• 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

1/822

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;

822 Cards in this Set

  • Front
  • Back
What is the distribution of cholinergic neurons?
Motor neurons to skeletal muscle; Parasympathetic pre- and postganglionic neurons; ; Sympathetic preganglionic neurons; Sympathetic postganglionic neurons to sweat glands and skeletal vesselsh
Which receptors mediate the actions of NE and Epi, and how do they differ?
α-adrenoceptors: α1=postjunctional/smooth muscle, α2=prejunctional/nerve terminals;
β-adrenoceptors: β1=heart, β2=smooth muscle, β3=lipocytes
How are receptor subtypes classified?
Based on relative affinities for agonist and antagonist drugs
What are nonadrenergic noncholinergic (NANC) mediators and what are some examples of them?
Substances released from nerve endings that act as neurotransmitters, co-transmitters, or neuromodulators – Examples: ATP, vasoactive intestinal peptide (VIP), substance P, neuropeptide Y (NPY), NO; all are released from motor or sensory nerves, except NO which is nitrergic
What is the distribution of nitrergic neurons?
Sympathetic and parasympathetic postganglionic neurons
What is the distribution of dopaminergic neurons?
Sympathetic postganglionic neurons to renal vasculature smooth muscle
What is the distribution of noradrenergic (aka adrenergic) neurons?
Most sympathetic postganglionic neurons
What neurotransmitters are associated with the sympathetic and parasympathetic nervous systems respectively?
Sympathetic: preganglionic=ACh, postganglionic=NE, adrenal medulla=ACh
Parasympathetic: : preganglionic=ACh, postganglionic=ACh
How does myelination differ between pre- and postganglionic nerves of the ANS?
Preganglionic nerves are myelinated whereas the postganglionic nerves are not
What happens at the varicosities of neurons?
Release of neurotransmitters
What do NO activate?
Not a receptor, but rather activates guanylate cyclase (GTP --> cGMP)
What are the sympathetic and parasympathetic effects on the iris/pupil?
Sympathetic: mydriasis (dilation); Parasympathetic: miosis (constriction)
Are the vascular endothelium and ventricular myocardium innervated by parasympathetic nerves?
No, but muscarinic receptors are present and the cells will respond when muscarinic agonists are administered
What describes the parasympathetic innervation of blood vessels?
Cholinergic innervation restricted to facial region (blush), uro-genital organs, GI mucosa, tongue and pharynx
What describes the sympathetic innervation of blood vessels?
Nearly all vascular beds receive noradrenergic innervation via the sympathetic nervous system – the exception is that skeletal muscle receives cholinergic innervation from the SNS which causes dilation of the blood vessels
What is the effect of sympathetic fibers on the heart?
Affect rate, conduction velocity, and contractility by innervating every aspect of the heart
What is the effect of parasympathetic fibers on the heart?
Affect rate and conduction velocity by innervating the SA node, AV node, and atria – they do not have an appreciable effect on the His-Purkinje system and ventricular myocardium!
What are the subtypes of dopamine receptors and how do they differ?
D1-4: renal vasculature; D2: prejunctional; D5: brain
What two subtypes of cholinoceptors exist?
Muscarinic: M1-5 (smooth muscle, heart, glands, endothelium); Nicotinic: Nn (parasympathetic and sympathetic ganglia – i.e. autonomic) and Nm (skeletal muscle end-plate – i.e. somatic)
Which part of the autonomic nervous system controls gluconeogenesis and glycogenolysis?
Sympathetic
Which physical effects are produced by both the sympathetic and parasympathetic nervous systems?
Secretion at the salivary glands; Contraction of the uterus
Which drugs reduce the synthesis and storage of NE and by what mechanism?
α-methyltyrosine: competes with tyrosine for tyrosine hydroxylase (used for pheochromocytoma);
α-methyldopa: taken up by the vesicle and is converted to α-methylnorepinephrine (=false transmitter used for the treatment of hypertension due to its effects on α2-receptors)
What effect can glucocorticoids have on noradrenergic nerve endings?
They can inhibit uptake 2 and potentiate Epi
What is the effect of Bretylium and Guanethidine?
They inhibit NE release (can cause some initial NE release) – used for arrhythmias
What drugs can block the postsynaptic NE receptors?
α- and β-adrenoceptor antagonists
What is the mechanism of action of Tyramine and what cautions should be provided with this drug?
It is an indirect-acting stimulator of NE release (it does so by getting into the vesicle and causing NE release) – Tyramine is found in many food stuffs (e.g. cheese, beer, red wine) and is normally degraded by MAO; however for people on MAO-inhibitors, consuming these foods can lead to high enough Tyramine levels to have an effect on the vesicles and cause NE release
What is the mechanism of action of Reserpine?
It inhibits the active transporter on the vesicle membrane that pumps dopamine in, and it inhibits the pumping of NE back into the storage vesicle as it is being recycled (i.e. there is a partial depletion of NE from the storage vesicle); toxicities of Reserpine prevent its widespread use
What is the mechanism of action of cocaine?
It blocks uptake 1, thus prolonging the presence of NE in the synaptic cleft
What is the terminal product of metabolism by COMT and MAO of DA?
Homovanillic acid (HVA)
What factors in CHF cause diastolic dysfunction?
Massive left ventricular hypertrophy; Myocardial fibrosis; Deposition of amyloid; Constrictive pericarditis
What is cor pulmonale?
Chronic severe pulmonary hypertension resulting in right-sided heart failure; can be caused by disease of the lungs or by left-sided heart failure
What usually causes right-sided heart failure and what changes are seen in the heart?
Usually caused by left-sided heart failure; Presents with right ventricular dilation and hypertrophy
What signs/symptoms are associated with left-sided heart failure?
Pulmonary congestion/edema; Alveoli with hemosiderin-laden macrophages; Prerenal azotemia; Hypoxic encephalopathy
What are causes of left-sided heart failure?
Ischemic heart disease; HTN; Aortic/mitral valvular disease; Non-ischemic myocardial disease
How does the size of a heart correlate with disease?
350-600 grams: pulmonary HTN, ischemic heart disease; 400-800 grams: systemic HTH, aortic stenosis, mitral regurgitation, dilated cardiomyopathy; 600-1000 grams: aortic regurgitation, hypertrophic cardiomyopathy
What is the difference between forward and backward failure with CHF?
Forward: diminished CO; Backward: damning back of blood in the venous system
What events follow cardiac dysfunction?
Increased cardiac output --> dilation of the ventricles --> left-ventricular hypertrophy --> heart failure
What is true according to the Frank-Starling mechanism?
Increasing preload (i.e. through dilation) will increase contractility
What is congestive heart failure preceded by?
Cardiac hypertrophy (in an attempt to maintain arterial pressure and perfusion of vital organs) – myocytes can hypertrophy, but they cannot increase in numbers
gWhat are the three variants of angina?
Stable: comes on with exertion; Prinzmetal: episodic, comes on at rest, due to coronary artery spasm, elevated ST segment on EKG; Unstable: occurs at rest, prolonged duration of chest pain, induced by disruption of atherosclerotic plaque with superimposed thrombosis, pre-infarction angina
What microscopic changes are seen after MI?
4-12 hours: coagulation necrosis (wavy fibers); 2-3 days: acute inflammation; 2-4 weeks: vascularized granulation tissue (fibrosis)
What macroscopic changes are seen after MI?
<12 hours: nothing; 12-24 hours: red-blue hue; 7-10 days: soft yellow-tan area; 10 days-2 weeks: hyperemic zone of granulation tissues
What parts of the heart are associated with the left anterior descending, right, and left circumflex coronary arteries, respectively?
Left anterior descending: anterior left ventricle, apex, anterior interventricular septum; Right: posterior left ventricle, posterior interventricular septum; Left circumflex: lateral left ventricle
What change is seen in the heart, 2 months or more after an MI?
Dense collagenous scar
What is the prominent mechanism of cell death associated with MI?
Coagulative necrosis (visible within 12-24 hours after MI)
Besides plaques, what are some other causes of MI?
Vasospasm, emboli, vegetative endocarditis, vasculitis, hematologic abnormalities, amyloid deposition
What signs/symptoms are associated with right-sided heart failure?
Congestive hepatomegaly; Passive congestion; Centrilobular necrosis; Cardiac sclerosis; Congestive splenomegaly; Ascites; Congested kidneys; Hypoxic encephalopathy; Pleural effusions; Subcutaneous edema
What are examples of acute coronary syndromes?
Acute MI; Unstable angina; Sudden cardiac death
At what point does an obstructive lesion prevent sufficient myocardial oxygen demand?
When it causes >75% obstruction; >90% stenosis will lead to inadequate blood flow at rest (unstable angina)
What clinical signs are associated with MI?
Rapid, weak pulse; Diaphoresis; Dyspnea; New Q waves on EKG – silent MI is common in diabetics and elderly
What complications are associated with MI?
Rupture of the ventricular wall (2-3 days post-MI) --> cardiac tamponade; Arrhythmias (most common cause of death post-MI)
Which enzymes involved in the metabolism of tyrosine to epi are found in the cytoplasm?
Tyrosine hydroxylase, Dopa decarboxylase, Phenylethanolamine-N-methyltransferase (adrenal);
Dopamine-β-hydroxylase is found inside the storage vesicle
What is the terminal product of metabolism by COMT and MAO of NE and Epi?
Vanillylmandelic acid (VMA)
What characterizes the MAO enzymes?
They are not very specific and will metabolize many types of amines by removing the amine from the α-carbon, which abolishes agonist activity; They are located inside mitochondria and are mostly found in noradrenergic nerve ending and in the liver (most important site); there are two isoforms
What type of compound is metabolized by COMT and where does this mainly happen?
Catechols (phenolic rings with 2 adjacent hydroxyls) – the most important site of COMT metabolism is in the liver; degradation by COMT reduces potency ~100 fold
By what mechanisms is NE removed after its release?
Taken up by effector cells (facilitated diffusion/uptake 2) and degraded by MAO or COMT;
Taken up by nerve terminal (active transport/uptake 1), moved to vesicles or degraded by MAO; Diffusion from synaptic cleft (if it diffuses to the liver, it can be degraded by MAO and COMT)
What are the catecholamines?
DA, NE, Epi
What molecule has an effect on Tyrosine Hydroxylase?
NE negatively inhibits this enzyme
What is the difference between autoreceptors and heteroreceptors found on varicosities?
Regarding the noradrenergic neuron, α2-autoreceptors will reduce subsequent NE release in response to NE, whereas heteroreceptors will modulate the release of NE in response to other neurotransmitters (e.g. ACh from adjacent neurons)
What important enzyme associated with the degradation of neurotransmitters is found inside the mitochondria?
Monoamine oxidase: involved in the breakdown of 5-HT, NE, Epi, DA
What enzymes are responsible for the metabolism of tyrosine into epinephrine?
Tyrosine --> (via tyrosine hydroxylase) Dopa --> (via dopa decarboxylase) Dopamine --> (via dopamine-β-hydroxylase) Norepinephrine --> (via phenylethanolamine-N-methyltransferase) Epinephrine (only in adrenal glands)
What is the terminal product of metabolism by COMT and MAO of DA?
Homovanillic acid (HVA)
What clinical signs are associated with aortic stenosis?
Heaped up calcified masses within the aortic cusps; Left ventricular hypertrophy – poor prognosis if angina, CHF, or syncope are present
What causes rheumatic heart disease?
Antibodies against M protein of Streptococci which cross-react with glycoprotein antigens in the heart
What causes mitral valve prolapse?
Enlarged, myxomatous, floppy mitral valve leaflets that balloon back into the left atrium during systole, causing a midsystolic click; often found in young females (incidental finding), and associated with Marfan syndrome
In what patients is mitral annular calcification seen and what problems are associated with these deposits?
Females > 60; can lead to mitral regurgitation, stenosis, and arrhythmias
What are causes of mitral regurgitation?
Abnormalities of leaflets and commissures (e.g. infective endocarditis, mitral valve prolapse);
Abnormalities of tensor apparatus (e.g. rupture of papillary muscle, rupture of chordae tendineae);
Abnormalities of left ventricular cavity/annulus (e.g. left ventricular enlargement, calcification of mitral ring)
What is the major cause of mitral stenosis?
Rheumatic heart disease
What problems arise in a patient with a congenitally bicuspid aortic valve?
Not symptomatic at birth, but they are predisposed to degenerative calcifications (on midline raphe of larger cusp)
What sign is usually responsible for bringing systemic hypertensive heart disease to the attention of medical personnel?
Atrial fibrillation
What are the most common causes of death in patients with hypertensive heart disease?
CHF, sudden cardiac death, renal disease stroke
What is the most common valvular abnormality and what is it often caused by?
Aortic stenosis; caused by post-inflammatory scarring
What age range does rheumatic fever usually present in?
5-15 years; it is very uncommon these days
What clinical findings are associated with infective endocarditis?
Fever, murmur, petechiae, splinter hemorrhages, Janeway lesions (lesions on palms/soles), Osler nodules (nodules in pulp of digits), Roth spots (retinal hemorrhages)
What valves are most commonly affected by infective endocarditis?
Aortic/mitral valve; Right heart valves in IV drug abuse
What organisms are mostly responsible for infective endocarditis?
Streptococcus viridans > Staphylococcus aureus (most common bug with IV drug abuse)
What factors contribute to the development of infective endocarditis?
Neutropenia, immunodeficiency, alcohol abuse, IV drug abuse (!), DM
What are the two forms of infective endocarditis and how do they differ?
Acute: highly virulent organism affecting previously normal valve, patient will have rapidly developing fever with rigor/malaise/weakness, death occurs in 50% of patients within days-weeks;
Subacute: low virulence organism affecting an abnormal/injured valve, patient will have less valvular destruction and will present with low-grade fever/weight loss/flu-like symptoms, disease has protracted course causing lower mortality rates
What causes infective endocarditis?
Microbial colonization of heart valves resulting in friable, infected vegetations, frequently causing valve damage
What part of the heart is mostly affected by rheumatic heart disease?
Valves
What is the morphology associated with rheumatic heart disease?
Aschoff bodies: foci of fibrinoid necrosis surrounded by leukocytes found in the pericardium, myocardium (mainly), and endocardium; Bread and butter pericarditis; Fibrinoid necrosis of cusps; Vegetations along lines of closure; MacCallum plaques: subendocardial collections of Aschoff nodules
What are the Jones criteria for rheumatic heart disease?
2 of 5 major manifestations: migratory polyarthritis of large joints, carditis (any layer), subcutaneous nodules, erythema marginatum (macular skin lesions in bathing suit distribution), Sydenham chorea (rapid, involuntary, purposeless movements);
2 minor manifestations: fever, arthralgia, increased blood levels of acute phase reactants
What complications are associated with artificial valves?
Thromboembolic complications, infective endocarditis, structural deterioration, hemolysis, obstruction, exuberant healing
What is the problem with arrhythmogenic right ventricular cardiomyopathy?
Right-sided failure and rhythm disturbances leading to ventricular tachycardia and sudden death; Right ventricular wall is thinned, with profound fatty infiltration (defect on chromosome 14)
What are the subtypes of restrictive cardiomyopathy?
Endomyocardial fibrosis: African children, ventricular subendocardial fibrosis, etiology unknown;
Loeffler endocarditis: occurs in temperate zones, endomyocardial fibrosis, eosinophilic leukemia;
Endocardial fibroelastosis: focal/diffuse fibroelastic thickening of the endocardium, most common in children younger than 2, often accompanied by congenital cardiac anomalies
What are possible causes of restrictive cardiomyopathy?
Idiopathic, radiation fibrosis, amyloidosis, sarcoidosis, tumors, inborn-errors of metabolism
What are possible causes of hypertrophic cardiomyopathy?
Genetic mutations in genes that encode sarcomeric proteins (e.g. β-myosin heavy chain, troponin T, α-tropomyosin, myosin-binding protein C)
What are possible causes of dilated cardiomyopathy?
Idiopathic, alcohol, peripartum, genetics (e.g. dystrophin), doxorubicin
What are the three different types of cardiomyopathy and how do they differ?
Dilated: most common, dilation of all chambers, impairment of contractility, death in <5 years;
Hypertrophic: myocardial hypertrophy, thickening of interventricular septum, “banana-like configuration”, impairment of compliance = poor diastolic relaxation, variable clinical features;
Restrictive: impairment of compliance = impaired diastolic filling = reduced CO
What causes nonbacterial thrombotic endocarditis?
Deposition of small masses of fibrin/platelets/blood components on valve leaflets; seen in patients with cancer (adenocarcinoma) and sepsis who have developed hypercoagulable states (e.g. DIC) – nonbacterial thrombotic endocarditis is also known as marantic endocarditis
What is Libman-Sacks disease associated with?
SLE – sterile vegetations mainly affecting the AV valves
What causes carcinoid heart disease and what does it affect?
Carcinoid tumors produce serotonin and other bioactive products which cause cardiac lesions, mainly affecting the tricuspid and pulmonary valves
What are common causes of pericarditis?
It is usually secondary to disorders involving the heart or adjacent mediastinal structures (e.g. MI, surgery, trauma, radiation, tumors, infections)
Which form of acute pericarditis is commonly associated with tuberculosis or mycotic infection?
Caseous pericarditis: caseation within the pericardial sac; leads to constrictive pericarditis
What characterizes hemorrhagic pericarditis?
An exudate of blood mixed with a fibrinous/suppurative effusion; follows cardiac surgery, tuberculosis, or malignancy
Which form of acute pericarditis is mostly associated with infection and what characterizes it?
Purulent/suppurative pericarditis: infection can reach pericardium via direct extension, seeding from blood, lymphatic extension, or during cardiotomy; associated with a creamy pus over red, granular coated serosal surfaces – can lead to constrictive pericarditis
What is the most common clinical form of acute pericarditis and what characterizes it?
Fibrinous/serofibrinous pericarditis: associated with a pericardial friction rub; often seen with MI (Dressler’s syndrome), but can also be due to any of the etiologies for serous pericarditis
What diseases are associated with acute serous pericarditis (slow accumulation of exudate)?
Rheumatic fever, SLE, scleroderma, tumors, uremia
When are pericardial effusions problematic?
When there is a rapid accumulation of fluid (can cause fatal cardiac tamponade)
What is myocarditis and what is the most common cause in the US?
Inflammatory process that injures cardiac myocytes – most common US cause is viral infections (mainly coxsackievirus A and B)
What clinical signs are associated with myocarditis?
Heart failure, arrhythmias, systolic murmur, sudden death – can mimic acute MI
What other causes are associated with myocarditis?
Infectious: Trypanosoma cruzi (Chagas disease in South America), parasitic disease (Toxoplasma gondii), bacterial disease (diphtheria, Lyme disease) – common in immunodeficient patients;
Noninfectious: rheumatic fever, SLE, drug allergies (doxorubicin/Adriamycin, lithium, cocaine), catecholamines, amyloidosis (seen with congo red dye), iron overload (due to hereditary hemochromatosis, hemosiderosis)
What is the most common primary tumor of the heart in adults?
