• 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

1/545

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;

545 Cards in this Set

  • Front
  • Back
What valves are open during systole?
The aortic and pulmonary valves
What valves are open during diastole?
The tricuspid and mitral (bicuspid) valves
What is the steady state equilibrium voltage for a myocyte
-90 mV
What ion is passively leaking out of the all myocyte while at rest? Do any special parts of the heart leak any other ions in or out at rest?
Cardiac myocytes passively leak K+ (maintaining their -90mV charge). The myocytes of the sinus and AV nodes, the His bundle, and Purkinje fibers leak Na+ inward decreasing their polarity and leading to their automaticity and pacemaker properties.
What ion is responsible for rapid depolarization of myocytes?
Na+ is responsible for phase 0. The opening of the ion channels is voltage-gated and occurs when the cell reaches a specific membrane threshold
What are the three possible states of Na+ ion channels during the cardiac cycle?
* Na+ channels start closed. * When a threshold potential is reached (obtained from neighboring cells), the voltage-gated channels open, allowing Na+ to rush into the cell.
* The channel will spontaneously inactivate, while depolarization is still occurring. The channel will remain inactive until the cell membrane is repolarized, returning the channel to the closed (resting) state.
How does the action potential get transmitted from cell to cell?
One myocyte gets depolarized to the point of its threshold, triggering the opening of its Na+ channels, rapidly depolarizing that cell. Gap junctions that connect cell to its neighbors and allows the depolarized cell to pass some positive charge to its neighbor until the neighbor reaches its threshold and opens its Na+ channels, pushing the neighbor cell to depolarize as well.
What ion is responsible for the transient partial repolarization of the myocyte?
The transient outward current of K+ ions (similar to what normally maintains the -90mV charge). This is phase 1 of the cardiac action potential
What ion is responsible for phase 2 of the cardiac action potential?
Phase 2 is the plateau phase, maintained by Ca2+ ions. Depolarization of the myocyte, in phase 0, triggers the opening of Ca2+ channels in the membrane. A slow, sustained influx of Ca2+ prolongs depolarization of the myocyte and triggers release of Ca2+ from the Sarcoplasmic Reticulum.
What ion is responsible for phase 3 of the cardiac action potential?
The voltage-gated K+ channels that remain open are responsible for the rapid repolarization of the myocyte. This repolarization is also what is required to turn the Na+ channels from an inactive to a resting state.
Which portion of the EKG represents the rapid depolarization of the heart?
The QRS segment
Which portion of the EKG represents the influx of Ca2+ into the heart?
The ST segment or the plateau
Which portion of the EKG represents the repolarization of the heart?
The T wave
Graphically, how does a Na+ channel blocker affect the cardiac action potential and EKG
* Slow the upstroke of phase 0
* Prolongs QRS
Graphically, how does a K+ channel blocker affect the cardiac action potential and EKG
* Prolongs the plateau of phase 2, delaying phase 3
* Prolongs the QT
What is the response to a premature beat during ERP?
The cell can provide no response
What is the response to a premature beat after the RRP?
The cell will produce a completely normal action potential
What is the response to a premature beat in between the ERP and the RRP?
The cell will produce an action potential with a slower-onset, but propagation to neighboring cells is normal
What cells normally have automaticity and how does it work?
The sinus node, AV node, His bundle, and Purkinje fibers all maintain a slow, inward leak of positive ions (mostly Na+) during phase 4, leading to a "pacemaker current".
How does automaticity differ between the sinus and AV nodes and the His bundle and Purkinje fibers?
The sinus and AV nodes are very leaky during phase 4. These cells never repolarize to -90mV, they only make it to -60mV rendering the Na+ channels irrelevant. Slow, steady Ca+ channels are responsible for phase 0 in these cells.
How do autonomics affect the normal cardiac action potential?
* Sympathetic influence increases the slope (leakiness) during phase 4, pushing the cell past threshold quicker, producing a quicker "pacemaker current".
* Parasympathetic stimulation decreases the slope (leakiness) during phase 4, slowing the cell's ability to overcome it's threshold, producing a slower "pacemaker current".
What neurotransmitters are responsible for a sympathomimetic/catecholaminergic response?
Epinephrine and Norepinephrine
What neurotransmitters are responsible for a parasympathomimetic/cholinergic response?
Acetylcholine
What are Ryanodine receptors?
They are the calcium-dependent calcium release channels found in the sarcoplasmic reticulum
Differentiate the distribution of L- v. T-type Ca2+ channels in the heart
* L-type (long-acting) are found in every heart cell and contribute to phase 2 (the plateau of depolarization) of the cardiac action potential
* T-type (transient) are concentrated in the SA and AV nodes, but are also found in the atria and Purkinje cells. T-type Ca2+ channels are involved in the automaticity of the nodes.
How does the funny current (If) in the heart and what type of channel regulates it?
The funny current is the slow, inward leak of Na+ that pushes the cell towards phase 0, and results in the pacemaker current. Positive ions flow in through Hyperpolarization-activate, Cyclic Nucleotide-gated (HCN) channels, which are open when the cell is polarized (phase 4)
How do autonomics, ß-blockers, and Ca2+ channel blockers interact with HCNs and what are the physiologic results?
* Sympathetics: Increases cAMP, which increases HCN activity, which increases the heart rate
* Parasympathetics: decreases cAMP, which decreases HCN activity, and slows the heart rate
* ß-blockers: decrease sympathetic stimulation, slowing the heart rate
* Ca2+ channel blockers: decrease HCN responsiveness to cAMP, slowing the heart rate
What are the components that make up thin filaments of muscle?
* G-actin (globular) polymerizes into f-actin (filamentous)
* Tropomyosin helps to hold f-actin together
* Troponin-T: binds tropomyosin, anchoring the troponin complex
* Troponin-C: binds Ca2+, initiating contraction
* Troponin-I: binds actin, inhibiting the actin-myosin interaction
What is the make up of thick filaments of muscle?
Each myosin molecule has 2 heavy chains and 4 light chains. The light chains are involved in structure and regulation. The heavy chains each have a globular head, with binding sites for ATP and actin.
What is the name of where thick and thin filaments attach and what is one contractile unit called?
Thin filaments, actin, bind to the Z-line. Thick filaments, myosin, bind tail-to-tail along the M-line. One contractile unit, or a sarcomere, runs from Z-line to Z-line.
Name and describe the stages of the sliding filament model.
* Attachment: The ATP absent myosin head is bound to actin
* Release: The myosin head binds ATP and releases the actin filament
* Bending: The breakdown of ATP to ADP + Pi (in the presence of Mg) causes the myosin head to bend
* Force generation: If Ca2+ is present, myosin will bind a new actin site and return to its unbent state, moving the actin filament. ADP is released from the myosin head.
Which portions of the sarcomere change in size with contraction
The I (actin only) and H (myosin only) bands both shrink as the thick and thin filaments overlap. the A band (total length of myosin, tail-to-tail) doesn't shrink
What ion is responsible for linking action potential to actual sarcomere filament interactions?
Ca2+ which rushes into the cell from outside and from the sarcoplasmic reticulum is responsible for unbending the myosin head during force generation, allowing muscle contraction.
How does Ca2+ regulation from the sarcoplasmic reticulum differ in skeletal muscle from cardiac muscle?
In skeletal muscle, Ca2+ is voltage-dependent. Confirmational changes in DHP receptors opens ryanodine receptors. In cardiac muscle, Ca2+ is Ca2+ activated. Ca2+, released by the DHP receptors binds the ryanodine receptors, triggering Ca2+ release.
How are cardiac cells coupled together?
Intercalated disks form linear cell-to-cell attachments to form muscle fibers, allowing cardiac action potential to be propagated in many directions.
What are the different types of junctions formed by intercalated disks?
* Adhering junctions tightly hold adjacent cells
* Desmosomes prevent cells from pulling apart with tension
* Gap/communicating junctions provide ionic continuity among adjacent cells so information can be passed between cells
Where are the SA and AV nodes located?
The sinoatrial node is located in the atrial wall, close to the entrance of the SVC. The atrioventricular node is located in the right atrium, near the lower part of the interatrial septum
What nerves regulate autonomic stimulation of the cardiac muscle?
CNX (vagus) regulates parasympathetic conduction at the SA and AV nodes, as well as in the coronary arteries. T1-T6 sympathetically innervates the SA and AV nodes, the myocardium, epicardium, and coronary arteries
How do autonomics affect heart contraction?
* ACh (parasympathetic) decreases heart rate by slowing depolarization at phase 4. It also reduces force of contraction (at muscarinic receptors), and constricts coronary arteries
* NE (sympathetic) increases heart rate, increses force of contraction (ß-receptors), and inhibits constriction of coronary arteries
What is the only electrical path from the atria to the ventricles?
The His bundle
How do the ERPs differ for atrial and ventricular myocytes?
Atrial myocytes have a short ERP, allowing them to repolarize quickly and sustain rapid arrhythmias
Why is the slow conduction through the AV node a protective mechanism?
Atrial myocytes with much shorter ERPs can sustain very rapid arrhythmias. The AV node, with a longer refractory period (only uses Ca2+ channels) will not propagate every beat during an arrhythmia.
What are the autonomic effects at the AV node?
Sympathetics (NE) speed up conduction through the AV node (+dromotropy). Parasympathetics (ACh) slow conduction through the AV node (-dromotropy)
Define chromotorpy, dromotropy, inotropy, and lusitropy
Chromotropy: automaticity (usually of the SA node).
* Dromotropy: the propagation of impulse (usually through the AV node).
* Inotropy: the strength of contraction (usually of the ventricles)
* Lusitropy: the ability to relax
What are two general mechanisms of bradycardia?
Failure to generate an impulse due to abnormal automaticity (fibrosis, ischemia, drugs). Failure to propagate an impulse from SA block, AV block, or a His bundle (infranodal)/bundle brach/purkinje fiber block
What are the normal automaticity rates for the latent pacemakers?
* Sinus node: 60-100bpm
* AV node and His bundle ("Junctional escape"): 40-60bpm
* Bundle braches, Purkinje system ("Ventricular escape"): 30-40bpm
What is an important difference between "pure" drugs that only work at ß1 and drugs that affect ß1 and a1 receptors.
The pure drug will only have positive chrono and dromo effects. The dirty drug will also stimulate a1, causing vasoconstriction (elevating BP).
What are common drugs for speeding up the heart rate?
Isoproterenol, a ß-agonist, and Atropine, a parasympathetic blocker
What coronary artery supplies the SA node? The AV node?
The right coronary artery usually supplies both the SA and AV nodes
What are the 3 primary mechanisms of tachycardia?
Abnormal automaticity, triggered activity (early afterdepolarizations - torsades, or delayed afterdepolarizations - intracellular Ca2+ overload), and Reentry
What are some causes of abnormal automaticity leading to tachycardia?
Ischemia, metabolic derangement, or ion channel abnormality that leads to phase 4 activity
What is the main cause for early afterdepolarization and what is a common result?
Phase 3 repolarization being interrupted by a depolarizing current, usually a reactivation of Ca2+ channels. This can be self-perpetuating and cause Torsades de Pointes
What condition, visible on an EKG, increases the likelihood of early afterdepolarization?
A prolonged QT interval like some drugs
What are the functional deficits found in congenital long QT syndrome and what type of arrhythmia does it predispose patients to?
Loss of K+ channel function and an increase in Na+ channel function. Prolonged QT can cause ventricular tachycardia.
What is Jervell & Lange-Nielsen syndrome and what is its key symptomatic association? Are there other forms of this syndrome?
* JL-N is a very malignant autosomal recessive syndrome resulting in a very long QT on EKG and a high risk of fatal arrhythmia. It is associated with a SENSORINEURAL deafness present at birth.
* Romano Ward & Andersen-Tawil (hypokalemic periodic paralysis) syndromes are autosomal dominant and less malignant
What are some reasons one pathway may conduct faster than another, contributing to reentry.
* Fast pathway cells may have a higher density of Na+ channels allowing for faster depolarization
* Fast pathways may be connected with more or better gap junctions
* Anisotropic conduction: conduction may be faster in one direction than another (due to myofibril alignment)
What direction is typical AV node reentry?
A slow pathway causes reentry into a fast pathway that has unidirectional block
What is the mechanism of Wolff-Parkinson-White syndrome?
These patients have a fast accessory pathway down the wall of a ventricle. A SA impulse can propagate through the AV node (slow) and reenter through the accessory pathway (fast) causing an Atrioventricular Reciprocating Tachycardia (AVRT)
What is AVNRT?
AV nodal reentry tachycardia where the impulse reenter within the AV node
Why is VT so bad following an MI?
The VT occurs because the the impulse is using mostly dead, scar tissue as slow pathways for reentry. The partial dead cardiac tissue cannot maintain necessary cardiac output.
Does the endocardium or the epicardium depolarize first? Repolarize?
The endocardium depolarizes first, the epicardium repolarizes first.
What is the general reason for a wide QRS?
Premature ventricular contraction (PVC). If the impulse originates outside the specialized conduction system, it will take longer to activate all of the ventricular cells
What is the EKG manifestation of WPW?
A delta wave and a very short PR interval
What portion of the EKG represents atrial contraction and which represents ventricular contraction?
Atria: The P wave
Ventricles: Onset of the QRS segment until the end of the T wave
Which are the inferior leads?
II, III, and aVF
Which are the lateral leads?
I and aVL
What does a deep pathologic Q wave represent?
Cell death like from an old MI
Differentiate ST elevation vs. ST depression
ST elevation represents a transmural injury. ST depression can represent subendocardial ischemia and NSTEMI or just reciprocal changes during an ST elevation
What are the numbers used to determine HR from an EKG?
300, 150, 100, 75, 60, 50
How is the 10-second rule used?
In EKG reading, to determine the HR if the rate is somewhat variable. Count the beats over the 10 second strip and multiple by 6.
