Pharm Final Flashcards
Define pharmacology/medical pharmacology
Study of the effects of drugs in the body
Define toxicology
Undesirable effects of chemicals on living systems, from individual cells to humans to complex ecosystems
What are the different types of drug groups?
Agonists, partial agonist, antagonist, inverse agonists, agonist mimics
Define pharmacodynamics
The effects of the drug on the body
Define pharmacogenomics
Looking at genetic profile to determine how you will respond to drug
The primary reason patients respond differently to the same drug
Define pharmacokinetics
Effects of the body on the drug (half-life, can it cross barriers, ADME, etc)
Drugs can either be an ______ or an ______
agonist; antagonist
Define agonist
Binding to specific site elicits a conformational change in the protein that is bound which activates the receptor, producing the same, or similar, effect to that of a native ligand
Define antagonist
Competitive in nature to agonists and they bind to a specific receptor before an agonist can, inhibiting the response to the amount of normal constituent activity
Define receptor
A large protein, usually on the cell surface, that can bind to drugs or endogenous ligands
Define endogenous
Produced inside the organism or cell
Define Exogenous
Growing or originating from outside the organism
Define poisons
Non-biological substances (arsenic or lead)
Define toxins
Biological substances from living organisms (mushrooms)
Define partial agonist
Like agonists in binding sites but can act like antagonists if in the presence of an agonist because it only produces a partial response and prevents the agonist from binding.
Define inverse agonist
Favor the inactive receptors which effectively makes them stronger forms of an antagonist because they lower the constitutive form of the receptor
Define stereoisomerism
Same chemical formula, but doesn’t behave the same in the body
Optical isomers (D:L; R:S) - applies to more than half the body
Define physiologic antagonist
two different drugs bind to different receptors and have opposite effects
Ex: epi binding to beta receptors and increasing HR, while acetylcholine binds to muscarinic receptors and decreases HR
Differentiate between competitive inhibitor and allosteric inhibitor
Competitive inhibitor: a drug that binds to the same active site as the agonist, competing for that binding site
Allosteric inhibitor: binds to a different site on the receptor, preventing the agonist from binding and eliciting a response, even at high agonist concentrations (can’t be surmounted)
What 4 things make up pharmacokinetics?
Absorption, distribution, metabolism, elimination
How does the Henderson-Hasselbach equation apply in pharm?
In practice, pKa is pH at which ionized and un-
ionized concentrations are equal
If pH < pKa; favors protonated form
If pH > pKa; favors unprotonated form
How do you solve for the therapeutic ratio?
TD50 / ED50
What is a dose-response curve?
A graph illustrating different pharmacologic potencies and different maximal efficacies
What is the goal of rational dosing?
To achieve desired beneficial effect
with minimal adverse effects
Bond strength is indirectly proportional to ____
Specificity
Flux is directly proportional to _______
Concentration
Kd is indirectly proportional to ________
Drug binding affinity
Drug safety is directly proportional to the _______
Therapeutic index (TI)
Volume of distribution (Vd) is directly proportional to the _____
Concentration of a drug outside of the systemic circulation
Define volume of distribution
The space available in the body to contain the drug
Define clearance
The ability of the body to eliminate the drug
Define rate of elimination
The rate at which the drug is eliminate from the body
Define half-life
The time it takes for the drug concentration to decrease by 50%
Define steady-state dosing, maintenance, and loading dose
Steady state: administering just enough to replace how much the body is eliminating per hr
Maintenance: maintain steady state of a drug by giving enough to replace eliminated drug
Loading dose: used when needing to reach steady state quickly
What are the parameters affecting passive diffusion?
Molecular Weight
pKa
Lipid solubility
Plasma protein binding
What are the 4 basic mechanisms of transmembrane signaling?