Myxoma: neoplastic lesion that occurs most commonly in the left atrium in the region of the fossa ovale; histologically they are composed of stellate/globular myxoma cells, endothelial cells, smooth muscle cells, and undifferentiated cells – can be identified by ECG, 10% is familial; can be seen in a Carney complex
What complications are associated with heart transplants?
Graft arteriosclerosis (progressive, diffuse intimal proliferation of the coronary arteries causing downstream myocardial ischemia = major limitation to long-term success); Infection; Malignancies (lymphoma related to EBV)
What are the most common reasons for heart transplants?
Dilated cardiomyopathy and ischemic heart disease
What tumors frequently metastasize to the heart?
Lung, breast, melanoma, leukemia, lymphoma
What is the most frequent primary cardiac tumor in children?
Rhabdomyoma: may cause valvular or outflow tract obstruction; they are hamartomas and may be associated with tuberous sclerosis; grossly they are grey-white ventricular wall masses; microscopically they are composed of “spider cells”: polygonal cells with glycogen-laden vacuoles
What are papillary fibroelastomas and where are they usually found?
Hair-like projections on the endothelial surface (resemble Lambl excrescences); usually found on right-sided valves in children and left-sided valves in adults
What is a lipoma and where in the heart are they most commonly found?
Excessive fat accumulation; more commonly seen in the left ventricle, right atrium, septum
What are the two forms of chronic/healed pericarditis and what characterizes each?
Adhesive mediastinopericarditis: pericardial sac is obliterated and the parietal layer is tethered to mediastinal tissue, leading to a strain on cardiac function; may follow suppurative/caseous pericarditis, cardiac surgery, irradiation to the mediastinum;
Constrictive pericarditis: heart is encased in dense fibrous/fibrocalcific sac, limiting diastolic expansion, restricting cardiac output; resembles a restrictive cardiomyopathy
What characterized rheumatic heart disease?
The most common finding is fibrinous pericarditis; there may be rheumatoid granulomatous nodules in the myocardium, endocardium, valves, and the root of the aorta
In a normal heart, which horizontal lead would show the largest depolarization?
Either V4 or V5
How does a right bundle branch block (RBBB) show up on EKG?
Widened QRS (>0.12 sec); rsR’ pattern in V1 (double peak)
What defines a third degree heart block?
No relation between atrial and ventricular activity (requires pacemaker)
What defines a second degree heart block, Mobitz Type II?
QRS wave dropped without PR interval lengthening (extreme bradycardia, requires pacemaker)
What defines a second degree heart block, Mobitz Type I (Wenckebach)?
PR interval lengthening until a QRS wave is dropped (causes mild bradycardia)
What defines a first degree AV block?
PR interval > 0.2 seconds; is not associated with any complications except in patients who are taking beta-blockers or calcium-channel blockers (since they can worsen the AV block)
What must be present to call something a sinus rhythm?
Upright p-wave in lead II (except in dextrocardia) that is related to the QRS complex
Why do both depolarization and repolarization of the heart show up as positive deflections on EKG?
Depolarization is a “positive deflection” flowing towards the electrode, whereas repolarization is a “negative deflection” flowing away from the electrode (two negatives make a positive)
What are the various waves seen on EKG associated with?
P wave: depolarization of the atria; QRS complex: ventricular depolarization; T wave: ventricular repolarization
In a normal heart, which vertical lead would show the largest depolarization?
Lead II, since it is oriented in the same direction as that of normal depolarization
What is the difference in atrial fibrillation and atrial flutter?
Atrial fibrillation: rapid, irregular P waves originating from many sites on the atrium;
Atrial flutter: fewer sites that generate P waves are present, leading to a more organized, “saw-tooth” appearance on EKG
Where is the indeterminate axis?
Between 180° and 270°
What are causes of right axis deviation (axis between 90° and 180°)?
Right ventricular hypertrophy, significant myocardial infarction, dextrocardia, posterior hemiblock
What are causes of left axis deviation (axis between 0 and -90°)?
Left ventricular hypertrophy, anterior hemiblock
What is meant by a rhythm described as bigeminy?
Alternating supraventricular/ventricular rhythms
What does a junctional rhythm look like on EKG?
Narrow QRS, no obvious atrial activity (check lead II), rate around 60 bpm (since the rhythm originates from the AV node)
What is the defect with a wandering pacemaker?
The site of the controlling pacemaker shifts from beat to beat, leading to changes in the P wave (switches from upright to inverted, and back)
How does a left bundle branch block (LBBB) show up on EKG?
Widened QRS (>0.12 sec); Completely upright QRS in V6 and I
What is seen on EKG with atrial fibrillation?
Rapid and irregular rhythm
What clinical implications are associated with atrial fibrillation/flutter?
Rapid ventricular response (= decreased filling, decreased output, increased oxygen demand);
Loss of atrial contraction (aka atrial “kick” = decreased ventricular filling, decreased output);
Risk of embolic events from stagnant atria
Which leads are used to find atrial abnormalities and what is considered abnormal?
Lead II and V1/V2; Abnormal: wide, tall, double-peaked or biphasic P waves
What are the lateral leads?
AVL/I/V5/V6
What are the septal and anterior leads, respectively?
Septal: V1/V2; Anterior: V2/V3/V4
Which leads are associated with the bottom of the heart?
III/AVF/II
What are the characteristics of ischemia, injury, and infarction on EKG, respectively?
Ischemia: T wave inversion; Injury: ST segment elevation; Infarction: larger Q waves
What is an indication of right ventricular hypertrophy on EKG?
Tall R in V1 (where you would expect a deep S)
What is the calculation for left ventricular hypertrophy?
Height of S in V1 + Height of R in V5/V6 (whichever is larger) > 35mm (patient has to be over 35 years of age)
What does a net positive deflection in lead I indicate?
Patient is either WNL or has a LAD; a net negative deflection would indicated RAD or indeterminate axis
How is the actual axis determined once the correct quadrant has been identified?
Find the isoelectric point and move 90° into the correct quadrant
What would a net positive deflection in AVF indicate?
Patient is either WNL or has a RAD; a net negative deflection would indicate LAD or indeterminate axis
What are the posterior leads?
V1/V2 – looking for ST depression and a tall R
What does ST depression in V1/V2 without any other abnormalities mean?
Subendocardial infarction
What is the blood supply associated with each of the leads?
Inferior (III/AVF/II): right coronary or LAD; Septal (V1/V2): LAD; Anterior (V2/V3/V4): LAD; Lateral (AVL/I/V5/V6): circumflex; Posterior (V1/V2): right coronary
How do agonistic effects differ between NE, Epi, and Isoproterenol (Iso) on the β-receptor and what are the β-receptor subtypes?
Iso > Epi ≥ NE; consist of β1 (Epi = NE), β2 (Epi > NE), β3-subtypes (Iso = NE > Epi)
What are the effects of catecholamines on other cardiac cells?
Shorten the effective refractory period (i.e. channel inactivation) and shorten action potential duration: lead to arrhythmias; Increase [Ca2+]i leading to an increase in contractility
What are the effects of catecholamines on pacemaker and latent pacemaker cells of the heart?
Increase phase 4 depolarization (more rapid succession of APs = increased HR); Increase phase 0 depolarization (increased conduction velocity)
What adrenoceptor is involved mainly in metabolic effects?
β2
What location in the body has β3 receptors?
Fat cells, stimulation causes activation of glycolysis
What are the actions of the catecholamines on cardiac muscle and smooth muscle, respectively?
Cardiac muscle (β1): excitation, causing an increase in contractility, HR, and conduction velocity; Smooth muscle: vasoconstriction of arterioles/pre-capillary sphincters/veins (via α1 and α2), vasodilation of skeletal/visceral beds (β2), pupillary dilation (α1), airway smooth muscle relaxation (β2), renin release (stimulation: β1, inhibition: α2), transmitter release inhibition (α2)
Besides stimulating Dopamine receptors, what other receptor does DA have an effect on?
β1-receptors
What are the sympathomimetic amines and how do they function?
DA, NE, Epi; their action mimics the effects of stimulating the sympathetic nervous system
How do agonistic effects differ between NE, Epi, and Isoproterenol (Iso) on the α-receptor and what are the α-receptor subtypes?
Epi ≥ NE >> Iso; consist of α1-receptors (A, B, D) and α2-receptors (A, B, C)
What kinds of receptors are adrenoceptors and why is that important?
G-protein coupled receptors: are able to detect very small differences between similar molecules
What receptor is important in renal vascular beds (as well as coronary/cerebral)?
D1 receptor: DA causes dilation
What are the effects of Phenylephrine on diastolic/systolic BP and HR?
Phenylephrine: causes vasoconstriction via α-receptors which increases diastolic and systolic pressures equally, which causes a decrease in HR due to the baroreceptor reflex (it has no direct effect on the heart)
What is the α- and β-specificity of the agonists Phenylephrine, NE, Iso, and Epi?
Phenylephrine: α1
NE: α1, α2, β1
Iso: β1, β2
Epi: α1, α2, β1, β2
Where are stretch receptors located and what happens when BP increases?
Carotid sinus and aortic arch; Increased BP stretches the receptors, which increases firing of afferents to the CNS
What effect does the sympathetic nervous system have on blood pressure?
Heart: increased HR and increased force of contraction (--> increased SV) will increase CO which leads to an increase in BP; Arterioles: increases vasoconstriction --> increased TPR --> increased BP; Veins: increased vasoconstriction --> increased venous return --> increased SV --> increased CO --> increased BP
What effect does the parasympathetic nervous system have on blood pressure?
It decreases HR (via the vagus nerve), which decreases CO, which leads to a decrease in BP; blood pressure is not affected by parasympathetic effects on the arterioles/veins (face, larynx, etc.)
What factors affect mean arterial blood pressure?
BP = CO x TPR; CO is dependent on HR and stroke volume, TPR is dependent on arteriolar radius and blood viscosity (not of much importance)
Which value gives an indication of total peripheral resistance?
Diastolic blood pressure
What two toxicities of catecholamines can lead to arrhythmias?
Activation of latent (ectopic) pacemakers; Shortened effective refractory period (ERP)
What is the difference in adrenoceptors present on the vascular beds in skin, splanchnic, and skeletal systems?
Skin: α1, α2 (Epi/NE cause constriction, Iso has no effect); Splanchnic: α1, α2, some β2 (Epi/NE cause constriction, Iso causes weak dilation); Skeletal: α1, α2, β2 (Epi/Iso cause dilation via β2, NE causes constriction since it has little effect on β2 receptors)
What are the effects of NE on diastolic/systolic BP and HR?
NE: diastolic pressure increases but systolic pressure increases more (since it also increases cardiac contractility via β-receptors), HR goes down due to baroreceptor reflex (despite increases in contractility)
Which G-protein is activated by α2 agonists and what are the effects of this activation?
Gi-protein: activation of Gi causes inhibition of adenylate cyclase whereas activation of the β- and γ-subunits of Gi-protein cause opening of K+ channels and closure of Ca2+ channels (thus leading to decreased neurotransmitter release in adrenergic neurons)
Which G-protein is activated by α1 agonists and what are the effects of this activation?
Gq-protein: directly activates MAP/PI-3 kinase, open calcium channels, and causes activation of phospholipase C which leads to hydrolysis of PIP2 forming DAG (activation of protein kinase C) and IP3 (raises intracellular calcium)
Which G-protein is activated by β1/β2 agonists and what are the effects of this activation?
Gs-protein: directly opens calcium channels (in cardiac muscle) and activates adenylate cyclase (which leads to a number of down-stream events such as the production of glucose)
What is responsible for the fact that the diastolic pressure in response to Epi drops below the baseline level after its initial increase?
As the concentration of Epi falls (i.e. as it is being metabolized), the preponderant effect of Epi is on the β-receptors, leading to a more significant drop in diastolic pressure (due to skeletal muscle vasodilation)
What are the effects of Epi on diastolic/systolic BP and HR?
Epi: causes a small increase in diastolic pressure (due to α-receptors) and a greater increase in systolic pressure (due to α- and β-receptors); although these pressures are increased, the baroreceptor reflex is countered by Epi’s potent effect on β-receptors of the heart which lead to increases in contractility and HR – Epi effects on diastolic pressure are dose-dependent
What are the effects of Isoproterenol on diastolic/systolic BP and HR?
Iso: causes a decrease in diastolic and systolic pressures (diastolic drop > systolic drop) due to skeletal muscle vasodilation combined with increases in contractility – HR is increased due to the baroreceptor reflex and direct cardiac stimulation
What is required chemically for direct action of a compound on adrenoceptors?
At least 2 OH groups among positions 3, 4, and the β-carbon – Dopamine is the exception
What kinds of compounds can displace and release NE from storage vesicles?
α-CH3 compounds (phenylisopropylamines)
What is required for metabolism by MAO?
NH2 group; no substitutions are allowed on the α-carbon, nor are substitutions of more than 2 carbons on the NH2
What is required for metabolism by COMT?
Both 3’ and 4’ OH groups – amphetamine and ephedrine are not substrates
What substitution confers isomerism?
β-carbon substitutions; most agonists are more potent as l-isomers; β-carbon substitutions may be required for storage of catecholamines (DA is the exception)
What happens when adding bulk to the amino terminal of adrenoceptor agonists?
2 or more carbon atoms will confer β-specificity – Phenylephrine somewhat violates that principle
What does loss of the 4’ OH group confer?
α-selectivity; whereas the presence of a 4’ OH group increases β-selectivity – the presence of a 5’ OH group specifically increases β2-selectivity
What is the adrenoceptor specificity of Albuterol and Terbutaline?
High affinity for β2 receptors (moderate β1)
What is the adrenoceptor specificity of Dobutamine?
High affinity for β1 receptors (moderate α1)
What is the adrenoceptor specificity of Clonidine and α-Methyl NE?
High affinity for α2 receptors (moderate α1)
What is the mechanism of action of Botulinum toxin?
It is a protease that is taken up into the presynaptic terminal of cholinergic neurons and interferes with the docking of ACh vesicles required for their release – only need one copy of the protease per nerve terminal to completely block ACh release
Which cholinergic receptors are found on the adrenal glands and sweat glands, respectively?
Adrenal glands: NN; Sweat glands: M3
Which muscarinic receptors are found on postganglionic autonomic targets and what is their purpose?
M1, M3: cause slow modulatory excitation of the response to NN activation
How can the muscarinic receptors be divided up and what actions are associated with each class?
Odd-numbered receptors (M1, M3, M5): activate Gq-protein leading to activation of calcium-dependent processes (e.g. contraction of smooth muscle, secretion); Even-numbered receptors (M2, M4): activate Gi-protein leading to activation of adenylate cyclase (which leads to a decrease of cAMP) and opening of K+ channels (e.g. hyperpolarization of heart SA/AV nodes)
What are the two types of nicotinic receptors and what is their pharmacologic difference?
NN: located on nerves, can be blocked by hexamethonium; NM: located on muscle, can be blocked by curare
What describes the nicotinic receptors?
Na+ channel that opens in response to the binding of two ACh molecules – continuous binding of ACh will cause a rapid desensitization, leading to closure of the channel
What is a common clinical application of Botulinum toxin?
Botox, used to fight wrinkles and muscle spasms
How is ACh synthesized?
By choline acetyltransferase (CAT) which combines acetyl CoA (synthesized in mitochondria) and choline (obtained by uptake from the extracellular space or by release from phophatidylcholine, a membrane phospholipid)
What is the only way that the action of ACh is terminated in the synaptic cleft?
By its degradation through acetylcholinesterase (AChE) bound to synaptic membranes – ACh can diffuse from the synaptic cleft and be degraded by plasma cholinesterases as well
What happens after ACh is degraded by AChE?
Choline is taken back up into the nerve terminal and is recycled
Which cholinergic receptor type is involved in memory?
Muscarinic receptors – whereas the NN receptor is involved in addiction to nicotine via pre-synaptic modulation
What characterizes the function of Methacholine?
It is ACh with an added methyl group; this causes it to be degraded slowly and be specific for muscarinic receptors only – the only clinical use it has is in diagnosing asthma; the drug is inhaled and if the person has had a break in the bronchial lining (i.e. due to developing asthma) they will have a strong response to the Methacholine and have trouble breathing
Why is therapeutic ACh not very helpful?
It is rapidly degraded by AChE and it is not specific for either nicotinic or muscarinic receptors – however, it can be useful for dilation of the eye
What is the key difference between direct- and indirect-acting cholinomimetic drugs?
Direct: ACh agonists; Indirect: AChE inhibitors
What differentiates the cardiac effects of a muscarinic agonist in low and high dose?
Low dose: tachycardia due to the response to dilation of vasculature; High dose: bradycardia due to the decreased rate of impulse generation and conduction in the SA/AV nodes
What do presynaptic nicotinic/muscarinic receptors lead to?
A decrease in the release of ACh
Which cholinergic receptor type is found on the vascular endothelium and what is the associated effect?
M3: leads to the release of NO with subsequent vascular smooth muscle release
What does activation of muscarinic receptors lead to on the iris and the lens?
Iris: contraction of pupillary sphincter muscle (miosis); Lens: contraction of ciliary muscle (accommodation to near vision)
Which muscarinic receptor type is responsible for the calcium-dependent smooth muscle contraction and secretory gland secretion?
M3
Which muscarinic receptor type is responsible for the hyperpolarizing effects on the SA/AV nodes and the atria?
M2
How does Cevimeline function?
As a M1, M3 agonist it can cause secretion from salivary glands and be used to treat xerostomia
What are the reversible inhibitors of AChE used in the treatment of Alzheimer’s disease?
Tacrine (no longer used), Donepezil, Galantamine, Rivastigmine – although they significantly improve the symptoms of early Alzheimer’s patients, they do not alter the course of the disease
What are characteristics of irreversible AChE inhibitors and what drugs belong to this classification?
Low molecular weight, lipid-soluble compounds that readily enter the CNS; they bind to the esteratic site on AChE with covalent attachment of a phosphate group to the enzyme’s active site; Examples: DFP (Isoflurophate), Echothiophate (only one used clinically), Parathion (insecticide, common with poisoning on the farm), Soman (nerve gas), Sarin (nerve gas) – treatment is possible with Pralidoxime (2-PAM), but must be done quickly before “aging” of the inhibited AChE occurs
By what mechanism do the Carbamate inhibitors function and what is specific about each?
These compound contain a carbamyl ester linkage that is hydrolyzed by AChE; after this hydrolysis, AChE will be carbamoylated and will not be able to hydrolyze ACh; this effect lasts 3-4 hours – Physostigmine: can enter CNS; Pyridostigmine/Neostigmine: cannot enter CNS; Carbaryl: pesticide
What are the competitive antagonists of AChE and how are they used clinically?
Edrophonium: used to help diagnose Myasthenia Gravis (patients will regain muscle strength upon administration, very short-term effect); Ambenonium: longer effect, used in the treatment of Myasthenia Gravis – both drugs do not contain an ester linkage and are not hydrolyzed
By what mechanism does AChE cause the breakdown of ACh?
AChE is a serine esterase; its anionic site attracts the positively-charged quarternary ammonium group of ACh and its esteratic site binds the acetyl portion causing hydrolysis by nucleophilic attack; After degrading ACh, AChE is acetylated, but rapid hydrolysis causes release of the acetyl group, regenerating the free enzyme
What are the naturally-occurring cholinomimetics and what characterizes each?