What is the normal length of a PR interval and what are some things that can cause the PR to deviate?
Less than 1 big box but more than 3 little boxes. Short PR is caused by AVRT (WPW). Long PR is the result of a heart block.
What is the difference between 1˚, 2˚, and 3˚ heart block?
1˚: All P wave produce a QRS
2˚: Only one P wave at a time is blocked
3˚: No P waves are conducted
What are the locational differences between Wenckebach and Mobitz II AV block?
Wenkebach is almost always a problem within the AV node (drugs). Mobitz II is usually infranodal, a problem in the His bundle or a L/R BBB
Do Wenckebach or Mobitz II blocks respond to medications?
Wenckebach will respond to atropine, isoproterenol, or dopamine. Mobitz II needs an urgent pacemaker, rarely responds to meds
How do the QRS complexes and PR intervals compare between Wenckebach and Mobitz II block?
Wenckebach will have a normal QRS and an increasingly prolonged PR. Mobitz II will have a wide QRS (infranodal) but a constant, prolonged PR
What constitutes a wide QRS?
More than 3 small boxes
What are the 3 characteristic EKG findings for WPW?
A wide QRS, a delta wave, and a short PR interval
What is the rule of thumb for determining a long QT?
QT is prolonged if it is more than half the RR interval
What are the septal leads?
V1 and V2
What are the anterior leads?
V2, V3, and V4
What does T wave inversion on a EKG suggest?
Ischemia
Name the three layers of the heart and describe their cellular makeup
* Endocardium: Single endothelial layer and underlying connective tissue
* Myocardium: Cardiac muscle
* Epicardium: Connective tissue, blood vessels, nerve fibers, covered by mesothelial cells
What layer of cardiac tissue do the heart valves come from?
Valves are extensions of the endocardium. The fibrous sheet is formed by merging fibroelastic supporting layers
What function do atrial muscle granules serve?
Granules contained in the atrials contain a natriuretic hormone that is secreted when the atrial fibers stretch. The hormone promotes the excretion of water, K+, and Na+ by the kidneys, and inhibits renin and aldosterone secretion, all of which help lower the blood pressure
Explain the orientation of the 3 layers of blood vessels
* Intima is closest to the lumen and is oriented along the direction of blood flow
* Media is oriented circumferentially and is able to regulate contractility of the vessels
* Adventitia is also longitudinal
What are some functional roles of the endothelium?
Secrete factors that prevents clotting, maintains the vascular tone of smooth muscle, and, when cytokines are activated, express adhesion molecules and allow WBC to stick
Which layers is the dominant layer in arteries? Which dominate veins?
The tunica media is the largest in arteries, the tunica adventitia is predominant in veins
Describe transport in and out of the three different types of capillaries.
* Continuous capillaries have endothelial cell connected with tight junctions. They use pinocytotic vessels to move material.
* Fenestrated capillaries, found in the pancreas, intestines, endocrine glands, and renal glomeruli pass material through diaphragms of extracellular material.
* SInusoids are discontinuous capillaries that do not have diaphragms and or a basal lamina. These are found in the bone marrow, liver, spleen, and lymphoid tissue and allow for the most transport.
How do you differentiate small lymphatic vessels from a venule?
Lymphatics will contain diifuse eosinophilic proteins. Venules will contain RBCs and some WBCs
What are the three functions of the lymphatic system?
Collect excess ECF and return it to cardiovascular circulation, immune defense filtering blood and ECF, transport fats
What are the 4 classes of antiarrhythmics?
Class I: Na+ channel blocker (1A, 1B, 1C)
Class II: ß-Blockers
Class III: K+ channel blockers
Class IV: Ca2+ channel blockers
What are the differences of ß1 v ß2 concentration?
ß1 receptors are found in the heart. ß2 receptors are found in vascular and bronchial smooth muscle
Which class of antiarrhythmics actually improve survival in patients with heart disease?
ß-blockers are shown to improve survival in patients with heart disease
What are the tropic effects of ß-blockers?
ß-blockers are negative chronotropic, negative dromotropic (block AVNRT & AVRT), and negative inotropic
What are the 2 categories of ß-blockers and how are they different?
Cardioselective drugs block only ß1 receptors. Nonselective drugs block ß1 and ß2 receptors and some also block a-receptors so they can cause bronchospasm, PVD, or Raynaud's
Are Atenolol, Metoprolol, and Esmolol cardioselective or nonselctive ß-blockers?
Atenolol, Metoprolol, and Esmolol are cardioselective ß-blockers
Are Popranolol, Labetalol, and Carvedilol cardioselective or nonselective ß-blockers?
Propanolol, Labetalol, and Carvedilol are nonselective ß-blockers. Labetalol and Carvedilol are also a-blockers, which inhibits vasodilation.
What are some common adverse effects of ß-blockers?
Effects of them being too efficient: sinus bradycardia (negative chromotrope), AV block (negative dromotrope), can worsen acute, severe decompensated heart failure (negative inotrope)
Which class of Ca2+ channel blocker is better for antiarrhythmia and which is for antihypertension?
Nondihydropyridines (Diltiazem and Verapamil) are better for antiarrhythmias. Dihydropyridines (Nifedipine, Amlodipine, Felodipine) are better for antihypertension
What type of Ca2+ channels on what type of cells do Ca2+ channel blockers work?
Ca2+ channel blockers work on the L-type Ca2+ channel blockers mainly in the AV node, but also the SA node.
What are the tropic effects of Ca2+ channel blockers?
Ca2+ channel blockers are negative chromotropic, negative dromotropic (1˚ purpose, to slow ventricular response rates during atrial tachyarrhythmias, also can interrupt a reentry loop), and negative inotropic
What are the adverse affects of dilatiazem and verapamil?
These Ca2+ channel blockers can be too tropic. Sinus node dysfuntion or a shut down junctional escape rhythm (chromotrope), AV block (dromotrope), or systolic heart failure (inotrope). These drugs also can cause hypotension and peripheral edema
What are some other terms used to describe Digoxin?
DIgoxin is a form of digitalis and is a cardiac glycoside
What is the function of digoxin?
* Digoxin augments vagal tone (PS) and slows conduction through the AV node
* Digoxin is also mildly a positive inotrope that disrupts the Na/K pump increasing the intracellular Na+ concentration, which leads to the Na/C exchanger bringing more Ca2+ into the cell. The increase in intracellular Ca2+ enters the sarcoplasmic reticulum increasing contraction potential
What is digoxin toxicity?
Nausea, vomiting, visual disturbances, high-grade AV block with atrial tachycardia, ventricular tachyarrhythmia (from delayed afterdepolarization), renal dysfunction
In what circumstance is Adenosine indicated?
Adenosine, a natural substance with a short half-life, is given as a rapid IV push to cause AV nodal block. It also is a vasodilator.
What are possible side effects of Adenosine?
Bronchospasm, atrial fibrillation, compensatory sinus tachycardia, and hypotension
What is the mechanism of Na+ channel blockers?
They limit the Na+ channels available for conduction, raise the threshold for action potential inititation, slow the upstroke of phase 0, and decrease the slope of phase 4.
How do Na+ channel blockers affect reentry?
* They slow the conduction of action potentials between cells. They can slow the "slow pathway" down so much that it converts the fast pathway unidirectional block into a bidirectional block and no conduction propagates.
* They also can slow either pathway just enough to make an incessant reentry circuit. DO NOT give to patient's with prior MI!!
What is the general effect of Quinidine, Procainamide, and Disopyramide?
Class I Na+ channel blocker. They prolong both the QRS and the QT. They are not "clean" and have a lot of side effects (GI, CNS, lupus-like syndrome, anticholinergic)
What is the general effect of Lidocaine and Mexiletine?
Class 1B Na+ channel blockers. They shorten the QT, only work in the ventricles, and preferentially target ischemic cells. Lidocaine is IV, Mexiletine is PO
What is the general effect of Flecainide and Propafenone?
Class 1C, most potent Na+ channel blockers. They are "clean" negative inotropes (contraindicated in heart failure), prolong the QRS with no effect on the QT interval. They are use-dependent and have mild side effects (fatigue and dizziness)
Are Na+ channel blockers use-dependent or reverse use-dependent?
Use-dependent
Are K+ channel blockers use-dependent or reverse use-dependent?
Reverse use-dependent
What is the general mechanism of K+ channel blockers and what is a major risk?
K+ channel blockers prolong the QT interval and torsades (polymorphic VT) is a risk, except with Amiodarone
What are the toxicities of Amiodarone?
Amiodarone makes you "blue, blind, and cough up your liver and thyroid".
* Eyes: corneal deposits, rare optic neuropathy
* Hypo/hyperthyroidism
* Hepatic injury
* Irreversible pulmonary toxicity
* Skin photosensitivity
* Interacts with Warfarin
What types of antiarrhythmic drugs are ok, relatively contraindicated, and definitely contraindicated in patients with systolic heart failure?
Ok: ß-blockers, Amiodarone, Dofetilide, Sotalol, Dronedarone, Digoxin
Relatively contraindicated: Verapamil, Diltiazem (from negative inotropy)
Definitely contraindicated: Any Class I (Na+) antiarrhythmic unless they already have an ICD
What antiarrhythmic drugs are ok and contraindicated for use in patients with previous MI and myocardial scarring, but no systolic heart failure?
Ok: ß-blockers, Amiodarone, Dofetilide, Sotalol, Dronedarone, Digoxin, Verapamil, Diltiazem
Contraindicated: Any Class I (Na+) antiarrhythmic unless they already have an ICD
Which class of antiarrhythmic is safest for patients with a structurally normal ventricle?
Class I (Na+). Especially 1C (Flecainide and Propafenone)
What are the 3 main mechanisms of peripheral artery occlusions?
Stenosis (atherosclerosis), thrombosis (underlying plaque or stenosis), and embolism (from thrombus in the heart)
How do the 3 main mechanisms of peripheral artery occlusions differ in their final presentation?
* Stenosis leads to chronic occlusion which happens slowly, allowing for bridging, collateral arteries to develop.
* Embolism leads to acute occlusion that can result in end-organ ischemia because there is no time to develop compensatory collateral blood flow
What are some common chief complaints referring to peripheral vascular disease?
Leg pain with or without ambulation, toe ulcers or gangrene, leg swelling, recent CVA or TIA, bruits in the neck, pulsatile abdominal mass, AAA
What are some risk factors or conditions associated with vascular disease?
Atherosclerosis and its risk factors (HD, stroke, smoking, DMII), altered exercise, clotting disorders, family history
What are the hallmark signs of acute limb ischemia?
6 "P's".
Pulselessness, pain, pallor, poikilothermia, paresthesia, paresis
What types of skin changes are associated with arterial pathology?
Decreased temperature, pallor, mottling, hair loss, ulceration, gangrene
What types of skin changes are associated with venous pathology?
Edema, varicositites, dermatitis, thickened skin, ulceration, gaiter distribution
Name 5 modalities commonly used to assess peripheral vascular disease
Ankle brachial index, duplex ultrasound, CT angiography, MR angiography, digital subtraction angiography
What is the normal cut-off for a normal ABI?
>0.9
What is the gold standard modality for screening carotid occlusive disease?
Duplex ultrasonography
What is the gold standard for screening Deep Venous Thrombosis?
Duplex ultrasound
What conditions are digital subtraction angiography used to diagnose, what are its risks, benefits, and contraindications?
Artery occlusive disease. It requires an artery puncture and so it carries a risk of bleeding. Since you are physically in the artery you can do an angioplasty or place a stent.
What are the different types of claudication?
* Intermittent is an arterial problem
* Spinal is a neurologic problem
* Venous is a postphlebitic or venous insufficiency problem
* Arthritis is a varying problem where you have good days and bad days
What are the 3 symptomatic characteristics necessary for intermittent claudication?
Exertional aching or cramping, that resolves with rest, and is reproducible over a consistent distance or level of exertion
What are some diagnoses that you need to rule out for intermittent claudication?
* Atherosclerosis
* Thrombangiitis obliterans (smokers, affects distal limbs)
Popliteal artery entrapment syndrome (cyclers)
* Fibromuscular dysplasia (beads on a string)
* Cystic adventitial disease of the popliteal artery (hypolucent tunica media outpouching into lumen)
* Cocaine-abuse
* Peripheral neuropathy, lumbar stenosis, degenerative hip or spine disease
How does smoking affect the arterial wall?
* Vasoconstriction, increased ACE activity
* Increased platelet aggregability, fibrinogen levels, secretion of adhesion molecules, growth of atherosclerotic plaques
* Atherosclerotic plaque stabilization decreases
What are the methods and indications for revascularization in PAD?
The two methods of revascularization are endovascular (arterial dilation ± stent placement, atherectomy) or surgical (endarterectomy or arterial bypass). Indications are symptomatic lifestyle limiting claudication or critical leg ischemia
What is the common combination therapy to improve claudication symptoms?
Medication + exercise + smoking cessation
Describe the believed pathogenesis of atherosclerotic plaque formation.
When there is an excess of plasma LDL/VLDL it can pass through the endothelial layer where it will interact with matrix proteoglycans. Those PGs facilitate LDL oxidation and aggregation, which upregulates the expression of adhesion molecules on the endothelium for circulating monocytes, promoting monocyte movement into the matrix and differentiation into macrophages. Those macrophages ingest the ox-LDL, recruiting more macrophages, and all of these macrophages necrose and release inflammatory cytokines. This process leads to endothelial damage which is repaired, creating a plaque
How do T-cells affect plaque formation?
T-cell cytokines inhibit smooth muscle proliferation and collagen production, promoting plaque rupture and thrombosis
What is the mechanism for plaque rupture?
Macrophage foam cells migrate towards the shoulders of plaques where they secrete matrix metalloproteinases (MMP) which breaks down collagen. T-cell cytokines also inhibit smooth muscle proliferation and collagen production. These all make plaques susceptible to rupture. When a plaque ruptures it exposes TF, causing platelet aggregation and thrombosis
How does lowering plasma LDL levels alter the progression of atherosclerotic plaques?
