• 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

1/454

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;

454 Cards in this Set

  • Front
  • Back
Preload
passive load that establishes the initial muscle length of the cardiac fibers prior to contraction

(left ventricular end-diastolic volume)
after load
sum of all loads against which the myocardial fibers must shorten during systole.

aortic impedance

arterial resistance

intraventricular pressure

mass and viscosity of blood in the great vessels
contractility
speed and shortening capacity at a given instantaneous load (ionotropy)
diastolic compliance
the ability to fill at a given diastolic pressure
heart rate
frequency of contraction
ejection fraction
end diastolic minus end systolic/end diastolic volume

most wildly used clinical indicator of LV function
P wave
atrial contraction (depolarization)
QRS wave
ventricular contraction (depolarization)

hides atrial repolarization
T wave
ventricular relaxation
systemic vascular resistance =
blood pressure/ cardiac output
Blood pressure =
= systemic vascular resistance (SVR) x cardiac output
cardiac output =
HR x stroke volume
Thin filaments components
G-actin (globular) polymerizes to form F actin (filamentous)

Tropomyosyin helps hold F-actin together
Troponin complex
Troponin T - binds tropomyosin, anchoring the troponin complex

Troponin C - binds Calcium

Troponin I - binds to actin, inhibiting actin-myosin interaction
Thick filaments of muscle
4 lights chains - structural role

2 heavy chains - each has biding site for ATP and a binding site for actin
also has ATPase activity/motor activity
Sarcomere Organization

Z band
Thin filaments bind to z line
sarcomere organization

H zone
middle of the sarcomere (z to z is one sarcomere)
Sarcomere organization

M line
thick bind to M line
Muscle contraction

stages:
Attachment

release

bend

force generation

Attachment

repeat
muscle contraction

attachment
ATP not present

get myosin head binding to actin
Muscle contraction

Release
ATP causes detachment of myosin head from actin

ATP causes a conformational change, myosin head loses affinity for actin
muscle contraction

bends
myosin head bends to original state

movement initiated by breakdown of ATP to ADP

bends towards Z line
muscle contraction

force generation
myosin head binds weakly to a new actin molecule (closer to Z line) only when calcium is present (calcium dependent)

phosphate is released, increasing the binding affinity, head returns to original position. As it returns, it pulls the thin filament along thick filaments (power stroke)
Regulators of contraction:
ATP

calcium

magnesium - requires ATPase activity

Na and channels

beta adrenergic receptors
Cardiac muscle contraction
action potential moves down T tubule

AP detected by DHP receptors that contain Ca channel

DHP opens to allow a small amount of extracellular Ca into the fiber

Ca binds to ryanodine receptors in the SR, opening SR to release large amount of calcium

Calcium release allows contraction of cardiac muscle
Cardiac AP and calcium influx
sodium initially flows in for AP

calcium then flows in, depolarization occurs

repolarization occurs with the outflow of potassium ions
Function of intercalated disks
attachments of cells to other cells

allows conduction of AP linearly

only found in cardiac muscle
structure of intercalated disks
(1) transverse component
major component is Fascia adherens - holds cells together to form fibers

(2) later component

each component has cell to cell junctions

fascia adherens

maculae adherens - help prevent cells from pulling apart under tension, in both components

gap junctions - major element of lateral component
provide ionic continuity among adjacent cardiac muscle cells, permit cardiac muscle cells to behave as syncytium
SA node cells
small
disorganized

no intercalated disks

slow depolarization in the late stage of each action potential
AV node cells
located in right atrium, near lower part of interatrial septum

small, few myofilaments, less organized, no intercalated disks

depolarize slowly (delay signal from SA node) - allows for delay in contraction of atria relative to ventricles....atria squeeze all the blood into the ventricles before ventricles contract
Purkinje and bundle fibers
from the AV node, transmit signal

transitional cells that end on myocardium

Purkinje cells - larger than cardiac muscle cells

large amounts of glycogen, few organized myofilaments

infrequent intercalated disks, frequent gap junctions
Neural regulation of heart
ANS -

parasympathetics - from vagus synapse on post synamptic in heart, terminate in SA/AV node

Para - use ACh to decrease HR

sympathetics from T1-T6

use NE to increase HR
Sequence of heart contraction
(1) SA node generates impulse
(2) AV node delays impulse
(3) impulse passes from atria to ventricles via the atrioventricular bundle of his (bundle to apex, and purkinje fibers to apex)
Systole
contraction
diastole
relaxation
ventricular filling
mid to late diastole

blood pressure low

Av valves open

then atrial systole
ventricular systole
atria relax

rising ventricular pressure results in closing of AV valves

isovolumetric contraction phase

ventricular ejection phase opens semilunar valves

isovolumetric relaxation occurrs
Factors affecting stroke volume

Frank Starling Law
(1)preload - amount ventricles are stretched by blood before contraction

increase SV with low heartbeat or exercise

decrease SV with extremely rapid HR or blood loss

(2) contractility - increase with some drugs/hormones...NE
decreased with acidosis, increased extracellular K, calcium channel blockers...
depolarization of SA node process
slow leak of calcium in, calcium released within the cell, potassium repolarizes cells

phase 0,3,4 only
Sodium propagation of signal
hits an L type calcium channel at teh bottom of the T tubule

allows calcium influx

calcium binds to ryanodine receptor on SR

calcium released within the cell

calcium binds to tropomyosin complex and shifts tropomyosin I (inhibitor) away from the myosin/actin binding site

myosin head interacts with actin

phosphate from hydrolyzed ATP is released, myosin head bends (get contraction)

calcium regulated, ATP dependent shortening of filaments
cardiac cycle
ventricle contracts - isovolumetric increase in pressure

when pressure equals aorta, aortic valve opens...ejection of blood

end of systole - myosin/actin dissociate

isovolumetric decrease in pressure

when pressure equal to atria, mitral valve opens

atria contract, ventricle full...repeat isovolumetric contraction
Regulation of cardiac cycle by brainstem
sympathetics - NE binds to beta 1 receptor...sends signals to increase sodium and calcium influx...causes an increase in depolarization (increased HR)

parasympathetics - ACh binds to muscaranic receptors and causes a decrease in Na/Ca...causes hyperpolarization
Frank Starling Mechanism and preload
if you stretch a muscle, you increase its performance

increase preload by aortic regurgitation

exercise - rapid relaxation and lower pressure increases stroke volume

valsalva - decreases

atrial fibrillation - decreases preload
Frank Starling Mechanism and afterload
afterload is the force that the ventricle must overcome to eject blood (mean arterial pressure) or aortic valve/outflow tract resistance

higher afterload results in longer isovolumetric contraction and slower rate of muscle fiber shortening
Frank Starling Mechanism and contractility
change in end systolic pressure volume relationship

increased contractility results in a greater stroke volume at any given preload or afterload
CHF physiology
rest: ventricle can still get low enough pressure to fill

exercise: can't relax as well...ventricle can't get low enough pressure, atrial pressure must increase

to improve stroke volume, must have higher end diastolic pressure

results in higher Left Atrial pressure, which results in pulmonary congestion, pulmonary congestion causes shortness of breath, shortness of breath increases disability
CHF pathology
usually starts with myocardial insult

myocardial dysfunction - reduced system perfusion

hemodynamic defense systems activated (normally suppose to be transient)...get altered gene expression, apoptosis, remodeling
Hemodynamic defense systems
(1) Sympathetic - NE

(2) rennin angiotensin

(3) aldosterone
Sympathetic as hemodynamic defense
NE

binds beta/alpha receptors

organs - cardiovascular, adrenal gland

cardiac - increase in automaticity, contractility, afterload, myocyte death
Renin angiotensin as hemodynamic defense
Angiotensin II

organs - kidney, lungs, cardiovascular

increases afterload and preload, myocyte hypertrophy
Aldosterone as hemodynamic defense
aldosterone

organs - kidney, heart

causes cardiac fibrosis
Staging of CHF
(a) high risk but no structural changes in heart

(b) structural changes without syndrome

(c) syndrome controlled by therapy

(d) syndrome uncontrolled despite therapy
CHF symptom staging
1 no limitation of activity

2 mild limitation improves with slowing down

3 marked limitation improves only with rest

4 severe limitation or symptoms at rest
CHF types
(1) diastole - thick non functioning heart, unable to fill

(2) systolic CHF - thin, large dysfunctional heart, unable to pump and unable to fill
Diastolic CHF
no change in heart volume

unable to fill

concentric hypertrophy of myocytes

near normal stroke volume

normal end diastolic volume

steep diastolic pressure preload volume
Systolic CHF
unable to fill and unable to pump

increase in heart volume

eccentric hypertrophy of myocytes

steep diastolic pressure preload volume

low stroke volume

high end diastolic volume
Organization of the heart wall
(1) endocardium - endothelial cells over connective tissue
collagenous connective tiisue
simple squamous epithelial cells on surface

