivates
Cytochrome P450 inhibition causes a decreased drug effect – IF metabolism ________ drug
activates
Define the role of drug efflux transporters.
Cell survival mechanism to pump unwanted substances out of the cell. Cell can increase the amount based on exposure over time.
1. Solute carrier (SLC) proteins - Passive transport via gradient
2. ABC gene family - ATP
Most drug efflux transporters are _______
ATP-binding cassette (ABC) transporters
3 major drug efflux transporters
B, C, G
List the components of the intact blood-brain barrier.
tight gap junctions
ABC transporters
Glial cells- astrocytes, podocytes
Where are neuron cell bodies of the autonomic system located?
outside the CNS (ganglia)
What are the subdivisions of the autonomic nervous system?
sympathetic, parasympathetic, and enteric
Chain ganglion are present in which NS? parasympathetic or sympathetic?
sympathetic
Where do sympathetic axons leave the CNS?
Thoracolumbar region
For sympathetic NS, preganglionic fibers are ________, while postganglionic fibers are _________.
preganglionic= short
postganglionic= long
For parasympathetic NS, preganglionic fibers are ________, while postganglionic fibers are __________
Preganglionic= long
postganglionic= short
Where do parasympathetic axons leave the CNS?
Craniosacral regions.
Most from brainstem, bladder and genitals from sacral
Which muscarinic receptors are excitatory?
M1, M3, M5
Which muscarinic receptors are inhibitory?
M2, M4
What neurotransmitter(s) acts on cholinergic receptors?
ACh
Which alpha receptors are stimulatory and what does it activate?
alpha 1, activates Gq protein, activates phospholipase C which activates secondary messengers IP3 and DAG
which alpha receptors are inhibitory and what does it inhibit?
alpha 2, inhibits adenylyl cyclase; leads to decreased cAMP
Where is the ganglia located in the sympathetic NS?
Close to the spinal cord
Where is the ganglia located in the parasympathetic NS?
In the visceral effector organs
What is the GPCR that alpha 1 activates? Effector?
Gq/G11 - activates phospholipase C
What is the GPCR that alpha 2 activates? Effector?
Gi - inhibits adenylate cyclase
What is the GPCR that beta activates? Effector?
Gs - stimulates adenylate cyclase
Where are alpha 1 receptors primarily found?
in the smooth muscle cell that surrounds the blood vasculature
What type of receptors are nicotinic receptors?
Ion channels
What type of receptors are muscarinic receptors?
GPCRs
Are Beta receptors inhibitory or stimulatory?
All beta receptors are stimulatory - stimulates adenylate cyclase, increasing cAMP
How does the activation of beta receptors work in the heart?
NE activates beta receptors - G stimulatory → stimulates adenylyl cyclase → increases cAMP → activates PK-A → more ICF Ca++ (from ECF and from sarcoplasmic reticulum) → contraction
How does the activation of beta 2 receptors work in the peripheral muscle?
NE binds to B2 in periphery → Increases cAMP → inhibit MLCK → no active myosin → relaxation - allows peripheral vessels to dilate
Compare the SNS vs PANS of the SA node and which receptors are present
SNS: accelerates SA node - B1 and B2
PANS: decelerates the SA node - M2
Compare the SNS vs PANS of the heart contractility and which receptors are present
SNS: increases heart contractility - B1 and B2
PANS: decreases heart contractility - M2
Compare the SNS vs PANS of bronchiolar smooth muscle and which receptors are present
SNS: relaxes bronchiolar smooth muscle - B2
PANS: contracts bronchiolar smooth muscle - M3
The ____ is the gap between the neuron and the cell
synapse
What is the role of the neuron?
Sends AP from neuron cell body to the telodendria (endpoints) capped with synaptic boutons (where neurotransmitters are stored)
info is coming into neuron through the _____
Dendrites
Action potentials are generated in the ________
axon hillock
What are the 6 Neurotransmitter Classes?
