Pharmacology Flashcards
In enzyme kinetics, competitive inhibitors _____ (resemble/do not resemble) the substrate while noncompetitive inhibitors _____ (resemble/do not resemble) the substrate.
Resemble; do not resemble
In enzyme kinetics, the value of Km reflects the _____ of the enzyme for its substrate.
Affinity
True or False? In enzyme kinetics, the lower the Km, the higher the affinity.
True
In enzyme kinetics, Vmax is directly proportional to the _____ _____.
Enzyme concentration
In enzyme kinetics, a graph of substrate concentration on the x-axis and velocity of the reaction on the y-axis has _____ (increasing/decreasing) velocity as substrate is increased.
Increasing, although it will plateau when the enzyme is saturated
When velocity is equal to one half of its maximum (Vmax), the corresponding concentration of substrate is equal to what value?
Km
In enzyme kinetics, the y-intercept of a graph that plots the inverse of velocity on the y-axis and the inverse of substrate concentration on the x-axis is equal to what value?
The inverse of Vmax = 1/Vmax
In enzyme kinetics, the x-intercept of a graph that plots the inverse of velocity on the y-axis and the inverse of substrate concentration on the x-axis is equal to what value?
The inverse of Km = 1/Km
In enzyme kinetics, the slope of a graph that plots the inverse of velocity on the y-axis and the inverse of substrate concentration on the x-axis is equal to what value?
Km/Vmax
In enzyme kinetics, a competitive inhibitor _____ (cannot/can) be overcome by increasing the concentration of substrate; a noncompetitive inhibitor _____ (cannot/can) be overcome by increasing the concentration of substrate.
Can; cannot. This is because competitive inhibitors bind the active site of the enzyme, competing with the substrate, whereas noncompetitive inhibitors bind elsewhere on the enzyme and so are not affected by substrate concentration
In enzyme kinetics, competitive inhibitors _____ (increase/decrease/do not change) the Vmax of the reaction, while noncompetitive inhibitors _____ (increase/decrease/do not change the Vmax of the reaction.
Do not change; decrease
In enzyme kinetics, competitive inhibitors _____ (increase/decrease/do not change) the Km of the reaction, while noncompetitive inhibitors _____ (increase/decrease/do not change the Km of the reaction.
Increase; do not change
What is the formula for calculating the volume of distribution of a drug?
Volume of distribution = amount of drug in the body / plasma drug concentration
Drugs with a low volume of distribution, such as 4-8 L, are found in the _____ (blood/extracellular space/tissues).
Blood alone; these drugs do not distribute outside the plasma
A drug with a volume of distribution of 15 L is most likely to be found in the _____ (blood/extracellular space/tissues).
Extracellular space; these drugs distribute throughout the total body water
In a 75 kg man, a drug has a volume of distribution of 40 L. It can be expected to be found in _____ (blood/extracellular space/tissues).
Tissues
What is the formula for calculating the clearance of a drug?
Clearance (L/min) = rate of elimination of drug (g/min) / plasma drug concentration (g/L)
What is the definition of the half-life of a drug?
The time required to reduce the amount of drug in the body by one half
How many half-lives of a drug must pass before a drug infused at a constant rate reaches approximately 94% of steady-state concentration?
Four
Given the volume of distribution and clearance of a drug, how does one calculate the half-life of the drug?
Half-life = (0.7 × volume of distribution) / clearance
After one half-life, given constant intravenous infusion of a drug, how close to steady-state is the concentration of the drug?
50% of steady-state concentration
After three half-lives, given constant intravenous infusion of a drug, how close to steady-state is the concentration of the drug?
87.5% of steady-state concentration
What is the formula for the loading dose of a drug?
Loading dose = (target plasma concentration × volume of distribution) / bioavailability
What is the formula for maintenance dose of a drug administered intravenously?
Maintenance dose = rate of elimination/bioavailability = (target plasma concentration × clearance) / bioavailability
How do loading and maintenance doses of drugs differ for patients with hepatic and renal disease?
For both hepatic and renal disease, loading dose does not change, but maintenance dose decreases
What is the bioavailability of a drug if it is administered intravenously?
100%
In zero-order elimination of drugs from the body, what is the relationship between the rate of elimination and the drug concentration?
The rate of elimination is constant regardless of drug concentration
What are three drugs that exhibit zero-order elimination?