(2) myocardium - cardiac muscle

(3) epicardium - connective tissue with blood vessels and nerve fibers, covered by layer of mesothelial cells
endocardium
endothelial cells over connective tissue

collagenous connective tissue

simple squamous epithelial cells on surface
myocardium
cardiac muscle with small amounts of connective tissue

richly vascularized
epicardium
connective tissue with blood vessels and nerve fibers

covered by layer of mesothelial cells

contains nerve bundles, larger blood vessels, and large fat deposits
fibrous skeleton of the heart location and function
(1) where ventricles/atria met

(2) where great vessels arise

function (a) extends partially into interventricular septum to anchor valves

(b) eletrically isolates right atrium from ventricles
gross structure of valves
form as extension of the endocardium

leaflets of collagenous tissue covered by endothelium that is continious with tha tof the chambers and the great vessels

central fibrous sheet (lamina fibrosa) is formed by margining of fibroelastic supporting layers beneath the endothelium

cardiac muscle of the myocardium does not extend into the valve, the valves are passive structures
histological structure of the valves
spongiosa - atrial side, underneath the endothelium

fibrosa - forms the bulk of the dense connective tissue in the valve

ventricularis - the dense connective tissue with layers of collagen and elastic fibers
Atrial cardiac cells
contain endocrine granules filled with atrial natriuretic hormone

secrete hormone when overly stretched

increases excretion of water, sodium, and potassium by kidneys

decreases BP by inhibitin rennin secretion and aldosterone secretion
vascularization of cardiac muscle
enter through epicardium

form many anastomoses
Structure of blood vessels
(1) tunica intima - closest to lumen.

(2) tunica media - middle layer with elements oriented circumferentially

(3) tunica adventitia - outermost layer with elemeents ordered longitudinally
Tunica intima
closest to lumen

endothelial cells oriented along direction of blood flow

endothelium with basal lamina

internal elastic membrane

variable subendothelial connective tissue

substanital in elastic arteries

smooth muscle cells produce the collagen and elastic fibers
tunica media
middle layer with elements oriented circumferentially

smooth muscle

variable amount of elastin and collagen

dominant layer in arteries
tunica adventitia
outermost layer with elements ordered longitudinally

collagenous fibers and a few elastic fibers

major component in veins with significant smooth muscle

contains vaso casorum and nervi vascularis in large vessels
endothelium
provides smooth, low resistance surface for flow

cell bound by junctional complexes and may have pinocytotic vesicles

secrete factors that maintain smooth muscle

can be activated by cytokines to express cell adhesion molecules that allow WBC to stick
atherosclerotic plaque
occupies tunica intima and expands it

contains connective tissue fibers, smooth muscle cells, lipid containing macrophages, necrotic material, cholesterl clefts
tunica media
shows greatest variation and structural specialization

large elastic arteries - media in concentric layers of elastic and smooth muscle

muscular arteries - have prominent media of smooth muscle (circumferential) bounded by internal and external elastic laminae

arterioles - few layers of smooth muscle, very responsive to vasoactive stimuli

elastic arteries - intima is longitudinally oriented with subendothelial connective tissue
general structure of muscular arteries
sparse endothelial connective tissue

prominent internal elastic membrane

prominent smooth muscle in t. media

longitudinally oriented, incomplete lamellae connective tissue in t. adventitia
arteriole structure
smallest branches of arteries

start of microvascular system

intimate endothelial cells with basement membrane

t. media has 1-2 layers of smooth muscle, smooth muscle becomes discontinous

t. adventitia is insignificant

small lumen relative to thickness of vessel wall

prominent endothelial nuclei
venules
postcap -> collecting -> muscular venules

large lumen compared to vessel thickness

post cap - site of migration of leukocytes out of blood stream and into peripheral tissue

regulated by factors produced by endothelial cells and cells outside of circulation
medium veins
irregular profile, larger lumen, thinner walls

more distinct t. intima, media and adventitia

sparse subendothelial connective tissue beneath endothelial cells

adventitia thicker than media

smooth muscle cells in media and adventitia
vein valves
in small/medium

develop as inelastic extensions of the t. intima but covered by endothelial cells
structure of large veins
t intima thin

t media circumferentially oriented smooth muscle cells, fibroblasts, and collagenous fibers

t adventitia thickest layer, prominent bundles of smooth muscle separated by connective tissue
microvasculature structure
capillaries arise from metarterioles

smooth muscle in t. media is discontinous form precapillary sphincters

contraction and relaxation of these sphincters regulates blood flow

pericytes help regulate capillary diameter
microstructure of capillaries
thin walls to permit exchange

capillary wall - endothelial cells, basal lamina covers pericytes, scattered contractile cells (pericytes)

endothelial cells make contact without gap junctions (selective permeability barrier)
capillary types
(1) continuous - form tight junctions
movement of material requires carried mediated transport in pinocytotic vesicles...
muscle, never, connective tissue

(2) fenestrated capillaries - contain fenestrae bridged by diaphagms of extracellular material
basal lamina is continuous
pancreas, intestines, endocrine glands, renal glomeruli
sinusoids
discontinuous capillaries

large diameter, thin walled

large fenestrae without diaphragms, gaps between endothelial cells, little basal lamina

greater amount of transport

found in bone marrow, liver, spleen, lymphoid organs
blood flow and capillaries
thorough fare channels bypass capillary beds - no sphincters

contraction of precap sphincters directs flow from bed to AV shunts or other way around

blood flow - greatest total cross sectional area, lowest velocity

velocity of flow depends on total cross sectional area, not individual diameters
Portal systems
vein or artery between to beds

hepatic portal system, hypothalamic-hyphophyseal portal system, kidney efferent arterioles carry blood from glomerulus to peritubular capillaries
capillary exchange
hydrostatic pressure drives fluid out of caps

highest on arterial side
bulk flow in on venule side

protein stays in, water/oxygen/glucose out

Co2, N wastes, water move in
lymphatic capillaries
single layer of endothelial cells with an incomplete basal lamina

endothelial cells overlap partially but have intercellular spaces for access to lumen

no fenestrae or tight junctions

anchoring elements terminate on abluminal plasma membrane

more fluid in tissues means higher extracellular hydrostatic pressure, forcing fluid into lymph

small/medium have closely spaced valves to ensure one way flow

flow maintained by muscle contractions
lymph drainage areas
(1)Rt side - above diaphragm, rt side head/neck..rt upper arm

drains into rt subclavian

(2) left - left head/neck arm. lower body

temporarily stored in cistern chyli

thoracic duct carries lymph to left lymphatic duct

drains into left subclavian
Transcytosis
process by which molecules are transported from blood plasma across endothelial wall
Pathogenesis of Artherosclerosis
(1)LDL/VLDL binds to charged proteoglycans instead of normally leaking back out to blood

(2)oxidized lipidsinduce the endothelial cells to begin producing adhesion molecules (selectins, VCAM1 and MCP1)

(3)selectins allow monocytes and T cells to roll along surface and invade arterial wall

(4)monocytes become macrophages and begin producing pro-inflammatory cytokines that attrct more cells to invade wall to consume lipids

(5) macrophages also produce oxidizing agents that modify LDL/VLDL

(6)SR-A receptor binds to modified lipids
macrophage to foam cell
LDL endocytosed

goes to lysosome

chopped into fragments

fragments are effluxed to HDL or
make into cholesterol esters

Esters build up in cell = foam cell
role of PDGF in artherosclerosis
produced by macrophages

tells smooth muscle cells to migrate from t. media to intima

begin producing fibromuscular cap
T cell cytokines and smooth muscle cells
cytokines inhibit smooth muscle cell proliferation and collagen production

destabilize plaque
fate of macrophage foam cells
some die and relase contents within the intima

activates neighboring macrophages to produce more pro-inflammatory cytokines
plaque rupturing
macrophage foam cells secrete MMP that breaks down collagen

foam cells tend to concentrate at edges of plaque...destabilizing edges

rupture interrupts the anti-thrombogenic endothelial cell surface and causes platelet aggregation
major risk factors for atherosclerosis
age

male gender
primary risk factors for atherosclerosis
high LDL

low HDL

hypertension

cigarette smoking

diabetes
secondary risk factors for atherosclerosis
obesity

sedentary life style
Role of female sex hormones in atherosclerosis
progesterone and estrogen given after menopause increase risk of stroke, breast cancer with no affect on CHD

estrogen given close to menopause reduces risk atherosclerosis

unknown: protective if given at time of menopause

if estrogen is only reason for less atherosclerosis in women
Primary prevention for atherosclerosis
reduction of LDL and plaque = primary prevention

most cholesterol comes from synthesis (not diet)

statin drugs reduce LDL and reduce initial event for CHD
secondary prevention for atherosclerosis
stabilize plaque

aggressive lowering of plasma cholesterol concentrations stabilizes plaque and reduces risk of rupture
HDL and atherosclerosis
HDL mediates cholesterol efflux
binds to macrophage ABCA1 and ABCG1 transporters

reverses cholesterol transport

Anti-inflammatory by reducing expression of adhesion molecules (p-selectin, VCAM1) on endothelial cells

stimulates nitric oxide production which stimulates vasodilation
Resistance to blood flow
vessel length

blood viscosity

and RADIUS
resistance in vessels in series vs parallel
series - changing resistance of one vessel, dramatically changes over all resistance

parallel - changing one vessel has little effect on total resistance
parasympathetic
preganglionic long, synapse near organ