- Direct crossing to intercellular receptor (lipid
soluble) - Enzymatic action mediated by ligand binding (Tyrosine kinase activated receptors)
- Ligand gated ion channel
- G protein receptor
Explain ligand-gated ion channels
Ligand gates ion channels open in response to the binding of a specific ligand
Explain GPCR structure
Receptor:
Seven transmembrane spanning domains, with an extracellular ligand-binding site (amino terminal) and an intracellular G protein-binding site (carboxy terminal). Several downstream effects possible
G-Proteins
- Trimeric: alpha, beta, gamma
- GDP (inactive) → GTP (active)
How many families/genes do drug efflux transporters have?
Seven families (A-G), over 50 genes
Define allosteric
The drug binds to a different site on the receptor (away from the active site)
Define orthosteric
The drug binds directly the the active site
Differentiate how a full agonist, partial agonist, antagonist, and inverse agonist would look on a graph
- Full agonist has the max response
- Partial agonist is slightly above constitutive activity, but below full agonist
- Antagonist is equal to constitutive activity
- Inverse agonist is below constitutive activity
See page 6 of review for reference
What is permeation? What are the 4 different types?
Ways to get the drug from outside the body to the inside of the body where it will eventually work
- Aqeuous
- Lipid
- Endo/exocytosis
- Special carriers
What is aqueous diffusion?
Water soluble drugs can cross membranes
What is lipid diffusion?
Lipid soluble drugs can cross membranes (steroid nuclei drugs)
What are special carriers?
Something that will bind to a large molecule, bring it into the cell and oftentimes may stay in the cell or then pushed outside of the cell
What is endocytosis?
Membrane engulfs drug, brings it in and spits it out the other side; usually due to clathrin coated pits in receptors that bind that drug and bring it in
This is an important mechanism for very large drugs to get across impermeable membranes
What is exocytosis?
Merging of the vesicle with the membrane
What are examples of aqueous components in aqueous diffusion?
Blood, ECF, ICF
What are things that can’t cross the membrane through aqueous diffusion?
Highly charge molecules: Will interact with the cell surface and won’t be able to cross the cell membrane
Bound to large proteins (carriers): can’t cross the barrier while bound to large proteins
Potency is measured by ____
EC50 (effective concentration fr 50% of the maximum effect)
A drug with a lower EC50 is considered more _____
Potent
List the components of the blood-brain barrier
Vascular endothelium with tight junctions, astrocytes, pericytes
These constrict the entry of most drugs in the brain
Describe drug biotransformation
Chemical modifications that a drug undergoes in the body, typically to make it more water-soluble and easier to excrete
Some metabolites become ____ after biotransformation
Active
Major phase 1 metabolic reactions include (4) _____ and are primarily catalyzed by _____
Oxidation, reduction, dehydrogenation, hydrolysis
Cytochrome P450
If a drug induces a metabolic enzyme it will ______ the metabolism and clearance of other drugs that are substrates of that enzyme
Increase
This potentially reduces efficacy
If a drug inhibits a metabolic enzyme, it will ______ the metabolism and clearance of other drugs that are substrates of that enzyme
Decrease
This potentially increases toxicity
What can induction of a metabolic enzyme do?
Enhance synthesis, inhibit degradation
What can inhibition of a metabolic enzyme do?
- Decrease or irreversible inhibit P450
- Competitive inhibition by co-administering drugs metabolized by the same P450
What are potential results of induction of p450?
- Decreased drug effect: if metabolism deactivates the drug
- Increased drug effect: if metabolism activates the drug
Define agonist mimic
Doesn’t bind to the active site, but can still elicit a response as if they did (cocaine, meth)
Differentiate between competitive and non-competitive antagonist
Competitive: bind and inhibit agonist response; can be countered by increasing amounts of agonist (surmountable)
Non-competitive: irreversible and insurmountable
What is an example of an organic compound?
Carbohydrate, lipid, protein, nucleic acids
What is an example of an inorganic compound?
Lithium, iron, hydrogen, oxygen
What are the receptor interactions?
Appropriate size, electrical charge, shape, composition
What is Bmax?
The maximum number of receptors available for the drug to bind to
What is EMax?
The maximum response or effect that can be achieved by the drug
What is Kd?
Drug concentration at which 50% of the receptors are occupied
What is TD50?