- Esters
- Monoamines
- Amino Acids
- Purines
- Peptides
- Inorganic gases
Give an example of an ester neurotransmitter
Acetylcholine (ACh)
Give an example of a monoamine neurotransmitter
NE, Serotonin, Dopamine
Give an example of an amino acid neurotransmitter
Glutamate, GABA, glycine
Give an example of a peptide neurotransmitter
Substance P, Endorphins - mediate pain and analgesia
Give an example of a purine neurotransmitter
Adenosine, ATP
Give an example of a inorganic gas neurotransmitter
Nitric oxide (NO)
NO is different than other neurotransmitters because it is not ______
stored - made as needed by nitric oxide synthase
Which drugs are esters of choline?
Ach, Methacholine, Succinylcholine (Sux), Carbachol, and Bethanechol
List Direct acting adrenergic agonists
albuterol
clonidine
dobutamine
dopamine
epinephrine
isoproterenol
norepinephrine
What drugs are indirect acting adrenergic agonists
amphetamine
What drug is both direct and indirect acting adrenergic agonist?
ephedrine
Which adrenergic receptor stimulates the heart?
B1
Which receptors does epinephrine act on
a1, a2, B1, B2
which receptors does norepinephrine act on?
a1, a2, B1
which receptors does isoproterenol act on?
B1, B2
which receptors does dopamine act on?
D1-5, higher doses: a1, B1
which receptors does dobutamine act on?
B1
Beta antagonists result in negative ________ & ________
inotropy, chronotropy
Differentiate between direct and indirect-acting cholinoceptor stimulants
Direct-acting - agonists of receptors (choline esters and alkaloids)
Indirect-acting - cholinesterase inhibitors, prolonging ACh in the synapse (reversible and irreversible)
Where are muscarinic receptors found?
Nerve, heart and smooth muscle, and glands and endothelium
Where are nicotinic receptors found?
Neuromuscular end plate, skeletal muscle, and autonomic ganglion cells
What are the indirect-acting cholinomimetics that we need to know?
Alcohols - edrophonium
Carbamates - neostigmine and pyridostigmine
Organophosphates - echothiophate
What is edrophonium used for and how long does it last?
Myasthenia gravis diagnosis, ileus, and arrhythmias; 5-15 min
What is the mechanism of indirect-acting cholinomimetics?
Targets ACh esterase (AChE), preventing the breakdown of ACh
How can an organophosphate covalent bond be broken?
Need a strong nucleophile - Pralidoxime
What are the 4 major therapeutic uses of indirect-acting cholinomimetics?
- Disease of the eye - glaucoma
- GI and urinary tracts - ileus
- Neuromuscular junction - MG and anesthesia
- Atropine overdose
What are the symptoms of overdose of direct-acting muscarinic stimulants?
SLUDGE-M: Need atropine
Salivation, lacrimation, urination, defecation, GI motility, emesis, myosis
What are the symptoms of overdose of cholinesterase inhibitors?
SLUDGE-M - need atropine and pralidoxime (organophosphate poisoning)
What are the indications for the use of atropine?
Blocks the parasympathetic response
- Bradycardia
- Poisonous mushrooms and organophosphate poisoning
What are the signs and symptoms of atropine overdose?
BRAND - blindness, redness, absent bowel sounds, nuts (CNS), dilated pupils
What is the treatment for atropine overdose?