Muscarine: selective agonist of muscarinic receptors, found in certain mushrooms;
Pilocarpine: selective agonist of muscarinic receptors, useful for the treatment of open-angle glaucoma (can trigger immense sweating when taken systemically);
Nicotine; selective agonist of nicotinic receptors, found in tobacco
What characterizes the function of Carbachol?
Substitutes a methyl group on ACh by an amino group; Not degraded by AChE; Not specific for either nicotinic or muscarinic receptors – its only clinical use is in treating open-angle glaucoma
Why should direct-acting cholinomimetic drugs not be allowed to enter systemic circulation?
They will act on the vascular endothelium and cause a dangerous drop in blood pressure
What characterizes the function of Bethanechol?
Combines both modifications seen in Methacholine and Carbachol, thus causing it to not be degraded by AChE yet be specific for muscarinic receptors – clinical uses include treatment of post-operative abdominal distention, promote gastric motility, promote voiding of bladder
Why are AChE inhibitors sometimes preferred over a drug like Bethanechol?
They do not have the effect of systemic activation of muscarinic receptors on the vascular endothelium
Which muscarinic antagonist is used to treat some of the symptoms of Parkinson’s disease?
Benztropine
Which muscarinic antagonist is used to prevent motion sickness?
Scopolamine (must be used prophylactically)
What are synthetic muscarinic antagonists and what are they used for?
Ipratropium, Tiotropium: quarternary ammonium salts used as inhalants for the treatment of COPD (same effects as atropine but without inhibition of mucociliary clearance);
Homatropine, Cyclopentolate, Tropicamide: shorter-acting production of mydriasis/cycloplegia;
Darifenacin, Solifenacin: selective M3 receptor antagonist used for the treatment of incontinence
What effects do Atropine and Scopolamine have, other than CNS effects?
Eye: mydriasis, cycloplegia (paralyzes accommodation), increases intraocular pressure;
Heart: tachycardia (with strong vagal tone – i.e. young people), blocks vasodilation/hypotension; GI: block of salivary secretion, block of gastric acid secretion (high dose), inhibition of peristalsis; Respiratory tract: bronchodilation, inhibits secretions and mucociliary clearance;
Urinary tract: urinary retention; Skin: inhibits sweating
What are the Belladonna Alkaloids and how do they function?
Atropine, Scopolamine; they are competitive antagonists of muscarinic receptors; Scopolamine enter the CNS more readily and produces CNS depression (low dose) or CNS excitation (high dose); Atropine can have a very long duration of effect (~1 week)
What can be used in the treatment of AChE inhibitor poisoning?
Atropine: block muscarinic receptors; Respiratory support; 2-PAM: for reactivation of phosphorylated AChE (2-PAM does not enter the CNS)
What are the three classes of effects of AChE inhibitors?
Muscarinic stimulation of organs; Stimulation/block of ganglionic/neuromuscular transmission; Stimulation/block of cholinoceptors in the CNS
What is the most dangerous effect of AChE inhibitors?
Paralysis of the diaphragm
What are therapeutic uses of AChE inhibitors?
Atony of the GI tract and urinary bladder; Glaucoma; Myasthenia Gravis; Termination of the effects of competitive neuromuscular blocking agents; Atropine poisoning; Alzheimer’s disease
What are contraindications and toxicities associated with muscarinic antagonists?
Contraindications: narrow-angle glaucoma; Toxicities: can aggravate cognitive problems in the elderly, and increase the risk of hyperthermia (unable to sweat)
What are the two classes of Neuromuscular Junction (NMJ) blocking drugs?
Non-depolarizing antagonists: d-Tubocurare, Pancuronium, Vecuronium, Atracurium; they competitively block the binding of ACh to the NM receptor, causing weakness/paralysis –
Depolarizing antagonists: Succinylcholine (2x ACh); causes persistent stimulation of NM receptors, leading to muscular fasciculations followed by weakness/paralysis
What therapeutic uses are associated with NMJ blocking drugs and what are some associated adverse effects?
Relax skeletal muscle and reduce the dose of anesthesia required; Relax skeletal muscle in orthopedic procedures; Facilitate intubation – Adverse effects: prolonged apnea, electrolyte imbalance due to excessive release of K+ (depolarizing antagonists only)
What is the difference between Phenylephrine and Oxymetazoline in treating nasal congestion?
Phenylephrine treats nasal congestion by working on the nasal veins (α1-selective), whereas Oxymetazoline works on the nasal arterioles (α2-selective)
What is unique about Amphetamine and what is it used for?
It is more potent in its dextro-form; used for appetite suppression, but has a high abuse potential
What is the isomer of Ephedrine and what is it used for?
Pseudoephedrine; used for nasal congestion (has less CNS activity and less β activity than Ephedrine)
Besides its indirect effects, what direct effects are seen with Ephedrine and what is it used for clinically?
Ephedrine has α and β activity; it is used for postural hypotension
What are the main uses of indirectly-acting sympathomimetics?
Alerting, sleep deferring; Narcolepsy; Appetite suppression; ADHD – since these drugs release NE, they can increase HR and BP (also, tolerance may develop)
What is Ritodrine given for?
It is β2-selective and is given IV as a uterine relaxant in premature labor
What is the key difference from Terbutaline and Albuterol?
β2-selectivity in Terbutaline is due to a 5’ OH group whereas β2-selectivity in Albuterol is due to a 4’ OH group
What is the adrenoceptor specificity of Methoxamine and what is it used for?
α1-selective; used for hypotensive emergencies
How do the different isomers of Dobutamine differ in effect and what is a racemic mixture used for?
+ isomer: β1-agonist, α1-antagonist
- isomer: α1-agonist; racemic mixture: α1 effects cancel out and β1 effect remains, used as a + inotropic drug in end-stage CHF
How does Clonidine cause its antihypertensive effects?
As a α1-selective agonists in the CNS; also has some peripheral effects which lower HNT, but they are not as significant as the central effects
What adverse effect occurred with Phenylpropanolamine?
Hemorrhagic stroke (taken off the market)
What is Phenoxybenzamine’s main site of action and what other actions does it have?
Main site: α-receptors (α1 > α2); Other actions: inhibits ACh, 5-HT, Histamine receptors, and blocks uptake I and II
What commonality in name do α1-specific antagonists have and what can happen with these drugs when given in high doses?
“-osin” as in Prazosin; At high doses they can cause cyclic nucleotide PDE inhibition which causes vasodilation, leading to orthostatic hypotension
What is the key difference between Prazosin and Phentolamine/Phenoxybenzamine?
Prazosin is α1-specific and will therefore not cause the release of NE associated with α2 antagonists
How does Phentolamine differ from Phenoxybenzamine?
It is equilibrium competitive and has equal affinity for α1 and α2; other actions include inhibition of 5-HT and histamine receptors, release of histamine (causes GI disturbances), it has some α efficacy and cholinomimetic activity; It’s hemodynamic effects are the same as Phenoxybenzamine except Phentolamine has a greater effect on NE release
What are the indications for Phenoxybenzamine?
Pheochromocytoma, Peripheral vascular disease (Raynaud’s)
What adverse reactions are seen with Phenoxybenzamine?
Orthostatic hypotension, tachycardia (due to baroreceptor reflex/α2 antagonism), miosis, nasal stuffiness, inhibited ejaculation
What hemodynamic effects are seen with Phenoxybenzamine?
Decrease in TPR/BP; Venodilation; Orthostatic hypotension; Cardiac stimulation: increased HR/contractility (baroreceptor reflex), increase in NE release (due to α2 antagonism)
What is the α-receptor antagonist selectivity of Phentolamine, Phenoxybenzamine, Prazosin, and Yohimbine?
Phentolamine: α1, α2; Phenoxybenzamine: α1, α2; Prazosin: α1; Yohimbine: α2
What kind of antagonism is associated with Phenoxybenzamine and does this mean for its duration of action?
Nonequilibrium competitive (i.e. irreversible covalent bond formation with receptors); has a long duration of action (days)
What is Yohimbine used for?
Impotence, although not much since there are better drugs available
What are the indications of α1-specific antagonists?
Mild to moderate hypertension; BPH
What are the ergot alkaloids and what is their use in medicine?
An old class of drugs with complex pharmacology (function as α-adrenoceptor antagonists); Ergotamine and Dihydroergotamine are used for the treatment of migraine (due to their interaction with 5-HT and DA receptors); Ergonovine is used in post-partum hemorrhage (due to its vasoconstrictive effects)
What happens when Epi is given after a person has taken Phentolamine?
It functions solely as a β-receptor agonist (like Isoproterenol) since the Phentolamine has block both α-receptors
What is special about Tamsulosin (Flomax)?
It is specific for the α1A-receptors which are clustered on the urinary bladder base and prostate smooth muscle, leading to a smaller incidence of vascular side effects
What is the β-adrenoceptor selectivity for Propranolol, Metoprolol, Butoxamine, and Labetalol?
Propranolol: β1, β2; Metoprolol: β1; Butoxamine: β2; :Labetalol: α1, β1
What characterizes Pindolol?
Some membrane-stabilizing activity; Slight agonist activity (may produce smaller fall in HR/BP)
How does Nadolol compare to Propranolol?
It is longer-acting; It has no membrane-stabilizing activity; like, Propranolol it does not have any agonist activity
In what patient population is Propranolol contraindicated?
Patients with asthma (since it is an antagonist of β2)
How does Propranolol function as an anti-arrhythmatic?
It is a membrane stabilizer and blocks β-receptors – also used as an anti-hypertensive agent and to decrease contractility
Besides blocking β-receptors, what other actions may be seen with beta-adrenoceptor antagonists?
Stimulate β-receptors as partial agonists, exhibit partial agonism, elicit NO release from endothelium (decrease BP), as antioxidants, block Ca2+ channels, block α-receptors
What is the clinical use of Labetalol?
It is used as an anti-hypertensive; the α1-blocker component causes vasodilation and thus a reduction in BP; the β1-blocker subsequently prevents the heart from becoming tachycardic in response to the baroreceptor reflex (i.e. there is a drop in BP without an increase in HR) – Labetalol has a slight β2 agonist activity
What direct effects on the heart are seen with β1-antagonists?
Slowed AV-conduction; Decreased phase 4 depolarization; Decreased contractility (due to antagonist or inverse agonist activity); Decreased SV, Increased residual volume; Decreased velocity of contraction; Decreased CO; Decreased oxygen consumption
What is particular about the use of Esmolol (β1-selective antagonist)?
It is fast acting and has a short duration of action, making it useful for the minute-minute control of hypertension
How does Metoprolol function?
It is a selective β1-antagonists that has no agonist activity, but has very strong inverse agonist activity
What is Metoprolol used for?
CHF, HTN, myocardial ischemia
What additional effect of Carvedilol functions as an important pharmacological effect?
It’s antioxidant activity
What is the β:α selectivity of Carvedilol?
10:1 – it has the same effect as Labetalol but is much more efficient; used for the treatment of CHF; (Carvedilol is a weak inverse agonist)
What is the function of Timolol (non-selective β-adrenoceptor antagonist)?
Reduction of anterior-chamber pressure in glaucoma (has no membrane-stabilizing activity or agonist effect)
What is the difference between the SR- and RR-isomers of Labetalol?
SR isomer: α1-blocker; RR isomer: β1-, β2-blocker – β:α = 1.5:1
What peripheral cardiovascular effects are seen with β1-antagonists?
Decreased BP (due to decreased CO); Decreased Renin release --> decreased Angiotensin II in blood; Decreased GFR --> Na+ retention and edema – (there is also a decrease in lipolysis)
What side effects are associated with β-adrenoceptor antagonists?
Rebound ischemia (long-term use increases the number of β-receptors, so sudden discontinuation can lead to this problem); Tiredness; Vivid dreams; Insomnia; Hallucinations; Exercise intolerance
What contraindications are associated with β-adrenoceptor antagonists?
CHF (severe); Asthma; AV-node conduction disturbances; Hypoglycemia
What are other uses associated with β-adrenoceptor antagonists?
CHF (mild); Hyperthyroidism; Performance anxiety
What are the primary uses of β-adrenoceptor antagonists?
Myocardial ischemia; MI; Supra-ventricular arrhythmias; HTN (CNS, ↓ Renin, ↓ CO); Glaucoma
What are the effects of β2-antagonists?
Vasoconstriction in skeletal beds; Bronchoconstriction; Decreased glycogenolysis; Decreased recognition of hypoglycemia; Decreased Insulin release
What characterizes the synthesis pathway of Epinephrine?
Tyrosine --> Dopa --> DA --> NE --> Epi
What characterizes the mesolimbic pathway?
Connect ventral tegmental area and limbic regions; Control affect, emotion, cognition; Excess activity causes schizophrenia; Stimulation is associated with award and addiction; Insufficient activity is seen with ADHD and depression
What characterizes the nigrostriatal pathway?
Project from substantia nigra to neostriatum; Involved in control of posture/voluntary movement; Loss occurs in Parkinson’s disease
What are the isoforms of MAO?
MAO-A: found in liver and catecholamine brain regions;
MAO-B: found in 5-HT brain regions and in the periphery (more selective for DA)
What are the substrates of MAO and where is it found?
Many types of amines – found in liver, and in mitochondria of nerve terminals/post-synaptic cells
What are the substrates of COMT and where is it found?
DA, NE, Epi, Levodopa – found in postsynaptic neurons and glia, as well as in the liver
How is the activity of catecholamines terminated?
Reuptake, breakdown by COMT, breakdown by MAO
What is the difference between metabotropic and ionotropic receptors?
Metabotropic: GPCR, slow response; Ionotropic: Ion channel receptor, fast response
What characterizes Acetylcholine (ACh)?
Project from ventral forebrain to cerebral cortex and hippocampus; Role in cognitive function and memory: loss in Alzheimer’s, Huntington’s; Role in control of movement: loss of DA neurons that inhibit ACh release in striatum (Parkinson’s), excess activates M1 receptors causing tremor
What are the receptor subtypes for ACh in the CNS?
M1, M3: metabotropic, excitatory; M2, M4: metabotropic, inhibitory; Nicotinic: ionotropic (cation channel), excitatory
In what diseased states is impaired function of NE seen?
ADHD, depressive disorders, anxiety disorders
What characterizes GABA?
Found throughout the brain and brainstem, major inhibitory neurotransmitter in the CNS
What are the Histamine receptor subtypes in the CNS?
H1: on neurons/blood vessels/glia, excitatory, sedation from antihistamines due to H1 antagonism;
H2: function unknown
What characterizes Histamine?
Project from hypothalamus to many brain areas; Involved in arousal, body temperature, vascular dynamics – maintain awake state
What receptor subtypes are associated with 5-HT?
5-HT1A: on somatodendritic autoreceptors, decreases neuronal firing, altered by SSRIs/anxiolytics;
5-HT1D: pre-synaptic, decreases 5-HT release, altered by antimigraine drugs (↓ pain perception);
5-HT1-7: post-synaptic; 5-HT2A: excitatory post-synaptic, excess activity in schizophrenia;
5-HT3: ionotropic, cation channel, postsynaptic excitatory, chemoreceptor trigger zone (n/v)
What characterizes Serotonin (5-HT)?
Project from dorsal raphe nucleus to amygdala, hippocampus, cerebral cortex; Associated with mood, fear/anxiety, emotional memory; Excess activity seen with schizophrenia; Impaired function seen with depressive and anxiety disorders – other projections to the spinal cord are involved in inhibition of physiological sexual responses; 5-HT receptors on the vagus and chemoreceptor trigger zone stimulate nausea/vomiting
What are the receptor subtypes of NE in the CNS?
α1: metabotropic, excitatory postsynaptic receptors; α2: metabotropic, presynaptic autoreceptors (decrease Ca2+ influx), inhibitory postsynaptic receptors; β: metabotropic, postsynaptic excitatory
What characterizes the tuberoinfundibular pathway?
DA inhibits prolactin secretion
What distinguishes the D1/D1-like receptors from the D2/D2-like receptors?
D1/D1-like receptors: GPCR, stimulate activity of adenylate cyclase, found post-synaptically;
D2/D2-like receptors: GPCR, inhibits activity of adenylate cyclase, found post-synaptically, as well as pre-synaptically and as somatodendritic autoreceptors (i.e. on the neuron cell body)
What characterizes NE?
Cell bodies located in locus ceruleus, project to cerebral cortex/thalamus/hypothalamus/ cerebellum/spinal cord; Released in response to external stimuli, arousal/attention, learning, memory, mood
What characterizes Glycine?
Released by interneurons, mainly in the spinal cord and brainstem; Inhibitory, involved in pain transmission/respiration/motor coordination – there are multiple subtypes with multiple subunits; ionotropic subtype binding causes hyperpolarization; Strychnine (a convulsant) is an antagonist
What are the receptor subtypes of Endocannabinoids in the CNS?
CB1: metabotropic, inhibitory, marijuana effects – CB1 antagonists are used for obesity and smoking cessation
What characterizes the Endocannabinoids?
Anandamide and others; Found throughout the CNS and body; Regulate mood/appetite/pain sensation/smooth muscle tone in GI/immune function
What are the receptor subtypes of Opioid peptides in the CNS?
Mu: narcotic analgesia (most); causes respiratory depression; Kappa: narcotic analgesia (some), dysphoria, less respiratory depression; Delta; Epsilon – all are metabotropic, inhibitory
What characterizes the Opioid peptides?
Enkephalins, β-endorphin, dynorphin, endomorphins; Found throughout the CNS; Opioids decrease pain perception/respiration/consciousness level; Opioid cause euphoria
What are the receptor subtypes of Glutamate in the CNS?
Group I: excitatory; Group II/III: inhibitory; associated with schizophrenia, pain, post-trauma cytotoxicity – Ionotropic glutamate receptors: Excitatory (increase Ca conductance, prolonged stimulation causes excitotoxicity), receptors named for agonists (NMDA, AMPA, Kainate)
What characterizes Glutamate?
Found in interneurons and relay neurons throughout the CNS; It is the principle fast excitatory neurotransmitter in the CNS; Excitotoxicity with prolonged high concentrations causing neuron death
What are the GABA receptor subtypes in the CNS?
GABAA: postsynaptic, Cl- channel, hyperpolarization, drug binding modulates activity, subunit composition determines pharmacology, deficit in anxiety disorders
What innervation is responsible for the release of NO in erection?
PNS activation via ACh causing NO release from the endothelium;
NANC activation (PNS) causing direct release of NO
What other drugs are used to treat ED?
An injection of PGE2 into the corpus cavernosum
How does Yohimbine treat ED?
It blocks the prejunctional α2-adrenoceptors causing an increase NE release which will increase penile blood flow – however, Yohimbine may also increase BP and HR and it is less effective than the PDE inhibitors
How does Vardenafil differ from Sildenafil and Tadalafil?
Vardenafil is more specific for PDE5 than for PDE6; it also has the fastest onset
Where is PDE6 found and why is this important to drugs treating ED?