Lowering LDL levels decreases macrophage foam cell presence in the plaque. This will decrease the inflammatory response and increase connective tissue proliferation, leading to plaque remodeling
What are the risk factors for atherosclerosis and vascular disease?
Age, gender, abnormal lipid levels, HTN, smoking, DMII, obesity, sedentary life style
How does hypertension exacerbate atherosclerosis?
* Flow/shear mediated damage to barrier function of endothelial cells
* Damage to endothelial cells due to greater stretching
What are some things that affect resistance?
* Greater viscosity (increased hematocrit)
* Longer vessels mean greater resistance
* Stenosis decreases the diameter of the vessel which greatly increases the resistance (r^4)
Name 2 things that affect turbulence and what finding they will affect.
* Increasing velocity of flow, via stenosis, or anything else that narrows vessels
* Decreasing viscosity of blood, via anemia.
* These both increase turbulence and make you more likely to hear a murmur
How do you calculate a mean arterial pressure?
MAP=Diastolic BP + 1/3 pulse pressure=Cardiac output * total peripheral resistance=(SV*HR)*TPR
How do you calculate cardiac output?
Cardiac output=Stroke volume* heart rate
What are some examples of patient presentation of symptomatic extracranial cerebrovascular disease?
TIA, amaurosis fugax, stroke, vertebrobasilar symptoms
How can someone be asymptomaticlly diagnosed with extracranial cerebrovascular disease?
Imaging findings (screening or incidental) or physical exam findings (bruit or Hollenhorst plaque)
What is the temporal difference between a TIA and a stroke?
Transient Ischemic Attack (TIA) is a focal neuro deficit that lasts less than 24 hours. A stroke (CVA) is a focal neuro deficit that last greater than 24 hours, representing neuronal cell death
What is amaurosis fugax and what is its mechanism?
Amaurosis fugax is temporary ipsilateral monocular visual loss and is the result of an embolization of the retinal artery or its branches (Hollenhorst plaques)
What are the two etiologies of stroke and which is most common?
Hemorrhage (aneurysm, trauma, anticoagulation, congenital abnormality) and Ischemia. Ischemia is the etiology for 80% of strokes
What are some risk factors for ischemic stroke?
Hypertension, increasing age, CVD (A.fib or extracranial cerebrovascular disease), smoking
What are two ways extracranial cerebrovascular disease can cause a stroke?
Embolism (most common) or a thrombosis/low flow state
How are statins believed to affect atherosclerotic plaques?
HMG-CoA (statins) are associated with fibrous remodeling and plaques stabilization
What are some non-atherosclerotic causes of extracranial cerebrovascular disease?
Arteritis (Takayasu, Giant cell), Dissection (Traumatic of spontaneous), Fibromuscular dysplasia, radiation
What is the etiology, symptoms, and treatment of giant cell arteritis?
Is most common in older caucasian women. It can lead to temporal headaches, jaw claudication, polymyalgia rheumatica, amaurosis, constitutional symptoms, elevated sedimentation rate. It is an infection treated with corticosteroids
What are the 2 mechanisms of carotid artery dissections and what is their common treatment?
Sponateous (cocaine/meth use, Horner's syndrome) or Traumatic compression injury occurring high in the neck (C2). They are treated with anitcoagulation treatment
What are the indications for procedural intervention in extracranial cerebrovascular disease?
Based on symptoms, degree of stenosis, patient's anatomy
What symptoms are commonly associated with vertebrobasilar disease?
"Posterior" Symptoms: dizziness, vertigo, diplopia, blurred vision, ataxia, drop attacks, bilateral sensory disturbances
What is the presentation of vertebrobasilar disease?
Defined precipitating factors: positional changes such as head turning or rising from sitting or standing, usage of upper extremities
What are 2 circulatory results of bilateral renal artery stenosis?
CHF and peripheral edema
Where are ATII-1 receptors distributed, what are some of their regulators, and what are their effects?
Type 1 Angiotensin II receptors are located in the kidney, vascualr wall, adrenals, heart, brain, lung, and liver. They are G-protein coupled receptors activated by angiotensin, LDL, Insulin, Estrogen, progesterone, and IFN-g. AT1R activation is vasoconstrictive, pro-inflammatory, and promotes volume expansion and sodium retention
What is the mainstay of treatment for renal artery stenosis
ACE inhibitors or ARBs. They improve survival, reduce cardiovascular events, and have renoprotective effects for diabetic patients
What are the 3 main indications for HMG-CoA inhibitors?
Statins. Vascular disease (CVD, CHD, MI), Chronic kidney disease (declining renal function), and Atherosclerotic renal artery stenosis
What is the common patient presentation for acute arterial mesenteric ischemia?
>60yo in severe acute abdominal pain and often nausea and diarrhea
What is the common patient presentation for chronic arterial mesenteric ischemia?
40-70yf in abdominal pain, often post-prandial (10-30 min) resulting in weight loss and food fear
What is the common patient presentation for acute venous mesenteric thrombosis?
Insidious onset, pain, nausea, vomiting, abdominal distension, fever, slow progression of symptoms
What is the common patient presentation for chronic venous mesenteric ischemia?
Portal hypertension that is usually only found as an incidental CT finding
What are risk factors of mesenteric venous thrombosis?
Hypercoagulable states, OCPs, neoplasm, IBD, smoking
What are the methods of treatment for acute and chronic mesenteric arterial and venous ischemia?
Acute arterial relies on open surgical management. Chronic arterial is either surgical revascularization or an endovascular procedure. Venous ischemia is treated by anticoagulation.
What are potential complications of infectious endocarditis?
Tamponade, CHF, rhythmic disturbances, heart block
What is the current epidemiology for infective endocarditis?
Older patients (~50yo) with predisposing cardiac lesions. IV drug use, indwelling catheters, & HA bacteremia are prevalent risk factors in developed countries.
What 2 events are necessary for infective endocarditis?
The formation of a nonbacterial thrombus (trauma, perisurgery, metabolic changes) followed by a transient bacteremia with an organism that can adhere to a platelet-fibrin matrix (usually a GPC)
What are the two most common pathogens responsible for infective endocarditis?
Staph aureus and Strep viridians
What are the most common symptoms and physical findings of infectious endocarditis?
Fever, heart murmur (new or changing regurgitant heart murmur), embolic phenomenon (skin, viscera, organs, brain), skin manifestations (Osler nodes, splinter hemorrhages, petechiae)
What are some major complications of infective endocarditis?
CHF (valvular destruction/regurgitation), systemic emboli, mycotic aneurysm, renal disease
What is the name of the criteria necessary for a definitive diagnosis of infectious endocarditis and what are the criteria?
The Duke criteria require appropriate microbiology (GPC 2+ times) and evidence of endocardial involvement (Valvular vegetation or a new regurgitant murmur via echo)
What is the normal treatment for infectious endocarditis and when is a higher level of intervention indicated?
Bactericidal antibiotics often in combination and for a prolonged period to achieve optimal killing. Surgery, such as a valve replacement is indicated with defined complications (HF, multiple emboli, persistent bacteremia, fungal infection, MDR infection, heart block)
What are the differences in common pathogens in early v. late prosthetic valve endocarditis?
Early (<2 months): Staph, GNR, fungi
Late (>2 months): Looks like native...S.aureus, coag (-) staph, strep
How does prosthetic valve endocarditis compare to native valve infectious endocarditis?
Prosthetic valve endocarditis is associated with a more varied microbiology, frequent metastatic complications, and greater need for surgical intervention, and a poorer outcome
What kind of patient is a candidate for endocarditis prophylaxis?
Patients with cardiac lesions that are at risk risk (prosthetic valve, prior IE) that are peridental procedures
What are common signs of infectious pericarditis?
Fever, chest pain, pericardial friction rub (sign of tamponade &/or constriction
What type of pathogen is most implicated in myocarditis and what are is signs?
Viral (enterovirus). Fever and signs of CHF, often in young males with new onset unexplained cardiac abnormalities and a recent URI or enteritis
What generates ventricular pressure during diastole? Systole?
During diastole, ventricular pressure is generated from the blood volume filling and stretching the ventricles. In systole, the pressure is from the ventricles contracting and ejecting its load
How does the total peripheral resistance react to a hemorrhage?
A hemorrhage greatly decreases the venous return to the heart, the body will react to this by vasoconstricting and increasing the TPR
How do cardiac output and venous return react to an increase in total peripheral resistance?
An increase in arterial pressure will increase the afterload, decreasing cardiac output and venous return
How does the total peripheral resistance react to exercise?
An increase in catecholamines increases contractility. Muscular arterioles dilate, decreasing the TPR, but allowing for greater cardiac output and venous return
What is preload?
Preload is the volume of blood and thereby pressure that fills and stretches the left ventricle during diastole
What is afterload?
Afterload is the pressure required to be generated by the left ventricle to overcome the mean arterial pressure and force the aortic valve to open, allowing for the ejection of blood
How do changing the preload or afterload affect the ESPVR and the EDPVR?
They do not. Those two relationships and the lines that describe them remain the same. The intrinsic properties of the heart are not changing.
How does changing the inotropy of the heart affect the ESPVR and the EDPVR?
A change in inotropy will affect ESPVR but keep the EDPVR the same
How does changing the lusitropy of the heart affect the ESPVR and the EDPVR?
A change in lusitropy will affect the EDPVR but keep the ESPVR the same
When does a higher HR not equal a higher cardiac output?
If the HR is going too fast, there will not be as much preload, decreasing the stroke volume and decreasing the cardiac output
What is the LVEF, how is it calculated, and what are clinically normal values?
The Left Ventricular Ejection Fraction is the fraction of blood actually pumped out during systole. LVEF=Stroke volume/End diastolic volume (~55-70%)
What are normal chamber pressures for both atria, both ventricles, the pulmonary artery, the pulmonary capillary wedge, and the aorta?
RA=0-8
RV=15-30/0-8
Pulmonary Artery=15-30/4-12
Pulmonary capillary wedge= 1-10
LA=1-10
LV=100-140/3-12
Aorta=100-140/60-90
What is the equation for cardiac output and where are each of the measures taken?
Cardiac output=rate of O2 consumption/(arterial O2 content - venous O2 content)
Arterial O2 content is measured from a peripheral artery. Venous O2 content is measured from the pulmonary artery
What are 3 major factors that affect mycardial O2 demand?
* Contractility: If the heart is squeezing harder it is doing more work and needs more O2
* Heart Rate: If the heart is beating faster it is doing more work and needs more O2
* Ventricular wall stress: More wall stress increases its need for O2
What are the 3 general principles of the Law of Laplace?
For any container wall stress is proportional to ventricular pressure and ventricular radius and inversely proportional to wall thickness
What is the general mechanism for nitroglycerine use for angina?
Nitroglycerin decreased preload, decreasing wall stress, decreasing myocardial O2 demand
What is the heart's physical response to chronic pressure overload?
Concentric hypertrophy. An increase in wall thickness but not in chamber size
What is the heart's physical response to chronic volume overload?
Eccentric hypertrophy. An increase in wall thickness and chamber size leading to the chamber dilating
What are some behavioral risk factors for hypertension?
Excessive alcohol consumption, sodium consumption, obesity/dietary excess, lack of aerobic exercise
What is the rule of 2?
* Stage 2 hypertension gets treated with 2 antihypertensive drugs (thiazide + another class)
* An increase of 20/10 will 2x your risk of CVD
What are some results of hypotension?
Shock, kidney failure, anoxic encephalopathy, lactic acidosis
What are some results of hypertension?
Intracranial hemorrhage, stroke, vascular disease, kidney damage, hypertensive heart disease, retinopathy
What are the systolic and diastolic cutoffs of normal BP? What treatment is indicated?
<120/<80. Treat with drugs for compelling indications
What are the systolic and diastolic cutoffs of prehypertension? What treatment is indicated?
120-139/80-89. Treat with drugs for compelling indications
What are the systolic and diastolic cutoffs of Stage 1 hypertension? What treatment is indicated?
140-159/90-99. Thiazide diuretic. Other drugs for compelling indications.
What are the systolic and diastolic cutoffs of Stage 2 hypertension? What treatment is indicated?
≥160/≥100. Stage 2=2 drugs. Thiazide diuretic+ ACEI, ARB, BB, or CCB.
Differentiate the function of Angiotensin II type 1 and type 2 receptors.
AT1 Receptors promote vasoconstriction, cell growth and proliferation, hypertrophy, aldosterone release, sympathetic activation, Na and H2O retention, and inhibit renin release. AT2 receptors cause vasodilation, promote excretion, and limit hypertrophy
Describe the epidemiology, presentation, and treatment of pheochromocytoma.
Pheochromocytoma is a chromaffin tumor that secretes catecholamines. Young - middle aged females that present with intermittent sympathomimetic symptoms (HTN, palpitation, sweating, anxiety). Must be treated 1 wk prior to surgical removal with Phenoxybenzamine, an a-blocker, to inhibit catecholamine secretion during surgery
Why is a coarctation of the aorta sometimes difficult to find?
The intercostal arteries can dilate and compensate for less flow through the abdominal aorta. A younger person can come in a have upper extremity hypertension and have normal blood pressure with diminished lower extremity pulses. Rib notching can also be seen on CXR.
What type of drugs are hydrochlorothiazide, chlorothiazide, metolazone, and indapamide, what is their mechanism, and what are their common side effects?
They are thiazide-type diuretics that act by inhibiting Na+ reabsorbtion at the distal tubule. Their side effects are decreased metabolites (K+, Mg, Na+), hyperglycemia, gout, dehydration, and rash
What type of drugs are spironolactone, eplerenone, triamterene, and amiloride, what is their mechanism, and what are their common side effects?