Physostigmine - can produce dangerous CNS effects
Differentiate between direct acting and indirect-acting adrenergic agonists
Direct agonist - bind to receptors and illicit response
Indirect agonist - Increase amount of catecholamines in synapse; either facilitating removal or preventing reuptake
Alpha 1 vs alpha 2 effects
- α1: Gq → phospholipase C → increases IP3 and DAG
- contracts vascular smooth muscle, prostate contraction, heart - increases force of contraction
- α2: Gi → decreases cAMP
- inhibited transmitter release at adrenergic and cholinergic nerve terminals
Compare the effects of beta 1, 2, 3
Gs - all increase cAMP
- β1: increases force and rate of contraction (heart) and increases renin release (juxtaglomerular cells)
- β2: promotes smooth muscle relaxation at respiratory, uterine, and vascular smooth muscle
- β3: activates lipolysis in fat cells
Describe the mechanism of action for alpha 1 receptors
Describe what happens in the cardiovascular system after the administration of an alpha-agonist
- Increases vascular resistance/tone
- HR decreases (indirect effect)
- BP increases
Describe what happens in the cardiovascular system after the administration of a beta-agonist
- Decreases vasculature resistance/tone
- Increased contractility and HR
- BP decreases overall
Describe the effects of epinephrine
- Potent vasoconstrictor
- α receptors
- Cardiac stimulant (β1)
- Positive inotropic (force)
- Positive chronotropic (rate)
- β2 activation in some vessels
- Dilation of skeletal muscle vessels
- Dilation in bronchioles
Describe the effects of norepinephrine
- Effects on α and β1
- Little effect on β2
- Results
- Increase in systolic and diastolic
- Vagal reflexes overcome chronotropic effects
Describe the effects of isoproterenol
- Potent β agonist - vasodilator - heart
- Little effect on α receptors
- Results
- Increase cardiac output
- Fall in Mean arterial pressure
- Slight decrease or increase in systolic BP
Describe the effects of dopamine
Triphasic response:
- Low dose: Activates D1 receptors - vasodilation,
decrease in peripheral resistance
- Higher dose: mimics action of epinephrine; β1 receptors in the heart
- Highest dose: activates alpha, increases BP
Describe the effects of dobutamine
- β1 selective agonist
- Cardiac shock, acute heart failure
What is angina caused by
accumulation of metabolites due to myocardia ischemia
What drug is given for immediate relief of angina?
nitroglycerin
what type of angina is also known as Prinzmetal angina or vasospastic?
Variant angina
O2 delivery decreased due to coronary vasospasm
Which type of angina is classic angina
“angina of effort”
O2 requirement increases with activity, but coronary blood flow not enough, leads to O2 debt and ischemia with toxic metabolites
What drugs are good for angina prophylaxis?
calcium channel blockers and beta blockers
What type of angina is also known as “angina at rest”?
Unstable angina
microvascular disease s/t small patelet clots and atherosclerotic plaque
Which type of angina is an emergency?
unstable angina
Which type of angina is most rare?
variant
What is treatment of classic angina?
reduction of demand through beta blockers, calcium channel blockers
what is treatment of variant angina?
primarily calcium channel blockers to prevent , vasodilators/nitrates
How does NO lead to vasodilation?
NO activates guanylyl cyclase, which converts GTP to cGMP. cGMP dephosphorylates Myosin-LC leading to relaxation
What are the good effects from Nitrates and Nitrites?
Increased venous capacitance
decreased ventricular preload
decreased heart size
decreased cardiac output
What are the bad effects of nitrates and nitrites?
Headache (most common), orthostatic hypotension, syncope, reflex tachycardia, hemoglobin interactions (methehemoglobin = low affinity for oxygen)
Describe how Ca++ contributes to blood vessel contraction
- Ca++ binds to calmodulin (protein) → activates MLCK → MLC-P interacts with actin to contract
- cAMP inhibits MLCK
What are the different drugs that induce relaxation of vascular tone?
Ca++ channel blockers (prevent contraction)
K+ channel blockers (prevent depolarization)
B2 agonists (increase cAMP)
Nitrates (increase cGMP)
How are Beta Blockers used for angina treatment?
Not vasodilators - decrease oxygen demand
↓ HR
↓ BP
↓ Contractility
- more B2 receptors in micro-arteries → dilation
How do Ca++ channel blockers treat angina?