Neurotrans = ACh
Receptor = nicotinic

postganglionic are short

NT = ACh
receptor = muscarinic
Parasympathetic nervous in cardiovascular
vagal fibers innervate the heart...originate in the dorsal vagal nucleus/nucleus ambiguous

have tonic tone - always on

vagal efferents from medulla travel to SA (rt vagal) and AV (left vagal)
sympathetic nervous system
preganglionic fibers are short

NT = ACh
receptor = nicotinic

postganglionics are long

NT= NE
receptor = Alpha/beta adrenergics
Sympathetics in cardiovascular system
adrenergic control of heart and blood vessels originates in medulla

spontaneous activity - always on

increases HR and contractility
Neural control of circulation

Arterial baroreflex - sympathetics
carotid sinus and aortic arch

stretch receptors

fire rate increases with increased pressure (stretch)

respond to mean pressure, changes in pulse pressure, and pressure over time

very sensitive to small changes
BP neural projections in the medulla
initially project to nucleus solitarius

then go to either (1)depressor center - caudal ventrolateral medulla (CVLM) and decreases BP

or go to (2)pressor center - rostral ventrolateral medulla (RVLM) and increase BP

CVLM inhibits RVLM
parasympathetic baroreflex
tonically active...arise in dorsal vagal/ambiguous

(1)excitatory connection to dorsl motor nucleus of vagas (DMN X) and the nucleus ambiguous (NA)

(2)inhibitory projections directly on end organs
Cardiopulmonary reflexes
stretch receptors in the heart - particularly atrium

relate blood volume

increase in blood volume leads to increased sympathetic activation and diuresis
Chemoreceptor reflexes
monitor oxygen, CO2 and H+

peripherally located in carotid and aortic body

also centrally in medulla

Regulate: respiratory activity to maintain normal pH, pO2 and pCO2

send to nucleus solitarius
Humoral control
(1) catecholamines

(2)Renin-Angiotensin-Aldosterone System

(3)Vasopressin

(4)Atrial natriuretic peptide
Humoral control

catecholamines
released by adrenal medulla (epinerpherine and NE)

stimulates renin release and subsequently AngII and aldosterone
humoral control

renin angiotensin aldosterone system
regulates blood volume and pressure (long term)

kidney main organ

regulated by: sympathetics, renal artery hypotension, decreased sodium

mechanism: renin converts angiotensinogen to A1, A1 becomes A2. A2 modulates many cardiovascular events

stimulates muscle mass in ventricles
stimulates vasoconstriction
stimulates thirst
stimulates aldosterone - kidney retains fluid
humoral control

vasopressin
released by posterior pituitary

acts at kidney

(1) increases fluid reabsorption
(2)vasoconstriction
humoral control

atrial natriuretic peptide
stored atrial myocytes

released by stretch and Ang2

counter balances RAAS by vasodilation and fluid release
vasodilator reserve
difference between basal and maximal flow
AV O2 difference
measure of oxygen extraction between arterial and venous sides

small differences suggest that additional oxygen can be extracted without greatly increasing flow
active hyperemia
increase in blood flow in response to metabolic demand
reactive hyperemia
blood flow increase in response to brief periods of ischemia
Tissue Factor control of vasculature
originate in surrounding tissue or paracrine cells

vasodilator or constrictor

interact with endothelium or smooth muscle
endothelial factors and circulatory control
endocrine, paracrine, or autocrine

physical factors - shear stress

may act on endothelial or on smooth muscle

vasodilator/constrictor
myogenic response
originates within the smooth muscle of arteries/arterioles

occur in response to sudden change in pressure

increased pressure, smooth muscle constricts to preserve vascular resistance and diameter
vasodilators
NE, PE, O2
special circulations
(1) coronary

(2)cerebral circulation

(3)skeletal muscle

(4)cutaneous blood flow

(5)splanchnic circulation

(6) renal circulation

(7) pulmonary circulation
special circulation

coronary
major vessels lie on epicardial surface = low resistance

branches dive into myocardim - act as regulators

Pulsatile perfusion - diastolic flow...compressive forces high during systole so not a lot of flow during systole

large vasodilator reserve

highly regulated by tissue metabolism (adenosine)

little oxygen extraction reserve
special circulation

cerebral circulatoin
circle of willis

rigid structure so in flow must equal out flow

endothelium has tight junctions and a basement membrane = BBB

blood flow tightly coupled to oxygen demand

predominant control of cerebral circulation is determined by local factors (CO2)
special circulation

skeletal muscle
blood flow to satisfy metabolic demand

vascular anatomy is highly organized - capillaries can be recruited if high demand

type of exercise changes blood flow - sustained contraction (reactive hyperemia) vs phasic (active hyperemia)

control: myogenic tone

muscle contraction can occlude vessels

metabolites - can override sympathetic control of vascular tone
special circulation

cutatenous blood floow
primary function = maintain body temp

regulation is neural

regulation at deeper AV anastomoses

Neural control: tonic sympathetic vasoconstriction

Raynaurds syndrome - too much constriction (ischemia, cold hands)
special circulation

splanchnic circulation
receives large portion of blood volume

can redistribute blood elsewhere (compensatory adjustments) if blood loss

neural control: sympathetic innervations of arterioles and veins

high resting sympathetic vasoconstriction tone
special circulation

renal circulation
receives relatively high blood flow but is constant

primary function - salt and water balance...long term BP control through blood volume

anatomy: two capillary segments in series...glomerular and peritubular

important for filtration and reabsorption


strong auto-regulation

doesnt respond to sympathetic stimulation
special circulation

pulmonary circulation
(a) pulmonary - supplies blood to alveoli for gas exchange

dervived from RT ventricle

low resistance, low pressure

hypoxia causes vasoconstriction to preserve blood flow to functional alveoli

(b) bronchial
derived from aorta

nutritive flow to trachea and bronchial structures
Epidemiology of PAD
8-12 million Americans
chronic process evolving over several decades

M=F
Area most commonly affected in PAD
abdominal aorta

lesion area greater in women

greater in smokers

pain when stenosis >75%
PAD presentation
gradual onset

exertional aching pain, cramping, tightness, fatigue

occurs in MUSCLE GROUPS not joint

reproducible pain (exact same amount of exercise every time)

resolves with rest
Types of claudication
(1) intermittent arterial claudication

(2) spinal neurological or pseudo claudication

(3) venous claudication

(4) arthritic claudication....good days/bad days
pseudoclaudication
musculosketal disease

location in the same place

pain similar

but problems still occur at rest

good days bad days

pain doesnt go away with standing
lifestyle consequences of claudication
functional ability similar to NYHA class III CHF
Buerger's Disease vs atherosclerotic disease
athero - have cardiovascular risks

smoking common in both

men = women

location - aorto iliac, coronaries, carotids

phlebitis - very rare
thrombosis vs embolism in claudication
embolism - 70%

sudden, no distal pulses, no collaterals, frequently bilateral

source: cardiac
atheroemboli
paradoxical - DVT

thrombosis - 30%
distal pulses, collaterals,
origin - aneurysm
arterial occlusive disease - hyper coag state or atherothrombosis/buerger's disease
Atheromatous embolization
causes of thrombembolus

trauma, invasive procedures

legs most common site (then kidneys)

blue toe sign

livedo reticularis
risk factors for PAD
Age
diabetes
smoking

lipids: high triglycerides
low HDL

family hx in young
Ankle Brachial Index (ABI)
= lower extremeity systolic/brachial systolic

use highest brachial

lowest calculated ABI for diagnosis of PAD
Non-invasive evaluation of lower extremity circulation
(1) ABI

(2) Resting segmental pressures

(3) resting pulse volume recordings

(4) Exercise testing

(5) Duplex utrasound

(6) Transcutaneous oximetry
Sites of extremity atherosclerosis
any bifurcation in arteries

CFA, SFA, profunda femoris
tibial, peroneal
aorto-iliac
Major collateral pathways in legs
(1) lumbar aortic branches (aortic disease)

(2) internal iliac system and pelvic circulation (CIA disease)

(3) circumflex femoral arteries (CFA disease)

(4) profunda femoris (SFA disease)

(5) geniculate arteries (PA disease)
Leriche's Syndrome
focal occlusive disease of distal aorta and proximal iliac

findings - small caliber abdominal aorta
multiple periaortic adhesions

calcified atherosclerotic plaque and thrombosis

operation - aorto iliac endarderectomy; aorto bi iliac bypass
Post op for all PAD patients
BP control
130/80 for renal and diabetics
140/90 for others

lipid control

diabetes control

smoking cessation

platelet control
Plavix vs aspirin platelet control in PAD
plavix
Medical intervention for PAD
exercise

smoking cessation

cilostazol - improves claudication, but dont use with heart failure

MAO: inhibits PDE III
blocks platelet aggregation, blocks thrombosis, vasodilation of collaterals
Cilostazol
improves caludication
dont use with CHF patients