They are K+ sparing diuretics and they inhibit K+/Na+ exchange at distal renal tubules. There common side effect is hyperkalemia, spironoLACtone can cause gynecomastia, and triamterene and amiloride can cause a rash.
What type of drugs are furosemide, bumetanide, torsemide, and ethacrynic acid, what is their mechanism, and what are their common side effects?
They are loop type diuretics and the inhibit NaCl reabsorption at the ascending Loop of Henle. Their side effects are decreased metabolites (K+, Mg, Na+), hyperglycemia, gout, dehydration, and rash (except for ethacrynic acid because it isn't made with sulfur
In what population are diuretics good and where should you use caution?
Diuretics work well in the african american population and caution must be exercised when administering to the elderly
What type of drugs are captopril, enalapril, lisinopril, and ramipril, what is their mechanism, and what are their common side effects?
* They are ACE inhibitors, inhibiting vasoconstriction, the release of aldosterone and vasopressin, and the decrease sympathetic stimulation.
* They also inhibit the breakdown of bradykinin into inactive peptides, causing a backup of bradykinin which can bind to BK-II receptors and release NO, cause vasodilation, and produce the side effect of a dry cough.
* Side effects: Hyperkalemia, increased serum creatinine, dry cough, angioedema
What type of drugs are losartan, valsartan, and olmesartan, what is their mechanism, and what are their common side effects?
* They are Angiotensin II receptor blockers, inhibiting vasoconstriction, the release of aldosterone and vasopressin, and the decrease sympathetic stimulation.
* Side effects: Hyperkalemia, increased serum creatinine, angioedema
What is important to know about Enalapril?
It is metabolized into enalaprilat which is also an active agent and can be given IV
What is important to know about lisinopril?
It is indicated as an antihypertensive and for CHF
What is important to know about ramipril?
It is indicated as an antihypertensive and can prevent HD by increasing blood flow
What is aliskiren indicated for and what is its mechanism?
Aliskiren is an antihypertensive and works by inhibiting renin's catalytic ability to activate angiotensinogen
What are contraindications for ACEI, ARBs and renin inhibitors?
Bilateral renal artery stenosis, pregnancy, angioedema to other ACEIs, hyperkalemia
What are some common side effects of ß-blockers used as antihypertensives?
Drowsiness, lethargy, confusion, bronchoreactive events, AV nodal blockade
What kind of drugs are atenolol, metoprolol, carvedilol, and labetalol, what is there mechanism, and what are their side effects?
* They are ß-blockers that can be used as antihypertensives
* Atenolol and metoprolol are cardiosensitive (ß1), labetalol and carvedilol are nonselective (a, ß1, ß2)
* Side effects: Drowsiness, lethargy, confusion, bronchoreactive events, AV nodal blockade
Are nondihydropyridines or dihydropyridines indicated for hypertension? Arrhythmia?
Nondihydropyridines (dilatiazem, verapamil) are used as antiarrhythmics. Dihydropyridines (Nifedipine, Nicadipine, felodipine, amlodipine) are used as antihypertensives
What types of antiarrhythmics are indicated for the compelling indication of heart failure?
ß-blockers, ACEIs, and ARBS
What types of antiarrhythmics are indicated for the compelling indication of post an MI?
ß-blockers, ACEIs, and aldosterone antagonists (ARBs)
What types of antiarrhythmics are indicated for the compelling indication of high coronary artery risk?
ß-blockers, ACEIs
What types of antiarrhythmics are indicated for the compelling indication of diabetes?
ACEIs, ARBs
What types of antiarrhythmics are indicated for the compelling indication of chronic kidney disease?
ACEIs, ARBs
What types of antiarrhythmics are indicated for the compelling indication of recurrent stroke prevention?
Diuretics, ACEIs
Which 2 valves are most commonly affected in Rheumatic Fever?
Mitral valve followed by aortic valve
What are the Jones criteria used to diagnose and what are they (11)
* Criteria used to diagnose Rheumatic Fever
* Polyarthritis, carditis, subcutaneous nodules, erythema marginatum, chorea, fever, arthralgia, increased ESR, Leukocytosis, heart block on ECG, hx of RF
What is the most common cause of mitral stenosis?
Rheumatic fever
What physiologic affects does mitral stenosis cause?
* Mitral stenosis causes elevated LA pressures, resulting in elevated pulmonary and right heart pressures
* Chronic pressure overload LA can cause A.fib which can worsen patient's symptoms
What are the physical exam findings with mitral stenosis?
* Loud S1 in early stages (pressure difference held longer), soft S1 in late stages
* Opening snap (after S2): the "sooner the worser"
* Diastolic rumble (longer duration, worse stenosis)
What are the treatments for mitral stenosis?
* Diuretics to reduce pulmonary congestion
* If A.fib occurs: control ventricular rate & start anticoagulant
* Percutaneous balloon mitral valvuloplasty is available
What is the most common etiology of mitral regurgitation?
Idiopathic chordae tendinae rupture
What is the result of acute mitral regurgitation?
Severe pulmonary edema. The LA is unprepared to deal with the extra blood flow and backs up into the pulmonary system
What are the physiologic changes of chronic mitral regurgitation?
* LA dilation minimizes pulmonary edema except in cases of transient decompensation (high salt intake, uncontrolled HTN)
* LA dilation can lead to A.fib
* LV is under chronic volume overload (normal blood plus regurgitated blood) and undergoes eccentric hypertrophy.
What is the primary symptom found in mitral regurgitation?
Fatigue, caused by decreased cardiac output (SV remains high, but is going in two directions)
How can mitral regurgitation sneak up on you?
LV eccentrically hypertrophys because it is chronically volume overloaded. This dilation leads to a decrease in inotropy. The afterload is also decreased though, due to blood exiting the LV in two directions, allowing the heart to compensate and keep the stroke volume artificially high. Cardiac output will be decreased.
When is surgery indicated for mitral regurgitation?
Severe symptomatic mitral regurgitation
What in the non-surgical treatment for mitral regurgitation?
* Medical therapy doesn't affect MR progression
* Diuretics to reduce pulmonary congestion
* Vasodilators to encourage more forward blood flow
What are the physical exam findings in mitral regurgitation?
* Holosystolic murmur, commonly with an S3
What are the gross and physical exam findings of mitral valve prolapse?
* A midsystolic click
* Thickened myxomatous connective tissue replaces dense collagen of the valve
What are the 2 most common etiologies of aortic stenosis and what age groups are affected by each?
* 40-60 yo present with atrial stenosis from a congenital bicuspid aortic valve
* >65 yo presents with degenerative aortic stenosis (similar changes to atherosclerosis)
What are the physiological effects of aortic stenosis?
Chronic pressure overload in the LV leads to concentric hypertrophy and decreased lusitropy. This decreases LV filling during diastole leading to increased importance on atrial contraction and the development of A.fib and a decreased cardiac output
What are the physical exam findings of aortic stenosis?
* A crescendo-decrescendo systolic ejection murmur (the stenosed valve maintains the pressure gradient between the LV and aorta longer)
* Pulsus parvus et tardus (weak and delayed pulse)
When is surgery indicated for aortic stenosis?
Once CHF, angina, or syncope develop
How do the afterload and stroke volume compare before and after surgery in mitral regurgitation and aortic stenosis?
* Before surgery: AS will have a high afterload and lower SV. MR will have a low afterload (blood flows into aorta AND back into LA) which artificially maintains a high SV
* After surgery: SV for AS will be drastically improved because of the decreased afterload. In MR the dilated LV now has a much higher afterload and the SV will drop sharply
What are the medical managements for aortic stenosis?
* No treatment will actually slow the progression of the pathology
* Statins may help reduce risk factors
* Diuretics may help (decreases TPR) especially with coexisting MR
* Vasodilators if hypertensive to decrease afterload. Be careful to NOT induce hypotension
What is the definition of an aneurysm and what are the two classifications?
A focal dilation of an artery, increasing the diameter at least 50%. True aneurysms involve all layers of the vessel wall while false aneurysms have none of the vessel layers
What are the components of vessel walls that are involved in AAA formation and how are they involved?
Elastin, matrix metalloproteinases, and collagen. Elastin is not produced in adults. Elastin concentrations decrease while matrix metalloproteinases increase degradation as we age. This elastin degradation (especially infrarenally) decreases vessel elasticity and integrity
What population most commonly is affected by AAA, what are common risk factors, and which one is the biggest risk factor?
* Elderly white men are 6 times more likely to get AAA.
* Risk factors: Smoking (biggest risk), family history, hypertension, white race
What size is necessary for surgery to be indicated in AAA?
5.5 cm for most patients, excluding petite women
What is the most common presentation for AAA?
Most patients with AAA are asymptomatic
What is the best screening/diagnostic modality for AAA?
Ultrasound
What are some symptoms associated with AAA at its different stages?
Acute expansion manifests as pain. An AAA rupture will present as abdominal or back pain, hypotension, syncope, abdominal distension, shock, and/or sudden death
What physics is responsible for AAA rupture?
The law of Laplace states that an increase in radius increases wall stress. Eventually, the wall stress will overcome the decreased wall integrity caused by decreased elastin, and the vessel wall will rupture
What AAA risk factors are implicated in increasing risk of AAA rupture?
Smoking and COPD are independent risk factors for increased rate of rupture
What treatments are indicated for AAA?
No medical therapy is proven to reduce or slow aneurysm growth
What are the 2 common surgical treatments for AAA?
Endovascular repair or open surgery
What is the pathophysiology of an aortic dissection?
Blood enters the aortic wall through an intimal tear creating a normal (true) lumen and an abnormal (false) lumen through which blood flows
What is the best treatment for AAA?
Reducing risk factors: HTN, DMII, smoking
What is the key histological finding in an aortic dissection?
Cystic medial necrosis
What are the 2 classifications of aortic dissection, what is their presentation,
* Stanford type A: Ascending aorta, proximal to the trunk
* Standford type B: Descending aorta, most often at the subclavian
* They both present with sharp, ripping, tearing pain
What congenital syndromes are associated with aortic dissection?
Connective tissue diseases: Marfan's, Turner's, Ehler-Danlos, Coarctations
What modality is best for picking up an aortic dissection?
Aortic dissection is best diagnosed with a CT scan. Second best would be an ultrasound revealing a widened mediastinum
What is the treatment for an aortic dissection?
* Immediately lower blood pressure. ß-blocker to reduce the HR and a vasodilator (nitroprusside or labetalol) to reduce afterload.
* Control pain
* Stanford type A is always a surgical emergency. Repair dissection, preserve coronary and valve function
Where are the most common spots for peripheral aneurysms and what other vascular problem are they commonly associated with?
The femoral and popliteal arteries are the most common peripheral aneurysms. They are usually bilateral and are STRONGLY associated with AAA. Screen for AAA
What is the most common site for visceral aneurysm and what medical condition puts patients at an increased risk for rupture?
The splenic (followed by renal) artery is the most common site of visceral aneurysm and pregnancy increases the risk of rupture, so surgery is indicted at a much smaller aneurysmal size
What is the difference between signs and symptoms?
* Signs are objective features of illness detected by a practitioner
* Symptoms are subjective features of an illness as related by the patient
What are the epidemiological differences between DHF and SHF?
DHF patients are older women and SHF patients are usually middle-aged men
What are the 4 characteristics that can be manipulated to regulate cardiac output?
Automaticity regulates the heart rate. Contractility, preload, and afterload all affect stroke volume
How does the heart immediately respond to an acute myocardial injury? How does it respond over time?
* Initially, the ejection fraction will decrease because the stroke volume decreases, but the heart will reflexively increase the HR to compensate for the decreased SV.
* After a period of decreased ejection fraction, the ventricle will dilate, increasing the preload to increase the stroke volume
How do sympathetic and parasympathetic drugs affect automaticity?
* Sympathetics bind ß-receptors that increase Na+ and Ca2+ flow into the cell, increasing the rate of depolarization, increasing the heart rate
* Parasympathetics bind muscarinic receptors, which increase the influx of K+ and block Ca2+ influx, hyperpolarizing the cell and decreasing the rate of depolarization and slowing the HR
What are some situations that affect preload?
Inspiration, exercise, aortic regurgitation, and a systemic shunt all increase preload. Valsalva, dehydration, A. fib, and some medications can decrease preload
What are the 3 hemodynamic defense systems, what organs do they regulate, what cardiac characteristics can they affect, and what would chronic upregulation result in?
* Sympathetic: Nor/Epi binds ß- and a-receptors in the cardiovascular system and adrenal glands. It increases automaticity, afterload, and preload. Chronic stimulation would lead to myocyte death and arrhythmia
* Renin-Angiotensin: Angiotensin II binds its receptors in the kidneys, lungs, and blood vessels and can increase afterload and preload. An increased stimulation would lead to myocyte hypertrophy and ischemia
* Aldosterone: Aldosterone binds mineralcorticoid receptors in the kidney and heart and increases afterload and preload. A chronic increase will cause cardiac fibrosis
What are the stages and classes of CHF?
Stage A: High risk but no structural changes
Stage B: Structural changes without syndrome
Stage C: Syndrom controlled with therapy
Stage D: Syndrome uncontrolled despite therapy
Class I: No activity limitation
Class II: Mild limitation that improves by slowing down
Class II: Marked limitation improved by rest
Class III: Sever limitation or symptoms even at rest
How is exercise handled differently by a healthy heart and a CHF heart?
Normally, with exercise the vessels dilate to try and get more blood to the tissues and the heart increases its stroke volume to compensate and meet the body demands. In HF, the heart is unable to increase the stroke volume enough to meet the body's needs
What is the impact of diastolic dysfunction?
With high LV pressure the LA pressure increases. This backs blood up and causes pulmonary congestion and shortness of breath with activity
What is the approach to acute therapy of CHF?
* Reduce congestion: reduce preload (diuretic) and afterload (vasodilator)
* Improve performance (inotrope)
* Provide vital organ support as needed
What are some signs and symptoms of cardiovascular congestion and what general therapy is used to treat it?