- L-type channel most dominant in cardiac and smooth muscle
- Drug binds to depolarized membranes
- Decreased opening frequency with drug binding
- relaxation and reduced BP in smooth muscle
- Heart: ↓ contractility, ↓ SA node pacemaker rate, ↓ AV node conduction velocity
What is an example of an irreversible alpha antagonist?
Phenoxybenzamine
What are the beneficial effects of beta blockers?
decrease oxygen demand,
decrease HR, BP and contractility
Calcium channel blockers target
L-type calcium channels of vascular smooth muscle and heart
Calcium channel blockers in smooth muscle cause
relaxation, reduce blood pressure
calcium channel blockers in heart cause
decrease contractility, decreased SA node pacemaker rate, decreased AV node conduction velocity
Which calcium channel blockers are more peripheral vascular selective?
Dihydropyridines (nifedipine, amlodipine, and nimodipine.)
which calcium channel blockers are more cardiac selective?
verapamil and diltiazem
What are toxicities of calcium channel blockers?
bradycardia, arrest
AV block
CHF
What are the 4 types of antihypertensive agents?
diuretics: deplete sodium
Sympathoplegics: decrease PVR, reduce CO
Direct vasodilators: relax VSM
Anti-angiotensins: block activity or production
What is the hydraulic equation?
BP= CO x PVR
Cardiac output is a function of
stroke volume, heart rate, venous capacitance (preload)
another word for preload is
venous capacitance
Which drugs are CNS sympathoplegics?
Methyldopa, clonidine
Which drugs are adrenoceptor antagonists
Propanolol, metoprolol, atenolol, prazosin, terazosin, doxazosin
Propanolol acts on which receptors?
B1 and B2
What are the effects of propanolol
Lowers BP, prevents reflex tachycardia
Decreases CO
Inhibits renin production
Which drugs are alpha 1 blockers?
Prazosin, Terazosin, Doxazosin
What are the effects of alpha 1 blockers?
block a1 receptors in arterioles and venules.
dilates both resistance and capacitance vessels
BP is reduced more in upright position
Which drugs are direct vasodilators
Minoxidil, hydralazine, sodium nitroprusside, fenoldopam
What is the mechanism of action of minoxidil
Opens K+ channels in smooth muscles, stabilizes
What are factors that affect blood pressure?
peripheral resistance
vessel elasticity
blood volume
cardiac output
Why is methyldopa better than clonidine for pregnant women?
Doesn’t cross the placental barrier like clonidine does
used for 2nd and 3rd trimester HTN
What are the 4 mechanisms of action of vasodilators? Give examples of drugs
What are the two main ways vasodilators lower BP?
- Relax smooth muscle of arterioles (all) and veins (nitroprusside and nitrates)
- Reduction of PVR and MAP - Elicits compensatory responses; Best when given in conjunction with other hypertensives that combat these mechanisms
Describe the effects of combination therapy of beta blockers and diuretics in lowering blood pressure
How does hydralazine lower BP?
Dilates arterioles – induces NO production in endothelium
What are the symptoms of hydralazine toxicity?
- HA, nausea, sweating, flushing
- Worse in slow acetylators (Symptoms resemble SLE)
How does Sodium Nitroprusside lower BP?
Relaxes vascular smooth muscle, dilating arterial and venous vessels
- Breaks down in blood to release NO
- Increases intracellular cGMP
What are the clinical indications for Sodium Nitroprusside?
HT emergencies and Cardiac failure
What is the upside and downside of Sodium Nitroprusside use?
Upside - Rapidly lowers BP; Effects disappear 1-10 min after d/c
Downside - CN accumulation, slowly eliminated by kidney
How does Fenoldopam lower BP?
Peripheral arteriolar dilator - Agonist of D1 receptors
- dilates renal vascular bed (lowers BP, diuresis)
What are the clinical indications for Fenoldopam use?
HTN emergencies, post-op HTN
What are the symptoms associated with Fenoldopam toxicity?
Reflex tachycardia, flushing, HA
How do Ca++ channel blockers lower BP?