MAO: inhibits phosphodiesterase III
blocks platelet aggregation, blocks thrombosis, vasodilation of collaterals
Surgical intervention for PAD
better improvement of clinical symptoms
screening for PAD
any patient with exertional leg pain

>50 with risk factors

>40 with strong risk factors

all patients over 70
Renin in renal stenosis
less blood flow, release Renin

Renin converts angiotensinogen to A1

ACE converts A1 to A2

A2 causes:

vasoconstriction
renal sodium retention
aldosterone secretion
LV Hypertrophy, remodeling, endothelin release
sympathetic nervous activation
Unilateral renal stenosis and renin
abnormal kidney activates RAAS

contralateral has increased perfusion, increases sodium excretion

net effect: plasma renin elevated
bilateral renal stenosis and renin
both activate renin

impaired sodium, water retention

once volume adequate get steady state: plasma renin is normal

patient is hypertensive
clinical implications of renal stenosis
paroxysmal hypertension - marked spikes in BP

treatment resistant hypertension

hypertensive nephropathy

renal failure

flash pulmonary edema

accelerated cardiovascular disease
children with renal lesions
correctable before 10 yo
prevalence of adults with hypertension
24% have renal vascular disease

older and BP>110 and renal insufficiency = more likely to have stenosis
causes of renal stenosis lesions
(1)atherosclerosis - 75%

(2) fibromuscular dysplasia - 20% and in younger people

(3) miscellaneous
fibromuscular dysplasia
(1) medial fibroplasias - 85%
more common in women

(2) intimal fibroplasias - 5%
focal and in children

arteries affected:
renal
carotid
iliac

can have aneurysms
tests for reno-vascular disease
(1) functional -

rapid sequence excretory urography

peripheral plasma renin

isotopic renography

(2) anatomic

angiography
MRI/MRA/CTA
isotopic renography
measures filtered radioactive tracer before and after ACE inhibitor

angiotensin II maintains GFR in kidney with renal stenosis via increased vasoconstriction in efferent arteriole

ACE inhibitor reduces synthesis of A2, diminishes efferent arteriole vasoconstriction and thus a decreased GFR

therefore less uptake of filtered radioactive tracer after ACE inhibitor
when to intervene in renal stenosis
severe

bilateral

rapidly deteriorating renal function

normal distal renal artery
visceral ischemic syndromes: mesenterics

acute vs chronic
acute - sudden onset
usually artery occlusion

mesenteric vein occlusion - more likely to cause gangrene of bowel

non-obstructive mesenteric ischemia - no lesion, large portion threatening to die off...just enough ishcemia to cause potential death

chronic - due to atherosclerosis
incidence of mesenteric ischemia
mesenteric artery occlusion - 75%

thrombus - 65%
embolus - 35%

mesenteric vein occlusion - 15%

non-obstructive mesenteric ischemia - 10%
risk factors for mesenteric ischemia
age >60

atrial fibrillation

CHF

recent MI

clotting disorder

hypercoaguable

hypOvolemia
embolus vs thrombosis in visceral ischemia
embolus - sudden onset
paucity of physical findings - extreme pain out of proportion

bowel evacuation

cardiac embolic risk

thrombosis -
insidious abdominal pain
systemic toxicity
mesenteric vein occlusion
insidious, diffuse abdominal pain

vague prodromal period
nausea, vomiting, distention
hypovolemic cardiovascular collapse

causes:
secondary
infection
clotting disorder
inflammatory bowel disease
NOMI
spasm of blood vessel in bowel
chronic mesenteric ischemia
fear of food
weight loss
post-prandial (after eating) abdominal pain
dx of ischemic visceral syndromes
CT
angiography
duplex
tx of ischemic visceral
can use local medical

embolus - surgery to remove
survival of visceral ischemia
remote organ dysfunction - systemic inflammatory response
main reason for death
renal insufficiency
hepatocellular dysfunction

survival - 20% in five years
venous anatomy of legs
deep system - within muscle and fascia

superficial system - drains into the deep

perforators connect the two
vein wall
has some components as an arterial wall (intima, media, adventitia)

muscular layer (media) not prominent in veins
venous physiology
post capillary blood collects in venules, drains into larger veins

superficial drains to deep
vein valves
allows flow towards larger tracts

prevent back flow

often found at junction and perforators

reduce column heihgt of blood to reduce STATIC PRESSURE
hydrostatic pressure in veins
static pressure of fluid column

transmitted right atrial pressure
overall volume status
thoracoabdominal pressure
muscle pump in veins
hydrodynamic pressure

calf muscles surround deep veins and the muscles are covered in fascial linings

muscle contraction increases the compartment pressure and forces blood out of the deep veins

valves prevent back flow to superficial system...propel blood to deep veins
pathophysiology of venous disorders
(1) valve failure - increased column height increases static pressure

(2) obstruction

(3) thromboembolus

effects: additional valve failure
capillary bed - edema inflammation
chronically - get structural changes and inflammation of surrounding tissue
varicose veins
genetic component
valve incompetence - saphenofemoral, saphenopopliteal, perforators

pain, aching sensation

Tx. compression stocking, elevation, removal of varicosities and any reflux
Telangiectasias - spider veins
same proces as varicose veins but on small scale

symptoms - cosmetic, occasional pain

tx - exclude larger vein reflux injection sclerotherapy, laser
skin changes with venous stasis
lipodermatosclerosis

pgimentation - hemosiderin deposition

eczema

ulceration - often perforating vein

gaiter area - common site
management of varicose veins
aggressive compression

elevation

antibiotics and debridement when infected

skin grafts
DVT

risk factors
local effects and embolization

age

prior hx

trauma

malignancy

exogenous estrogens

obesity

surgery - hip, knee, pelvic

malignancy - sometimes get DVT many years later get a malignancy
DVT dx, hx, pe
warmth, redness

homan's sign

d-dimer labs

imaging is the best...venous duplex
DVT sequlae
local - swelling distal
long term potential for dysfunction (morbidity)

systemic - postphlebitic syndrome or thrombo-emboli (mortality)
DVT prevention/tx
mechanical compressors

pham - anticoags

tx. anti coags and compression socks
Phlegmasia Cerulea Dolens
limb threatening extensive venous trhombosis

often associated with cancer

limb threatening, need emergency surgery
lymphatic disorders
extensive interstial fluid enters lymphatic capillaries

have valves too, usually fail

drainage dependent on compression forces

80% drain into left chest
lymphedema
protein rich

interstial volume overload

failed lymph drainage
edema
increased interstial fluid volume
primary lymphedema
inherent defect within lymph conduits
secondary lymphedema
acquired damage to lymph conduits
types of primary lymphedema
(1) congenital - milroy's disease

(2) lymphedema praecox - most common <35 yo
usually unilateral, adolescent women
hypoplastic lymphatics

(3) lymphedema tarda - congenitally weakened lymphatics event triggers onset >35 yo
secondary lymphedema
obstruction

most common in the world = filariasis

most common in US = treatments from other malignancy
skin changes with lymphedema
fibrosis

hyperkaratotic
tx for lymphedema
no cure

elevation minimal effect

exercise helpful

compression with massage

manual lymph drainage
Stroke -
focal neurlogical deficit related to neuronal cell death with signs and symptoms lasting greater than 24 hrs
TIA
a focal neurological deficit with signs and symptoms lasting less than 24 hrs
risk factors for stroke
Hypertension - most major modifiable risk factor

increasing age

cardiovascular disease: atrial fibrillation

smoking

carotid stenosis
stroke etiology
ischemia - 80%
embolism - major cause

hemorrhage - 20%
stroke pathophysiology
cardiac most common source - atrial fibrillation

atherothrombotic carotid stenosis

lacunar infarcts - diabetes and hypertension
atherothrombotic stroke
major cause of preventable stroke

carotid disease

vertebrobasillar disease

hypotension - watershed strokes
carotid disease and stroke
anatomy - disease of the carotid bulb
rich source of emboli
embolic risk related to plaque burden

predominantly atherosclerotic

other causes - fibromuscular dysplasia

arthritis - takayasu's
giant cell arthritis

dissections

radiation
risk factors of carotid disease and stroke
advanced age

cigarette smoking

HYPERTENSION

cardiovascular disease

hyperlipidemia
carotid disease stroke presentation
TIA

amaurosis fugax

completed stroke

vertebrobasillar symptoms
TIA presentation
contralateral paresis

contralateral numbness

aphasia
Amaurosis fugax
temproary ipsilateral monocular visual loss due to embolization of retinal artery or branches

visual field defect

not scotomas or floaters
dx of carotid disease
carotid duplex

CTA arteriography

MRA
carotid disease natural hx
depends on presentation

asymptomatic

symptomatic - 2 year stroke risk of 15-30%
carotid disease tx
secondary prevention/medical management

hypertension, smoking lipid, diabetes, antiplatelets
surgical recommendations for carotid disease
based on severity of stenosis and symptoms

(a) optimal medical therapy without revascularization in symptomatic patients with <50% stenosis