Phenytoin and ethanol; aspirin at toxic concentrations
In first-order elimination of drugs from the body, what is the relationship between the rate of elimination and the drug concentration?
The rate of elimination is directly proportional to the drug concentration; a constant fraction (rather than a constant amount) is eliminated
In zero-order elimination of drugs from the body, how does the plasma concentration of a drug change over time: linearly or exponentially?
Linearly
In first-order elimination of drugs from the body, how does the plasma concentration of a drug change over time: linearly or exponentially?
Exponentially
Weak acids get trapped in _____ (acidic/basic) environments.
Basic
Weak bases get trapped in _____ (acidic/basic) environments.
Acidic
What substance is given to enhance the renal clearance of weakly acidic drugs such as phenobarbital, methotrexate, and aspirin?
Bicarbonate
What substance is given to enhance the renal clearance of weakly basic drugs such as amphetamine?
Ammonium chloride
Ionized species become trapped in urine because they are not _____ _____.
Lipid soluble; therefore, they cannot cross cell membranes
A 24-year-old man attempts suicide by consuming a small bottle of aspirin. After three hours he thinks better of it, and comes to the emergency room. He is put in your care, and you start him on intravenous saline with bicarbonate. By what mechanism does this help him?
Bicarbonate alkalinizes the lumen of his nephrons, which traps acetylsalicylic acid within the lumen because it is a weak acid and is ionized in a basic environment
What three types of biochemical reactions are involved in the phase I metabolism of drugs?
Reduction, oxidation, and hydrolysis
What is the polarity of the drug products that result from phase I metabolism?
The products are slightly polar
True or False? Drug products that result from phase I metabolism are water soluble.
True
True or False? Drug products that result from phase I metabolism are often still active.
True
What enzyme system mediates the phase I metabolism of drugs in the body?
Cytochrome P-450
What three types of biochemical reactions are involved in the phase II metabolism of drugs?
Acetylation, glucuronidation, and sulfation; these are conjugation reactions
Do geriatric patients lose the ability for phase I or phase II drug metabolism first?
Phase I
Phase I metabolism of drugs yields _____ (nonpolar/slightly polar/very polar) molecules that are _____ (inactive/often still active), whereas phase II metabolism of drugs yields _____ (nonpolar/slightly polar/very polar) molecules that are _____ (inactive/often still active).
Slightly polar; often still active; very polar; inactive
The products of phase II metabolism of drugs are excreted by what organ?
Kidneys
What is the definition of efficacy?
Maximal effect a drug can produce
What is the definition of potency?
Amount of drug needed for a given effect
A drug that requires a very low dose to achieve its desired effect is considered _____.
Potent
True or False? In pharmacodynamics, when a competitive antagonist is given, the maximal effect of an agonist is decreased regardless of how much additional agonist is given.
False; the maximal effect of an agonist is still achievable in the presence of a competitive antagonist if increased amounts of the agonist are given
What is the effect of a noncompetitive antagonist on the position of an agonist’s dose-response curve?
It vertically shrinks; the agonist’s efficacy is decreased
In pharmacodynamics, the addition of a noncompetitive agonist _____ (increases/decreases/does not change) the efficacy of the agonist.
Decreases
How does the efficacy of a partial agonist relate to the efficacy of a full agonist of the same receptor?
A partial agonist has a lower maximal efficacy than a full agonist
How does the potency of a partial agonist relate to the potency of a full agonist of the same receptor?
A partial agonist may be more potent than, less potent than, or equally as potent as a full agonist
What property of a drug is determined by its therapeutic index?
Safety; drugs with higher therapeutic indices are less likely to cause toxicities
What is the formula that describes the therapeutic index of a drug?
TI (therapeutic index) = LD50 (median toxic dose) / ED50 (median effective dose) (remember: TILE)
Safer drugs have _____ (higher/lower) therapeutic index values.
Higher
From which regions of the central nervous system do parasympathetic nerves originate?
Cranial and sacral regions
From which regions of the central nervous system do sympathetic nerves originate?
Thoracic and lumbar regions
What types of nerves arise from the spinal cord and innervate skeletal muscle directly?
Somatic nerves
How many neurons are involved in parasympathetic transmission from the spinal cord to the target organ?
Two
True or False? Craniosacral parasympathetic axons synapse on neurons in the peripheral ganglia.