* Orthopnea, paroxysmal nocturnal dyspnea, S3, rales, JVD, hepatomegaly, edema
* Reduce fluids: Diuretics (Furosemide) or vasodilators (Nitroglycerine)
What are some signs and symptoms of low perfusion?
* Cool extremities, hypotension with and ACEi, sleepy, obtunded, low serum sodium
* Increase contractility: Inotropes (Dobutamine, milrinone)
What is the medical therapy for CHF?
* ACEi to block renin vasoconstriction and fluid retention
* ß-blocker to block sympathetic increase in automaticity, kidney fluid retention, and vessel vasoconstriction
What is the most important risk factor for sudden cardiac death?
Left ventricular ejection fraction
What are the different vascular responses to injury in the different vessel layers?
* Intima: thrombi, thickening and spasm, causing pain
* Media: aneurysm and rupture, leading to hypotension and pain
* Adventitia: weakening, false aneurysms causing pain
What are the different vascular responses to injury by different sizes of vessels?
Large: aneurysm and rupture
Medium: thickening, stenosis, thrombosis, causing ischemia
Small: narrowing causing hypertension
Capillaries: petechial hemorrhage, microthrombi, narrowing
What is the common site of atheroma?
Atheromas usually develop in the intimal layer of arteries (aorta and it's major branches). Atheromas form at sites of low shear force, opposite branch sites
What are complications of arteritis?
Inflammation from the arteritis causes:
* Weakened vessel walls leading to aneurysm or rupture
* Narrow lumen causing ischemia
* Damage to the endothelium and leading to thrombosis
What is the epidemiology, target vessel, presentation, and results of Kawasaki's disease?
Muco-cutaneous lymph node syndrome causes arteritis in children less than 4 years old and it's cardiac symptoms can present years after the infection. It causes fever, RASH, conjunctival or oral erythema and erosion, erythema of palms/soles, and enlarged cervical lymph nodes. It targets coronary arteries and can lead to aneurysm, AMI, or sudden death.
What is the epidemiology, pathophysiology, and presentation of Takayasu arteritis?
Takayasu arteritis thickens the aortic arch of women under 40. It leads to decreased pulses in the upper extremities, cold or numb fingers, occular disturbances, HTN, and neurological defects. Microscopically it looks like giant cell arteritis.
What is the epidemiology, pathophysiology, results, and presentation of Thrombangiitis obliterans?
Buerger disease affects young adult males less than 35. It almost exclusively affects tobacco users and the effects resolve following cessation. Inflammation and thrombosis target small & medium sized arteries and lead to fibrosis around nerves and lymphatics. This results in pain, ischemia, and gangrene in the legs and arms
Where is the most common site of AAA and what is the physiological reason for this distribution?
AAAs are most commonly in the infrarenal aortic section. This is because elastin is the tissue responsible for restricting AAA development and there is a 58% decrease from suprarenal aorta to infrarenal aorta
Aortic aneurysm secondary to what infectious disease is describe as "tree-barking"?
Syphilis causes inflammatory destruction of the thoracic aorta. It also causes chronic inflammation around the vaso vasorum
What is the pathophysiology of mycotic aneurysm?
An infection causes destruction to the media, resulting in aneurysm
In what vessel pathology is the degradation of elastin important?
Vessel aneurysms
What are the 3 syndromes on the spectrum of acute coronary syndrome?
Unstable angina, non-STEMI, and STEMI
How do an unbalanced myocardial oxygen supply and demand lead to angina?
If the oxygen demand (heart rate and wall force) is greater than the oxygen supply (arterial PO2, Hb, coronary flow, microciirculation, O2 extraction) it leads to ischemia and pain
What classes of drugs increase coronary flow, increasing myocardial oxygen supply, lessing angina?
Nitroglycerin and dihydropyridines
What classes of drugs prevent thrombus formation, increasing myocardial oxygen supply, lessing angina?
Aspirin and thienopyridines (clopdogrel)
What classes of drugs decrease ventricular wall force, decreasing myocardial oxygen demand, lessing angina?
ß-blockers and ACEi/ARBs
What classes of drugs decrease heart rate, decreasing myocardial oxygen demand, lessing angina?
ß-blockers and Nondihydropridines
What classes of drugs decrease preload, decreasing myocardial oxygen demand, lessing angina?
Nitroglycerins
Name 3 cardioselective and 3 nonselective ß-blockers and explain how they are different
* Cardioselective: Atenolol, Metoprolol, Esmolol
* Nonselective: Labetalol, Carvedilol, Propanolol.
* Nonselectives bind ß2 receptors in the lungs causing bronchospasm
What are contraindications for ß-blockers used to treat angina?
Hypotension, bradycardia, decompensated HF, and active bronchoconstriction
What is the mechanism of nitroglycerin in the treatment of angina?
Nitroglycerin increases intracellular cGMP causing venodilation (decreasing preload) and vasodilation (increasing myocardial oxygen supply)
What are the 3 nitrates, what is an important administrating characteristic to note, and when are nitrates contraindicated?
Nitroglycerine, isosorbide mononitrate, and isosorbide dinitrate. There are enzymes necessary to activate the nitrate and a "nitrate tolerance" can develop when those enzymes are depleted. Patients should remove treatment for 8-10 hours every day to allow for enzyme repletion. Nitrates are contraindicated with ED drugs which are also vasodilators, with RV infarcts, and critical aortic valve stenosis
Name some dihydropyridines. What is their mechanism, when do they have compelling indications, and what physiologic reflex can occur with admission?
* Nifedipine, Niccardipine, Idradipine, Nisolodipine, Felodipine, Amlodipine
* They inhibit Ca2+ transport across the membrane
* They have compelling indications with chronic kidney disease
* They will elicit a reflex tachycardia and greatly decreased vascular resistance
Are nitrates used for chronic angina treatment or for breakthrough pain?
Breakthrough pain from their chronic ß-blocker treatment
What is Prinzmetal's angina and how is it treated?
Prinzmetal's angina is a vasospastic angina not associated with thrombus formation and is sometimes associated with atherosclerosis. DO NOT GIVE ß-BLOCKERS, they will trigger bronchospasm and make things worse. Give long-acting nitrates or Ca2+ channel blockers (Diltiazem, verapamil, amlodipine, felodipine)
What anti-anginal drugs are preferred with a HTN comorbidity?
ß-blockers and Ca2+ channel blockers
What anti-anginal drugs are preferred with a migraine comorbidity?
ß-blockers and Ca2+ channel blockers
What anti-anginal drugs are preferred with an asthma comorbidity?
Nondihydropyridines. NO ß-blockers
What anti-anginal drugs are preferred with a hyperthyrodism comorbidity?
ß-blockers
What anti-anginal drugs are preferred with a diabetes comorbidity?
ß-blockers cautiously and Ca2+ channel blockers
What anti-anginal drugs are preferred with a SHF comorbidity?
ß-blockers, nitrates, maybe add-on a dihydropyridine (amlodipine or felodipine). Do NOT give nondihydropyridines
What anti-anginal drugs are preferred with a DHF comorbidity?
ß-blocker or nondihydropyridine. Do not give dihydropyridines
What anti-anginal drugs are preferred with an A.fib comorbidity?
ß-blocker or nondihydropyridine
What anti-anginal drugs are preferred with a bradyarrythmia comorbidity?
DIhydropyridines. Do NOT give ß-blockers or nondihydropyridines
What anti-anginal drugs are preferred with a supraventricular arrythmia comorbidity?
ß-blockers or nondihydropyridines
What anti-anginal drugs are preferred with a ventricular arrythmia comorbidity?
ß-blockers
Name some thienopyridines and explain their mechanism of action
Thienopyridines (clopidogrel, prasugrel, ticagrelor[reversible]) irreversibly bind P2Y12 receptors on platelets, inhibiting platelet aggregation
What is aspirin's mechanism of action?
Aspirin irreversibly blocks cyclooxygenase, blocking arachadonic acid conversion into Thromboxane A2, inhibiting platelet aggregation
What is the mechanism of action for GpIIb/IIIa inhibitors?
GpIIb/IIIa inhibitors displace fibrinogen in thrombi and prevent further cross-linking and platelet activation
What is a common side effect of oral anti-platelets that is unrelated to bleeding?
Rash
What are the treatment recommendations for chronic stable angina?
* Aspirin (or clopidogrel if ASA intolerant)
* ß-blocker (metoprolol, carvedilol) to vasodilate and slow HR
* Consider adding a long-acting nitrate, long-acting Ca2+ channel blockers, or an ACEi (Lisinopril: LV dysfunction benefit, decreases diabetes comorbidities)
* Manage lipids with a statin
What is the prominent wall layer in venous vessels?
Tunica adventitia
What is responsible for forcing blood out and through the venous veins?
Calf muscles contraction increases the compartment pressure and pushes blood along the venous system
What are some causes of increased hydrostatic pressure in the venous system?
* Venous valve failure allows blood to flow from deep veins back into superficial veins and increases column height which increases static pressure.
* Obstruction causes distal distention which induces valve failure
What are some effects of high venous pressure?
* Venous flow holds valve leaflets apart longer adding to value failure
* Fluid backs up into capillary beds increasing edema, extravasation of proteins, and subsequent inflammation which induces wall and valve structural changes
Dysfunction at which venous valves contribute to varicose veins, what are the symptoms and treatment of varicose veins?
* The saphenofemoral and saphenopopliteal junctions and the perforators are failing
* Varicose veins present as a pain, aching sensation that is better in the morning and worsens as the day progresses
* Treat with compression stockings, elevation, and treat source of reflux
What skin changes are associated with venous stasis?
GAITER distribution. Lipodermatosclerosis, hemosiderin deposition causing pigmentation, eczema, and ulcerations at the site of perforators
What is the management of venous stasis ulceration?
Aggressive compression, elevation, antibiotics and debridement if infected or necrotic, skin grafts
What are some causes of venous stasis?
Immobility on auto or plane trips, valvular dysfunction, pregnancy (pelvic vein compression)
What modality is used to diagnose DVT?
Venous duplex
What is phlegmasia cerulea dolens?
A limb threatening, surgical emergency of extensive venous thrombosis often associated with cancer
What are the most common causes of secondary lymphedema in the developing v industrialized world?
* Developing: Wuscheria bancrofti (filariasis)
* Industrialized: Malignancy and associated treatments
What is the flow of fetal circulation and how does it changes postpartum?
* Oxygenated blood flows into the RA via the umbilical vein and the IVC. From the RA it flows through the foramen ovale and into the LA, down into the LV, and out the aorta. Less oxygenated blood flows from the RA to the RV, out the pulmonary valve and through the fetal pulmonary system.
* After birth, the umbilical vein is gone and the foramen ovale closes, producing a normal blood flow pattern
How do you determine respiratory distress in infants?
Follow up on feeding problems or changes
What is the most common congenital heart defect, what is its pathophysiology, common presentation, and surgical indications?
* Ventricular septal defects are the most common congenital heart defect.
* The hole in the ventricular septum causes a left to right shunt.
* Children will present as not thriving, a normal sized heart, and a holosystolic murmur of varying loudness & pitch.
* Surgery is indicated if there is a huge hole with uncontrollable CHF or pulmonary HTN.
What cardiac defect causes a fixed widely split S2?
Atrial septal defects because chronic right-sided volume overload decreases resistance in the pulmonary vascular system and this delays the pulmonary portion of S2 even more than normal
Which valves is most likely to have a congenital stenosis?
The pulmonary valve is most likely to have a congenital stenosis
What is the pathophysiology, physical exam findings, and treatment of pulmonary stenosis?
Pulmonary stenosis is a lack of separation of the valve cusps. It can cause a hypertrophied RV and a systolic ejection murmur can be heard at the left upper sternal border. Balloon valvuloplasty is indicated in severe stenosis or with a very sick infant
What is acrocyanosis?
Sluggish circulation in the periphery allows for extensive removal of oxygen in the capillary beds. Common in chilled children
Name the 6 cyanotic heart diseases
Hey Harry This Turkey Tastes Terrible.
* Hyperplastic Right Heart Syndrome (tricuspid/pulmonary atresia)
* Hyperplastic Left Heart Syndrome
* Transposition of the great arteries
* Transposition of pulmonary veins (Total anomalous pulmonary venous return - TAPVR)
* Tetralogy of Fallot
* Truncus arteriosus
What is the most common cyanotic defect and what defects are associated with it?
Tetralogy of Fallot. Ventricular septal defect, with an overriding aorta, pulmonary stenosis, and right ventricular hypertrophy
What component of tetralogy of Fallot is the variable component responsible for severity?
Pulmonary stenosis determines the obstruction of blood flow to the lungs. Mild PS = minimally cyanotic (pink). Severe PS = critically cyanotic (blue)
What is the treatment for a tetralogy of Fallot?
Palliation until the child is stable , usually a surgical shunt to get more blood to the lungs. Then repair by closing the VSD with a patch and opening the pulmonary outflow tract
What is the big picture cause of a myocardial infarction?
Inadequate tissue perfusion results in myocardial necrosis
How is a myocardial infarction a chronic an acute pathology?
Myocardial infarctions a the acute ischemia and necrosing of myocardium following the chronic progression of an atherosclerotic disease
What is the most common cause of a vasospasm myocardial infarction?
Cocaine use
What are some causes of plaque rupture or fissure formation?
Changes in intraluminal pressure, bending or twisting of arteries during heart contraction, high lipid content within plaque, the thickness of the fibrous cap, the plaque shape, and mechanical injury
What are some risk factors for CAD and acute MI?
Smoking, HTN, dyslipidemia, DMII, a positive family history, sedentary lifestyle, obesity, age
What cardiac markers are used to detect irreversibly injured ischemic myocardium and when is their elevation detectable?
Troponin I, Troponin T, creatine kinase, aspartate serum transaminase, lactate dehydrogenase and their peak elevation is around 24 hours post event
What are the T wave, Q wave, and ST segment changes in an acute MI?