(b) medical when asymptomatic in patients <60% stenosis

(c) carotid endarterectomy plus medical in symptomatic 50-99% stenosis

(d) endarterectomy plus medical in asymptomatic 60-99% stenosis and low perioperative risk
vertebrobasilar disease
embolization or low flow

symptoms - dizziness, vertigo, diplopia, blurred vision, ataxia, drop attacks

epidemiology - much less common than carotid disease
risk factors for vertebrobasillar disease
`age

smoking

cardiovascular disease
symptoms of vertebrobasilar disease
more common from flow related phenomena

systemic hypotension, brady/tachy. autonomic dysfunction/compression
arteries potentially affected in vertebrobasillar disease
innominate

subclavian

vertebral

carotid
presentation of vertebrobasillar
dizziness

vertigo

diplopia

blurred vision

ataxia
vertebrobasillar and precipitating factors
need to know

problems when head turning?

problems when rising/standing?

problems when using upper extremities?
dx of vertebrobasilar
pulse examination of upper extremity blood pressures

duplex - transcranial doppler

imaging
tx of vertebrobasilar
hypertension management

smoking

lipid, diabetes, antiplatelet agents

increase pressure to basilar
- subclavian or vertebral artery
tx of extracranial occlusive disease with global hypoperfusion
treat anterior carotid disease first
Giant cell arteritis (temporal arteritis)
temporal headaches

jaw claudication

polymyalgia rheumatic

amaurosis

constitutional symptoms

dx temporary artery biopsy

tx corticosteroids
carotid artery dissections
spontaneous

traumatic

can lead to pseudoaneurysms
aneurysm
focal dilation of an artery involving an increase in diameter of at least 50% as compared to expected diameter

true - have the vessel layers

false - have none of vessel wall layers
aortic aneurysms
75% of all aneurysms

male> female

atherosclerotic

below renals
pathogenesis of aortic aneurysms
(1)smooth muscle

(2)matrix proteins - elastin, collagen
decreasing matrix concentrations from proximal to distal aorta

(a) elastin - load bearing protein
(b)collagen - safety net...resists AAA rupture

(3) up regulation of matrix metalloproteinases

(4) infection - chlamydia
(5) autoimmune/inflammation
(6) genetic
Epidemiology of AAA
3-10% of people over 50yo
risk factors for AAA
elderly white males...peak at 80yo

M>F

white>AA
presentation of AAA
asymptomatic

occasionally feel pulsation
rupture of AAA symptoms
most symptomatic

abdominal or back pain

most palpable
dx of AAA
imaging

ultrasound or CT

angiography not good for screening...use CTA more often
AAA screening
males >65

smokers

family hx
risk of AAA rupture
larger

>4 cm, screen every 6 months

5.5 cm surgery suggested
when to do surgery for AAA
>5.5 cm diameter

life expectancy is greater than 2 years

balance rupture risk, operative risk, life expectancy
co-morbidities of AAA
coronary artery disease

pulmonary disease

renal failure
complications of AAA repair
early - MI

late - graft infection
Aortic Dissection
non radiating chest pain

separation of aortic wall layers by extra luminal blood...usually through tear in intima

blood may circulate between aortic lumen and abnormal channel

uncommon clinical event

survival low
risk factors for Aortic dissection
male>female

location - ascending in 50-55yo
descending aorta 65-70yo

chronic systemic hypertension

aortic diseases

bicuspid aortic valve

trauma

cocaine

pregnancy

rebound from abrupt discontinuation of beta blocker therapy
Marfan syndrome
hereditary connective tissue disorder

auto dominant

defect in fibrillin 1

affects skeleton, heart, major vessels, lungs, eyes

get dilation of aorta
DeBakey classification of Aortic Dissection
type 1 - starts at valve goes all the way down

type 2 - just valve

type 3 - descending aorta
Tx of aorta dissection
ascending - surgery + medical control of bp (beta blockers)

descending - control with medical therapy for BP (beta blockers)
clinical manifestations of aortic dissection
sharp pain

doesnt radiate

hypertensive

diastolic murmur
dx of aortic dissection
imaging - CT, MRI
peripheral and visceral aneurysms
femoral - most common false aneurysms

popliteal - most common true aneurysm

popliteal - tend to be limb threatening...more likely to thrombose of embolus then rupture
more likely atherosclerotic

male>female

strongly associated with aneurysms elsewhere in body
popliteal vs femoral aneurysm
popliteal - most common true anuerysm

usually atherosclerotic


femoral - more common false aneurysm

both more likely to embolus/thrombos then to rupture
visceral aneurysms
splenic artery most common

biggest concern is rupture

increased risk with pregnancy

diameter 2-3cm
BP =
(left ventricle end diastolic volume - left ventricle end systolic volume) x HR x SVR

depends on:

how full at end of diastole

how empty at systole - strength of contraction, difficulty in ejecting blood

heart rate

resistance in vasculature
alpha 1 receptors
peripheral and splanchnic vasculature in smooth muscle

few in heart muscle

stimulation: contraction of smooth muscle = vasoconstriction
alpha 2 receptors
peripheral and splanchnic vasculature

fewer than alpha 1

in smooth muscle, cause contraction

on presynaptic neurons, has negative feedback so less NE released with stimulation....net effect = less effect of future drugs

also present in CNS causes sedation and pain relief
beta 1 receptors
cardiac tissue

inotropic effect - increased contractility

chronotropic effect - increased HR

lusitropic effect - relaxes faster in systole

peripheral - causes smooth muscle relaxation/vasodilation....minor role

presynaptic receptors: causes positive feedback...increases enhanced release of future stimulus
beta 2 receptors
cardiac tissue - much more beta 1 though

ratio of beta 1 to beta 2 increases in CHF (lose beta 1s)

same effect as beta 1:
inotropic
chronotropic
lusitropic

peripheral - minor vasodilation

bronchial smooth muscle - causes bronchodilation/relaxation
dopamine receptors
cardiac tissue: 2 types
minimal effect but what it does is:

iontropic

CHRONOTROPIC

splanchnic vasculature- causes vasodilation...major role of dopamine receptors

CNS
vasopressin receptors
peripheral vasculature - marked vasoconstriction

splanchnic vasculature - marked vasoconstriction
beta receptor complex:
drug binds to receptor

receptor interacts with G protein

G protein interacts with intracellular bound adenylcyclase

adenylcyclase converts ATP to cAMP

cAMP activates numerous cascades...one activates a phosphorylase that binds to calcium channels

increaess amount of calcium that can come in, this increases amount of SR released calcium

get strong contraction, also allows for calcium to get pumped out faster so relaxation faster

phosphodiesterase degrades cAMP
preload manipulation
can't measure directly

increase: give fluids

alpha agonists in low doses
afterload manipulation
left ventricle end systolic volume

not measured = amount coming out of ventricle and resistance to pumping out

estimated with Hx and PE

rx: vasodilators

ACE inhibitors, nitroprusside
PDE inhibitors
contractility manipulation
also part of end systolic volume

not measured

estimated by hx and PE

= force generated by LV

Rx. inotropes
give beta
PDE inhibitors
HR manipulation
chronotropy

easily measured

at high rates, cant fill fast enough

rx. chronotropes
beta, atropine, aminophyline, pacemaker
SVR manipulation
vascular resistance

not measured, calculated

rx. vasoconstrictors - alpha and vasopressin
epinepherine
beta effects predominate (has some alpha at higher doses)

inotropic and chronotropic (Beta)

use - sepsis, cardiogenic shock
dobutamine
inotrope with vasodilator (reduces afterload)

beta mostly

increases contractility and HR

vasodilates and reduces afterload

Use - MI
norepinephrine
alpha effects - overwhelming vasoconstrictor

some beta high doses

use - sepsis helps support heart in high doses
phenylephrine
pure alpha, but weak

vasoconstriction and increased SVR

mild sepsis, hypotension
dopamine
use for renal support

beta and alpha at very high doses

one drug, every receptor
vasopressin
intense vasoconstriction, no alpha or beta

use - severe sepsis
phosphodiesterase inhibitors
vasodilators and inotropes

augements beta stimulation

smooth muscle relaxation

keeps cAMP around longer
MI pathology general
chronic - coronary atherosclerosis

acute - changes in plaque
Dx of MI
coronary arteriogram
Acute symptoms of MI
unstable angina

Non ST- elevation myocardial infarction

ST elevation myocardial infarction
Acute Coronary syndromes
result from diminished coronary perfusion relative to demand

usually a hemorrhagic rupture of a fat laden, unstable atheroma with development of an occlusive platelet thrombus

plaque rupture exposes interior to blood (1) platelet aggregation (2) coagulation thus get thrombus formation
MI pathology specific
Initially, a sudden change occurs in the morphology of an atheromatous plaque - intraplaque hemmorrhage, ulceration, or fissuring
Platelets are exposed to subendothelial collagen and necrotic plaque contents, leading to adhesion, activation, and aggregation of platelets. This leads to buildup of platelet mass which gives rise to emboli or potentiates occlusive thrombosis.
Simultaneously, tissue thromboplastin is released, activating the extrinsic pathway and fibrin deposition.
Adherent activated platelets release thromboxane A2, serotonin, and platelet factors . These predispose to coagulation, favors vasospasm, and adds to the bulk of the thrombus.
The thrombus evolves to become occlusive(often within minutes).
uncommon MI pathology
erosion of endothelium exposing interior plaque

vasospasm from cocaine

emboli from left atrium
factors affecting MI plaque rupture
Sudden changes in intraluminal pressure or tone
Bending and twisting of an artery during each heart contraction
Lipid content of plaque
Thickness of fibrous cap
Plaque shape
Mechanical injury
myocardial response to coronary occlusion
Get ischemia in region supplied
Starts at subendo and progresses to epicardium
Get anaerobic glycolysis…leads to inadequate production of ATP and accumulation of noxious breakdown products