The point where we see 50% of toxic side effects
What is important about a dose-response curve in regards to therapeutic index?
The distance in between the ED50 line and LD50 line is the therapeutic index
An agonist + a noncompetitive antagonist will _______ the agonist effect
Decrease
On a dose-response curve, the higher the response, the _____ efficacious a drug is
More
Define target concentration
The desired concentration of the drug in the body to achieve the desired effect
Define bioavailability
The fraction of the administered dose that reaches the systemic circulation
A high volume of distribution (Vd) means the drug is distributed to the tissues ___________
Outside of the blood
A high clearance means the drug is eliminated from the body _________
More rapidly
Differentiate between first order and zero order elimination
First order: clearance is constant, rate of elimination varies with concentration
Zero order: rate of elimination is constant, clearance varies with concentration
First-order elimination
Occurs with most drugs
Zero-order elimination
Occurs when the body’s ability to eliminate a drug has reached it’s maximum capability
As the dose and drug concentration increase, the amount eliminated per hr does not
What is a racemic mixture?
Refers to a combination of optical isomers
Remember R-ketamine and S-ketamine, where S-ketamine is four times more potent than R-ketamine but has increased risk of dissociative effects and hallucinations.
What are the 4 main causes of drug variation?
Genetic factors
Physiologic factors (age, sex)
Environmental factors (diet, smoking)
Pharmacokinetic factors (ADME)
Describe the cell signaling process
- signaling molecules (drug, endogenous ligand) binds to the receptor
- the receptor undergoes a confirmational change
- activation of signal transduction proteins (G proteins, kinases)
- Generation of second messengers
- Activation effector proteins that elicit the cellular response
Phosphorylation cascade
Inactive Protein is activated by a drug or endogenous ligand → Kinase 1 (active) → Kinase 2 (active) possibly 2 or more kinases→ Kinase 3 (active) possibly more than the previous number of activated kinases→ Eventually reaches effector protein (active response)
Describe RTK structure
Extracellular ligand-binding domain, a single transmembrane domain, and an intracellular tyrosine kinase domain.
What is an example of a ligand gated ion channel?
Nicotinic ACh receptor; the receptor opens when ACh binds to it
Process of a ligand gated ion channel
- The signal molecule binds to the receptor
- Surrounding ions go through the channel and elicit a response
Explain the mechanism of GPCR signaling
- Ligand binds to the GPCR, causing a conformational change.
- The conformational change activates the associated G protein.
- The activated G protein dissociates into its α and βγ subunits.
- The Gα subunit releases its bond to GDP and binds with GTP which then activates an effector protein (e.g., adenylyl cyclase).
- The effector protein generates second messengers (e.g., cAMP, IP3).
- The second messengers initiate downstream signaling cascades.
What do second messengers do?
Amplify and propagate the signal initiated by the first messenger (drug, ligand)
What is desensitization?
The process where the cell reduces its responsiveness to a stimulus overtime
What is a voltage gated ion channel?
Ion channels that open and close in response to changes in the membrane potential
Define the generic pathway of CYP450 metabolism
The generic pathway of CYP450 metabolism involves the drug binding to the enzyme, followed by a series of oxidation and reduction reactions followed by dehydrogenation, and then release of the metabolite via hydrolysis.
Define the role of drug efflux transporters
They actively pump drugs out of cells, reducing intracellular drug concentrations. This can contribute to drug resistance.
Ex: ATP-binding cassette (ABC)
What are the major drug efflux transporters?
ABCB1: broadest substrate specificity; wide distribution and are critical to maintenance of the blood brain barrier
ABCC: antineoplastics
ABCG2: breast cancer resistance protein; also an efflux transporter of folate
What is the difference in inotropic and metabotropic ion channels?
- Ionotropic ion channels directly allow the passage of ions across the membrane.