* Hyper acute T waves are the earliest sign of injury. T wave inversions suggest ischemia
* ST depression in an anatomical distribution suggest ischemia. ST elevation in a consistent anatomical distribution suggests an ongoing myocardial injury
* Diagnostic Q waves in a consistent anatomical pattern suggest prior MI
What is the physiologic difference between a STEMI and a NSTEMI?
STEMIs result from a complete vessel obstruction. NSTEMIs result from an incomplete blockage or recanalization of a thrombotic occlusion
What type of MI shows ST elevation in II, III, and aVF and what artery is implicated?
II, III, and aVF detect inferior MI. The right coronary artery supplies that portion of the heart
What type of MI shows ST elevation in V1-V3 and what artery is implicated?
V1-V3 suggests an anterior MI which implicates the LAD artery
How much fluid is in the pericardium and what is it's purpose?
10-50 mL and it serves as a lubricant, minimizing frictional forces between the heart and other structures
What radiologic finding is found with a congenitally absent pericardium?
Pericardial cysts
What are the main sources of infections pericarditis?
Viral (coxsakie A&B, echovirus, mumps, adenovirus, HIV), mycobacterium tuberculosis, bacterial (Pneumococcus, strep, staph, legionella), fungal (histoplasmosis, coccidioidomycosis, candidiasis, blastomycosis)
What are some non-infectious causes of pericarditis?
Idiopathic, neoplasm, renal failure, irradiation, MI, hypothyroidism, aortic dissection, trauma
What are some causes of hypersensitive pericarditis?
Collagen vascular diseases (systemic lupus, rheumatoid arthritis, scleroderma, acute rheumatic fever), drug induced (procainamide, hydralazine, isoniazid), post-MI or cardiac surgery
What is the presentation of acute pericarditis?
Sudden, severe onset retrosternal or left precordial pain, low-grade fever, tachycardic, frictional rub on ausculataion, possibly pleuritic
What are the EKG findings for acute pericarditis?
DIFFUSE ST elevation that is anatomically inconsistent with MI and T wave inversion
What is the pathophysiology of cardiac tamponade?
An accumulation of fluid in the pericardial space increases pressure until cardiac compression results in impaired diastolic ventricular filling, depressing cardiac output
What is the presentation for cardiac tamponade?
Cardiac tamponade mimics heart failure: dyspnea on exertion, orthopnea, tachycardia, tachypnea, JVD, hypotension and poor peripheral perfusion, shock and hemodynamic collapse, pulsus paradoxus
What is the treatment for a cardiac tamponade?
Supportive measures (IV fluids and pressors). Remove the fluid (pericardiocentesis, surgical pericardectomy)
What is the pathophysiology of constrictive pericarditis and what are some common causes?
* A dense fibrosis forms and causes adhesion of the parietal and visceral layers, creating a rigid "case" around the heart. This case decreases diastolic ventricular filling because of the restraint of the thick, rigid, calcified pericardium
* Mediastinal radiation, history of cardiac surgery. Can also be neoplasm, rheumatoid arthritis, or recurrent pericarditis
What is the presentation of constrictive pericarditis?
Right-sided heart failure: Weakness, dyspnea, orthopnea, anorexia, peripheral edema, hepatosplenomegaly, ascites, pericardial knock, JVD with prominent y descent
What is the treatment for constrictive pericarditis?
Surgical stripping of the pericardium
Name the 2 types of cardiac trauma and give examples of both
Penetrating: Gunshot or stab wound
Non-penetrating: Blunt force, rapid deceleration, transections
What is the presentation of stable angina?
Substernal pain that radiates to the left arm. Often described as a dull, constrictive pressure usually precipitated by exertion. It must present as an unchanged pattern for 60 days or more
What is the presentation of unstable angina?
New onset angina of less than 2 months. Angina will present at low activity levels or rest and will have increased in frequency or severity from previously stable angina
What are the big picture steps in angina therapy?
* First, reduce myocardial oxygen demand (ß-blockers, nitrates, ACEi)
* Then stabilize the process exacerbating the problem: coronary artery spasm (Ca2+ channel blockers, nitrates), platelet activation (aspirin, clopidogrel, GpIIb/IIIa receptor blockers), thrombus formation (heparin, LMWH, or thrombolytics if indicated)
What are some diagnostic options for diagnosing ischemic heart disease and angina?
A good history & physical, an electrocardiogram, an exercise stress test, cardiac catheterization, or coronary angiography
What are the 3 therapeutic goals for angina?
Relieve the angina, prevent myocardial infarction, and promote longevity
What are the 3 main treatment options for angina?
Medical therapy, PCI-DES, CABG
What veins are most often used to perform CABGs?
Saphenous vein, internal mammary artery, radial artery, or gastroepiploic artery
What are the options for angioplasty stents and what are 2 considerations when deciding between the 2?
Either way the patient needs to be on life-long aspirin
* Drug-eluting stents have a lower restenosis rate (5-10%)
* Bare metal stents require less time on anti-platelet therapy (restenosis rate: 15-25%)
What are the treatment options for 50-75% and >75% stenotic angina?
* 50-75%: try medical therapy, if angina persists consider PCI-DES or CABG
* >75%: if 1-3 vessels, individualize PCI-DES or CABG based on morphology, LV function. if diffuse, 3 vessels with poor LV and diabetes then use CABG. If it's the left main artery then do CABG
Describe changes in the cardiovascular system due to aging
* Arterial and myocardial stiffness, impaired ß–adrenergic responsiveness and endothelial function, and reduced sinus node function.
* There is also decreased maximal HR and CO during exercise
How is HTN controlled in the elderly?
Mainly with lifestyle modification (sodium restriction, weight control, physical activity, moderation of alcohol, smoking cessation, control co-morbid risk factors). First-line therapy is a thiazide diuretic. Statin.
What factors contribute to adverse outcomes in older patients with acute coronary syndrome?
Age related myocardial changes, increased comorbidity, atypical symptoms & non-diagnostic ECG, extensive coronary artery disease, prevalence of prior myocardial damage, risk of complications from therapy
What is the atypical presentation of MI in the elderly?
More common in older women: confusion, dyspnea, lassitude, new onset CHF, new arrhythmia, syncope, stroke
What are the population differences between DHF and SHF?
DHF is more older women with HTN, but without CAD. SHF is associated with middle-aged men with HTN & CAD comorbidities
What are the atypical presentations of hypo and hyperthyroidism in older adults?
Hypothyroidism is under-diagnosed and is often confused with normal aging. Hyperthyroidism will present as a sedated, apathetic patient without a goiter
What is the atypical presentation of acute abdominal disease in older adults?
Patients may not feel early pain and the frequency of NSAID use in this population may mask clinical features
What is the atypical presentation of infections in older adults
Infections can go undetected due to a lack of fever and no leukocytosis
What is the pathophysiology of aortic regurgitation?
If aorta chronically regugitates, the LV will eccentrically dilate from volume overload in an attempt to lessen pulmonary edema progression
What are the physical exam findings in aortic regurgitation?
A diastolic "blowing" murmur, an Austin-Flint murmur (diastolic rumbling w/out and opening snap), and a widened pulse pressure (back flow increases preload and increases stroke volume)
What is the treatment for aortic regurgitation?
Nothing will slow progression, only help symptoms. Vasodilators to encourage forward flow
Which valve is the most common target for stenosis from carcinoid syndrome?
The tricuspid valve
What are the major differences between a mechanical and bioprosthetic valves?
Mechanical valves will last the lifespan of the patient but lifetime anti-coagulation is necessary. Bioprosthetic valves need to be replace every 10 years but do not require anti-coagulant therapy
What are the 2 systolic ejection murmurs and how are they distinguished?
Aortic (RUSB) and Pulmonic (LUSB) Stenosis
What are the 2 holosystolic murmurs and how are they distinguished?
Mitral regurgitation (apex) and Ventricular septal defect (LSB)
How do you distinguish between mitral regurgitation and aortic stenosis?
Ask the patient to clench their fists, increasing their afterload. An increase in afterload will increase the regurgitated volume making the murmur louder. The maneuver will have no impact on aortic stenosis
Where are a-receptors localized and what does their stimulation do?
* A-receptors are in the peripheral and splanchnic vasculature.
* They cause smooth muscle contraction and vasoconstriction.
* They are under negative feedback control and are located pre-synaptically.
Where are ß1-receptors localized and what does their stimulation do?
* ß1-receptors are located in cardiac tissue and peripheral vasculature.
* They increase cAMP effects, increasing inotropy, chromotropy and lusitropy.
* They are under positive feedback control and are located pre-synaptically.
Where are ß2-receptors localized and what does their stimulation do?
* ß2-receptors are located in cardiac tissue, peripheral vasculature, and bronchial smooth muscle.
* In cardiac tissue, they increase ino- chromo- and lusitropy. In the peripheral vasculature, they relax smooth muscle and are vasodilators. In the bronchial smooth muscle, they are relaxants and bronchodilators
Where are dopamine receptors localized and what does their stimulation do?
* Cardiac tissue: Inotropic, chromotropic
* Peripheral and splanchnic vasculature: vasodilation
* Renal: diuretic, natriuretic
* CNS
Where are vasopressin receptors localized and what does their stimulation do?
* Peripheral & splanchnic vasculature: marked vasoconstriction
What classes of drugs are used to increase preload?
Administer fluids, a-agonists in low doses
What classes of drugs are used to decrease afterload?
Vasodilators: ACEi, Nitroprusside, phosphodiesterase inhibitor
What classes of drugs are used to increase contractility?
Inotropes: ß-adrenergics (dobutamine), phosphodiesterase inhibitor (milrinone)
What classes of drugs are used to increase heart rate?
Chronotropes: ß-adrenergics (dobutamine), atropine (vagolytic), pacemaker
What classes of drugs are used to increase systemic vascular resistance?
Vasoconstrictors: a-adrenergics, vasopressin agonists
What is epinephrine's mechanism of action and what are its indications?
Epinephrine is a ß-agonist. It is inotrophic and chronotropic. It is indicated in cardiogenic shock, post CABG, and with sepsis
What is dobutamine's mechanism of action and what are its indications?
Dobutamine is a ß-agonist. It is an inotrope (increasing contractility) and chromotrope (increasing HR) and a vasodilator (reducing afterload). It is indicated in acute MI, and mild cardiogenic shock
What is norepinephrine's mechanism of action and what are its indications?
Norepinephrine is an a-agonist. It is a vasoconstrictor and increases systemic vascular resistance. It is indicated in sepsis and distributive shock with good CO
What is phenylephrine's mechanism of action and what are its indications?
Phenylephrine is a pure a-agonist. It is a vasoconstrictor and increases systemic vascular resistance. It is indicated in mild sepsis and hypotension with good CO
What is dopamine's mechanism of action and what are its indications?
Dopamine has dose-dependent pharmacology, at low doses it is dopamine agonist and as doses increase it moves to ß- and then a-activity. It is no longer used.
What is vasopressin's mechanism of action and what are its indications?
Vasopressin only binds its own receptors. It causes intense vasoconstriction and gut ischemia is a concern. It is indicated in advanced cardiac life support, severe shock, and CABG (when on ACEi)
What are milrinone and amrinone's mechanism of action and what are their indications?
Milrinone and amrinone are phosphodiesterase inhibitors allowing cAMP and cGMP to stay in circulation longer. They are smooth muscles relaxers, vasodilators and inotropes and augment ß-adrenergic stimulation. They are indicated in CABG and situation of vasoconstriction with low CO
How is left atrial pressure measured?
Directly by punching a catheter through the foramen ovale, or indirectly by using a Swan-Ganz cathetor and using the pulmonary capillary wedge pressure
What is the problem in right sided heart failure, what are some possible causes, and what is the clinical result?
* A decreased RV SV leads to a compensatory increase in RA pressure
* Increase in afterload (PS or pulmonary HTN) or decreased contractility (RV infarct or infiltrative disease)
* The result is peripheral edema
What is the problem with left heart failure, what are its possible causes and what is the clinical result?
* LV SV decreases and that leads to a pressure build up in the LA
* Caused by and increased afterload (aortic stenosis, systemic HTN, aortic coarctation) or decreased contractility (LV infarct or infiltrative disease)
* The result is pulmonary edema
What percentage of women suffer sudden cardiac death without having any symptoms?
2/3
What is the temporal differences in the clinical manifestation of CHD between men and women?
Women develop clinical manifestations 10 years later than men
What are the risk factors associated with the Framingham RIsk Score?
Age, gender, total cholesterol, HDL, smoking, systolic blood pressure, current HTN treatment
What are the 2 classes of lifestyle interventions for preventing heart disease in women?
* Class I: Smoking cessation, physical activity, diet intake, weight control
* Class II: Omega-3 fatty acids
What are the components of metabolic syndrome?
* Waist circumference >35in
* Fasting triglycerides >150 mg/dL
* HDL cholesterol <50 mg/dL
* HTN (≥130/≥85) or on HTN drugs
* Fasting glucose ≥100 mg/dL
What age group currently has an increasing death rate?
Women 35-54
What are some potential factors affecting diagnostic accuracy of the exercise tolerance test in women?
Hormonal influences, reduced functional capacity, resting ST-T wave abnormalitities
How does CAD grossly look in women?
Women's CAD is more often a circumferentially diffuse disease without luminal narrowing and less often obstructive plaques
Where in the cardiac anatomy would infarcts in the LAD, LCX, and RCA affect?
* LAD: anteroseptal LV
* LCX: lateral LV
* RCA: posteroseptal LV
What is the time frame for an early acute MI, what does it look like grossly and microscopically?
* 6-24 hrs
* Subtle changes, patchy pallor, slight hyperemia
* Thin wavy fibers, eosinophilia, few PMNs
What is the time frame for an acute MI, what does it look like grossly and microscopically?