Eventually get necrosis
In absence of reperfusion – hypoxia and noxious metabolites leads to impaired ventricular function…predisposing to lethal arrhythmias
Get transient diminished diastolic relaxation, abnormal systolic contraction, abnormal wall motion, diminished stroke volume
dx of MI
With symptoms of ischemia
ECG
Pathological Q waves
presentation of MI
Acute onset of chest pain radiating to left arm and neck, shortness of breath

Atypical chest or epigastric discomfort
risk factors for MI
Hypertension
Smoking
High ldl, low hdl
DM
Family hx
Sedentary lifestyle
Obesity
Age
cardiac markers for MI
Detection in plasma released by injured myocardium
Troponin most specific
Can use CK-MB
AST, LDH
Look at MB to CK, peaks early
LDH peaks late
Troponin peaks a few days after MI

Troponin and CK don’t begin evaluating until at least 4 hrs

Troponin correlated with outcome
ECG findings in MI
ST elevation
T waves get very high..earliest sign of injury
Ongoing injury will get ST elevation
Thought to be persistent occlusive disorder…no flow restored

Non ST Elevation MI
More likely to have multivessel CAD
Prior MIs
Diabetes
fields of coronary arteries
LAD – anterior, spetum/anteroseptum, anterolateral

L Circumflex – lateral or posterior

RCA – inferior
Prognosis of MI
Early – presence of arrhythmias

Late – determined by left ventricular function
complications of MI
Arrhythmias

CHF
Cardiogenic shock

Pericarditis

Scar or ventricular aneurysm
CHD epidemiology in women
leading cause of mortality

major cause of disability - 55% over 75yo

prevalence equal with men at 40-59yo
Gender differences in MI
Men younger

Women less likely to undergo cardiac workup when presenting with chest pain

Non-invasive studies have decreased sensitivity and specificity in women

Higher in hospital stay

Worse long term mortality

Women have more risk factors

Women have higher silent MI

Women have atypical pattern for angina such as neck/jaw pain, dyspnea or fatigue
coronary artery disease risk factors
Hypertension

Diabetes mellitus - relative risk increases with heart disease

Dyslipidemia

Smoking

Age (men >45 women >55)

Family hx
obesity in women and MI
increasing problem in the US

women started fatter, getting fatter

looking at wast circumference to hip
higher ration, higher risk of CHD
Smoking and women with MI
women smoke less than men

but still risk factor
guidelines for LDL in MI
<160 with 0-1 risk factors

<100 with >2 risk factors

<70 with CHD
C reactive protein and CHD in women
CHD - sensitive marker for inflammation

associated with CHD

statins will reduce
Alcohol risk vs benefit in MI in women
drinking less than 2 a day is slightly beneficial in no risk factors

>1 risk factors, even better improvement

>2 drinks, increases risk in women
gender differences in risk factors
Diabetes much stronger predictor in women than men

High triglycerides worse for women

Physical inactivity more prevalent in women

Smoking may be stronger

CRP levels higher in women
Sex hormones in women with MI
estrogen - modifies lipid, arterial vasodilation, clot formation

progesterone unclear

HERS study: HRT was associated with increase in invasive breast cancer without CAD benefit


increased stroke and PE
types of atherosclerosis lesions
fatty streak - innocuous, small, intracellular fat in intima

atheroma - raised, lipid center

fibrous plaque - innocuous, intimal scar...problem when it ruptures
Atheroma
sites: intimal, may involve media
arteries - aorta and its major branches

elements: necrotic center of extracellular lipid
fibrous cap - in contact with blood

proliferation cells - myofibroblasts


complications: within plaque - ulceration, fissuring
thrombosis - often caused by ulceration

medial damage - causing aneurysms

hemorrhage into plaque

calcification

insidious: usually asymptomatic until 75% stenosis
atheroma effects and clinical complications
Aortic branches: aneurysm, mural thrombus, rupture

Cardiac: infarct, ischemia, dysrhythmia, sudden death

GI: ischemia, infarction

Brain: infarction (Stroke), ischemia

Peripheral: ischemic atrophy, claudication and gangrene

Clinical complications of atherosclerosis:

Carotids and vertebral: cerebrovascular disease
Narrow, ulcerate, embolize, cause ischemia or infarct

Coronary arteries: ischemic heart disease
Narrow, ulcerate, or thrombose, causing ischemia, infarct, arrhythmia, heart failure

Peripheral arteries: PVD
Ischemia, gangrene, usually lower limbs or intestinal

Aorta: aneurysm, rupture, atheroembolism, mural thrombus with thrombembolism
properties of ion channels
selective but passive

unidirectional
properties of exchanger channels
selective and passive

exchanged based on differences in concentration...not necessarily 1 to 1, can change direction depending on relative gradients
properties of pump channels
selective and actively transport ions across membrane against concentrations or voltage gradients

ATP used

in heart

increase intracellular potassium and decrease intracellular sodium/calcium
myocyte ion concentrations at steady state
K in

ca/Na out

negatively charged inside (proteins inside are negative) -90mV
cardiac myocyte channels at rest
Na closed
Ca closed
some K closed
some K open

cell is actively pumping Na/Ca out

K is leaking out passively
Cardiac AP

Phase 4
resting state

Na closed

Ca closed

K closed, some leaking out

-90mV
Cardiac AP

Phase 0
rapid depolarization of Na into cell
Cardiac AP

Phase 1
very brief

some transient outward current corrects for over shoot of depolarizing
Cardiac AP

Phase 2
plateau phase

calcium channels open - calcium into cell

Na closed

calcium takes long time to come in, the other receptors hold depolarized state

Calcium binds to SR, letting out more calcium

(Calcium activated calcium release)
Cardiac AP

Phase 3
repolarization

calcium channels close

Na closed

K only ones open

lose positive charge

get to -90mV
QRS and cardiac cycle
phase 0
ST and cardiac cycle
phase 2
T wave and cardiac cycle
phase 3
Refractory periods of cardiac cycle
(1)absolute refractory period

(2) effective refractory period

(3)relative refractory period
Absolute refractory period
all Na inactivated

Ca open

cell is completely inexcitable

Phase 2

cant make a new AP
Effective Refractory Period
absolute + part of phase 3

Na channels still closed

Some Ca channels closed - meaning they can reopen

any inducible AP will be too weak to propagate

Effectively cell is inexcitable
Relative refractory period
cell is beginning to repolarize

not at rest yet (in phase 3)

Na channels are still inactivated

Ca channels are mostly closed but some can be reopened

strong stimulus can induce an AP that will be lower in amplitude and slower in velocity...strong enough to propagate
Automaticity
ability of cell to generate a tone rhythm

in cardiac cells - phase 4 resting takes external stimulus to get to threshold

but some cells have inward leak of positive ions in phase 4 (pacemaker current - SA, AV, His/purkinje bundles

mostly sodium, some calcium

continueal leak of opsotive ions does bring cell to threshold, generating automatic rhythm

SA/AV so leaky cant actually get to -90 or even threshold

depolarization is driven by Ca channels because they do not have Na channels
difference in nodal cell AP and cardiac AP
phase 0 - slower in nodal

phase 1 - does not occur in nodal (no Na to overshoot)

phase 2 - Ca drive phase 0 instead

phase 3 - occurs normally
autonomic nervous system on cardiac AP
affect slope of phase 4 depolarization

sympathetic increase slope (epi) - mediated by calcium channels

parasympathetics decrease slope and slightly hyperpolarize at end of phase 3...mediated through calcium channels
Atrial myocytes and current flow
repolarize quickly

able to sustain rapid arrhythmias (refractory period)
His, bundle, and purkinje and current flow
ERP longer than atrial

transmit signal through ventricles

all basically same type of cells

transmit individual heart beat quickly

have more Na channels - each individual cell activates faster, plus more gap junctions, plus better gap junctions
AV node and current flow
some automaticity, but real purpose is to conduct SA node

moves slowly - protects from atrial fibrillation becoming ventricular fibrillation
dromotropic
alters current propagation
sympathetic stimulation of cardiac cycle
positive chronotropic, positive dromotropic
parasympathetic stimulation of cardiac cycle
negative chronotropic

negative dromotropic
Bradyarrhythmias
(1) failure to generate
SA node automaticity failure - ischemia, fibrosis, metabolic disorders

(2) failed impulse propagation
SA block - current blocked
AV block - physiologic or pathologic