Found in the retina – most of the ED drugs are not specific for PDE5, but rather act on both forms; their action on PDE6 can cause abnormal color perception
What enzyme causes degradation of cGMP?
Phosphodiesterase 5 (PDE5); converts cGMP to GMP – this is the enzyme that ED drugs inhibit
What does the release of NO lead to?
NO reacts with guanylate cyclase and GTP to for cGMP; cGMP then causes muscle relaxation
What are the effects of post-synaptic D1 stimulation?
Moderate stimulation: suppresses noise; Excess stimulation: suppresses signals and noise;
Inadequate stimulation: causes excessive phasic DA release which suppresses signal and noise
What side effects are associated Amphetamines?
Nervousness/insomnia (very common, don’t give close to bedtime), anorexia (very common), abdominal pain, HA, emotional lability, dullness/fatigue, lack of spontaneity in social situations, dry mouth
What is the mechanism of action of the Amphetamines?
Reverse the direction of the nerve terminal reuptake transporter, causing NE/DA release;
Minor mechanism: inhibit NE/DA reuptake and MAO – overall effect: increased synaptic NE/DA
What characterizes the Amphetamines used in the treatment of ADHD?
Amphetamine mixed salts: amphetamine + dextroamphetamine; Dextroamphetamine: less commonly prescribed; Lisdexamfetamine: prodrug (lysine + dextroamphetamine), designed to decrease risk of overdose and abuse
What CNS alterations occur in ADHD regarding NE?
Polymorphisms in α2A result in a lower affinity for NE; this leads to a decreased enhancement of the signal – only true for a minority of ADHD cases
What CNS alterations occur in ADHD regarding DA?
Deficit in prefrontal cortex function due to a decrease in the tonic DA pool; this leads to less stimulation of autoreceptors, excess release of DA in response to stimuli, excess stimulation of post-synaptic D1 receptors (signal/noise suppressed), attention drifts/all stimuli demand attention
What are the effects of pre-synaptic α2A stimulation?
Moderate stimulation: enhances signal; Excess stimulation: impairs prefrontal cortex;
Inadequate stimulation: causes excessive phasic NE release which impairs prefrontal cortex
What are the symptoms of ADHD?
Inattention, hyperactivity, impulsivity – must be excessive, pervasive, long-term, and not due to something else
What happens when DA/NE bind to the axon terminal autoreceptors (D2/α2)?
They cause a decrease in the amount of DA/NE released by each AP – tonic levels of DA/NE present in synapses control the amount of impulse-associated DA/NE release (i.e. phasic levels)
What are the major neurotransmitters in the prefrontal cortex?
NE/DA: involved in regulation and inhibition of behavior, attention, impulse control – act on postsynaptic D1 and α2A receptors
What absolute contraindications are associated with Amphetamines?
Eating disorders, substance abuse, advanced cardiovascular disease, moderate/severe HTN, structural heart defects, hyperthyroidism, glaucoma, breastfeeding, abrupt discontinuation
What characterizes Atomoxetine?
Selective NE reuptake inhibitor, low affinity for DA/5-HT transporters; onset of action is 1week; Non-stimulant, used specifically for ADHD; no abuse potential, not a controlled substance –
less potent than other ADHD drugs
What absolute contraindications are associated with Methylphenidate?
Severe anxiety/psychosis, substance abuse, glaucoma, breastfeeding
What adverse effects are associated with Methylphenidate?
Mental effects: nervousness, insomnia, dullness, fatigue, emotional lability worsening of tics; growth inhibition, psychological dependence (less physical dependence than Amphetamines)
What is the mechanism of action of Methylphenidate?
Blocks DA reuptake in CNS – no effect on NE (thus less peripheral/cardiovascular effects)
What characterizes Methylphenidate?
Adverse effects primarily seen are more mental and less peripheral/cardiovascular; Methylphenidate has a shorter duration of action than mixed Amphetamines
What drug interactions are associated with Amphetamines?
Sympathomimetics and drugs that increase synaptic NE levels (e.g. MAOIs) cause additive CNS and cardiovascular stimulation; Lithium and antipsychotics antagonize Amphetamine’s actions
What are less common side effects of Amphetamines?
Worsening of tic disorders, increased BP, growth inhibition (rebound if drug is discontinued in adolescence), sudden unexplained death (caution in those with structural heart defects, patients should be screened before starting stimulants), dependence (physiological/psychological), abrupt discontinuation causes withdrawal symptoms
What are the effects of toxic doses of Amphetamines?
Cardiac arrhythmias, heart block, circulatory collapse, rhabdomyolysis, seizures, coma, death – toxic effects in children occur over a wider dose range
What are signs of a supra-therapeutic dose of Amphetamines?
Supra-normalization, anxiety, psychosis, blurred vision, dilated pupils, sweating, flushing, pallor, hyperthermia, labile vital signs, purposeless movement, tremor
What are the absolute contraindications of Atomoxetine?
Glaucoma, jaundice
What are some general principles of ADHD therapy?
Therapy is highly individualized; Continually monitor effectiveness of drug therapy (feedback from home/school); Periodically discontinue drugs to check for remission; Efficacy of drugs is increased by psychological therapies
What characterizes the use of Modafinil in ADHD?
Stimulant for narcolepsy – significant adverse effects
What characterizes the use of Clonidine in ADHD?
Agonist of α2 autoreceptors – normally used as antihypertensive; has significant adverse effects, rarely used
What characterizes the use of TCAs in ADHD?
Onset of action is weeks; Beneficial for anxiety/depression; Doesn’t worsen tics; Unexplained cardiac deaths in children; Fatal in overdose
What are the second-line/off-label drugs used in ADHD?
TCAs and other antidepressants (Bupropion/Buspirone), Clonidine, Modafinil
What drug interactions are associated with Atomoxetine?
MAOIs: cardiovascular stimulation; CYP interactions
What adverse effects are associated with Atomoxetine?
In children: GI upset, increased suicidal thoughts/behaviors; In adults: insomnia, anticholinergic effects, GI upset, sexual effects
What is the purpose of the juxtaglomerular apparatus?
By sensing Na+ levels and blood pressure, the juxtaglomerular apparatus can adjust flow through the release of renin and vasoconstriction of the afferent arteriole
What are the carbonic anhydrase inhibitors?
Acetazolamide, Dichlorphenamide, Methazolamide; they work in the proximal tubule by inhibiting carbonic anhydrase
What toxicities/adverse effects are associated with osmotic diuretics?
Heart failure, pulmonary congestion; Urea: impaired liver function; Mannitol/urea: cranial bleeding (can cause reoccurrence of hemorrhage due to increased extracellular volume)
What are the uses of osmotic diuretics?
Treatment of acute renal failure (can convert oliguric renal failure into polyuric renal failure); Prophylaxis of acute renal failure (e.g. with radiocontrast or crush syndrome); Cerebral/ocular edema (due to surgery or head trauma)
What are the effects of osmotic diuretics?
Pull fluid into the extracellular space (at the expense of intracellular fluid); Increase excretion of electrolytes; Increase renal blood flow – they increase Na+ excretion to about 5%
What is the mechanism of action of the osmotic diuretics?
They have an osmotic effect in the proximal tubule (i.e. they keep fluid in the tubule), which leads to a washout of NaCl and urea in the loop of Henle
What are the osmotic diuretics?
Glycerin: can be given orally, but gets metabolized; Mannitol: most common, given IV, not metabolized); Urea: given IV
What is the function of diuretics?
Increase urine flow rate and Na+ excretion
What happens in distal portions of the nephron with increased Na+ levels?
It can be involved in Na+ ↔ K+ and Na+ ↔ H+ exchange
Where in the nephron is Na+ reabsorbed?
65% in the proximal tubule (some by Na+ ↔ H+), and 25% in the ascending limb of the loop of Henle (thick portion especially)
What toxicities are associated with carbonic anhydrase inhibitors?
Metabolic acidosis; Stone (due to increased phosphate excretion); Potassium wasting; Contraindicated in hepatic cirrhosis (due to decreased NH4+ excretion); Hypersensitivity reactions (since they are sulfonamide drugs) – they are self-limiting since they will no longer work when HCO3- is depleted
What toxicities/adverse effects are associated with loop diuretics?
Depletion of Na+, loss of ECF volume, circulatory collapse; Hypokalemia, hypomagnesemia, hypocalcemia (can be used in patients with hypercalcemia when given in conjunction with saline); Hyperuricemia (since they compete with the organic anion transporter); Ototoxicity (Ethacrynic acid and Furosemide to a lesser degree); Increase LDL/triglycerides, decrease HDL, hyperglycemia (precipitates diabetes) – latter effects are less severe than with thiazide diuretics
What pharmacokinetics are associated with loop diuretics?
Highly protein bound; Tubular secretion (in PCT); Short half-life
What are the uses of loop diuretics?
Acute pulmonary edema; CHF (venous capacitance); Acute renal failure (not effective if renal failure is due to damage to the proximal convoluted tubule); HTN
What is the effect of loop diuretics?
Increased excretion of Na+, K+, H+, Ca2+, Mg2+, Cl-;Furosemide will also increase excretion of HCO3- and H2PO4- (due to its carbonic anhydrase inhibitor activity) and increase RBF;
~25% FENa (which is why they are called high-ceiling diuretics)
What is the mechanism of action of the loop diuretics?
They inhibit the Na+-K+-2Cl- symporter in the thick ascending limb; they require transport by an organic anion transporter in the proximal convoluted tubule to get access to the tubular lumen
What are the loop/high-ceiling diuretics?
Furosemide (Lasix), Bumetanide, Torsemide, Ethacrynic acid
What does inhibition of carbonic anhydrase result in?
In the lumen: inhibits conversion of H2CO3 to CO2 + H2O, which prevents their reabsorption into the proximal tubule cell; In the proximal tubule cell: inhibits conversion of CO2 + H2O to H2CO (and subsequently to HCO3- + H+), which prevents HCO3- from being symported with Na+ into the blood and prevents Na+ in the lumen from being antiported into the proximal tubule cell by H+
What are the uses for carbonic anhydrase inhibitors?
Glaucoma; Acute mountain sickness (prophylaxis/treatment) by decreasing CSF formation and CSF pH (causes increased ventilation); Treatment of alkalosis from increased H+ excretion (due to other diuretics); Cause urinary alkalinization
What is the effect of carbonic anhydrase inhibitors?
Increased excretion of HCO3-, Na+, K+, H2PO4-; ~5% FENa
What are the uses of thiazide diuretics?
Edema, HTN
What toxicities/adverse effects are associated with ENaC inhibitors?
Hyperkalemia (especially with ACE inhibitors, NSAIDs, β-blockers), GI disturbances; Triamterene: impaired glucose tolerance, musculoskeletal effects; Amiloride: headaches
What are the uses of ENaC inhibitors?
In combination with a loop/thiazide diuretic to maintain plasma K+; Treatment of HTN due to ENaC polymorphism (which makes it constitutively active); Amiloride can be used as an aerosol for the treatment of cystic fibrosis (makes mucus thinner)
What are the effects of ENaC inhibitors?
Increased excretion of Na+, Cl-; Decreased excretion of K+, H+, Ca2+, Mg2+; ~2% FENa
What are the ENaC inhibitors and what is their mechanism of action?
Triamterene, Amiloride – MOA: inhibit epithelial Na+ channel in the late distal tubule and early collecting duct (prevents activation of K+ secretion)
What are the two types of potassium-sparing diuretics?
ENaC inhibitors; Mineralocorticoid antagonists
What toxicities/adverse effects are associated with thiazide diuretics?
Hyperuricemia (with chronic use --> gout); Hypokalemic metabolic alkalosis; Hyponatremia; Increased LDL/total cholesterol/triglycerides; Decreased insulin secretion (impaired glucose tolerance); Erectile dysfunction; Hypersensitivity reactions (rare)
What are the thiazides and thiazide-like diuretics?
Chlorothiazide/Hydrochlorothiazide (HCTZ), Chlorthalidone, Indapamide, Metolazone
What are the effects of thiazide diuretics?
Increased excretion of Na+, K+, H+, HCO3-, Cl-, H2PO4-; There is a decrease in Ca2+ excretion with long-term effects (can be used in women with osteoporosis); ~5% FENa; Has no effect on the juxtaglomerular apparatus (and thus no effect on renal blood flow)
What is the mechanism of action of the thiazides?
Inhibit Na+-Cl- symporter in the distal convoluted tubule; they require transport by an organic anion transporter in the proximal convoluted tubule to get access to the tubular lumen
What are the mineralocorticoid antagonists and how do they differ?
Spironolactone: not very selective; Eplerenone: selective aldosterone receptor antagonist
What are the toxicities associated with the mineralocorticoid antagonists?
Hyperkalemia; Spironolactone: gynecomastia, impotence, decreased libido, hirsutism, diarrhea, gastritis, gastric bleeding, peptic ulcers (contraindication); Eplerenone: GI effects, interaction with CYP3A4 inhibitors (since it is metabolized by 3A4)
What are the uses of mineralocorticoid antagonists?
In combination with a loop/thiazide diuretic to maintain plasma K+; Hyperaldosteronism (1°/2°); Edema from hepatic cirrhosis
What are the effects of the mineralocorticoid antagonists?
Increased excretion of Na+, Cl-; Decreased excretion of K+, H+, Ca2+, Mg2+; ~2% FENa (overall effect depends on how much aldosterone is normally active in the nephron)
What is the mechanism of action of the mineralocorticoid antagonists?
Antagonists for aldosterone receptor in the late distal tubule and early collecting duct
What enzyme converts angiotensin I to angiotensin II and where is it mainly found?
Angiotensin converting enzyme (ACE): present on lumenal surface of vascular endothelial cells, particularly in the lungs
How does sympathetic stimulation affect renin release?
It stimulates renin release by activating β1 receptors
By what mechanism does NaCl reabsorption across the macula densa affect renin release?
Decreased reabsorption of Na+ will cause the release of renin using prostaglandins as a messenger; Increased reabsorption of Na+ will inhibit renin release using adenosine as a messenger
What can be a complication of atherosclerosis of the renal artery?
It can decrease blood flow to the glomeruli, thus activating the renin-angiotensin system which will work to elevate a normal blood pressure
Where is renin released and what is the baroreceptor stimulus responsible for its release?
By the juxtaglomerular cells in response to the baroreceptor function of the afferent arteriole: decreased stretch leads to increased renin release, and vice versa
When the conversion of angiotensin I to angiotensin II is prevented by an ACE inhibitor, what compounds are generated instead?
Angiotensin (1-7) and angiotensin IV (they have opposite effects to that of angiotensin II)
What is the half-life of angiotensin II?
Very short (~1 min.) – it is degraded by aminopeptidase and angiotensinase to peptide fragments
What characterizes the synthesis of angiotensinogen?
It is constitutively synthesized
by liver, leading to its abundance in plasma
What substances can increase angiotensinogen levels?
Corticosteroids, estrogen, pregnancy, estrogen-containing contraceptive, angiotensin II
What is the rate-limiting step in the formation of angiotensin II?
The conversion of angiotensinogen to angiotensin I by renin (released from the kidney)
What are the rapid responses of angiotensin II that involves increases in blood pressure?
Direct vasoconstriction of peripheral vasculature (very potent); Enhanced peripheral noradrenergic transmission; Increased CNS sympathetic tone (increases baroreceptor set-point); Increased release of catecholamines from the adrenal medulla
What are the effects associated with kinins?
Vasodilation (leads to increased blood flow and reduced effects of the renin-angiotensin system); Increased sodium excretion, diuresis; Powerful algesic; Mimic inflammation, produce edema, wheal and flare; Play a role in inflammatory disease (IBD, arthritis, asthma) – they have a very short half-life and thus very localized effects
What characterizes the kinin receptors?
They are GPCRs; B1: limited distribution, upregulated in inflammation; B2: activated by bradykinin and kallidin, mediates the majority of effects in the absence of inflammation, stimulation leads to production of NO and prostacyclin
What factors increase the activation of kallikreins and thus the production of kinins?
Factors that oppose renin release: increased ECF volume, BP, Na+ retention, mineralocorticoids, and angiotensin II
What are the precursors to kallidin and bradykinin and how are they degraded?
Kallidin is produced from LMW kininogen by tissue kallikrein, whereas bradykinin is produced from HMW kininogen by plasma kallikrein; both are degraded by Kininases I&II, which are ACEs
What effects of angiotensin II are due to its mitogenic actions?
Hypertrophy of vascular and cardiac cells, thickening of walls, cardiac fibrosis, increased preload/afterload
What are the slow responses of angiotensin II that involves increases in blood pressure?
Increases Na+ reabsorption in proximal tubule (direct effect); Increased release of aldosterone; Altered renal hemodynamics: direct renal vasoconstriction (especially on the efferent arteriole, thus maintaining glomerular pressure), enhanced noradrenergic neurotransmission in kidney, increased renal sympathetic tone
What is meant by the short loop feedback of the renin-angiotensin system?
Angiotensin II produced in response to renin release, will work to lower the secretion of renin
What characterizes the angiotensin receptors?
They are GPCRs; AT1: mediates most actions of angiotensin II, high affinity for the antagonist Losartan, Gq-receptor (PLC/IP3/DAG), stimulates gene expression/cell growth/mitogenic effect;
AT2: role poorly defined, opposite effects to AT1, highly expressed in fetus
Drugs with what action will cause the release of renin?
Drugs that decrease blood volume/blood pressure, and drugs that cause β1 stimulation
What are some common ACE inhibitors?
Captopril: active form with an active metabolite; Alopril: ester pro-drug which is converted to Enalaprilat, the active metabolite; Lisinopril: active form is excreted
What side effects/adverse reactions are associated with drugs that inhibit the renin-angiotensin system?
Severe hypotension in patients who are hypovolemic, salt-depleted, or have CHF;
Hyperkalemia in patients with impaired renal function; Acute renal failure in patients with bilateral renal artery stenosis; Cough, angioedema (due to increased kinins with ACE inhibitors); Fetopathic after first trimester of pregnancy
What therapeutic applications are associated with the drugs that inhibit the renin-angiotensin system?
HTN: do not trigger reflex tachycardia, can be effective even with low renin;
CHF: improved hemodynamics/left ventricular systolic function, slows hypertrophy/remodeling;
MI: prevent excessive remodeling of cardiac tissue, begin immediately after an infarction;
Slowing of diabetic nephropathy: ↓ arterial BP; ↓ afferent arteriolar resistance; ↓ intraglomerular pressure
What effects do ACE inhibitors, AT1 receptor antagonist, and Aliskiren have in common?
They decrease PVR/BP, increase renal blood flow, and decrease secretion of aldosterone
How do the effects of Aliskiren differ from those of an ACE inhibitor?
It does not increase angiotensin (1-7), does not increase renin activity, and does not increase kinins
How do the effects of an AT1 receptor antagonist differ from those of an ACE inhibitor?
They do not increase angiotensin (1-7), do not increase kinins, and do not decrease stimulation of AT2 receptors
What are some common competitive AT1 receptor antagonists?
Losartan, Irbesartan, Valsartan
What happens to bradykinin levels when a patient is given an ACE inhibitor?