- Metabotropic ion channels indirectly regulate ion movement through second messenger signaling. (E.g. GPCR activates cAMP which opens an ion channel)
What are common examples of second messengers
cAMP
IP3 (Inositol triphosphate)
Diacylglycerol
Calcium
cGMP
Describe the structure of a neuron
A cell with a nucleus with dendrites (fingers on the outside), connected to an axon. At the end of the axon, there is a telodendria (looks like little fingers at the end). Telondendria are also known as synaptic boutons
Refer page 24 in the review
Neurons send action potentials/signals from the __________ to the _________
Neuron cell body
Telondendria
Where are neurotransmitters stored?
Telodendria
Where does the information coming into the neuron come in?
Dendrites
Describe the neuron process:
- Once information is in dendrites it can send a “go” or “stop” signal to nucleus of neuron to either activate or suppress activation of neuron
- Decision in cell body can activate the axon hillock and this will generate action potential
- Action potential can go faster if myelin sheath is present; this is known as saltatory conduction.
- Action potential will reach the telodendria (capped with synaptic boutons)
- Synaptic boutons release neurotransmitter into the synapse and the neurotransmitter binds to receptors on the postsynaptic cell
- Neurotransmitters are stored in synaptic terminal/boutons but they are actually made in the neuron itself
What is a synapse?
Gap between the neuron and the cell
What are the types of synapses?
Chemical: release neurotransmitters into the synapse (ACh, GABA)
Electrical: pass electrical current from one cell to the other through gap junctions (commonly in the heart)
What are the 6 main classes of neurotransmitters?
- Esters (ACh): cholinergic
- Monoamines (norepinephrine, serotonin, dopamine, many catecholamines): adrenergic
- Amino acids (glutamate, GABA)
- Purines (adenosine, ATP)
- Peptides (substance P, endorphins)
- Inorganic gases (Nitric oxide)
Why are inorganic gasses considered a neurotransmitter?
It is released by effector cell and has effect on postsynaptic cell
Afferent is going ____ the CNS; Efferent is going _____ from CNS
Towards
Away
What are the efferent divisions?
Somatic nervous system: controls skeletal muscle; conscious control
Autonomic nervous system: includes parasympathetic and sympathetic; no conscious control over
The autonomic nervous system is seperated into:
Parasympathetic, sympathetic, and enteric
Neuron cell body clusters in the CNS are called _____
Nuclei
Neuron cell body clusters in the PNS are called ________
ganglia
Describe the sympathetic nervous system
Known for fight or flight (increasing HR, bronchiole dilation, shunt blood to skeletal muscles and away from GI)
Affects a lot of our CV system. Widespread impact, reaches organs and tissues
Describe the parasympathetic nervous system
Rest and digest system
This brings you back down to normal. It conserves energy and shunts blood to GI and endocrine system. Innervates only specific visceral structures, effects are shorter lived
The gut has its own nervous system called
Enteric nervous system: associated with causing “gut feelings”
Where do the fibers origin in the sympathetic nervous system?
Thoracolumbar region of the spinal cord
Where do the fibers origin in the parasympathetic nervous system?
Brain and sacral spinal card
What is the length of fibers in the sympathetic nervous system?
short preganglionic and long postganglionic
What is the length of fibers in the parasympathetic nervous system?
Long preganglionic and short postganglionic
Where is the location of the ganglia in the sympathetic nervous system?
Close to the spinal cord
Where is the location of the ganglia in the parasympathetic nervous system?
In the visceral effector organs
What is the difference in receptors in the SNS and PNS?
SNS: beta and alpha receptors - GPCR
PNS: muscarinic (GPCR) and nicotinic receptors (ion channel)
How does the alpha and beta receptor work in the SNS?
SA node is primary node that sets the pace for the heart – if we release norepinephrine or epinephrine, it binds to beta 1 and 2 receptors and increases heart rate and heart contractility
Where does the PNS primarily work through?