Dilate peripheral arterioles by inhibiting Ca2+ influx in arterial smooth muscle
Which Ca++ channel blockers target the heart vs the periphery?
- Verapamil - more targeted to heart
- Diltiazem - both heart and periphery
- Dihydropyridine family - more targeted to periphery
What are the types of Inhibitors of Angiotensin?
ACE inhibitors (-pril)
Angiotensin competitive inhibitors (angiotensin receptor blockers, ARB) (-artan)
______ is the prototype drug of ACE inhibitors
Captopril
Renin release in kidney stimulated by what 4 mechanisms?
- Reduced arterial pressure
- Reduced sodium delivery
- Increased sodium concentration
- Sympathetic stimulation (beta receptors)
Why is a persistent, non-productive cough a symptom for ACE inhibitors but not ARBs?
ARBs have no effect on bradykinin - no cough
ACE inhibitors - inhibit breakdown of bradykinins, which stimulate PG synthesis - excess bradykinin causes non-productive cough
Which angiotensin inhibitors do we need to know?
ACE inhibitors - Captopril
Angiotensin receptor blockers – Losartan, Valsartan
What are the first line drugs for HTN?
- Low dose diuretic
- Beta blocker
- CCB
- Dual therapy
Define heart failure
Heart fails to meet the metabolic demands of tissues - CO inadequate
What is the most common cause of heart failure?
Coronary artery disease
Differentiate between systolic and diastolic heart failure
Systolic failure – reduced cardiac function
- Acute; heart walls thinned (less effective pumping)
↓ CO, ↓ Ejection fraction
Diastolic failure – reduced cardiac filling (can be peripheral)
- Chronic; heart more stiff/thicker walls (chronic HTN)
- ↓ CO, Normal Ejection fraction
- Does not respond well to positive inotropic drugs
Differentiate between symptoms of right vs left ventricular failure of CHF
Right ventricle – peripheral congestion
Left ventricle – pulmonary congestion
What are the 4 factors of cardiac performance?
CO = SV x HR
- Preload
- Afterload
- Contractility
- Heart Rate
Heart failure where the normal CO not sufficient for demands of body
“High-output” failure (rare)
- Hyperthyroidism
- Beriberi
- Anemia
- Arteriovenous shunts
Responds poorly to inotropic agents – treat underlying cause
In systolic heart failure, CO is ______ and ejection fraction is _______
decreased, decreased
In diastolic heart failure, CO is ______ and ejection fraction is _______
decreased, normal
Systolic heart failure responds ______ to positive inotropes
well
Diastolic heart failure responds _______ to positive inotropic drugs
poorly
Digoxin mechanism of action
Directly inhibits Na/K ATPase
maintains normal resting potential, positive inotrope
Digoxin has a ____ therapeutic index
narrow
What are ways to decrease preload?
salt restriction, diuretics, venodilation (Nitroglycerin)
Discuss the normal control of cardiac contractility
- Trigger Ca++ enters cell
- depends on number of L type channels, duration of channel opening, and sympathetic stimulation - Binds to channel is SR, release stored Ca++
- depends on amount stored and amount of trigger Ca++ - Frees actin to interact with myosin
- Removal of Ca++
- SR Calcium ATPase
- Na+/Ca2+ antiporter – sodium gradient
- Na+/K+ ATPase: removes sodium
What are the EKG changes that occur with digoxin toxicity?
tachycardia, fibrillation, arrest
What effect does hyperkalemia have on digoxin effects?
- Potassium competes with digoxin
- Excess K+, decreased effect
What effect does hypomagnesia have on digoxin effects?
Increased risk of digoxin induced arrhythmias
How do phosphodiesterase inhibitors help treat HF?
Enzymes that inactivate cAMP and cGMP - positive inotropic effects, vasodilation
Increase contractility w/o inhibiting Na+/K+ ATPase (increase/prolong Ca++)
PDE3 specific
The pacemaker of the heart is _______ and is located in the ________
SA node, right atrium