True
What neurotransmitter mediates parasympathetic nervous system function?
Acetylcholine
What neurotransmitter receptor mediates parasympathetic nervous system function at the peripheral ganglia?
Nicotinic acetylcholine receptors
What neurotransmitter receptor mediates parasympathetic tone in the cardiac muscle?
Muscarinic acetylcholine receptors (specifically, M2)
What neurotransmitter receptor mediates parasympathetic tone in the smooth muscle?
Muscarinic acetylcholine receptors (specifically, M3)
What neurotransmitter receptor mediates parasympathetic tone in the glandular cells?
Muscarinic acetylcholine receptors (specifically, M1 and M3)
Somatic nerves that arise from the spine innervate skeletal muscle. What neurotransmitter receptor, which is located on skeletal muscle, receives this input?
Nicotinic acetylcholine receptors
How many neurons are involved in sympathetic transmission from the spinal cord to the target organ?
Two
Where is the first synapse after the spinal cord in sympathetic innervation of an organ?
Preganglionic sympathetic axons synapse on neurons in the paravertebral ganglia
True or False? Preganglionic sympathetic axons synapse on neurons in the peripheral ganglia.
False; preganglionic sympathetic axons synapse on neurons in the paravertebral ganglia
At the paravertebral ganglia, the neurotransmitter _____ acts on _____ receptors to mediate sympathetic nervous system function.
Acetylcholine; nicotinic acetylcholine
What neurotransmitter mediates sympathetic nervous system function at the sweat glands?
Acetylcholine
What neurotransmitter receptor mediates sympathetic nervous system function at the sweat glands?
Muscarinic acetylcholine receptors
What neurotransmitter mediates sympathetic tone in the cardiac muscle, smooth muscle, and glandular cells?
Norepinephrine
What are four cell types in which α- and β-adrenergic receptors mediate sympathetic tone?
Cardiac muscle, smooth muscle, glandular cells, and terminal ends of neurons
What neurotransmitter mediates sympathetic tone in the renal vascular smooth muscle?
Dopamine
What neurotransmitter receptor mediates sympathetic tone in the renal vascular smooth muscle?
D1 receptors
What two substances are released into the blood from the adrenal medulla after the activation of the sympathetic nervous system?
Epinephrine and norepinephrine
How many synapses are involved in activation of the adrenal medulla?
One; the adrenal medulla releases epinephrine and norepinephrine into the blood
Are nicotinic acetylcholine receptors ligand-gated sodium-potassium channels or G-protein coupled receptors?
Nicotinic receptors are ligand gated sodium-potassium channels
Are muscarinic acetylcholine receptors ligand-gated sodium-potassium channels or G-protein-coupled receptors?
Muscarinic acetylcholine receptors are G-protein-coupled receptors that act through second messengers
To what class of G-proteins are α1-receptors linked?
q
To what class of G-proteins are α2-receptors linked?
i
To what class of G-proteins are β1-receptors linked?
s
To what class of G-proteins are β2-receptors linked?
s
To what class of G-proteins are M1-receptors linked?
q
To what class of G-proteins are M2-receptors linked?
i
To what class of G-proteins are M3-receptors linked?
q
To what class of G-proteins are D1-receptors linked?
s
To what class of G-proteins are D2-receptors linked?
i
To what class of G-proteins are H1-receptors linked?
q
To what class of G-proteins are H2-receptors linked?
s
To what class of G-proteins are V1-receptors linked?
q
To what class of G-proteins are V2-receptors linked?
s
What are the major effects of α1-receptor activation?
It increases vascular smooth muscle contraction, and increases pupillary dilator muscle contraction (mydriasis)
What are the major functions of α2-receptor activation?
It decreases sympathetic outflow and decreases insulin release
What are the major functions of β1-receptor activation?
It increases heart rate and contractility, increases renin release from the kidneys, and increases lipolysis of adipose tissue
What is the major function of β2-receptor activation on the body’s vasculature?
Vasodilation
What is the major function of β2-receptor activation on the respiratory system?
Bronchodilation
What effect does β2-receptor activation have on glucagon release?
It increases glucagon release
Where are M1-receptors located?
The central nervous system
What effect does M2-receptor activation have on cardiac function?
It decreases heart rate and contractility
What are the effects of M3-receptor activation?