Vagus nerve: PNS stimulation is not constant, it returns to homeostasis and shuts off
Define sympathomimetics
drugs that mimic the sympathetic nervous system (mimic fight or flight)
Define cholinomimetic (aka parasympathomimetics)
drugs that mimic acetylcholine
Define parasympatholytics (aka antimuscarinics/parasympathoplegic)
Drugs that block parasympathetic nervous system
Define sympatholytics/Sympathoplegic (alpha or beta blockers/sympathoplegic)
drugs that block sympathetic nervous system response
List the ANS receptors
Cholinergic (receptors that bind to and are activated by ACh)
Adrenergic (respond to norepinephrine and epi)
What are the adrenergic receptors divided into?
Alpha 1/Alpha2
Beta 1-3
Dopamine (1-5)
What does alpha 1 do?
activates Gq → GDP to GTP → activates phospholipase C → activates IP3 and DAG (second messengers)
What does alpha 2 do?
activates Gi → inhibits adenylyl cyclase → inhibits cAMP
What does beta 1-3 do?
activates Gs → stimulates adenylyl cyclase → produce cAMP
What does IP3 do after it’s activated?
Goes to sarcoplasmic reticulum where calcium is produced in the cell (binds to calcium channels) → Calcium is mobilized into the cell → activates protein kinase → MLCK activated → interacts with actin to contract the muscle cell
What does DAG do after it’s activated?
activates protein kinase C: this is something that inhibits myosin light-chain phosphatase → MLCK doesn’t get stripped up the phosphate
When you think of beta 1, think of the _____
heart
What is the pathway for beta 1 (and beta 2)?
GDP to GTP → stimulates adenyl cyclase → ATP to cAMP → activates protein kinase A → (2 effects) 1. More calcium gets in from the outside 2. Protein kinase A is going to stimulate calcium release from sarcoplasmic reticulum
In the heart, beta 2 = ______. In the periphery, beta 2 = _________
Contraction
Relaxation
How does beta 2 work in the periphery?
Increases in cAMP → inhibit myosin light-chain kinase (MLCK is going to phosphorylate myosin and make it so that it can interact with actin in a form of contraction.) → relaxation
What are the cholinergic receptors divided into?
Nicotinic and muscarinic (1-5)
Which muscarinic receptors are stimulatory?
1, 3, 5
What will the muscarinic Gq activate?
Gq activates phospholipase activation. → increase IP3 and DAG
Which muscarinic receptors are inhibitory?
2 and 4
What does muscarinic Gi inhibit?
Adenylyl cyclase
Where can our cholinergic nicotine receptors be?
Ganglionic, skeletal muscle, neuronal CNS
What is different about nicotinic receptors vs muscarinic receptors?
Muscarinic receptors are GPCRs, nicotinic are ion channels
Where are the muscarinic receptor subtypes found?
Pacing centers of the heart
Smooth muscle
Nerves
Glands
Endothelium
Where are our dopamine receptors?
Most are in the brain, but there are some in the CV system and smooth muscle in the kidney
Effects of alpha 1 (sympathetic)
Smooth blood vessels: contract
GI Sphincter smooth muscle: contract
Kidney/urinary sphincter: contract
Liver: glycogenolysis
reference page 29
Effects of alpha 2 (sympathetic)
GI tract wall smooth muscle: relax
reference pg 29
Effects of beta 1 (sympathetic)
SA node: accelerates
Contractility: increases
Kidney: renin release
reference pg 29
Effects of beta 2 (sympathetic)
SA node: accelerates
Contractility: increases
Skeletal blood vessels: relax
Bronchiolar smooth muscle: relax
GI tract wall smooth muscle: relax
Bladder: relax
Liver: glycogenolysis
reference pg 29
Effects of M2 (parasympathetic)
SA node: decelerates
Contractility: decreases
Effects of M3 (parasympathetic)
Smooth blood vessels: relax
Bronchiolar smooth muscle: contract
GI wall smooth muscle: contract
GI spinchter smooth muscle: relax
GI secretions: increase
Bladder: contract
Urinary spinchter: relax
What is the autonomic feedback loop?
- Baroreceptors sense increased pressure and send it to the vasomotor center
- If MAP is high, it will send to PNS and decrease CO and HR
- If MAP is too low, it will send to SNS → sends norepi which binds to beta 1 to increase CO; norepi will also bind to alpha to increase BP
- If MAP is low, SNS will also increase HR, contractility, and venous tone; so works on chronotropic and inotropic
What is the hormonal feedback loop?