It will be increased, thus causing more vasodilation and a lowering of blood pressure; however, it will also act as an irritant which may cause cough
By what four ways can the activity of angiotensin II (AT-II) be inhibited?
Inhibit the release of renin (β-blockers); Inhibit the activity of renin (Aliskiren);
Inhibit the formation of AT-II by ACE inhibitors; Block the AT1 receptors for AT-II
Which calcium channel type is affected by the calcium-entry antagonists?
Slow channels (L-type)
What are the effects of the dihydropyridines?
They cause prominent vasodilation (peripheral/coronary artery) which triggers sympathetic reflexes, which in turn cause increases in heart rate and contractility; they have no effect on AV conduction (β-blockers can prevent the cardiac effects without reducing vasodilatory effects)
How does depolarization differ between vascular smooth muscle and cardiac muscle, and what does this mean for the action of dihydropyridines?
Vascular smooth muscle is found to be in a chronic state of depolarization, which means that its calcium receptors will spend a much greater amount of time in the inactivated-closed state than cardiac muscle will; this means that dihydropyridines will have a much greater effect on vascular smooth muscle than on cardiac muscle
Which antagonists bind to calcium channels in the inactivated-closed state?
Dihydropyridines
What happens to the fraction of calcium channels that are blocked by Verapamil/Diltiazem when heart rate increases?
It will go up, since the calcium channels will be in their open state more frequently (i.e. use-dependent block: the higher the heart rate, the more effective the drug becomes at lowering it)
Which antagonists bind to calcium channels in the open state?
Verapamil, Diltiazem
What three groups can the calcium-entry antagonists be divided up in?
1-4 dihydropyridines (Nifedipine, -dipine); phenylalkylamines (Verapamil, D-600);
benzothiazepines (Diltiazem)
In what three states can the calcium channel be found?
Open; Resting-closed; Inactivated-closed
What happens when calcium channels are depolarized?
They fluctuate between open and resting-closed (thus allowing for the influx of calcium ions), until they assume the inactivated-closed state; at this point, they have to be hyperpolarized again before being able to go to the open state once more
What calcium channel states are favored by hyperpolarization vs. depolarization?
Hyperpolarization: resting-closed; Depolarization: open/inactivated-closed
For what type of angina is Bepridil used and what side effect is associated with it?
Angina that is refractory to other calcium antagonists; however, it can broaden the cardiac action potential (long QT) and be pro-arrhythmic
What should be kept in mind when giving an elderly patient Verapamil/Diltiazem?
Whether their AV conduction is already compromised or if β-blockers are in use (contraindication)
What must be done with patients who recently had a heart attack and are now taking Nifedipine?
They must be given a β-blocker as well to avoid the positive chronotropic/inotropic effects on the heart
What are the main adverse effects seen with dihydropyridines and Verapamil, respectively?
DHP: hypotension, HA, peripheral edema, tachycardia, gingival hyperplasia
Verapamil: hypotension, AV block, bradycardia, constipation, gingival hyperplasia
Which of the calcium channel blockers is used in the treatment of Raynaud’s phenomenon?
Dihydropyridines
What common indication is seen with all three classes of calcium channel blockers?
HTN: Nifedipine would be most useful for treating HTN alone, whereas Verapamil/Diltiazem are most useful in a patient with HTN and angina/arrhythmia)
What type of arrhythmias are treated by Verapamil/Diltiazem?
Supraventricular tachycardias (AV node reentry, atrial flutter/fibrillation) – the dihydropyridines have no use in treating arrhythmias
What are the effects of Verapamil/Diltiazem?
They increase vasodilation (peripheral/coronary artery), but their most prominent effects are in reducing heart contractility, rate, and AV conduction (Verapamil > Diltiazem)
What calcium-entry antagonists are recommended for the treatment of variant/unstable angina?
All three classes of calcium channel antagonists are effective
What calcium-entry antagonists are recommended for the treatment of exertional angina?
Verapamil/Diltiazem; can also give Nifedipine and a β-blocker as co-treatment
What are some common indications for Heparin?
Pre- and post-surgical management of DVT and pulmonary thrombosis as well as arterial and heart valve emboli; Venous stasis; Unstable angina and post-MI; DIC – Heparin does not break down clots, it just inhibits their formation
How does the mechanism of action of LMWH differ from that of Heparin?
LMWH has the pentasaccharide sequence that binds to ATIII, but not the 18 saccharide sequence found in Heparin that binds to thrombin --> this means that LMWH affects Xa without affecting thrombin – Fondaparinux only has the pentasaccharide sequence needed to bind ATIII
How are Low Molecular Weight Heparins (LMWH) recognized?
They end on –parin: Enoxaparin, Fondaparinux
How does low-dose Heparin differ in effects from high-dose Heparin?
Low-dose: prevents thrombin formation only; High-dose: prevents thrombin formation and inhibits the actions of already formed thrombin
What contraindications are associated with Heparin and most other anticoagulant drugs?
GI ulcerative lesions/occult bleeding, severe hypertension, recent eye/brain/spinal surgery, visceral carcinoma, threatened abortion; Anti-platelet drugs (caution required); Presence of thrombocytopenia (may result in serious bleeding)
What is the Heparin “antidote” and how does it function?
Protamine: basic (+) low molecular weight proteins that bind to negatively charged Heparin to neutralize its anticoagulant effects – does not affect Fondaparinux (LMWH)
What toxicities are associated with Heparin?
Primary: hemorrhage, thrombocytopenia, HIT; Secondary: hypersensitivity, fever, anaphylaxis
What is the mechanism of action of unfractionated Heparin?
It binds to Antithrombin III (ATIII) and accelerates the inactivation of factors IIa, IXa, Xa, and XIIa by ATIII
What is the difference in IV vs. SC administration of Heparin?
IV: immediate anticoagulation effect; SC: anticoagulation is delayed due to absorption
What pharmacokinetics are associated with Heparin?
Not bound to plasma proteins – Metabolism: initial, rapid zero-order mechanism (cellular uptake and depolymerization) followed by slower first-order renal clearance
Which factor is affected the earliest by Warfarin?
Factor VII, since it has the shortest half-life; With chronic doses, however, all of the Vitamin K dependent factors will be affected
What are indications for antiplatelet drugs?
Prosthetic heart valves; MI (prophylaxis and prevention of re-thrombosis); Combination therapy with anticoagulant and fibrinolytic drugs; Percutaneous transluminal angioplasty (PCTA or PCI) and stenting
What class of anticoagulants is used in a patient with thrombocytopenia?
Direct thrombin inhibitors: all end on –rudin (Lepirudin, Bivalirudin, Desirudin)
What drug is an oral alternative to Warfarin?
Rivaroxaban: small molecule, active site inhibitor of factor Xa
How can Warfarin toxicity be countered?
No “antidote” available; Can administer Vitamin K1 but takes hours to have an effect; In emergencies, FFP or concentrates containing functional factors can be administered
What are the toxicities associated with Warfarin?
Same as Heparin, but also contraindication in pregnancy
What pharmacokinetics are associated with Warfarin?
>95% bound to plasma proteins; Crosses the placenta; CYP450 metabolism
How do the pharmacokinetics of LMWH differ from that of Heparin?
Greater bioavailability; Longer lasting; 1st order pharmacokinetics only
What is a key clinical difference between Warfarin and Heparin?
Warfarin can be given PO, wheras Heparin is only given IV/SC
What is the mechanism of action of Warfarin?
Warfarin is a Vitamin K antagonist that inhibits an epoxide reductase reaction required to recycle Vitamin K --> this lack of recycling means that Vitamin K is unable to perform γ-carboxylation of glutamic acid residues on factors II, VI, IX, X, as well as Proteins C and S --> this lack of γ-carboxylation means that the factors are unable to complex with Ca2+ during coagulation
What are the pharmacokinetics of Ticlopidine and Clopidogrel?
Bind plasma proteins, metabolized by the liver; Onset of Ticlopidine is slow (days) whereas that of Clopidogrel is much faster
What is the indication for Urokinase?
Pulmonary embolism – Urokinase is not antigenic
What characterizes Streptokinase?
Not an enzyme; Forms a complex with plasminogen, exposing an active site that converts additional plasminogen into plasmin; Obtained from β-hemolytic streptococci, making it antigenic; Antigenic effects can be minimized by giving the drug as an intracoronary injection
What are the indications of plasminogen activators?
Ischemic stroke, arterial thrombosis, MI (early therapy is essential, must establish flow first), DVT, pulmonary embolism
How do first, second, and third generation plasminogen activators differ?
First: Streptokinase, Urokinase --> not fibrin-specific; Second: Alteplase (t-PA) --> causes less fibrinogenolysis; Third: Reteplase, Tenecteplase --> improved fibrin specificity and kinetics
What is the difference between fibrinolysis and fibrinogenolysis?
Fibrinolysis: breakdown of a fibrin clot by plasmin; Fibrinogenolysis: breakdown of clotting factors in the plasma by plasmin (prevented by α2-antiplasmin)
What drugs function as GPIIb/IIIa inhibitors and how do they differ?
Abciximab: monoclonal antibody that noncompetitively inhibits GPIIb/IIIa (indication: PCI);
Eptifibatide, Tirofiban: competitive inhibitors of GPIIb/IIIa (contain “fib” and “ban” portions)
How does a baby Aspirin/day affect coagulation?
It prevents TXA2 formation in platelets more than it prevents PGI2 formation in endothelial cells (since endothelial cells can synthesize COX de novo and platelets can’t), thus tipping the balance towards anticoagulation
What are examples of ADP receptor inhibitors and how do they function?
“clop” drugs: Ticlopidine, Clopidogrel (Plavix); They irreversibly inhibit platelet ADP receptors and thus inhibit ADP-induced exposure of platelet membrane GPIIb/IIIa and fibrinogen binding to activated platelets
What are the indications for Ticlopidine and Clopidogrel and what are associated toxicities?
Indications: prevention/treatment of thrombotic stroke, PCTA/PCI; Toxicities (more so with Ticlopidine): GI disturbances, potential life-threatening neutropenia, blood dyscrasias
What characterizes Alteplase?
Single chain peptide that is converted to a two-chain t-PA dimer upon exposure to fibrin; 30 min half-life (may require multiple administrations)
What is the mechanism of action of Antifibrinolytic drugs (e.g. Aminocaproic acid, Tranexamic acid) and what are they used for?
They block the lysine-binding interactions between plasminogen/plasmin and fibrin, thus inhibiting plasminogen activation and plasmin fibrinolysis – used for dental procedures in Hemophiliacs and to control bleeding during surgery
How are Reteplase and Tenecteplase improvements over Alteplase?
They contain only the domain necessary for binding to fibrin and the protease domain of t-PA; this allows for a more rapid onset and potent effect than is seen with Alteplase; However, they still have some fibrongenolytic properties
How does Anistreplase function?
It is a noncovalent complex of streptokinase and plasminogen which is catalytically inert but is deacetylated to give rise to active plasminogen; has a prolonged fibrinolytic effect, but is still antigenic
What is the drug of choice for HTN in African-Americans?
Thiazides (CCBs also work very well)
What name is associated with the α-blockers and why are they α1-specific?
-osin (e.g. Prazosin); they are α1-specific so that they don’t cause the increase in NE release associated with α2-blockers; since α-blockers cause vasodilation, they will have a reflex tachycardia and cause activation of the RAS – this is why they are usually prescribed along with a β-blocker and diuretics
What are the key differences between pure β-blockers (BB) and those with ISA?
Pure BB decrease CO by decreasing both HR and SV, and they inhibit RAS; pure BB cause a reflex increase in TPR (can lead to cold hands and extremities);
BB with ISA do not decrease CO (since inotropic activity is balanced by the partial agonist activity), but they do decrease TPR (due to β2 ISA); BB with ISA do not inhibit RAS;
Which β-blockers have intrinsic sympathomimetic activity (ISA)?
Pindolol, Penbutalol, Carteolol, Acebutolol – they have weak partial agonist activity
What does activation of the SNS stimulate regarding control of BP?
β1 activation in the heart; β1 activation in the kidney (activates RAS); α1 activation in the vasculature
What two major systems are involved in the regulation of BP?
SNS (short-term) and RAS (long-term)
Which classes of drugs used in the treatment of HTN are K+ sparing?
Aldosterone antagonists, ACE inhibitors/ARB, Triamterene/Amiloride
What is the primary class of diuretics used in HTN?
Thiazides (whereas loop diuretics are the primary class used in CHF)
What is the long-term effect of thiazide diuretics?
Plasma volume returns to normal, but TPR remains decreased
Do diuretics cause a baroreceptor reflex?
No, since their effects take days-weeks to take place (same goes for RAS inhibitors – they reset the baroreceptor reflex)
What does Guanethidine lead to?
Depletion of NE – mechanism not well understood
What drugs are used for hypertensive emergencies?
Sodium nitroprusside, Labetalol (IV bolus), Fenoldopam, Diazoxide, Nicardipine, Hydralazine
What is the mechanism of action of Minoxidil?
It is a K-channel opener, meaning it will cause hyperpolarization of the cell; therefore it will make cells much less likely to contract --> causes dilation of arteries
What should hydralazine always be combined with?
BB (to prevent reflex tachycardia) and a diuretic (to reverse edema caused by RAS activation) – the same is true for Minoxidil, although the effects are more severe (contraindicated in CHF)
What are the two most important side effects of an ACE inhibitor?
Cough, angioedema (these side effects are not as prominent with ARBs)
Why is the combination of a thiazide diuretic with an ACE inhibitor so effective?
The thiazide makes the HTN more renin dependent, which increases the efficacy of the ACE inhibitor; also, ACE inhibition preserves K, which offsets the K loss causes by the thiazide
What contraindication is given for all drugs that inhibit the RAS?
They should not be used in pregnancy (since AT-II is used in fetal development)
Which drugs inhibit both α- and β-receptors?
Carvedilol, Labetalol (they are not first-line therapy, due to orthostatic hypotension/bradycardia)
Which drugs are centrally-acting inhibitors of the SNS that used to be popular for treating HTN, and how do they exert their effects?
α-methyldopa (metabolized to α-methylNE in the brain) and Clonidine (direct-acting agonist) – they are α2-selective agonists that only work in the brain; in this fashion, they lower NE outflow to the heart, vasculature, and kidney (side effects, like sedation/drowsiness, has made them mostly obsolete)
What is the mechanism of action of Reserpine?
It blocks NE uptake into the storage vesicle, causing it to be degraded by MAO – knocks out the sympathetic nervous system
What type of drug is Fenoldopam?
Selective DA1 agonist; it dilates arterioles in renal, mesenteric, and cerebral vascular beds;
it increases RBF while lowering BP (useful for patient with renal insufficiency in hypertensive emergencies)
What is the mechanism of action of Diazoxide?
K-channel opener (like Minoxidil, but IV), so it dilates arterioles – reflex tachycardia is one of the major adverse effects (caution in ischemic heart disease)
Considering the fact that CHF is a neurohormonal disorder, what classes of drugs would be beneficial for its treatment?
ACE inhibitor/ARB (for AT-II), and a β-blocker (for NE) – also effective would be inotropic agents (to increase CO), arterial vasodilators (decrease afterload), venodilators (decrease preload), and diuretics (eliminate edema, decreases preload)
What makes cardiac glycosides especially dangerous?
They have a low therapeutic index; Side effects include AV block, arrhythmias, anorexia, nausea, vomiting, fatigue, visual disturbances
What are the CNS effects of cardiac glycosides?
They increase vagal tone which slows HR, and at high levels can cause an AV block (can be desired in atrial fibrillation)
How do cardiac glycosides cause ectopic contractions?
Excessive increases in intracellular calcium lead to Ca-induced-Ca release from SR, which lead to calcium-dependent after-depolarizations (= ectopic contractions)
Why are the cardiac glycosides pro-arrhythmic?
Because they inhibit the Na/K-ATPase, which causes extracellular potassium to increase;
this causes a partial membrane depolarization, which has a pro-arrhythmic effect
What is the mechanism of action of the cardiac glycosides?
They inhibit the Na/K-ATPase --> increases intracellular sodium --> decreases the driving force for the Na/Ca exchanger --> increases intracellular calcium --> increases inotropy
What are the cardiac glycosides (inotropes) used in chronic CHF?
Digitalis glycosides: Digoxin, Digitoxin, Ouabain
How are inotropy, afterload (TPR), and preload (LVEDP) affected in CHF?
Inotropy is decreased, whereas afterload and preload are increased
What is meant by the statement that CHF is a neurohormonal disorder?
A failing heart activates the renin-angiotensin system (AT-II/Aldosterone) and causes sympathetic nervous system activation: β1 (heart/renin), β2 (vasodilation), α1 (vasoconstriction)
What is meant by the vicious cycle in CHF?
Outer loop: left ventricular failure (e.g. due to ischemic heart disease) --> decrease in CO --> reflex increase in sympathetic nervous system activity --> vasoconstriction --> increase in BP --> increase in afterload --> worsens left ventricular failure;
Inner loop: ventricular failure --> decrease in CO --> activation of renin-angiotensin system --> fluid retention --> increase in preload --> worsens ventricular failure
What do the cardiac glycosides and diuretics have in common, concerning the treatment of CHF?
They do not reduce mortality, but they do improve the QOL for patients (reduce hospital visits)
What are the adverse effects associated with Isosorbide Dinitrate?
HA, dizziness, flushing, orthostatic hypotension, drug rash
How does Isosorbide Dinitrate reduce the load on the heart?
It causes venodilation which reduces preload, thus decreasing LVEDP – it also has less activation of the SNS than Hydralazine
What are the adverse effects associated with Hydralazine?
HA, flushing, nasal congestion, reflex tachycardia/palpitations (less common in CHF), fluid retention, myocardial ischemia/infarction (due to increased cardiac work); also Lupus syndrome at higher doses
How does Hydralazine increase CO?
It causes arterial vasodilation which decreases afterload, thus decreasing the impedance to LV ejection
How do they aldosterone antagonists differ in their effects on CHF when compared to other diuretics?
They do reduce mortality because they cause potassium retention (thus preventing hypokalemia!)
Which diuretics are used in CHF?
Loop diuretics (since they are the only ones powerful enough to decrease edema and pulmonary congestion
What is the danger in giving a patient cardiac glycosides in combination with diuretics?
Diuretics can cause hypokalemia which will worsen the pro-arrhythmic toxicity associated with cardiac glycosides (!)
What are the notable differences between Digitoxin, Digoxin, and Ouabain?
Digitoxin: longest half-life; Digoxin: intermediate half-life, most common;
Ouabain: shortest half-life, given IV
What is the treatment for digitalis toxicity?
Stop digitalis, control arrhythmias, give potassium, use anti-glycoside antibodies
What are the ONLY β-blockers effective in the treatment of CHF?
Carvedilol, Metoprolol (long-acting), Bisoprolol – there is not a class effect!
Which of the sympathomimetic amines cause the greatest increase in RBF?
DA (since it acts on DA1 receptors); Dobutamine will cause a slight increase (due to increase CO), and Isoproterenol can cause either increase or decrease depending on which receptor activity dominates (β2 vs. β1)
How do the effects of DA, Dobutamine, and Isoproterenol differ in regards to BP and preload?