Increased exocrine gland secretions, gut peristalsis, bladder contraction, bronchoconstriction, miosis, and accommodation
What effect does D1-receptor activation have on renal vasculature?
It relaxes renal vascular smooth muscle
What are the effects of H1-receptor activation?
Pruritis, pain, nasal and bronchial mucus production, contraction of bronchioles
What is the effect of H2-receptor activation?
It increases gastric acid secretion
What effect does V1-receptor activation have on vascular smooth muscle?
It increases vascular smooth muscle contraction
The activation of what two types of G-protein-coupled receptors can increase vascular smooth muscle contraction? Which receptors mediate vascular relaxation?
α1- and V1-receptors increase contraction; relaxation is mediated by β2, and D1 (renal only)
What is the effect of V2-receptor activation? Where are they located?
It increases water permeability and reabsorption in the collecting tubules of the kidney
What five types of receptors are coupled with Gq proteins?
α1, M1, M3, H1, and V1
What five types of receptors are coupled with Gs proteins?
β1, β2, D1, H2, and V2
What three types of receptors are coupled with Gi proteins?
a2, M2, and D2
What enzyme is activated directly downstream of Gq-coupled receptors?
Phospholipase C
What enzyme is activated directly downstream of Gs-coupled receptors?
Adenyl cyclase
What enzyme is inhibited directly downstream of Gi-coupled receptors?
Adenyl cyclase
Adenyl cyclase catalyzes the conversion of adenosine triphosphate into what molecule?
cAMP
What final effector enzyme is activated by receptors that are coupled with Gs proteins?
Protein kinase A
What final effector enzyme is inhibited by receptors that are coupled with Gi proteins?
Protein kinase A
Phospholipase C catalyzes the cleavage of membrane lipids into what molecules?
Inositol trisphosphate3 and diacylglycerol
What is the effect of increased inositol triphosphate on the intracellular concentration of calcium?
It increases the intracellular calcium concentration
What enzyme is activated by diacylglycerol?
Protein kinase C
What pharmacologic agent blocks the uptake of choline into cholinergic nerve terminals?
Hemicholinium
What enzyme is responsible for the formation of acetylcholine? What are its two substrates?
Choline acetyltransferase; Acetyl-CoA and choline
What pharmacologic agent blocks the transport of acetylcholine into the presynaptic vesicles in nerve terminals?
Vesamicol
The entry of what ion into the nerve terminal induces the release of acetylcholine into the synaptic cleft?
Calcium
What toxin inhibits the calcium-induced release of acetylcholine from the cholinergic nerve terminals?
Botulinum
What enzyme breaks down acetylcholine in the synaptic cleft? What two products result from this reaction?
Acetylcholinesterase; choline and acetate
Tyrosine transporters are located in the nerve terminals of what type of cells?
Noradrenergic cells; tyrosine is the precursor of norepinephrine
Tyrosine is a precursor to the formation of which neurotransmitters? What is the order of their synthesis?
Tyrosine, DOPA, dopamine, norepinephrine, epinephrine
What pharmacologic agent blocks the conversion of tyrosine to DOPA?
Metyrosine
Tyrosine is converted into dopamine via what intermediate precursor?
DOPA; DOPA can be used as a pharmacologic agent to increase central nervous system dopamine
What pharmacologic agent blocks the transport of dopamine into the presynaptic vesicles in nerve terminals?
Reserpine
Dopamine is converted into norepinephrine in the ______ (cytoplasm/presynaptic vesicle).
Presynaptic vesicles
The entry of what ion into the nerve terminal induces the release of norepinephrine into the synaptic cleft?
Calcium
What pharmacologic agent inhibits the calcium-induced release of norepinephrine from the noradrenergic nerve terminals?
Guanethidine
What pharmacologic agent stimulates the release of norepinephrine from the noradrenergic nerve terminals?
Amphetamine
How is norepinephrine cleared form the synaptic cleft?
Diffusion, metabolism (monoamine oxidase A), and reuptake
What pharmacologic agents inhibit the reuptake of norepinephrine into the nerve terminals?
Cocaine, amphetamine, and tricyclic antidepressants
What three receptor types modulate the presynaptic release of norepinephrine from the noradrenergic nerve terminals?