- MAP low → kidney sees this (juxtaglomerular apparatus) → release renin → changes angiotensinogen to angiotensin (1,2) → aldosterone → constrict blood vessels, take up more water, produce less urine
What are the direct acting adrenergic agonists?
Albuterol
Clonidine
Dobutamine
Dopamine
Epi
Isoproterenol
Norepi
What are the indirect acting adrenergic agonists?
Amphetamines
What is a direct and indirect acting adrenergic agonist?
Ephedrine
What organ system to beta receptors determine direct effects?
Heart
What does stimulation of beta receptors in the heart do?
Increased CO
Decreased peripheral resistance
How do you solve for CO?
SV (70ml/beat) X HR (75 beat/min) = CO (ml/min)
This would equal 5250 ml/min
What are examples of catecholamines?
Epi
Norepi
Isoproterenol
Dopamine
Dobutamine
What receptors does epi work on?
Alpha 1 & 2, Beta 1 & 2
What is the structure of a catecholamine?
Catechol group and amine group
What receptors does norepinephrine work on?
Alpha 1 & 2, Beta 1
What receptors does isoproterenol work on?
Beta 1 & 2
What receptors does dopamine work on?
D 1-5; higher doses alpha 1 and beta 1
What receptor does dobutamine work on?
Beta 1
When would you use an adrenoceptor antagonist as treatment?
HTN related to phenochromocytoma
What are examples of reversible adrenoceptor antagonist drugs?
Phentolamine, tolazoline, prazosin, labetalol, propanolol, metoprolol, atenolol, terazoin, doxazosin
What is an example of an irreversible adrenoceptor antagonist? Why is it irreversible?
Phenoxybenzamine
It forms a covalent bond; requires new receptors
What do beta antagonists do in the heart?
Negative inotropic
Negative chronotropic
What do beta antagonists do in the blood vessels?
Opposes B2 mediated vasodilation
Acute: increased peripheral resistance
Chronic: decreased peripheral resistance (mechanism unclear)
What are examples of beta antagonist drugs?
Propanolol
Metoprolol
Atenolol
Esmolol
Where does propranolol work?
Beta 1 and 2
Where does metoprolol and atenolol work?
Mainly B1 selectivity
What 2 diseases is it safer to use metoprolol or atenolol when picking a beta blocker?
COPD, diabetes
Esmolol is _________ acting
ultra short
What are cholinomimetics (direct acting) mode of action?
- Bind to and active M or N receptors
- Esters of choline, alkaloids
What are cholinomimetics (indirect acting) mode of action?
Inhibit hydrolysis of ACh
- inhibit action of acetylcholinesterase
- prolongs effects of ACh released at junction
What are cholinomimetic effects on the eye?
- muscarinic agonists: miosis
- increase intracocular drainage
What are cholinomimetic effects on the CV system?
- reduction in peripheral vascular resistance
- vasodilation (reduction in BP; reflex tachycardia)
In large doses, can cause bradycardia
What are examples of indirect acting cholinomimetics?
Simple alcohols
- quaternary ammonium group (ex: edrophonium)
Carbonic acid esters of alcohols
- quaternary or tertiary ammonium group (ex: carbamates and neostigmine)
Organic derivatives of phosphoric acid
- Ex: organophosphate
What are the major therapeutic uses of indirect cholinomimetics?
Disease of the eye
GI and urinary tracts
Neuromuscular junction (myasthenia gravis)
- autoimmune against ACh receptor
Atropine OD
S/S of organophosphate exposure? What is the treatment?
SLUDGE-M
Tx: atropine, pralidoxime
What is edrophonium used for?
Diagnostic test for MG
What does SLUDGE-M stand for?
Salivation
Lacrimation
Urination
Defecation
GI motility
Emesis
Miosis (constriction of the pupil)
S/S of muscarinic excess? What can cause this, and what is the treatment?