DA: increase in BP/preload; Dobutamine: no change in BP/increase in preload;
Isoproterenol: decrease in BP/preload
Which receptors are affected by DA, Dobutamine, and Isoproterenol, besides their common effects on β1?
DA: α1, DA1; Dobutamine: β2, α1; Isoproterenol: β2
What are the goals of therapy with cAMP elevators?
Improve LV function, increase CO, improve circulatory status and tissue perfusion, decrease pulmonary congestion
Which drugs are used in acute CHF?
cAMP elevators (inotropes): sympathomimetic amines (DA, Dobutamine, Isoproterenol) and phosphodiesterase III inhibitors (Milrinone, Amrinone); Short-acting vasodilators: Sodium nitroprusside, Nesiritide
What adverse effects are associated with the use of β-blockers in CHF?
May worsen CHF, especially acutely (must be carefully titrated on initiation of therapy); Bradycardia; Hypotension; Worsen asthma/COPD; Increase glucose (diabetogenic);
Increase VLDL/decrease HDL; Fatigue; Insomnia
In which patients is a combination treatment of Hydralazine and Isosorbide Dinitrate indicated?
African-Americans (due to reduced effectiveness of inhibitors of RAS and β-blockers)
When a patient taking an ACE inhibitor complains of excessive coughing (due to excess bradykinin), what drug can be substituted?
AT1 receptor antagonist
What drugs are part of the standard therapy for CHF?
Digoxin, diuretic, ACE inhibitor, β-blocker (certain kinds)
Which sympathomimetic amine would be given to a patient with acute CHF who has low BP and low RBF?
DA: increases BP/RBF – if the patient’s BP is normal but preload too high, Dobutamine would be the best choice; if the patient has high BP, Isoproterenol would be the drug of choice
What are the adverse effects associated with Nesiritide?
Hypotension, insomnia, dizziness, vomiting
What is the mechanism of action of Nesiritide?
As a recombinant form of BNP it increase cGMP and produces vasodilation (both arterial and venous); it has no direct inotropic effect but increase CO due to reduced LV impedance; may also produce a mild diuresis
What are the adverse effects associated with Sodium nitroprusside?
HA; n/v; Thiocyanate toxicity on longer infusions (!) which can lead to metabolic acidosis, confusion, coma, and death
What is the mechanism of action of Sodium nitroprusside?
It is metabolized to NO which is a potent vasodilator (both arterial and venous); vasodilation decreases LV impedance, and thus CO is increased – can also cause some reflex tachycardia, renin release and fluid retention (not severe)
What are the adverse effects associated with PDE III inhibitors?
Arrhythmias (also GI/hepatic effects and thrombocytopenia)
What is the mechanism of action of phosphodiesterase III inhibitors in acute CHF?
They inhibit PDE III’s degradation of cAMP (which was generated by the activation of β-receptors), thus prolonging the effect that cAMP has on the phosphorylation of calcium channels which increases calcium flow into the cell causing increased force of contraction
Case #1: What drug would be useful in a patient with post-operative atrial fibrillation?
Digitalis glycoside
Case #6: What can be given to a patient with Class IV heart failure, who is on Digoxin, Captopril, Furosemide, Carvedilol, and Spironolactone?
Any drug used for acute decompensation (i.e. β1-agonist, PDEIII inhibitors, short-acting vasodilators); patient needs a “tune-up”
Case #5: What can be done for a patient who has an ACE inhibitor-induced cough?
Replace the ACE inhibitor by an ARB
Case #4: What can be given to a patient who is hemodynamically unstable, for CHF symptoms and survival improvement?
Aldosterone antagonist: Spironolactone/Eplerenone
Case #3: What can be given to a patient who is hemodynamically stable, for CHF symptom and survival improvement?
β-blocker: Carvedilol
Case #2: Why can a patient on Digoxin, who is also given Verapamil, develop toxicity?
Anytime another drug is added to Digoxin, it can increase Digoxin levels; also, if the patient has diarrhea, a loss of potassium can exacerbate cardiac effects
What are the stages of hypertension?
Pre-hypertension: 120-139/80/89; Stage 1: 140-159/90-99; Stage 2: >160/100
What is the common treatment for HTN without compelling indications?
Thiazide diuretics (ACE inhibitors with combined treatment)
What are some of the causes of secondary HTN?
Primary renal disease, renovascular disease, oral contraceptive, pheochromocytoma, primary hyperaldosteronism, Cushing’s, coarctation of the aorta, sleep apnea
What are the Sokolow and Lyon criteria for LVH?
S on V1 + R on V5/6 > 35 mm
What is the Keith, Wagener, Barker classification regarding hypertensive retinopathy?
Group 1 (benign HTN): mild narrowing/sclerosis of retinal arterioles;
Group 2 (more marked hypertensive retinopathy): moderate/marked sclerosis of arterioles, exaggerated arterial light reflex, A-V crossing changes (nicking!);
Group 3 (mild angiospastic retinopathy): retinal edema, cotton-wool spots!/hemorrhages, sclerosis/spastic lesions of arterioles;
Group 4 (malignant HTN): group 3 with papilledema
What factors are important in getting an accurate BP measurement?
Patient should avoid exercise, eating, smoking, caffeine 60 min. before; They should sit quietly for 5 min; check both arms
What treatment is suggested for patients with pre-hypertension?
Lifestyle changes
What are the criteria required for a diagnosis of hypertension?
Systolic>140 OR Diastolic>90 on 2+ readings after the initial visit
What are the desired systolic and diastolic pressures for patients with chronic kidney disease and in diabetics?
Systolic<130; Diastolic<80
What percent of hypertensive patients are properly controlled?
One-third
What are the organic nitrates used with angina?
Nitroglycerin (most common); Amyl nitrite (inhalant, drug of abuse); Isosorbide mono/dinitrate (long-acting)
What drug interactions are associated with organic nitrates?
PDE5 inhibitors for impotence; leads to profound hypotension
Which enzyme is responsible for the breakdown of organic nitrates?
Hepatic glutathione organic nitrate reductase (which is why sublingual administration of nitroglycerin is effective – no first-pass effect)
How does tolerance come about with organic nitrates?
Through depletion of –SH groups (i.e. cysteine) and by down-regulation of aldehyde dehydrogenase (involved in metabolism of nitroglycerin to NO)
What side effects are associated with organic nitrates?
HA, postural hypotension, methemoglobinemia (Fe2+ --> Fe3+)
By what three mechanisms do the effects of the organic nitrates take place?
1° mech.: preload reduction by preferential dilation of venous capacitance vessels (↓ LVEDP);
2° mech.: large coronary artery dilation, causing a redistribution of blood to the ischemic zone;
3° mech.: afterload reduction due to arterial dilation, causing decreased vascular wall compression
What is the mechanism of action of organic nitrates?
They stimulate guanylate cyclase to produced cGMP which causes a decrease in intracellular calcium and thus vascular smooth muscle relaxation; organic nitrates can do this either through the formation of NO directly, or through NO binding with cysteine which causes formation of
S-nitrosothiols
When is left coronary blood flow maximal?
During diastole
What substances cause vasodilation in coronary vessels as part of autoregulation?
pO2, adenosine, NO
What factors affect myocardial O2 consumption?
Wall tension (LVEDP), myocardial contractility, HR
Why should β-blockers not be used in Prinzmetal angina?
Since they cause the unmasking of α-receptors, they can worsen vasospasms by causing vasoconstriction in affected vessels
Why are calcium channel blockers effective in the treatment of variant angina?
Because they inhibit coronary vasospasm and relax vasospastic vessels – the direct calcium channel blocker’s effects on the heart are not important
What does a blockade of calcium channels lead to in the heart?
SA-node: reduced HR, slowed conduction; Myocardial cell: reduced contractility
How do the effects of slow calcium currents differ between myocardial cells and SA-nodal cells?
In myocardial cells they are responsible for the plateau phase of the action potential, whereas in the SA-nodal cells they are responsible for the upstroke of the action potential
What are the indications of calcium channel blockers in myocardial ischemia?
Medium to long-range prophylaxis of variant angina and stable angina (in conjunction with nitrates and β-blockers)
Why is the combination of organic nitrate and β-blocker therapy so effective?
β-blockers offset the baroreceptor response caused by the nitrate-induced hypotension;
Nitrates offset the increase in LVEDP caused by β-blockers by decreasing venous return
What can occur if a β-blocker is withdrawn suddenly?
Rebound ischemia
What is the indication of β-adrenoceptor antagonists in myocardial ischemia?
Used for medium to long-range prophylaxis (not for acute attacks)
By what mechanisms of action do β-blockers treat myocardial ischemia?
By blocking β1-receptors they decrease HR/contractility which directly decreases oxygen demand and indirectly increases oxygen supply (increase diastolic perfusion/decrease wall compression);
By unmasking α-receptors in the normal zone, flow is shunted away from healthy vessels toward the ischemic zone, thus increasing its oxygen supply
What β-blockers are approved for the treatment of myocardial ischemia?
β1/β2-nonselective: Propranolol, Nadolol, Pindolol; β1-selective: Atenolol, Metoprolol
What is the tissue selectivity of each of the classes of calcium channel blockers?
Verapamil: AV node > myocardium > vascular smooth muscle, used for stable angina;
Diltiazem: vascular smooth muscle > myocardium; some effect on AV node, intermediate use;
Nifedipine: vascular smooth muscle > myocardium; no effect on AV node, used for variant angina
What is the mechanism of action of Ranolazine?
It blocks late Na+ channels, which in effect decreases intracellular Na+, and subsequently decreases intracellular Ca2+ via the Na/Ca exchanger (since extracellular Na+ is increased);
It is also a partial fatty acid oxidation inhibitor, meaning that it enhances glucose metabolism at the expense of lipid metabolism, which translates into more efficient oxygen consumption
What is the indication for Ranolazine?
Chronic angina in patients in whom other anti-angina drugs have not been effective
What drug combinations are useful with calcium channel blockers?
Verapamil & nitrates: nitrates offset congestion caused by Verapamil (by reducing pre/afterload);
Diltiazem & β-blockers: additive effects; Nifedipine & β-blockers: β-blockers offset the baroreceptor reflex caused by Nifedipine’s vasodilatory actions
What maternal factors are associated with congenital heart disease?
Rubella infection during 1st trimester; Drugs (thalidomide, alcohol, phenytoin, lithium, estrogenic steroids); Diabetes
What is formed when the septum secundum grows down to join with the endocardial cushions?
Foramen ovale (a small hole in the septum secundum allowing blood to flow through it and through the ostium secundum from the right atrium into the left)
What does the septum primum join with as it extends downward and what is formed in the process?
Septum primum joins with the endocardial cushions thus closing off the ostium primum; however, as this happens, a hole is formed in the septum primum (called the ostium secundum)
What are congenital heart defects called that have a permanent right-to-left shunt?
Cyanotic: tertralogy of Fallot, truncus arteriosus, tricuspid atresia, complete transposition of the great vessels, total anomalous pulmonary venous connection
What are some examples of heart defects that are cyanose tardive?
PDA, ASD, VSD, AVSD
What are the congenital heart defects called that cause a reversal of flow later in life?
Cyanose Tardive: an initial left-to-right shunt progressively increases pulmonary vascular resistance to such levels that right ventricular pressure exceeds that of the left ventricle, at which point the left-to-right shunt is reversed and becomes a right-to-left shunt (patient becomes cyanotic)
What are congenital heart defects called that do not have abnormal communication between the two circulations?
Acyanotic: coarctation of the aorta, right-sided aortic arch, Ebstein-malformation
During what period of embryogenesis do congenital heart defects form?
3-8 weeks
What happens around the 5th week of gestation regarding the heart?
Single chamber heart is divided by interatrial and interventricular septa; also, the formation of atrioventricular valves from endocardial cushions occur
Deletion of what gene leads to failure of cardiac myocyte specification?
Csx/Mkx2-5 (member of NK homeobox gene family); it is possible for cardiac myocytes to form even with the deletion, but this can lead to a host of other cardiac defects
Where do sinus venosus ASDs occur and what other problem is seen with them?
Near the entrance of the SVC or IVC; they can be accompanied by an anomalous connection of the right pulmonary veins to the SVC or right atrium
What defect can lead to a AVSD?
Failure of fusion of the superior and inferior endocardial cushions – leads to inadequate formation of the tricuspid and mitral valves
What substance is used to keep a PDA open in infants with multiple congenital heart defects?
Prostaglandin E1 (Alprostadil) – NSAIDs are used to close an isolated PDA (Indomethacin)
What murmur is associated with a PDA?
Harsh, “machine-like” murmur
What results in a patient with a PDA?
Oxygenated blood from the aorta flows into the deoxygenated blood of the pulmonary artery
What is the most common congenital cardiac anomaly?
VSD – almost all are associated with the membranous ventricular septum
What structures form the interventricular septum?
Upward growth of the muscular ventricular septum and downward growth of the membranous ventricular septum
What clinical signs are associated with an ASD?
Murmur; Volume hypertrophy of the right atrium/ventricle; Irreversible pulmonary hypertension (less common)
What is the most common ASD?
A defect of the ostium secundum leading to a deficient/fenestrated fossa ovalis
In patients with what syndrome are AVSDs common?
Down syndrome
What other defects can be found in a patient with Tetralogy of Fallot?
ASD, right aortic arch; also, the aorta becomes larger in diameter while the pulmonary arteries become smaller and thin-walled
How can a patient with Tetralogy of Fallot still get blood to the lungs?
Via a PDA or dilated bronchial arteries
What clinical findings are associated with Tetralogy of Fallot?
Boot-shaped heart (on x-ray); Physical development may be retarded; Cerebral tromboses; Bacterial endocarditis; Brain abscess
What causes Tetralogy of Fallot?
Anterosuperior displacement of the infundibular septum (leads to stenosis at or around the pulmonary valve)
What is the Tetralogy of Fallot?
Pulmonary stenosis (obstruction of the right ventricular outflow tract); Right ventricular hypertrophy; Overriding aorta; VSD
What is the key clinical sign associated with right-to-left shunts?
Cyanosis – “blue babies”
What can cause a partial AVSD?
Primum ASD (leads to mitral insufficiency)
What are fully developed combined atrial and ventricular septal defects called?
Persistent common AVSD
In what congenital heart defect do all four heart chambers freely communicate?
Complete AVSD
What causes Truncus Arteriosus?
Developmental failure of separation of the embryologic truncus arteriosus into the aorta and pulmonary artery
What other heart defects are found with TAPVC?
ASD/PFO; Volume/pressure hypertrophy of the right atrium/ventricle; Dilation of right atrium/ventricle/pulmonary trunk; Hypoplastic left atrium; (left ventricle is normal)
What causes a Total Anomalous Pulmonary Venous Connection (TAPVC)?
The common pulmonary vein fails to develop or becomes atretic; so, instead of pulmonary circulation flowing into the left atrium, they flow into the left innominate (brachiocephalic) vein or to the coronary sinus
What other heart defects are found with Tricuspid Atresia?
Hypoplasia of the right ventricle; ASD/patent foramen ovale (maintained by right-to-left shunt); VSD
What causes Tricuspid Atresia?
Unequal division of the AV canal during development; leads to complete occlusion of the tricuspid valve
What clinical signs are associated with a Truncus Arteriosus?
Systemic cyanosis; Irreversible pulmonary hypertension; CHF; Harsh holosystolic murmur; Widened pulse pressure; Bounding arterial pulses
What other heart defect is associated with a Truncus Arteriosus?
VSD
What are “tet spells”?
Acute hypoxia characterized by sudden cyanosis and syncope; patient will feel better in a squatting position since it will shunt more blood to the brain instead of the extremities
What signs are associated with TGA?
Prominent right ventricular hypertrophy; ASD (present in all patients); VSD and PDA are also common
What causes Transposition of the Great Arteries (TGA)?
Abnormal formation of truncal and aortopulmonary septa during development; causes the aorta to come off the right ventricle whereas the pulmonary artery comes off the left ventricle
What causes an Ebstein Malformation?
Downward displacement of an abnormal tricuspid valve into an underdeveloped right ventricle; there will be functional hypoplasia of the right ventricle, and tricuspid regurgitation or stenosis
How does pulmonary stenosis usually present?
As part of a complex anomaly (e.g. Tetralogy of Fallot)
What clinical signs are associated with aortic coarctation without PDA?
Enlarged intercostal/internal mammary arteries; Notching of the undersurfaces of ribs (on x-ray); Systolic murmur; Thrill; Cardiomegaly
How does the presentation of aortic coarctation differ with and without a PDA?
With PDA: presents early in life with cyanosis to lower half of the body (require immediate intervention); Without PDA: asymptomatic until adulthood, will have hypertension in upper extremities and weak pulses in lower extremities
What differentiates between the infantile and adult forms associated with coarctation of the aorta?
Infantile: tubular hypoplasia of the aortic arch; Adult: discrete ridge-like infolding of the aorta
What genetic abnormality is associated with coarctation of the aorta?
Turner syndrome
What signs/symptoms are associated with an Ebstein malformation that can be potentially lethal?
Attacks of supraventricular tachycardia; Wolfe-Parkinson-White syndrome (direct connection between SA node and ventricle, bypassing the modulatory actions of the AV node) – can lead to sudden death
What causes Hypoplastic Left Heart syndrome?
Severe underdevelopment of the left side of the heart, leading to hypoplasia of the left ventricle and ascending aorta, hypoplasia of the left-sided valves, and severe aortic valve stenosis/atresia;
How does blood get into systemic circulation with Hypoplastic Left Heart syndrome?
After flowing into the left atrium, a left-to-right shunt forces flow back into the right atrium; from here it goes into the right ventricle, out the pulmonary artery, and then through a PDA into systemic circulation
How does an infant with Hypoplastic Left Heart syndrome present?
They will initially appear well, but quickly become short of breath; most will die within the first month if they don’t receive surgical treatment
What are the three types of aortic stenosis and how do they differ?
Valvular: due to hypoplastic/dysplastic cusps or cusps abnormal in number, seen in Hypoplastic Left Heart syndrome; Subvalvular: due to a thickened ring/collar, dense endocardial stenosis;
Supravalvular: inherited form with ascending aorta wall thickening (due to defect in elastin gene)
What is dextrocardia?
Displacement of the heart to the right, which can be due to complete transposition of the right and left chambers; often associated with severe cardiovascular anomalies
What causes Endocardial Fibroelastosis?
Fibroelastic thickening of the endocardium in the left ventricle
What clinical signs are associated with aortic stenosis?
Prominent systolic murmur; Thrill; Pressure hypertrophy of left ventricle; Sudden death can occur with exertion
What keeps electrical signals in the heart from traveling both ways through their gap junctions?
Their inherent refractory period
What does lengthening of the PR interval imply?
Slowing of AV conduction
What does a broadened QRS wave imply?
Slowing of ventricular depolarization (ex. through drugs that inhibit the opening of voltage-gated sodium channels)
What does lengthening of the QT interval imply?
Lengthening of the ventricular action potential
What are the three types of aortic stenosis and how do they differ?