* 1-6 days
* Obviously pale, yellow
* Necrotic myocytes, many PMNs
What is the time frame for an organizing MI, what does it look like grossly and microscopically?
* 1-3 weeks
* Red-brown edges around a pale center
* Granulation tissue, acute and/or chronic inflammation
What is the time frame for an remote MI, what does it look like grossly and microscopically?
* 3 mos+
* Firm, white scar with a contracted, thin wall
* Collagen inside a thinning wall
What is the difference between transmural and subendocardial infarcts?
Transmural involves an infarction of the full ventricular wall thickness. Subendocardial is limited to the inner 1/3 to 1/2 of the ventricular wall
In what case would a complete vessel occlusion not cause an MI?
If there was sufficient collateral blood flow
Plaques with what type of architecture is most vulnerable to rupture?
Plaques with a soft lipid core and a thin fibrous cap is the most vulnerable to rupture
What is the pathogenesis for subendocardial myocardial infarcts?
Areas of diffuse CAD & global perfusion deficit undergo periods of increased demand, vasospasm, or hypotension
What are common complications of myocardial infarctions?
Death, congestive heart failure, rupture (free wall, septum, papillary muscles), mural thrombus, LV aneurysm, dysrrhythmias, pericarditis, pulmonary or leg thromboembolism
What are the major manifestations of right-sided heart failure?
* Portal, systemic & peripheral (feet, ankles, sacrum) congestion and edema. Pleural & peritoneal effusions/ascites
* Hepatomegaly with severe hypoxia and subsequent central fibrosis
* Congestive splenomegaly with focal hemorrhage and hemosiderin deposits
* Renal congestion with acute tubular necrosis
What are the pathological manifestations of acute rheumatic fever?
Pancarditis. Rare endocarditis leading to stenotic murmurs. Myocarditis that causes cardiac enlargement and failure, dilation of ventricle and mitral ring. Mitral regurgitation & Aschoff bodies (focal, granulomatous inflammation) are found in the myocardium
What are the pathological changes seen in chronic rheumatic heart disease?
* Thickening of left-sided valve leaflets (predominantly mitral valve) especially along closure lines. Commissures fuse, chordae tendinae thicken, shorten, and fuse. Result is mitral stenosis, insufficiency, or both
* Mitral stenosis causes LA hypertrophy and dilation, a pulmonary congestion leading to RVH, and chronic L heart failure leading to R heart failure and eventually total heart failure
* Mitral regurgitation causes LV hypertrophy and dilation leading to LA dilation
* Can cause aortic stenosis and regurgitation as well
What gross pathological finding is often associated with aortic disease from chronic RHD?
The "fish-mouth" bicuspid deformity of the aortic valve is commonly seen in chronic RHD
What are the 3 general pathologies of aortic stenosis and compare their epidemiology, mechanism, and murmur presentation?
* Acquired: Middle-aged women get fused commissures and thickened closure lines at the aortic and mitral valves almost always from rheumatic fever. Murmur presents in mid-adult life
* Degenerative: Elderly men suffer hemodynamic injury and dystrophic calcification deep in sulci of only the aortic valve. Late onset of murmur.
* Congenital: Bicuspid, uneven tricuspid, or unicommissural aortic valves develop with calcium deep in sulci AND on cusps of only the aortic valve. Childhood murmur but adult stenosis
What are the different mechanisms of aortic regurgitation?
* Disease of the valve: rigid (rheumatic, degenerative), destruction (microbial endocarditis), collase (prolapse through VSD or with myxomatous degeneration)
* Weaking of the valve ring leading to dilation and a valve insufficient to cover increased area (Marfan, dissecting aneurysm, cystic medial degeneration, syphilitic aortitis)
What are the infectious and non-infectious causes of endocarditis?
* Non-infective: Verrucous (ARF), atypical verrucous (SLE), non-bacterial thromboembolism
* Infective: Viral, bacterial, fungal destruction of valve tissue. Thrombus w/ invasive organisms
What is the natural history of subacute endocarditis?
Enteric organismal (S. viridans) infection of previously damaged valve. An insidious onset of low grade fever, malaise, fatigue, anemia, murmur for months. Diagnosed with a series of positive blood tests and treated with IV bacteriocidal antibiotics for 4-6 weeks
What is the natural history of acute endocarditis?
Virulent, focally destructive infection (S. aureus, S. faecalis) that targets previously normal valves. A sudden onset of high fever, weakness, and murmur that lasts a few weeks. Death from heart failure, valve perforation, or sepsis. Treat with IV antibiotics and resect infected valve
What organism is commonly implicated in endocarditis in drug addicts?
S. aureus or fungi from skin, contaminated drugs or cutting materials. Mostly left-sided endocarditis, but more likely than non-addicts to get right-sided endocarditis
What are common complications of infectious endocarditis and what is the most common cause of death?
* Intra-cardiac: Perforated cusps or leaflets, rupture of chordae tendinae, abcess, fistula, obstruction of valve or outflow tract, embolization to coronary arteries. Above mechanisms lead to CHF which leads to death
* Extra-cardiac: Emboli to other organs, mycotic aneurysm, glomerulonephritis (Type III immune reaction)
What is the presentation of infective endocarditis?
FROM JANE. Fever, roth's spots, osler's nodes, murmur, janeway lesions, anemia, nail bed hemorrhage, embolism
What is the pathophysiology of non-infective endocarditis (marantic endocarditis)?
Small, sterile fibrin & platelet thrombi adhere to valves at closure lines of the mitral valve. Often due to systemic hypercoagulability and seen in cancer patients 2˚ to DIC. Can embolize systemically
What valves are targeted in Libman-Sacks endocarditis and what changes are found?
Libman-Sacks endocarditis is a valvulitis targeting the mitral and tricuspid valves in the context of SLE & anti-phospholipid syndrome. Fibrinous vegetation builds on either side of the valve leading to necrosis and valve deformations
What is the pathophysiology of carcinoid heart disease?
Elaboration of bioactive products (serotonin, bradykinin, histamine, prostaglandin) cause right-sided valvular lesions
What is the epidemiology, morphology, presentation, and complications associated with mitral valve prolapse?
Females aged 20-40 can develop stretched, redundant myxomatous valves with interchordal hooding. Mid-systolic click (atypically dyspnea, fatigue, psychologic manifestations) or asymptomatic. Complications include mitral regurgitation, 2˚ infective endocarditis, or sudden death
What are the diagnostic criteria, morphology, and clinical features of hypertensive cardiovascular disease?
HTCVD is diagnose when there is concentric LVH with a history of HTN and an absence of other LVH-causing lesions. It is concentric LVH and cardiomegaly enlarged by myocytes and nuclei leading to diffuse interstitial fibrosis, focal degeneration, LV dilation and thinning.
What is cor pulmonale and what condition is it associated with?
The right-sided counterpart of systemic HTCVD. RVH or dilation 2˚ to chronic pulmonary HTN (disorders affecting lung structure or function). Commonly caused by COPD. EXCLUDE RVH 2˚ CHD or LV disease
What is the morphology of general myocarditis?
Soft myocardium w/ 4 chamber dilation and matchy hemorrhagic mottling. Mural thrombi may form. Myocardial inflammation with infiltration, myocyte necrosis and degeneration. Focal lesions
What is the natural history of hypertrophic cardiomyopathy?
Hypertrophic cardiomyopathy is often familial. A thickening of the septum narrow the LV outflow tract leading to impaired ventricular compliance, pump failure, systolic murmur, dysrhythmia, and sudden death. Microscopic myofiber disarray
What are common complications of infectious endocarditis and what is the most common cause of death?
* Intra-cardiac: Perforated cusps or leaflets, rupture of chordae tendinae, abcess, fistula, obstruction of valve or outflow tract, embolization to coronary arteries. Above mechanisms lead to CHF which leads to death
* Extra-cardiac: Emboli to other organs, mycotic aneurysm, glomerulonephritis (Type III immune reaction)
What is the presentation of infective endocarditis?
FROM JANE. Fever, roth's spots, osler's nodes, murmur, janeway lesions, anemia, nail bed hemorrhage, embolism
What is the pathophysiology of non-infective endocarditis (marantic endocarditis)?
Small, sterile fibrin & platelet thrombi adhere to valves at closure lines of the mitral valve. Often due to systemic hypercoagulability and seen in cancer patients 2˚ to DIC. Can embolize systemically
What valves are targeted in Libman-Sacks endocarditis and what changes are found?
Libman-Sacks endocarditis is a valvulitis targeting the mitral and tricuspid valves in the context of SLE & anti-phospholipid syndrome. Fibrinous vegetation builds on either side of the valve leading to necrosis and valve deformations
What is the pathophysiology of carcinoid heart disease?
Elaboration of bioactive products (serotonin, bradykinin, histamine, prostaglandin) cause right-sided valvular lesions
What is the epidemiology, morphology, presentation, and complications associated with mitral valve prolapse?
Females aged 20-40 can develop stretched, redundant myxomatous valves with interchordal hooding. Mid-systolic click (atypically dyspnea, fatigue, psychologic manifestations) or asymptomatic. Complications include mitral regurgitation, 2˚ infective endocarditis, or sudden death
What are the diagnostic criteria, morphology, and clinical features of hypertensive cardiovascular disease?
HTCVD is diagnose when there is concentric LVH with a history of HTN and an absence of other LVH-causing lesions. It is concentric LVH and cardiomegaly enlarged by myocytes and nuclei leading to diffuse interstitial fibrosis, focal degeneration, LV dilation and thinning.
What is cor pulmonale and what condition is it associated with?
The right-sided counterpart of systemic HTCVD. RVH or dilation 2˚ to chronic pulmonary HTN (disorders affecting lung structure or function). Commonly caused by COPD. EXCLUDE RVH 2˚ CHD or LV disease
What is the morphology of general myocarditis?
Soft myocardium w/ 4 chamber dilation and matchy hemorrhagic mottling. Mural thrombi may form. Myocardial inflammation with infiltration, myocyte necrosis and degeneration. Focal lesions
What is the natural history of hypertrophic cardiomyopathy?
Hypertrophic cardiomyopathy is often familial. A thickening of the septum narrow the LV outflow tract leading to impaired ventricular compliance, pump failure, systolic murmur, dysrhythmia, and sudden death. Microscopic myofiber disarray
What is the pathogenesis and morphology of dilated/congestive myocarditis?
* Alcohol, peripartum, sarcoid, autoimmune, and chemotherapy
* Cardiomegaly, mural thrombi, normal valves, subtle microscopic changes of mycotic hypertorphy and interstitial fibrosis
What is the natural history of restrictive myocarditis?
Amyloid infiltration of the myocardium with a characteristic CONGO RED STAIN. Amyloid can sometimes be limited to heart and vessels. Can mimic ischemic heart disease (failure, EKG changes)
What are the 3 types of obliterative myocarditis?
* Endomyocardial fibrosis: children and young adults in Africa. Subendocardial fibrosis decreasing ventricular chamber volume
* Fibroblastic endocarditis (Loeffler's): Endomyocardial fibrosis, mural thrombi, peripheral eosinophilia, eosinophilic infiltration, rapidly fatal!
* Endocardial fibrosis: children <2 yo
What is the pathophysiology of arrhythmogenic myocarditis?
* Autosomal dominant: RV adipose or fibro-adipose replacement
* Autosomal recessive: RV aneurysms
* Myocarditis: LV dilation
* Causes serious dysrhythmia and sudden death
What is the common physiological problem with dilated cardiomyopathy?
A decreased left ventricular ejection fraction
What are some common etiologies of non-ischemic dilated cardiomyopathy?
Tachycardia-meidated, excessive alcohol, peripartum, chemotherapy toxicity (especially adriamycin), infectious (chagas, lyme, rocky mountain spotted fever, rheumatic fever)
What are some risk factors for peripartum cardiomyopathy?
Maternal age >30 yo, african descent, previous twin or triplet pregnancy, previous preeclampsia or pregnancy-induced hypertension, long term tocolytic treatment, cocaine use
What viruses are commonly indicated in viral myocarditis and what populations are particularly at risk?
* Coxsakievirus! parvovirus, HHV6, influenza, adenovirus, echovirus, CMV, HIV
* Infants and pregnant women
What mutations are commonly associated with hypertrophic cardiomyopathy?
ß-myosin heavy chain! Troponin T or a-tropomyosin
How do the two pathological findings in hypertrophic cardiomyopathy lead to its clinical manifestations?
* Thickened muscle decreases its ability to relax, decreasing its diastolic function and increasing the LVEDP
* Myofibrillar disarray increases risk of dangerous, deadly ventricular arrhythmia
What is the pathogenesis and morphology of dilated/congestive myocarditis?
* Alcohol, peripartum, sarcoid, autoimmune, and chemotherapy
* Cardiomegaly, mural thrombi, normal valves, subtle microscopic changes of mycotic hypertorphy and interstitial fibrosis
What is the natural history of restrictive myocarditis?
Amyloid infiltration of the myocardium with a characteristic CONGO RED STAIN. Amyloid can sometimes be limited to heart and vessels. Can mimic ischemic heart disease (failure, EKG changes)
What are the 3 types of obliterative myocarditis?
* Endomyocardial fibrosis: children and young adults in Africa. Subendocardial fibrosis decreasing ventricular chamber volume
* Fibroblastic endocarditis (Loeffler's): Endomyocardial fibrosis, mural thrombi, peripheral eosinophilia, eosinophilic infiltration, rapidly fatal!
* Endocardial fibrosis: children <2 yo
What is the pathophysiology of arrhythmogenic myocarditis?
* Autosomal dominant: RV adipose or fibro-adipose replacement
* Autosomal recessive: RV aneurysms
* Myocarditis: LV dilation
* Causes serious dysrhythmia and sudden death
What is the common physiological problem with dilated cardiomyopathy?
A decreased left ventricular ejection fraction
What are some common etiologies of non-ischemic dilated cardiomyopathy?