SLUDGE-M
Caused by poisonous mushrooms
Tx: atropine
S/S of atropine OD? What can cause this, and what is the treatment?
BRAND
Caused by belladona
Tx: physostigmine
What does BRAND stand for?
Blind, red, absent bowel sounds, nuts, dry
What type of receptors are all adrenergic receptors?
GPCR
In angina classification, what is the difference in stable, unstable, and variant?
Stable: angina of effort (classic)
Unstable: acute coronary syndrome
Variant: Prinzmetal, angina inversa
What are the causes of stable, unstable, and variant angina?
Stable: plaque
Unstable: plaque
Variant: hyperreactive vessels
What are the precipitating factors of stable, unstable, and variant angina?
Stable: exercise, stress
Unstable: resting
Variant: resting/vasospasm
Stable angina is ______ and may be relieved by ______
Brief
Rest
Unstable angina is an __________
Emergency
Variant angina is considered ______ with only _______ of anginas being this type
Rare
2%
What are NO, nitrates, nitrites actions on vascular smooth muscle?
Activate GC, increase cGMP: relaxation
Good: increase venous capacitance, decrease ventricular preload, decrease heart size, decrease CO
Bad: orthostatic hypotension, syncope, HA, reflex tachycardia
What are beta-2 agonists actions on vascular smooth muscle?
GPCR, cAMP, relaxation (mainly respiratory)
What are beta blockers actions on vascular smooth muscle?
Decrease demand (HR)
What are calcium channel blockers actions on vascular smooth muscle?
Less total calcium: relaxation
What is sildenafil actions on vascular smooth muscle?
Block PDE5, increase cGMP: relaxation
Describe the pathway of blood vessel contraction in periphery
Involves an influx of calcium
1. Calcium is released from SR and binds to calmodulin
2. Calcium calmodulin activates MLCK (MLCK is responsible for adding phosphate group to myosin light chain)
3. Phosphorylated myosin can interact with actin, causing contraction
Pathway of relaxation in the periphery
- Beta 2 agonists increase cAMP production
- cAMP deactivates MLCK
- Increase in cGMP will dephosphorylate MLCK (cGMP can be increased by NO)
- This will cause relaxation
What is the other way that relaxation in the periphery can occur (in terms of guanylyl cyclase)?
- NO activates guanylyl cyclase
- GC turns GTP into cCMP
- gCMP desphosphorylates MLCK, causing relaxation
What type of drug is sildenafil, and what is its MOA?
PDE inhibitor
Inhibits breakdown of cAMP and cGMP by blocking phosphodiesterase
Postive inotropic effects
What type of channels do calcium channel blockers work on?
L-type in the vascular smooth muscle and heart
Relaxation:
- some effects on GI, GU, uterine
Long lasting smooth muscle relaxation, reduce BP
What do CCB do in the heart?
Decrease contractility
Decrease SA node pacemaker rate
Decrease AV node conduction velocity
When it comes to selectivity, what are the two main types of CCB and what is their focus? sorry I know this is worded weird
Dihydropyridines: more peripheral vasculature
Verapamil and Diltiazem: more cardiac
What is the toxicity associated with CCB?
Serious cardiac suppression (rare)
Bradycardia
AV block
CHF
Small facts about beta blockers
- not vasodilators
- used in angina of effort and silent (ambulatory) ischemia
Beneficial effects of beta blockers
Decrease oxygen demand
- decrease HR
- decrease BP
- decrease contractility
What are the 4 anatomical control sites that antihypertensives work on?
- diuretics: deplete sodium
- sympathoplegics: decrease PVR and CO
- direct vasodilators: relax vascular smooth muscle
- anti-angiotensins: block activity or production
Antihypertensives can act on 1 or more of these
What is the hydraulic equation?
BP = CO X PVR
Cardiac output is a function of:
stroke volume
heart rate
venous capacitance (preload)
What drugs are CNS sympathoplegics?
methyldopa
clonidine
What do CNS sympathoplegics do? (clonidine and methyldopa)
Primary antihypertensive activity due to alpha agonist activity in the brainstem; decreases sympathetic stimulation
Bind more tightly to alpha 2 than alpha 1
Propanolol MOA and what does it do?