Valvular: due to hypoplastic/dysplastic cusps or cusps abnormal in number, seen in Hypoplastic Left Heart syndrome; Subvalvular: due to a thickened ring/collar, dense endocardial stenosis;
Supravalvular: inherited form with ascending aorta wall thickening (due to defect in elastin gene)
What are the phases of the ventricular myocyte AP associated with?
Phase 0: opening of Na channels; Phase 1: opening of K channels; Phase 2: opening of Ca channels (plateau); Phase 3: inactivation of Ca channel/opening of K channels; Phase 4: myocytes are waiting for stimulus
What are the phases of the SA/AV nodal cell AP associated with?
Phase 0: opening of Ca channels; Phase 3: opening of K channels; Phase 4: If/“funny current”/Na-K leakage current
What anatomical defect can cause arrhythmias?
Wolff-Parkinson-White syndrome: accessory pathways
What are early afterdepolarizations (EADs) and what can they lead to?
A spontaneous depolarization that occurs during the latter part of the repolarization; caused by a lengthening of the QT interval (e.g. by blocking K channels) which delays the inactivation of Na/Ca channels – EADs can lead to torsades de pointes
How do functionally defined reentrant signals cause tachyarrhythmias?
A region of Purkinje fibers is damaged (e.g. due to MI) and does not allow the propagation of active Na APs through that region --> this means that the anterograde signal coming from the Purkinje fibers encounters resistance as it travels down to innervate the myocardium --> at the same time, signals that traveled down through non-impeded Purkinje fibers have already activated the myocardium and these signals now reach the opposite side of the damaged region --> the signal from the myocardium is much larger (since there is more tissue on that end) and is able to passively cross the damaged region in a retrograde manner to cause a functionally defined loop
How do anatomically defined accessory pathways (e.g. with Wolff-Parkinson-White) cause tachyarrhythmia?
The signal from the SA node travels through both the AV node and the accessory pathway --> in the AV node, conduction is slowed whereas in the accessory node it is not --> once the signal exists the AV node, the accessory node has already recovered from the AP and will now conduct the AV node signal in a retrograde fashion --> this sends the signal back up to the AV node and in effect creates a continuous loop of APs that is independent of the SA node which causes tachycardia
What do ectopic pacemakers cause?
Impulses initiated in locations other than the SA node
What does Sick sinus syndrome cause?
Alternating periods of bradycardia and tachycardia caused by a dysfunctional SA node
What does hypokalemia cause?
Broadening of the AP/QT interval
What does responsiveness refer to?
Rate of depolarization of Vm during phase 0
What does conduction velocity depend on?
Number of open channels per unit area
What are two common types of mutations that cause arrhythmias?
Gain of function: Na channels that don’t inactivate; Loss of function: K channels that don’t conduct
What are strategies for decreasing arrhythmic activity, using drugs that decrease activity in reentrant circuits?
Decrease conduction velocity: Na/Ca channel blockers (cause complete block in reentrant circuit);
Increase refractory period (in accessory pathways): K channel blockers
What are the effects of Class III antiarrhythmic drugs?
K channel blockade, leading to slowed repolarization (↑QT), ↑AP duration, and ↑refractoriness
How do Class II antiarrhythmic drugs exert their effects and what are those effects?
As β-blockers they decrease cAMP in the cell and in effect decrease Ca channel recruitment; this leads to an overall decrease in intracellular Ca; because of this mechanism, Class II drugs will ↓AV conduction (= ↑PR), ↓HR, ↓Ca overload, and ↓triggering by DAD
What are the subdivisions of the Class I antiarrhythmic drugs and what characterizes each?
Class IA: medium-affinity block of Na and K channels (class III effect), ↑QRS and ↑QT
Class IB: low-affinity block of Na channels, use-dependent, ↓QT
Class IC: high-affinity block of Na channels, ↑QRS, small effect on repolarization
What are the main classes of the Singh-Vaughan Williams classification of antiarrhythmic drugs?
Class I: Na channel blockers
Class II: β-blockers
Class III: K channel blockers
Class IV: Ca channel blockers
What are strategies for decreasing arrhythmic activity, using drugs that functionally isolate the ventricular rhythm from atrial arrhythmias?
Decrease AV conduction: CCBs, β-blockers, adenosine, and vagal stimulation
What are strategies for decreasing arrhythmic activity, using drugs that decrease triggering (i.e. DADs)?
Increase threshold potential: Na channel blockers; Decrease cytosolic Ca: CCBs and β-blockers
What causes delayed afterdepolarization (DAD)?
Ca overload causes excess activation of the Na/Ca exchanger which in turn generates a Na current that creates an AP; can occur with β-receptor stimulation and with digitalis glycosides
What causes atrial flutter/fibrillation?
Reentrant circuits in the right atrium
What are strategies for decreasing arrhythmic activity, using drugs that decrease automaticity?
Increase max. diastolic Vm (i.e. hyperpolarize membrane during diastole): adenosine and vagal stimulation (both hyperpolarize cells by opening K channels, although by different mechanisms);
Decrease phase 4 slope (If): β-blockers and vagal stimulation; Increase threshold potential: Na/Ca channel blockers; Increase refractory period: K channel blockers
Which S-V W Classes are useful in treating atrial fibrillation and why?
Class III: ↑refractoriness; Class IA/IC: slow conduction; Class II/IV/Digitalis/adenosine: ↓AV conduction – adenosine is added here since it is given IV (emergency situations)
Which drug can cause an increase in DADs and why?
Digitalis, since it increases intracellular Ca indirectly by blocking the Na/K-ATPase which decreases the effectiveness of the Na/Ca exchanger
Which S-V W Classes are associated with toxicities leading to EADs/torsades de pointes and increased reentry, respectively?
EADs/torsades de pointes: IA/III, since both cause K channel blockade which leads to ↑QT;
Increased reentry (proarrhythmia): IC, since it can slow conduction in less damaged regions, forming another reentry circuit
Which treatments are useful for torsades de pointes and why?
Remove QT-prolonging drug; If genetic: Mg (required for processing of K)
Which S-V W Classes are useful in treating VT caused by DADs and why?
Class II/IV: ↓intracellular Ca
Which S-V W Classes are useful in treating ventricular tachycardia (VT) and why?
Class IA/IB/IC: blocking of Na channels (slows conduction); Class III: ↑refractoriness
Which S-V W Classes are useful in treating AV nodal reentry and why?
Class II/IV/Digitalis/adenosine: ↓AV conduction; Class IC: high-affinity block of Na channels in the case of an accessory pathway
What are the effects of Class IV antiarrhythmic drugs?
Ca channel blockade, leading to decreased intracellular Ca; this will be effective in slow-response cells (SA/AV nodes), causing ↓responsiveness, ↓AV conduction (= ↑PR), ↓DAD, ↑refractoriness; can uncouple ventricular rhythm from atrial arrhythmia
Which S-V W Classes are useful in treating nodal automaticities and why?
Class II/IV: both cause a decrease in intracellular Ca and Ca influx (↓AV conduction)
Which S-V W Classes are useful in treating atrial flutter and why?
Class III: ↑refractoriness decreases reentrant circuits; Class II/IV/Digitalis: ↓AV conduction
Which S-V W Classes are associated with toxicities leading to depression of LV function and why?
Class II/IV: ↓intracellular Ca
What is the usefulness of digitalis regarding arrhythmias?
Its “vagotonic” actions promoting the release of ACh from vagal nerve terminals; (not its effects on phase 4 slope and automaticity) – digitalis does produce harmful DADs
What is the usefulness of adenosine regarding arrhythmias?
It has a very short half-life (<10 sec), so it can be given IV to inhibit AV nodal arrhythmias, and once the arrhythmia has been treated, the effects of the drug will cease within seconds
Which drugs are mainly associated with a Class IV effect regarding the S-V W system?
Verapamil/Diltiazem (suppress DADs, negative inotropy, hypotension)
Which drugs are mainly associated with a Class III effect regarding the S-V W system?
Sotalol (d isomer), Amiodarone (many effects, long half-life, little risk of torsades de pointes), Dofetilide/Ibutilide (very K channel specific, high risk of torsades de pointes)
Which drugs are mainly associated with a Class II effect regarding the S-V W system?
Propranolol, Esmolol (IV only), Sotalol (l isomer: class II/III, d isomer: class III; both ↑EADs)
Which drugs are mainly associated with a Class I effect regarding the S-V W system?
Class IA: Quinidine (many effects, high risk of torsades de pointes), Procainamide, Disopyramide;
Class IB: Lidocaine (rapid metabolistm, given IV, ↑mortality post-MI), Mexiletine (given PO);
Class IC: Flecainide (does not produce EADs), Propafenone (Propranolol deriv., Class II effects)
Which S-V W Classes are associated with toxicities leading to sinus bradycardia and AV block, respectively?
Sinus bradycardia: Class II/IV due to ↓AV conduction, Class III due to ↑refractoriness;
AV block: Class II/IV due to ↓AV conduction
Which S-V W Classes are associated with toxicities leading to infranodal conduction block and why?
Class IA/IB/IC: blocking of Na channels can lead to a block of ventricular signal propagation, which can be lethal
Which S-V W Classes are associated with toxicities leading negative inotropism and why?
Class IA/IB/IC: ↓rate of depolarization; Class IV: ↓Ca entry
What adverse effects are associated with Amiodarone?
Pulmonary fibrosis (can be fatal), phospholipidosis; increases Digoxin levels; thyroid/hepatic dysfunction; CNS disturbance; Photosensitive reactions
What non-drug therapies have been successful in the treatment of arrhythmias?
Radio-frequency catheter ablation; Implantable converter/defibrillator
Which antiarrhythmic drugs are known to reduce mortality with chronic treatment?
β-blockers and (maybe) Amiodarone
What did the CAST clinical trial elicit?
Class IC drugs (e.g. Flecainide) uncover latent reentrant circuits and can increase mortality
What did the AFFIRM clinical trial elicit?
Treatment of rate control in atrial fibrillation is more important than rhythm control
What drugs should not be used in patients with Wolff-Parkinson-White syndrome with atrial fibrillation and why?
Digitalis, Verapamil, Diltiazem – since blocking AV conduction in this case will shunt all of the electrical signals from the atrial fibrillation through the accessory pathway, thus significantly increasing the rate of ventricular contraction
What adverse effects are associated with Digitalis?
Hyperkalemia, GI disturbance
What adverse effects are associated with Quinidine?
Diarrhea, cinchonism (tinnitus, HA, nausea, dizziness), hypersensitivity reactions
What adverse effects are associated with Procainamide, Disopyramide, Lidocaine/Mexiletine, and Propafenone?
Procainamide: Lupus-like syndrome; Disopyramide: anti-muscarinic effects; Lidocaine/Mexiletine: neurologic effects; Propafenone: 2D6 metabolism
What is the most important ADH receptor in maintaining fluid balance?
V2 receptors in the collecting duct --> leads to aquaporin-2 expression --> increased water permeability
Besides extrarenal and diuretic causes of hypovolemic hyponatremia, what other less common issues can be responsible?
Salt-losing nephropathy (advanced kidney disease with renal Na/K wasting); Mineralocorticoid deficiency (urine sodium>20, high serum K); Osmotic diuresis (glucosuria, urea diuresis after obstruction relief); Cerebral salt wasting
How can hypovolemic hyponatremia present with high urine sodium?
If the patient is on diuretics (thiazides > loop diuretics), a loss of fluid volume can cause hypovolemic-stimulated ADH release which will conserve free water but not sodium
How can hypovolemic hyponatremia be caused?
High GI loss of water or third-spacing along with free water intake (i.e. patient is losing isotonic fluids and taking in hypotonic fluids) --> this leads to nonosmotic release of ADH --> causes the kidney to conserve water as well as sodium and chloride (i.e. patient will have low urine sodium)
What causes pseudohyponatremia?
Errors in the lab calculation of sodium levels; can be due to severe hypertriglyceridemia or paraproteinemias
What is translocational hyponatremia?
Hyponatremia due to substances that increase osmolality (e.g. glucose); not a true hyponatremia since it is caused by sodium compensation due to the increase of another osmole
What causes hypo- and hypernatremia, respectively?
Hyponatremia: defect in urine-diluting capacity coupled with excess water intake;
Hypernatremia: defect in urine-concentrating capacity coupled with inadequate water intake
How is plasma osmolality calculated?
2x[Na+] + glucose/18 + BUN/8 --> normal range is 275-290 mosm/kg
What two stimuli have a role in ADH secretion?
Osmotic stimuli: effective osmoles (since they enhance water movement out of the cell, decreasing cell volume); Nonosmotic stimuli: decreased effective circulating volume, nausea, postoperative pain, pregnancy
What is the difference between effective and ineffective osmoles?
Effective osmoles (hypertonic saline, mannitol) cannot cross the cell membrane whereas ineffective osmoles (urea, methanol, ethanol) can
What are causes of euvolemic hyponatremia?
Glucocorticoid deficiency: impaired water excretion, but normal sodium excretion;
Severe hypothyroidism: reduction in cardiac output is a nonosmotic stimulus for ADH release;
Drugs; SIADH: defect in osmoregulation leading to inappropriate ADH secretion
What is the treatment associated with SIADH?
Water restriction; saline can worsen hyponatremia (since the kidney is handling salt normally, but not water)
What is the treatment associated with hypervolemic hyponatremia?
Water removal via loop diuretics; no additional sodium given; patient’s water intake is restricted
What is the treatment associated with hypovolemic hyponatremia?
Normal saline (since this will correct the hypovolemia --> halts excessive ADH secretion --> normalizes sodium levels)
What can be inferred if the urine osmolality of a patient is <100 mosm/L?
They are not producing ADH (everything else being equal)
What physical signs give a clue regarding the patient’s volume status?
Mucus membranes, skin turgor; Edema; Orthostatic drop in blood pressure; Weight gain
What are the clinical signs of SIADH?
Low serum osmolality (<270); Inappropriate urine concentration (>100); Clinical euvolemia; Elevated urine sodium levels – must rule out other causes of euvolemic hyponatremia
How is hypervolemic hyponatremia caused in a patient with CHF?
Low CO --> reduced effective circulating volume --> nonosmotic ADH stimulation --> increased free water retention and sodium excretion; patients will have also have a strong thirst stimulation in response to their decreased CO
How can nephrotic syndrome and advanced chronic kidney disease lead to hypervolemic hyponatremia?
Nephrotic syndrome: either through a renal defect in sodium excretion or through protein loss which leads to lowered plasma oncotic pressure and intravascular volume depletion;
Advanced chronic kidney disease: can lead to an increase in sodium excretion to counteract the loss of nephrons
How can liver disease lead to hypervolemic hyponatremia?
There is vasodilation and AV fistulas which leads to a decrease in mean arterial BP and effective CO --> this is a nonosmotic stimulus for ADH secretion
What formula can be used to determine the change in sodium after giving them 1L of saline?
[ ([Na]fluid – [Na]serum)/(0.6 x weight + 1) ]; 0.6 is used for men/0.5 for women
How can Diabetes Insipidus cause euvolemic hypernatremia?
Since there is a defect in ADH action (central/secretion vs. nephrogenic/receptor), the patient will have polyuria; they compensate for this through polydipsia; however, if they cannot take in their excess amount of water (e.g. during a hospital stay) they will become hypernatremic
What are causes of euvolemic hypernatremia?
Lack of access to free water (young, elderly); Extrarenal causes (insensible losses: respiratory, dermal); Renal causes (hypodipsia, Diabetes Insipidus)
What are causes of hypervolemic hypernatremia?
Hypertonic solutions (3% saline, NaHCO3), salt tablets
What are causes of hypovolemic hypernatremia?
Loss of hypotonic water: kidney loss (osmotic/loop diuretics, postobstructive injury), extrarenal loss (burns, diarrhea, fistulas)
What normally prevents people from becoming hypernatremic?
Thirst – people do not typically become hypernatremic if they have access to water
What is the major problem with overly rapid correction of hyponatremia?
Since the brain cells have adapted to the hyponatremia, correcting it too rapidly may result in cerebral dehydration and demyelination --> central pontine myelinolysis --> paraparesis, quadriparesis, dysarthria, dysphagia, coma
What is the maximum rate of correction for sodium levels and what is given to hyponatremic patients in an emergency setting?
Max. rate: <12meq/L/day; Emergency: hypertonic saline (max. rate still applies)
How is the amount of saline required to correct a patient’s hyponatremia determined?
[Total body water x Na+ deficit/L] – can use 3% saline (513meq/L), normal saline (154meq/L), ½ saline (77meq/L) or D5W (0meq/L)
How is chronic asymptomatic hyponatremia treated?
Fluid restriction
How is the diagnosis of Diabetes Insipidus made?
Polyuria, polydipsia, urine osmolality<serum osmolality; By giving the patient dDAVP (synthetic vasopressin) and seeing if the kidney responds (central problem) or not (nephrogenic problem)
What happens when the sodium levels are corrected too rapidly in hypernatremia?
Rapid correction will lead to cerebral edema (seizures, neurological damage, death); correction rate should be 0.5meq/L/hour
What is the treatment for hypernatremia?
Apply same concepts as for hyponatremia (i.e. change in Na after 1L formula)
What drugs are used for slowing ventricular rate?
BB (Metoprolol), Diltiazem (and some Verapamil), Digoxin – a combination of a BB and Digoxin is most effective
What drugs used for the maintenance of sinus rhythm are used in a patient with HTN and LVH?
Amiodarone; or catheter ablation if treatment is unsuccessful
What drugs used for the maintenance of sinus rhythm are first-line in a patient no heart disease?
Flecainide, Propafenone, Sotalol; if those fail, Amiodarone, Dofetilide, or catheter ablation are useful – same drugs are also used in patients with hypertension, but with no substantial LVH
What is the biggest danger in giving a patient Dofetilide?
QT prolongation (since it has Class III effects) – however, Dofetilide is safer than other drugs in patients with CAD or heart failure; with CAD Sotalol/Amiodarone can also be used, and in heart failure Amiodarone is an option (not Sotalol due to its negative inotropic effects)
Which drugs are used for rhythm control of A-fib?
Dofetilide, Sotalol, Amiodarone, Propafenone, Flecainide; Ibutilide is for acute treatment only
What is the danger of blocking AV conduction in a patient with WPW in A-fib?
Signals traveling through the AV node go to the accessory pathway and cause a “road block”; by removing this mechanism, signals from the A-fib will be able to travel through the accessory pathway unobstructed, leading to a much greater ventricular rate
What is required along with ablative therapy for A-fib?
A pacemaker to take over the ventricular rhythm
What antiarrhythmic drugs are used in emergency situations?
Amiodarone (cardiac arrest with VT or V-fib), BB/Diltiazem (rate-control of A-fib),
Adenosine (termination of SVT: AV nodal or AP reentry)
What is the therapeutic approach for A-fib?
Slow ventricular rate (AV blocking drugs); Restoration of sinus rhythm (antiarrhythmic drug/DC shock); Anticoagulation (Heparin/Warfarin)
What symptoms are associated with atrial fibrillation?
Palpitations, dyspnea, fatigue, chest discomfort, lightheadedness, syncope – major risk with A-fib is thromboemboli in the left atrial appendage due to stagnating blood (consider anticoagulation)
What drug might be considered in people with benign premature ventricular beats?