His bundle block - infranodal
Escape rhythms
cells with normal automaticity - sinus node, AV node and his bundle, bundle branches, purkinje system

faster one wins
normal sinus node beat
60-100 bpm
normal AV node/His bundle beat
40-60 bpm
normal bundle branches/purkinje system beat
30-40 bpm
ectopic atrial pacemakers
40-90s

blurs line between normal and abnormal

usually not referred to as an escape rhythm

reliable rhythm
AV node/His bundle (AV junction)
junctional escape

good but less reliable, more easily suppressed
Purkinje Network
includes bundle branches

ventricular escape

not reliable, easily suppressed
Tacharrhythmias
mechanism: abnormal automaticity

triggered activity

reentry

(1) abnormal automaticity - ischemia or metabolic derangement transforms phase 4

activity of atrial or ventricular myocyte

(2) triggered activity

after depolarizations
had normal, then after you get something extra...if big enough get another depolarization

early - phase 3
can be self perpetuating
likely to get Torsades de Pointes - prolonged QT interval

delayed - phase 4
intracellular calcium overload
digoxin toxicity

(3) reentry
Typical AV node reentry tachy
normal: two pathways are present but during normal sinus rhythm wave fronts hit each other when cells are in refractory period so conduction is extinguished

slow pathway
fast pathway

abnormal: fast pathway is blocked
slow pathway continues past where it would normally be extinguished, if it arrives at the right moment, cells can be reactivated and AP loops back onto the slow pathway

unidirectional block
premature beat
beat comes in very soon after preceding beat
atypical AV node reentry
get block in slow pathway

fast pathway circles around, and signal reenters through fast pathway
Wolff-Parkinson White syndrome
accessory conduction pathway penetrates fibrous skeleton of heart

AV would be slow pathway

accessory would be fast pathway
Atrioventricular reentrant tachycardia
with a block in the accessory (fast pathway), signal goes down AV node and comes back up through accessory
VT after MI
primary mechanism of death from MI

scar becomes slow pathway

normal becomes fast

get reentry through infarct
Class 2 antiarrhymthic drugs
beta blockers

Beta 1 in heart
Beta 2 in vascular smooth muscle, bronchial smooth muscle

MOA: block catecholamines of ANS

(1)negative chronotropic effect - slow automaticity in sinus node and AV node by slowing phase 4

(2) negative dromotropic effect - slows velocity of conduction through AV node. slow ventricular response rate during atrial tachycardia

(3) negative inotropic effect

Types (a) cardioselective - beta 1 only
(b) non-selective - 1, 2 and some alpha

intrinsic sympathomimetic activity - block and stimulate beta receptors

adverse: sinus bradycardia
AV block

can worsen patients who already have severe decompensated heart failure
cardioselective beta blockers
atenolol

metoprolol

esmolol
atenolol
class 2 antarrhymthmic

beta blocker

cardioselective


adverse: sinus bradycardia
AV block

can worsen patients who already have severe decompensated heart failure
metoprolol
class 2 antarrhymthmic

beta blocker

cardioselective


adverse: sinus bradycardia
AV block

can worsen patients who already have severe decompensated heart failure
esmolol
class 2 antarrhymthmic

beta blocker

cardioselective

adverse: sinus bradycardia
AV block

can worsen patients who already have severe decompensated heart failure
nonselective beta blockers
propranolol

lebetalol - also block some alpha

carvedilol - also block some alpha

adverse: sinus bradycardia
AV block

can worsen patients who already have severe decompensated heart failure
propranolol
class 2 antiarrhymthmic

non selective beta blocker

blocks 1 and 2

adverse: sinus bradycardia
AV block

can worsen patients who already have severe decompensated heart failure
lebetalol
class 2 antiarrhymthmic

non selective beta blocker

blocks beta 1 and 2 and alpha


adverse: sinus bradycardia
AV block

can worsen patients who already have severe decompensated heart failure
carvedilol
class 2 antiarrhymthmic

non selective beta blocker

blocks beta 1 and 2 and alpha

adverse: sinus bradycardia
AV block

can worsen patients who already have severe decompensated heart failure
Class 4 antiarrhymthmic
calcium channel blockers

Types - dihydropyridine (DHP)
non dihydropyridine (non-DHP)

diltiazem
verapamil

block Ca channels in heart (DHP)
only affect sinus and AV nodal cells (mostly AV)

effects: negative chronotropy - sinus node slowed down
AV - junctional escape shutdown

Negative dromotropy
effect on AV node: terminate reentry circuit

negative inotropy
contraindicated for patients with systolic heart failure

adverse:
too much negative chronotropy, dromotropy, inotropy
hypotension
peripheral edema
drug interactions
diltiazem
block Ca channels in heart (DHP)
only affect sinus and AV nodal cells (mostly AV)

effects: negative chronotropy - sinus node slowed down
AV - junctional escape shutdown

Negative dromotropy
effect on AV node: terminate reentry circuit

negative inotropy
contraindicated for patients with systolic heart failure

adverse:
too much negative chronotropy, dromotropy, inotropy
hypotension
peripheral edema
drug interactions
verapamil
Class 4
block Ca channels in heart (DHP)
only affect sinus and AV nodal cells (mostly AV)

effects: negative chronotropy - sinus node slowed down
AV - junctional escape shutdown

Negative dromotropy
effect on AV node: terminate reentry circuit

negative inotropy
contraindicated for patients with systolic heart failure

adverse:
too much negative chronotropy, dromotropy, inotropy
hypotension
peripheral edema
drug interactions
digoxin
form of digitalis, cardiac glycoside

positive inotrope: poisons Na/K pump
increases Na concentration inside cell
Na/Ca exchanger keeps more Ca in cell to balance
net: more calcium enter SR so more Ca released

Use: increases vagal tone

AV node primary target - no hypotension, mild positive inotrope
less effective than older drugs at slowing AV nodal conduction
narrow therapeutic index

adverse:nausea vomiting, visual disturbances (yellow, green halos)
high grade AV block along atrial tachycardias
ventricular tachy due to delayed afterdepolarizations
renal toxicity
adenosine
natural occurring substance
Give as rapid IV, very short half life
Bronchospasm in an asthmatic does not go away
Large doses can cause atrial fibrillation
Primary use:
To block AV node
Slight affect on sinus node
Terminate reentry rhythms that include AV node
Class 1 antiarrhythmias
Na channel blockers


Drug slows upstroke of phase 0
Prolongs QRS duration
Blockade of sodium channels: less available for conduction
Raise the threshold for initiation of action potential
Decrease slope of phase 4
Takes larger stimulus to excite each cell (larger threshold)
Slows or terminates automaticity
Effect on conduction:
Na channel blockade causes each cell to activate a bit more slowly
Cumulative effect is to slow conduction through the affected tissue

Use: convert unidirectional block to bidirectional block
Slow the slow pathway so much that it no longer conducts
Bad effect on reentry:
Slow either pathway just enough to make the reentry circuit virtually incessant

Don’t give to prior MI unless they have defibrillator
Class 1 A:
prolong QRS and QT

quinidine
procainamide
disopryamide

advese: Torsades de pointes
quinidine
class 1A antiarrhythmic

blocks Na channels

Major side effects: GI nausea vomiting
Diarrhea
CNS: cinchonism – confusion, visual, tinnitus
Autoimmune thrombocytopenia
Major interaction with digoxin
procinamide
Type 1A antiarrhythmic

Na channel blocker

IV only
Breakdown product = NAPA
Active metabolite
Metabolism depends on rate of acetylation
Blood level = check both procainamide + NAPA
Lupus-like syndrome
disopyramide
Type 1A antiarrhythmic

Na channel blocker

Strong anticholinergic effect – dry mouth, urinary retention, constipation, dry eyes
Negative inotrope – can use with hypertrophic cardiomyopathy
Class 1B antiarrhytmic
Na channel blocker

lidocaine

mexiletine
Lidocaine
Class 1B antiarrhythmic

block Na channels

Shorten QT interval
Target ventricular cells, ischemic tissue
Lidocaine: IV only
Cleared by liver…careful with liver disease
With heart failure, liver doesn’t have good perfusion
CNS side effects: can be serious if levels get too high…confusion, seizures
mexiletine
Class 1B antiarrhytmic

Na channel blocker

Oral
Mixes well with other antiarrhythmics
Basically oral lidocaine…liver toxicity, cns
Class 1C antiarrhythmic
Na channel blockers


Flecainide

Propafenone

prolong QRS

no difference in QT

negative inotropes
flecainaide
Class 1c
Na channel blockers

Most potent Na channel blockers
Markedly decerase upstroke of phase 0
Clean durgs – no effect on K channels
No direct effect on QT interval
Can cause bad arrhythmias, but not Torsades

Negative inotropes – contraindicated in systolic heart failure
Primarily used for atrial arrhythmias (AFib)
accessory pathways

Side effects are common but not severe
Fatigue
Visual disturbances
propafenone
Class 1c

Na channel blockers

Most potent Na channel blockers
Markedly decerase upstroke of phase 0
Clean durgs – no effect on K channels
No direct effect on QT interval
Can cause bad arrhythmias, but not Torsades

Negative inotropes – contraindicated in systolic heart failure
Primarily used for atrial arrhythmias (AFib)
accessory pathways