Tachycardia-meidated, excessive alcohol, peripartum, chemotherapy toxicity (especially adriamycin), infectious (chagas, lyme, rocky mountain spotted fever, rheumatic fever)
What are some risk factors for peripartum cardiomyopathy?
Maternal age >30 yo, african descent, previous twin or triplet pregnancy, previous preeclampsia or pregnancy-induced hypertension, long term tocolytic treatment, cocaine use
What viruses are commonly indicated in viral myocarditis and what populations are particularly at risk?
* Coxsakievirus! parvovirus, HHV6, influenza, adenovirus, echovirus, CMV, HIV
* Infants and pregnant women
What mutations are commonly associated with hypertrophic cardiomyopathy?
ß-myosin heavy chain! Troponin T or a-tropomyosin
How do the two pathological findings in hypertrophic cardiomyopathy lead to its clinical manifestations?
* Thickened muscle decreases its ability to relax, decreasing its diastolic function and increasing the LVEDP
* Myofibrillar disarray increases risk of dangerous, deadly ventricular arrhythmia
What is the difference between HCM and HOCM?
If the ventricular septum hypertrophies asymmetrically it can cause the anterior leaflet of the mitral valve to get sucked towards it and obstruct flow into the aorta causing "Dynamic LV outflow obstruction"
What makes dynamic LV outflow obstruction worse or better?
* Worse: decrease LV volume (dehydration) or increased contractility (exercise or inotropic drugs)
* Better: increase volume (hydration) or decrease contractility (ß-blocker or NDHP Ca2+ channel blocker)
What 2 exam maneuvers are important in diagnosing a HOCM?
The squat to stand and the valsalva.
* Patient squats increasing venous return and increasing the LVEDP, keeping the mitral away from the septum and decreasing or eliminating the murmur
* When the patient stands, it decreases their venous return and LVEDP, with less volume to keep the mitral leaflet from obstructing the HOCM murmur will get louder
* With valsalva, increased intra-abdominal pressure decreases venous return and LVEDP, the murmur will get louder
How do you distinguish between an aortic stenosis and a HOCM murmur?
* HOCM (dynamic LV outflow obstruction) will get louder when then volume (venous return/LVEDP) decreases, Valsalva and standing back up will make HOCM murmurs get louder.
* Aortic stenosis murmurs get louder when there is an increase in venous return and LVEDP. Squatting will make an aortic stenosis murmur get louder
What are some potential treatments for HOCM?
* Negative inotropes (ß–blockers, NDHP Ca2+ channel blockers)
* Surgical myomectomy (cutting out part of the septum)
* Intentional septal infarct
What are the 3 mechanisms of restrictive cardiomyopathy? Give examples
* Fibrosis or scarring like in scleroderma
* Infiltration of myocardium by amyloid, sarcoid, or rarely, cancer
* Deposition of hemochromatosis or glycogen
Is systole or diastole affected in a restrictive cardiomyopathy?
Diastole, because the "cage" restricts the ventricles ability to relax
What are the common signs and symptoms of Rheumatic fever?
FEVERSS.
* Fever
* Erythema marginatum
* Valvular damage (vegetation and fibrosis)
* Erythocyte sedementation rate increase
* Red-hot joints (migratory polyarthritis)
* Subcutaneous nodules
* St. Vitus' dance (chorea)
What size vessels control vascular resistance?
Small arteries and arterioles
Where in the circulation is the greatest cross-sectional area?
Capillaries and venules
What are the two systems that coordinate together to regulate global circulatory homeostasis?
Autonomic innervation (baro and chemoreceptors) and circulating hormones (ANP, ADH, bradykinin, PG, TxA)
Where are the three types of baroreflexes located and what do they affect?
* Arterial baroreceptors trigger the brainstem vasomotor center to change total peripheral resistance, cardiac performance, sympathetic kidney drive, and venous compliance
* Cardiopulmonary receptors act at the brainstem vasomotor center affecting venous compliance, sympathetic kidney drive, total peripheral resistance, and cardiac performance and stimulates the hypothalamus to release antidiuretic hormone (vasopressin)
* Intrarenal baroreceptors stimulate the RAS, GFR, and salt and water reabsorption
What affect does increasing blood pressure have at baroreceptors?
Increasing blood pressure will increase baroreceptor firing which will increase parasympathetic stimulation to the heart and decrease sympathetic stimulation to the heart and vessels
What do chemoreceptors monitor and how do they respond?
Chemoreceptors in the carotid and aortic bodies monitor oxygen, hydrogen (pH), and carbon dioxide. They are stimulated by decreased blood flow or increased CO2, and decreased pH and O2. They respond by activating a sympathetic respiration response
What catecholamine is released more by the adrenal medulla and which is released by sympathetic nerve activation at the blood vessel?
Epinephrine is released in higher percentages by the adrenal medulla. Sympathetic nerves release more Norepinephrine
Name 3 situations where renal renin release is stimulated
Sympathetic stimulation, hypotension, and decreased sodium delivery to the distal tubules
What are the effects of the RAS at the brain, the kidney, the vasculature, and the adrenal gland?
* Brain: thirst & sympathetic activity
* Kidney: water & salt retention
* Vasculature: vasoconstriction & increased blood pressure
* Adrenal: aldosterone release
Where is vasopressin produced and what does it do?
Anti-diuretic hormone (vasopressin) is released by the posterior pituitary and stimulates thirst, acts at the kidneys to increase blood volume, and vasoconstricts blood vessels
What is atrial natriuretic peptide and what does it do?
Atrial natriuretic peptide is released by the atrial myocytes when they are stretched and counter-balance the RAAS. ANP increases GFR decreasing the blood volume, the cardiac output, and eventually the arterial pressure
What are some common tissue factors that contribute to local blood flow regulation?
* CO2, H+, K+, lactate, adenosine.
* Histamine, bradykinin, prostacyclin, leukotrienes
* They originate in tissue (often as metabolic products) and act as either constrictors or relaxants
What are some common endothelial factors and how do they contribute to local blood flow regulation?
* Nitric oxide and prostacyclins are dilators
* Endothelin, leukotrienes, and thromboxanes are constrictors
* They are released during physical stress (shear) and act via endothelial receptors or directly on smooth muscle
Which vasodilators are receptor dependent and which are receptor independent?
* Acetylcholine, bradykinin, serotonin, histamine, substance P are all vasodilators that circulate in the lumen, bind a receptor and trigger NO release.
* Sodium nitroprusside is a direct NO donor and does not need to bind to endothelial receptors to have a vasodilating effect
Are prostaglandins (PGI2) vasodilators or vasoconstrictors, what is their source, and where do they have their effect?
Prostaglandins are vasodilators that are released by endothelial cells and have their effect at vascular smooth muscle
Is adenosine a vasodilator or vasoconstrictor, what is its source, and where does it have its effect?
Adenosine is a vasodilator that is released by parenchymal cells and has its effect at vascular smooth muscle
Is epinephrine a vasodilator or vasoconstrictor, what is its source, and where does it have its effect?
Epinephrine is a vasodilator that is released by the adrenal medulla and has its effect at ß2-receptors and vascular smooth muscle
Are H+, CO2, and K+ vasodilators or vasoconstrictors, what is their source, and where do they have their effect?
H+, CO2, and K+ are vasodilators that are released by skeletal muscle and have their effect at vascular smooth muscle
Is endothelium derived hyperpolarizing factor a vasodilator or vasoconstrictor, what is its source, and where does it have its effect?
Endothelium derived hyperpolarizing factor is a vasodilator that is released by endothelial cells and has its effect at vascular smooth muscle and K+ channels
Where is vasopressin produced and what does it do?
Anti-diuretic hormone (vasopressin) is released by the posterior pituitary and stimulates thirst, acts at the kidneys to increase blood volume, and vasoconstricts blood vessels
What is atrial natriuretic peptide and what does it do?
Atrial natriuretic peptide is released by the atrial myocytes when they are stretched and counter-balance the RAAS. ANP increases GFR decreasing the blood volume, the cardiac output, and eventually the arterial pressure
What are some common tissue factors that contribute to local blood flow regulation?
* CO2, H+, K+, lactate, adenosine.
* Histamine, bradykinin, prostacyclin, leukotrienes
* They originate in tissue (often as metabolic products) and act as either constrictors or relaxants
What are some common endothelial factors and how do they contribute to local blood flow regulation?
* Nitric oxide and prostacyclins are dilators
* Endothelin, leukotrienes, and thromboxanes are constrictors
* They are released during physical stress (shear) and act via endothelial receptors or directly on smooth muscle
Which vasodilators are receptor dependent and which are receptor independent?
* Acetylcholine, bradykinin, serotonin, histamine, substance P are all vasodilators that circulate in the lumen, bind a receptor and trigger NO release.
* Sodium nitroprusside is a direct NO donor and does not need to bind to endothelial receptors to have a vasodilating effect
Are prostaglandins (PGI2) vasodilators or vasoconstrictors, what is their source, and where do they have their effect?
Prostaglandins are vasodilators that are released by endothelial cells and have their effect at vascular smooth muscle
Is adenosine a vasodilator or vasoconstrictor, what is its source, and where does it have its effect?
Adenosine is a vasodilator that is released by parenchymal cells and has its effect at vascular smooth muscle
Is epinephrine a vasodilator or vasoconstrictor, what is its source, and where does it have its effect?
Epinephrine is a vasodilator that is released by the adrenal medulla and has its effect at ß2-receptors and vascular smooth muscle
Are H+, CO2, and K+ vasodilators or vasoconstrictors, what is their source, and where do they have their effect?
H+, CO2, and K+ are vasodilators that are released by skeletal muscle and have their effect at vascular smooth muscle
Is endothelium derived hyperpolarizing factor a vasodilator or vasoconstrictor, what is its source, and where does it have its effect?
Endothelium derived hyperpolarizing factor is a vasodilator that is released by endothelial cells and has its effect at vascular smooth muscle and K+ channels
Are angiotensin(1-7), bradykinin, and substance P vasodilators or vasoconstrictors, what is their source, and where do they have their effect?
Angiotensin(1-7), bradykinin, and substance P are vasodilators that are released by blood and local tissue. They have their effects at NO or PG receptors and on vascular smooth muscle
Is Angiotensin II a vasodilator or vasoconstrictor, what is its source, and where does it have its effect?
Angiotensin II is a vasoconstrictor that is released by the kidneys, blood, and local tissue. It has its effects at ATII receptors and on vascular smooth muscle
Is Arginine vasopressin a vasodilator or vasoconstrictor, what is its source, and where does it have its effect?
Arginine vasopressin is a vasoconstrictor that is released by the posterior pituitary. It has its effects at vasopressin receptors and on vascular smooth muscle
Are Norepinephrine and Epinephrine vasodilators or vasoconstrictors, what is their source, and where do they have their effect?
Norepinephrine and Epinephrine are vasoconstrictors that are released by the adrenal medulla. They have their effects at a-receptors and on vascular smooth muscle
Is endothelin a vasodilator or vasoconstrictor, what is its source, and where does it have its effect?
Endothelin is a vasoconstrictor that is released by the endothelial cells and has its effects at ET receptors and on vascular smooth muscle
What is autoregulation of blood flow and what organs have this feature
Autoregulation is an intrinsic characteristic to the brain, kidneys, and heart and involves being able to maintain a constant blood pressure despite changes in perfusion pressure
Do the coronary arteries have a high or low vasodilator reserve?
The coronary arteries have a high vasodilator reserve
When do the coronary arteries fill and the left ventricular receive its perfusion?
The coronary arteries fill during diastole and they deliver that pulsatile perfusion to the LV
What is the pathology behind Raynaud's Syndrome?
Excessive sympathetic vasoconstriction in response to cold
What are the two capillary beds in the kidneys and what are their distinct functions?
The glomerular capillaries are at high pressure to drive filtration. The peritubular capillaries are at low pressures to facilitate reabsorption
What are the two circulations in the lungs, what are their functions, and which side of the heart do they receive blood from?
* The bronchial circulation provides nutritive flow to the trachea and bronchial structures from the aorta.
* The pulmonary circulation supplies blood to the alveoli for gas exchange. It pumps right ventricle blood at low resistance and pressure. Unlike other organs, hypoxia will cause vasoconstriction (body needs more O2). Sympathetic stimulation increases resistance and mobilizes blood into the systemic circulation
What is the greatest contributor to racial disparities in life expectancy?
Chronic heart disease
From a population perspective, what are the leading risk factors for heart failure?
Hypertension leading to LVH, coronary artery disease, diabetes
What are the 4 leading risk factors for CVD in the US?
Smoking, diabetes, high cholesterol, hypertension. Obesity, diet, physical activity, education, genetics, socioeconomic status, and public policy all play factors as well
Are clinical interventions or risk factor reductions contributing more to the reduction in heart disease deaths?
Both clinical intervention and risk factor reduction are both contributing about 50% to heart disease death reduction
What are the 1˚, 2˚, and 3˚ measures for doctors trying to prevent CVD death?
1˚: Screen for risk factors, educate and counsel patients, treat with drugs and lifestyle modifications
2˚: Manage survivors to prevent recurrence (HTN, lipids, ASA)
3˚: Treat CVD, CVA, HF acutely using evidence-based strategies
What is the acronym for the 4 things patients should focus on to decrease their risk of CHD?
ABCS. Aspirin, blood pressure control, cholesterol control, smoking cessation
What is the silver bullet strategy vs the polypill strategy?
* Silver bullet: Pick drugs that are effective, combine them into one pill, with each drug at half dose to minimize side effects and give it to all with CVD and all adults over a certain age, regardless of risk factors
* Polypill: A statin, 3 BP lowering drugs (thiazide, ß-blocker, ACEi), folic acid, and aspirin. Start at 55 or CVD.
What are health disparities?
Population-specific differences in the presence of a disease, health outcome or access to healthcare