BB (Propranolol), although therapy is not needed nor indicated
Which antiarrhythmic drugs are associated with renal and hepatic elimination, respectively?
Renal: Sotalol, Dofetilide, Digoxin; Hepatic: Propafenone, Amiodarone, Lidocaine
What drug interactions are associated with Amiodarone, Dofetilide, and Propafenone?
Amiodarone: increases Warfarin, Digoxin, Cyclosporine; Dofetilide: metabolized by CYP3A4 and P-glycoprotein transport; Propafenone: increases Digoxin, Warfarin, and increased by Cimetidine
What treatment is effective in V-fib?
Asynchronous shock; if not effective, include Epinephrine and subsequently Amiodarone
Which drugs reduce the need for ICD shocks?
Sotalol and Amiodarone
What treatment is effective in sustained ventricular tachycardia in patient presenting with hypotension, angina, or heart failure?
Direct current (DC) shock after sedation; if chronic: ICD
What treatment is effective in non-sustained ventricular tachycardia?
ICD (implantable cardioverter-defibrillator)
What treatment choice is more effective in atrial flutter as compared to A-fib?
Catheter ablation (since destroying one reentry loop is easier than taking out multiple entities)
What is the main therapy for Paroxysmal Supraventricular Tachycardia?
Adenosine (slows AV conduction) – avoid with asthma, dipyridamole (increases effect by blocking cell uptake), theophylline (reduced effect by blocking receptors)
What is the calculation used for BMI?
for kg/m2: Weight/(Height)2; for lb/in2: (Weight x 703)/(Height)2
Where and when is Ghrelin produced?
By the stomach when it is empty
What type of drug is Rimonabant?
Endogenous canabinoid
What main side effect do appetite suppressants have in common and what is the exception?
They are stimulants (HR, BP, insomnia) – the exception is Orlistat which is a lipase inhibitor; its main side effects are gastric problems (diarrhea, flatulence, bloating, etc.)
What symptoms are often seen in conjunction with mitral stenosis?
Atrial fibrillation (palpitations), thromboembolism (stroke), endocarditis, right heart failure, hoarseness (recurrent laryngeal nerve paralysis)
What is factor determines the severity of symptoms of a patient with mitral regurgitation?
LV function
What is the difference between acute and chronic mitral regurgitation?
Acute: inadequate compensation, causes an increase in LA pressure without dilation, which leads to pulmonary congestion/edema; Chronic: time for compensation to occur (increased LA size, LV dilation/eccentric hypertrophy, increased LVEF with normal SV) but eventually leads to progressive worsening LVEF/CO
What is the pathophysiology of mitral regurgitation?
LV volume overload --> portion of LVSV ejected into low pressure LA --> enlarged LA/LV (eccentric) --> increased SV/EDV
What is the etiology for mitral regurgitation?
Degenerative (prolapse, Marfan); Papillary muscle dysfunction; Dilated LV; Infective endocarditis; Mitral annular calcification; Rheumatic heart disease
What physical findings are associated with mitral stenosis?
Increased S1 (more prominent) due to high LA pressure; Opening snap prior to diastole (audible leaflet opening); Diastolic rumble (low pitch, occurs during diastole, severity is related to length of rumble but not intensity, best heard in L lateral decubitus position); L parasternal heave (with increased P2); Right heart failure
What describes the latent period of mitral stenosis?
It is very long (~20 years)
What is the most common etiology for mitral stenosis?
Younger people: rheumatic fever; Older people: calcific
What symptoms are associated with mitral stenosis?
Dyspnea (early: dyspnea of exertion, later: paroxysmal nocturnal dyspnea), fatigue, hemoptysis
What symptoms are associated with aortic stenosis?
Dyspnea (due to CHF); Angina (often with CAD); Syncope (with exertion)
What is the natural history of aortic regurgitation?
Depends on rapidity of onset; LV function preserved early --> gradual fall of EF seen (but EF fall may precede symptoms)
What is the difference between acute and chronic aortic regurgitation?
Acute: inadequate compensation, causes an increase in LV diastolic pressure and LA pressure without dilation, which leads to pulmonary congestion/edema; Chronic: time for compensation to occur (LV dilation/eccentric hypertrophy, increased LVEF/SV, widened pulse pressure), but eventually leads to progressive worsening of LVEF/CO
What is the pathophysiology of aortic regurgitations?
Regurgitation from aorta to LV during diastole --> volume overload of LV --> eccentric hypertrophy (increased LV/LA size, increased SV/EDV)
What etiologies are associated with aortic regurgitation?
Abnormalities of leaflets (bicuspid, endocarditis, rheumatic); Dilation of the aortic root (degenerative/ectasia, aortic dissection, Marfan)
What physical findings are associated with aortic stenosis?
Systolic ejection murmur; Decreased A2 (leaflet immobility); Delayed carotid upstroke (may go unnoticed in elderly due to carotid hardening); Carotid shudder/suprasternal notch thrill (seen with bicuspid aortic valve)
What must be done when the patient with aortic stenosis becomes symptomatic?
Immediate aortic valve replacement
What physical findings are associated with mitral regurgitation?
Holosystolic murmur (in MV area); Displaced PMI (point of maximal impulse);
L parasternal heave; MV prolapse (systolic click and late systolic murmur)
What is the pathophysiology of aortic stenosis?
Pressure overload of LV due to increased afterload  concentric hypertrophy --> LV to aorta pressure gradient; there will be normal chamber size (early on)
What etiologies are associated with aortic stenosis?
Calcific: senile/degenerative (>65); Congenital bicuspid valve (<65); Rheumatic disease (95% with MV involvement as well)
What physical findings are associated with aortic regurgitation?
Low blowing diastolic murmur; Widened pulse pressure (bounding pulses); Displaces PMI (if significant/chronic)
What characterizes pulmonary regurgitation?
Can be caused by pediatric congenital surgeries; also, secondary to pulmonary hypertension; Physical findings will be similar to AR (e.g. RV eccentric hypertrophy)
What characterizes pulmonary stenosis?
Usually congenital; gradient between PA and RV in systole; Physical findings can include a systolic ejection murmur that increases with inspiration
What characterizes tricuspid regurgitation?
Usually due to pulmonary hypertension or RV annulus dilation; symptoms will be similar to those of right heart failure; Physical findings will be similar to MR except sounds increase on inspiration; also, neck vein distention (with prominent v waves) and a pulsatile liver can be seen
What characterizes tricuspid stenosis?
Often associated with rheumatic disease; Physical finding similar to MS except sounds increase with inspiration (due to the increase in blood volume that occurs during inspiration); also, neck vein distention can be seen
How is the quality of pain associated with angina pectoris described?
Pain, pressure, burning, tightness
What are the most common GI causes of chest pain?
Esophageal reflux (GERD, rarely radiates) > Esophageal spasms (mimics angina) > Peptic ulcers > Biliary disease
Besides the most common cause of angina pectoris (atherosclerosis), what other disease can cause these symptoms?
Syndrome X (ischemia, normal arteries); Coronary spasm (epicardial arteries); Aortic valve disease (stenosis)
What is the difference between stable and unstable angina?
Stable: due to stable coronary artery plaques (i.e. reduced flow related to stenosis), presents on exertion, it is predictable, and its course unchanging; Unstable: presents at rest, nocturnally, or with minimal exertion, it is unpredictable, and its course changing
What other discomfort is relieved by nitroglyerin?
Esophageal spasm
How long does it take for nitroglycerin to take effect?
1-5 minutes (it’s not instant and will not last longer than 20 min) – must be sure to see whether the nitroglycerin is still good (i.e. it hasn’t been exposed to the environment for too long)
What activities is the onset of angina associated with and how long does it normally last?
Related to exertion, eating, emotion, coitus, cold exposure – usually lasts 30 sec. to minutes
What are the key differences between superficial and visceral chest pain?
Superficial: pain specific fibers, felt as “pain”, well localized, pain related to movement;
Visceral: autonomic fibers, felt as a vague sensation which may not be “pain”, poorly localized
What is the definition of angina pectoris?
A visceral quality discomfort anywhere between the nose and the umbilicus that comes on with exertion, emotion, or other activity that either increases HR, BP, inotropic state of the heart, or heart size or decreases oxygen supply; and that abates with rest, subsidence of emotion, or resolution of either the increases in the determinants of myocardial oxygen demand or decreases in oxygen supply
What techniques should be done on physical exam for chest pain?
Check for chest wall tenderness; Passive limb movement; Auscultate the chest
What are some common pulmonary causes of
chest pain?
Pneumonia (fever, sputum); Pulmonary embolus (dyspnea); Pneumothorax (absence of breath sounds); Pleurisy (URI)
What are some miscellaneous causes of chest pain?
Pericarditis (relieved by sitting up and leaning forward, pericardial friction rub, EKG changes); Aortic dissection (tearing quality pain with radiation into the back, abnormalities of pulses)
What are some common musculoskeletal causes of chest pain?
Costochondritis (most common, tender); Xiphodynia; Precordial catch (relief by chest splinting)
What defines situational couple violence?
Violence that arises out of conflicts that lead to arguments and then to violence (most common form in every day life)
What questions should be avoided during the domestic violence interview?
“What keeps you with someone like that”; “What did you do that caused him to hit you”; “How long are you going to put up with this”; Why don’t you just leave”; “Do you realize you are a battered woman” (avoid stigmatisms)
What behavioral health effects are associated with domestic violence?
Chronic pain syndromes, substance abuse, sleep disturbances, chronic fatigue, anxiety/panic disorders PTSD, hypervigilance, depression, suicidal ideation/attempts
What adverse health effects are associated with domestic violence?
Homicide, acute injury, chronic headaches, chronic abdominal pain, pelvic pain, recurrent STDs/UTIs, musculoskeletal complaints, eating disorders
What is a key difference between the female and male victims of intimate terrorism?
Women are more likely to be injured by the violence
What is the prevalence of intimate terrorism (when looking at rape and/or physical assault)?
In the past year: women 1.5%, men 0.9%; During lifetime: women 25%, men 7.6%
What defines mutual violent control?
Both partners are violent and controlling, while battling for control in a relationship (not sure this exists, very rare)
What are the typologies of partner violence?
Intimate terrorism, violent resistance, situational couple violence, mutual violent control
What defines violent resistance?
Violence used in resistance to an intimate terrorist (can be self-defensive, like payback)
What defines intimate terrorism and what characterizes it?
One partner uses violence along with emotional/psychological abuse to maintain control over the other; most commonly done by the male, more likely to escalate over time, most often seen by police/hospitals
What is the role of the physician in patients with domestic violence?
Communicate concern, offer information, provide medical treatment, review options (refer as necessary), initiate safety planning
What is the role of the physician in patients with domestic violence?
Communicate concern, offer information, provide medical treatment, review options (refer as necessary), initiate safety planning
What does the mnemonic RADAR stand for?
Remember to ask about domestic violence; Ask direct questions; Document findings;
Assess safety; Review options – also, know the local resources available to the patient
What does the mnemonic RADAR stand for?
Remember to ask about domestic violence; Ask direct questions; Document findings;
Assess safety; Review options – also, know the local resources available to the patient
What are the rules regarding the reporting of domestic violence to law enforcement?
Domestic violence is not a reportable condition (unless there is an attempt at lethal violence)
and should only be reported if the victim requests it
What are the rules regarding the reporting of domestic violence to law enforcement?
Domestic violence is not a reportable condition (unless there is an attempt at lethal violence)
and should only be reported if the victim requests it
What should be included in the medical documentation of domestic violence cases?
Written information recorded in quotations; Body charts and injury maps; Photographs –
Do not speculate, be vague, victimize the victim
What should be included in the medical documentation of domestic violence cases?
Written information recorded in quotations; Body charts and injury maps; Photographs –
Do not speculate, be vague, victimize the victim
What information should be provided to the patient?
IPV is common, IPV often escalates over time, IPV has adverse effects on children, there are resources in the community to help the patient
What information should be provided to the patient?
IPV is common, IPV often escalates over time, IPV has adverse effects on children, there are resources in the community to help the patient
What are clues uncovered during the physical examination that may indicate domestic violence?
Frontal injuries (from direct blow), perforated tympanic membrane (from slap), injuries in various stages of healing, explanation inconsistent with injury, delay in help-seeking
What are clues uncovered during the physical examination that may indicate domestic violence?
Frontal injuries (from direct blow), perforated tympanic membrane (from slap), injuries in various stages of healing, explanation inconsistent with injury, delay in help-seeking
What are examples of atypical symptoms associated with MI?
Abdominal pain, jaw pain, altered mental status (in elderly patients)
What is the treatment for acute MI?
If at a center where an interventional cardiologist can perform a PTCA, this would be the preferred method; however, if out in the field, give thrombolytics and then get the patient to a center where PTCA can be performed
What is the purpose of GP IIb/IIIa inhibitors?
Keep a vessel open after thrombolytic therapy
What are indications for the use of thrombolytics?
ST segment elevation greater than 0.1 mV in 2+ contiguous leads; New LBBB; Anterior ST depression consistent with posterior infarction
What should always be done when giving a patient nitrates in the ED?
Get an IV ready to push fluids in case they become hypotensive (since nitrates reduce preload)
What are the indications for Heparin and LMWH regarding MI?
Heparin for catheterization (since its effects can be stopped); LMWH in NSTEMI/unstable angina
In the treatment of MI, what may be used as an alternative to aspirin?
Clopidogrel
What are the criteria associated with air medical transport?
When the outcome of the patient is dependent on rapid transport or the patient requires the expertise of the flight team at the scene or en route (medical transport is activated by EMS personnel)
How is chest pain of substernal origin described by patients?
Often as a pressure sensation, but can be described as any kind of pain
If an EKG does not clearly show ST segment elevation, what can be looked at for a more definitive diagnosis?
Reciprocal ST segment depression (i.e. ST segment depression in the other areas of distribution)
What is meant by the statement that traumatic death is tri-modal?
Patients either die immediately, within 6 hours, or within 2-7 days
What is the leading cause of adult disability in the US?
Stroke (men are at higher risk)
What should be given to a patient needing a massive transfusion?
FFP/cryo
What is meant by the secondary survey?
Head-to-toe examination for injury (most important is the head)
What does tachycardia/hypotension mean until proven otherwise?
Blood loss
What is meant by the ABCDEs of emergency medicine (primary survey)?
A (airway): provide suppl. oxygen  non-definitive (jaw thrust, suction), definitive (intubate);
B (breathing): check rate, depth, work, sounds, symmetry, if inadequate, intubate;
C (circulation): BP, HR, perfusion, control bleeding (pressure dressing), IV access;
D (deficit/deformity): neurologic assessment (GCS: 3-15), major deformities (face, pelvis, back);
E (exposure/environment): take everything off patient for examination, keep patient warm!
What is an essential approach when evaluating a trauma patient?
Use a systematic approach so that nothing big is missed
What other disease processes can cause ST segment elevation?
Ventricular aneurysm (anterior ST elevation); Acute pericarditis (diffuse ST elevation)
What is the first sign that changes in a trauma patient?
Heart rate
What are the three considerations to be looked at with a trauma patient?
Mechanism, anatomy, physiology
What is the purpose of a MERCI catheter?
Physically remove a clot from the patient’s artery (funnel shaped, spiral wire, hooks onto clot)
What is the purpose of a MERCI catheter?
Physically remove a clot from the patient’s artery (funnel shaped, spiral wire, hooks onto clot)
What are causes of left ventricular systolic dysfunction in CHF?
Impaired contractility (MI, mitral/aortic regurgitation, dilated cardiomyopathy) and/or excessive afterload (aortic stenosis, uncontrolled HTN) – will see ESPVR shifted downward, SV down, ESV up, and EDV/LVEDP up
What are causes of left ventricular systolic dysfunction in CHF?
Impaired contractility (MI, mitral/aortic regurgitation, dilated cardiomyopathy) and/or excessive afterload (aortic stenosis, uncontrolled HTN) – will see ESPVR shifted downward, SV down, ESV up, and EDV/LVEDP up
What increases SV?
Increased
preload/contractility, decreased afterload
What increases SV?
Increased
preload/contractility, decreased afterload
What does ESV depend on?
Afterload, contractility
What does ESV depend on?
Afterload, contractility
What does ejection fraction depend on?
Preload, afterload, contractility
What does ejection fraction depend on?
Preload, afterload, contractility
What is the Law of LaPlace and what does it relate to?
[stress = (pressure x radius)/(2 x wall thickness)]; used to calculate afterload
What are non-invasive measures of looking at left ventricular preload and right ventricular preload, respectively?
Left-ventricular: echocardiogram, cardiac MRI; Right-ventricular: jugular venous pressure
What is the Law of LaPlace and what does it relate to?
[stress = (pressure x radius)/(2 x wall thickness)]; used to calculate afterload
What will be affected in a patient with a stroke in the basilar artery (posterior circulation)?
Vision, balance, consciousness, breathing, CN X-XII
What are non-invasive measures of looking at left ventricular preload and right ventricular preload, respectively?
Left-ventricular: echocardiogram, cardiac MRI; Right-ventricular: jugular venous pressure
What are the 5 aspects of the NIH stroke scale?
Wakefulness, Visual, Motor, Sensation, Language
What will be affected in a patient with a stroke in the basilar artery (posterior circulation)?
Vision, balance, consciousness, breathing, CN X-XII
What is the time-frame for the use of IV tPA?
Must be administered in the first 3 hours (tPA is associated with a 10x increase in hemorrhage)
What are the 5 aspects of the NIH stroke scale?
Wakefulness, Visual, Motor, Sensation, Language
What is the time-frame for the use of IV tPA?
Must be administered in the first 3 hours (tPA is associated with a 10x increase in hemorrhage)
What is the hallmark of eccentric ventricular hypertrophy?
Ventricular dilation (due to volume overload, new sarcomeres in series with old) – there is no chamber dilation in concentric ventricular hypertrophy (due to pressure overload, new sarcomeres in parallel with old)
What physical findings are associated with RV CHF?
Jugular venous distention (increase RA pressure), hepatomegaly, peripheral edema
What physical findings are associated with LV CHF?
Diaphoresis, tachycardia, tachypnea, crackles/wheezes, dullness over lung bases (effusion), abnormal cardiac impulse (cardiomegaly), S3 (S4) gallop (abnormal filling sound)
What symptoms are associated with RV CHF?
Peripheral edema (better in morning), weight gain, RUQ discomfort (swollen liver), anorexia/nausea (bowel edema)
What symptoms are associated with LV CHF?
Orthopnea (sleep with pillows), paroxysmal nocturnal dyspnea, nocturnal cough, fatigue, nocturia
How does ethanol affect the heart?
It is a negative inotropic
compound
What are causes of left ventricular diastolic dysfunction in CHF?
Impaired relaxation or increased stiffness (left ventricular hypertrophy, hypertrophic cardiomyopathy, restrictive cardiomyopathy, transient myocardial ischemia) – will see lower part of PV loop shifted upwards, increased LVEDP but lower EDV
What is the problem with mitral stenosis, regarding the PV loop?
It lowers CO by reducing a to b time and volume (i.e. there is less LV filling time and thus less LV volume)