Antagonizes beta 1 and 2 receptors
Lowers BP, decreases CO, inhibits renin
What is propanolol toxicity associated with?
Beta blockade
What are the alpha 1 adrenoceptor antagonist? How do they work?
Prazosin, terazosin, doxazosin
Block alpha 1 at arterioles and venues
Dilates both resistance and capacitance vessels
BP is more reduced in upright position
Vasodilators example
Minoxidil
Hydralazine
Sodium nitroprusside
Fenoldopam
What is minoxidil MOA?
Opens K+ channels in smooth muscle
- stabilized potential, less likely to contract
Dilates arteries and arterioles
What is hydralazine MOA?
Dilates arterioles (NO production)
What is the toxicity associated with hydralazine?
HA, nausea, sweating, flushing
What is sodium nitroprusside used for?
HT emergencies, cardiac failure
Dilates arterial and venous vessels
MOA of sodium nitroprusside
Relaxes vascular smooth muscle
- breaks won in blood to release NO
- increases intracellular cGMP
What is fenolopam used for?
HTN emergencies, post op HTN
Peripheral arteriolar dilator
MOA of fenoldopam
Agonist of D1 receptors
What does D1 and D5 work on?
Brain, effector tissues, smooth muscle of the renal vascular bed
What does D2 work on?
Brain, effector tissues, smooth muscle, presynaptic nerve terminals
What does D3 work on?
Brain
What does D4 work on?
Brain, CV system
What do ACE inhibitors do?
Block ACE, which is what converts angiotensin 1 to angiotensin 2
This causes decreased BP
What do ARBS do?
Block angiotensin II so it can’t convert to aldosterone
What does aldosterone do?
Increased sodium and water retention
Definition of heart failure
When the heart fails to meet the metabolic demands of the tissues or not pumping properly (inadequate CO)
Causes of HF
Most common: CAD
Chronic BP
Uncontrolled hyperthyroidism
Graves disease
How does CAD cause HF?
Coronary artery disease → angina→ MI (death of cardiac myocytes→ cannot proliferate→ scar tissue replaces them making heart pump ineffectively) → scarring/remodeling→ heart failure
What are the two types of HF?
Systolic: reduced cardiac function
Diastolic: reduced cardiac filling (peripheral), cardiac hypertrophy
Describe systolic HF
Heart is not pumping due to thin heart muscle walls
Decreased CO and EF
Describe diastolic HF
Heart muscle is too thick, so it’s pumping effectively but unable to pump as much blood because there isn’t room
Decreased CO with normal EF
What is congestive heart failure?
Increased left ventricle pressure at end diastole
Results in increased pulmonary pressure (pulmonary edema)
Pathway of normal cardiac contractility
- Trigger calcium enters cell
- binds to channel in SR, release stored calcium
- Frees actin to interact with myosin
What are the 4 factors in cardiac performance?
Preload
Afterload
Contractility
HR
Define preload
Measure of stretch, not volume
If there is increased blood volume or venous tone, this will increase preload
Define after load
Force that the heart has to pump against
Essentially is our BP: the higher the BP, the higher the after load
Define contractility
Contraction of myocytes (inotropy)
MOA of digoxin
Inhibits Na/K ATPase pump to maintain normal resting potential
Positive inotrope
What is the EC50 and TC50 of digoxin?
EC50: 1 ng/ml
TC50: 2 ng/ml
What are the effects of digoxin on other organs?
Affects all excitable tissues d/t inhibition of Na/K pump
- Smooth muscle
- CNS
What is an example of PDE inhibitor?
Milrinone
What is the crescent shaped node in the right atrium?
SA node / pacemaker
What is the rate of contraction at the SA node?
75 beats/min
What is the node at the junction of the aria and ventricles?
AV node
What is the bundle at the interventricular septum? Another name for this?
Atrioventricular bundle
Bundle of His