M2-receptors, angiotensin II receptors, and α2-receptors
What effect does the activation of α2-receptors in presynaptic sympathetic nerve terminals have on norepinephrine release?
It inhibits norepinephrine release
What effect does the activation of angiotensin II receptors in presynaptic sympathetic nerve terminals have on norepinephrine release?
It stimulates norepinephrine release
What effect does the activation of M2-receptors in presynaptic sympathetic nerve terminals have on norepinephrine release?
It inhibits norepinephrine release
The norepinephrine-mediated activation of α2-receptors on presynaptic sympathetic nerve terminals is an example of a mechanism of what type of feedback?
Negative feedback
Name four direct cholinergic agonists.
Bethanechol, carbachol, pilocarpine, methacholine
What is the clinical application of bethanechol?
Treatment of postoperative and neurogenic ileus and urinary retention (remember: Beth Anne, call (bethanechol) me if you want to activate your Bowels and Bladder)
What is the mechanism of action of bethanechol?
Bethanechol is a direct cholinergic agonist resistant to acetylcholinesterase that works on receptors in the bowel and bladder
What two direct agonist cholinomimetic drugs can be used to treat glaucoma?
Carbachol and pilocarpine
Carbachol and pilocarpine are effective for the treatment of open-angle glaucoma because they activate what muscle?
The ciliary muscle of the eye
What is a methacholine challenge test?
A test in which methacholine is inhaled to stimulate muscarinic receptors and induce bronchoconstriction to diagnose asthma
Pilocarpine is effective for the treatment of narrow-angle glaucoma because it activates what muscle?
The pupillary sphincter
True or False? Pilocarpine is susceptible to acetylcholinesterase.
False; pilocarpine is resistant to acetylcholinesterase
Name five indirect cholinergic agonists.
Neostigmine, pyridostigmine, edrophonium, physostigmine, echothiophate
What are the clinical indications for use of neostigmine?
The treatment of postoperative and neurogenic ileus
True or False? The treatment of myasthenia gravis is a clinical application of pyridostigmine.
True
Which anticholinesterase is used to diagnose myasthenia gravis? Why?
Edrophonium; the effects last for minutes and if weakness is transiently reversed it is diagnostic of myasthenia gravis
True or False? The treatment of glaucoma is a clinical application of physostigmine.
True (remember: “PHYS is for the EYES”)
Which pharmacologic agent is used to treat atropine overdose?
Physostigmine, because it crosses the blood-brain barrier and is able to reverse central nervous system as well as peripheral nervous system effects
What is the clinical indication for use of echothiophate?
The treatment of glaucoma
Indirect cholinergic agonists increase endogenous acetylcholine by inhibiting what enzyme?
Acetylcholinesterase
Why is pyridostigmine used to treat myasthenia gravis?
It increases the amount of acetylcholine in the neuromuscular synapse, thereby increasing muscle strength
What effect does neostigmine have on the central nervous system?
None; it does not penetrate the blood-brain barrier (remember: NEO CNS = NO CNS)
What is the clinical application and mechanism of action of topical atropine, homatropine, and tropicamide?
These drugs antagonize muscarinic receptors in the eye to produce mydriasis and cycloplegia
What is the mechanism and clinical application for benztropine?
It is a muscarinic antagonist used to reduce symptoms of Parkinson’s disease
What is the mechanism and clinical application for scopolamine?
It is a muscarinic antagonist used to treat motion sickness
What is the mechanism and clinical application for ipratropium?
It is a muscarinic antagonist used to treat asthma and chronic obstructive pulmonary disease (remember: I PRAY I can breathe soon!)
What is the mechanism and clinical application for methscopolamine?
It is a muscarinic antagonist used to treat peptic ulcers
What is the mechanism and clinical application for oxybutynin?
It is a muscarinic antagonist used to reduce urgency in mild cystitis and reduce bladder spasms
What is the mechanism and clinical application for glycopyrrolate?
It is a muscarinic antagonist used to reduce urgency in mild cystitis and reduce bladder spasms
What is the mechanism and clinical application for pirenzepine?
It is a muscarinic antagonist used to treat peptic ulcers
What is the mechanism and clinical application for propantheline?
It is a muscarinic antagonist used to treat peptic ulcers
Which muscarinic antagonist can be used to reduce urgency in patients with mild cystitis?
Oxybutynin (also glycopyrrolate)