Has beta blocking properties
Mild CNS symptoms – dizziness

Use dependence:
Na channel blocking drugs bind preferentially to open channels
More open, more being blocked
Means they are more effective at faster HR
Class 3 antiarrhythmics
K channel blockers

sotalol

dofetilide and ibutilide

amiodarone

dronedarone

Reverse use dependence:
Greater affect the slower HR
Less useful for Afib (good for prevention, not stopping)
Also means that bad arrhythmias usch as Torsades are more likely during slower HR if QT is prolonged
Prolong phase 3…lengthening refractory period
Torsades: more likely when
Too much drug
Hypokalemia
Bradycardia
Sotalol
class 3 antiarrhythmic

K channel blocker

Also has beta blocker activity
Cleared by kidney – renal toxicity
QT must monitored periodically

Reverse use dependence:
Greater affect the slower HR
Less useful for Afib (good for prevention, not stopping)
Also means that bad arrhythmias usch as Torsades are more likely during slower HR if QT is prolonged
Prolong phase 3…lengthening refractory period
Torsades: more likely when
Too much drug
Hypokalemia
Bradycardia
Use with patients with MI scar
dofetilide and ibutilide
Class 3
ibutilide = IV

convert Afib
torsades in 10%

dofetilide = oral

cleared by liver and KIDNEY
relatively safe for heart failure
Reverse use dependence:
Greater affect the slower HR
Less useful for Afib (good for prevention, not stopping)
Also means that bad arrhythmias usch as Torsades are more likely during slower HR if QT is prolonged
Prolong phase 3…lengthening refractory period
Torsades: more likely when
Too much drug
Hypokalemia
Bradycardia
Use with patients with MI scar
Amiodarone
Class 3

Safest antiarrhythmic drug for your heart, most dangerous for rest of body

Prolongs QT but rarely causes Torsades
Don’t need to monitor QT
Rarely causes Torsades

Almost never causes ventricular arrhythmias

Adverse:
Multiple toxicities require routine monitoring
Eyes – corneal deposits, optic neuropathy
Hypothyroidism – common easily remedied
Hyperthyroidism rare
Hepatic injury
Pulmonary toxicity –irreverisble but usually after several years (look at age of pt)
Skin photosensitivity – bluish
Interacts with warfarin

Has some properties of all antiarrhythmics
K, Na, Ca channel blockers
dronedarone
class 3 antiarrhythmic

Supposedly like amiodarone without toxicities

But need to monitor QT, torsades probably low

Interferes with creatinine secretion – raises serum creatinine level but renal function fine
idiopathic necrotizing arteritis
(1)Kawasaki's disease

(2) takayasu arteritis

(3) Thromboangiitis obliterans (Buerger's Disease)
kawasaki's disease
idiopathic necrotizing arteritis

young children under 5yo

high fever

enlarged lymph nodes

rash

10% get coronary artery autoimmune attack

weakens wall, get aneurysm or dilation
Takayasu arteritis
Thickening of aortic arch
Blindness, cns deficits, decreased pulses in upper extremities
Microscopic = giant cell artitis
Women most commonly affected
Thromboangiitis obliterans (Buerger's Disease)
Pain and ischemia in legs/arms
Relatively young men
Leads to gangrene
Genetic predisposition
Almost exclusively smokers
Inflammatory with thrombosis
Lesion – cellular thrombosis with inflammation
Phlebitis: inflammation of veins
Usually with thrombosis
Caused by injury
Pylephlebitis – portal vein…associated with appendicitis
Dural sinuses
Pulmonary veins in pneumonia

Examples: iliofemoral veins in puerperal sepsis
Hepato-veno-occlusive disease from chemo
Migratory thrombophlebitis – Trousseau Syndrome
Idiopathic trhombophlebitis
Phlebothrombosis
venous thrombosis without inflammation

Usually in deep veins or pelvic veins
Exact incidence uncertain
Pain, tenderness, swelling or may be symptomatic
Complications: pulmonary embolus – 50%
Clots usually form in relation to valve cusps, propagate upwards
vericose veins
Abnormal dilated, tortuous veins
Produced by prolonged increase in intraluminal pressure
Often in relation to injury or defect of the vein proximally
Sites: legs in estimated 20% of population
Clusters in families
Complications- inflammation, thrombosis, stasis dermatitis, ulceration of skin
No pulmonary embolism

Sites: perianal – hemorrhoids
Testis – varicocele
Esophageal varices hepatic cirrhosis, portal hypertension…can hemorrhage
Atherosclerotic aneurysm
Age: >50, males 5:1, relatively common
Location : abdominal aorta (97%)
Usually below renals, may involve iliacs, but may occur in thoracic aorta
Mechanism: atheromasmedial destruction, ulcerated plaques

Presentation: pulsatile abdominal mass or pain
Rupture causes pain, swelling and shock
Incidentally on x ray (calcified)

Complications: rupture (retroperitoneal hemorrhage)
Stenosis of ureter by pressure or fibrosis
Occlusion of aortic branch (renal, mesenteric)
Embolism
Rupture: related to size
Greater than 7cm 80% rupture
Use ultrasound for cut-off
Therapy: surgery
syphilitic dissection
Location: ascending thoracic aorta and arch
Mechanism: tertiary syphilis involves vasa vasorum causes vasculitis and medial damage of aorta
Presentation: dyspnea, stridor, dysphagia, cough, pain
Congestive heart failure due to aortic insufficiency
Aneurysm can compress esophagus
Causes: heart failure or hemorrhage
Therapy: surgery
mycotic aneurysm
Mycotic refers to any infection, not necessarily fungal
Mechanism: infection causes destruction of media
Complication: infarcts distally, septic or bland
Cause: sequel of infective endocarditis or endarteritis…can embolism to brain carrying infection
dissecting hematoma of aorta
Age: 40-60 males
Location: intimal tear in ascending aorta
Types: A: most common involves ascending aorta and may extend distally
B: begins distal to subclavian artery, extends distally – better prognosis than A

Mechanism: hypertension
Trauma
Creates false lumen
Presentation: 85% pain in chest or radiating to back
Usually normal or elevated blood pressure
May have MI
Therapy: anti hypertensives, surgery
Stenotic lesions cause what kind of hypertrophy
cause pressure overload, and myocardial hypertrophy = concentric hypertrophy
regurgitant lesions cause what kind of hypertrophy
volume overload, eccentric hypertrophy
Rheumatic heart disease
uncommon in Us
most common valvular lesion in US
aortic stenosis
Aortic stenosis and bicuspid valve
Bicuspid Aortic valves 1-2% population
Frequent cause of symptomatic aortic stenosis in young patients
Associated with other congenital abnormalities in 20% of cases
80% of coarctation patients have Bicuspid
aortic stenosis etiology
Young patient: think bicuspid
Male more than female
Old patient – think degenerative: MOST COMMON in older patients
aortic stenosis symptoms
Cardinal symptoms
Chest pain (angina)
Reduced coronary flow reserve
Increased demand – high afterload
Syncope
Dyspnea on exertion
Impaired exercise tolerance
Heart failure
GI bleeding possible
classification of aortic stenosis symptoms
Classification of AS severity
Normal aortic valve area 2-3cm
Mild AS >1.5cm
Moderate As 1.0-1.5 cm
Severe AS <1.0 cm
when to treat aortic stenosis
Asymptomatic patients – follow expectantly
Endocarditis prophalaxis
Survey with echo annually when severe AS or symptoms change

Surgery – severe and symptomatic AS
If need heart surgery (bypass, etc.) for another reason, replace even if asymptomatic
aortic regurgitation causes
Causes:
Bicuspid – coarctations and aortic root dilation
Infective endocarditis
Rheumatic heart disease
Radiation
Subvalvular causes (septal defects)
Etiologies:
Chronic: rheumatic disease
Congenital bicuspid
Myoxamtous disease
Marfan’s
Syphilis
Acute: dissection
Infective endocarditis
Traumatic
symptoms of aortic regurgitation
Chronic AR – asymptomatic for a long time
Symptoms – exertional dyspnea, orthopnea, PND
Syncope and angina are less common
Uncomfortable awareness of heart beat especially lying down
aortic regurgitation PE
PE – apical impulse diffuse
Tachycardia
Diastolic descrendo murmur, begins just after second heart sound at apex
S3 and S4
Severity of AR correlates with duration of murmur
Murmur best heard along RIGHT sterna border
Austin Flint murmur – apical rumble due to regurgitation trying to close mitral valve (valve normally trying to open)…blowing diastolic murmur

Bobbing of head
Capillary pulsations
Clubbing of fingers
treatment for aortic regurgitation
Dilated aortic root or mild LV dysfunction – class I indication for surgery
Don’t do surgery if: normal systolic and asymptomatic with Chamber size normal
mitral valve stenosis etiology and prevalence
Primarily result of rheumatic fever
Rarely congenital
Slightly female to male
Don’t see in the US much unless poor access to care
mitral valve stenosis downstream affects
IN crease in left atrial pressure, can cause increase in pulmonary congestion and edema
Dypsnea common
Mitral valve stenosis associated with diastolic murmur because of turbulence
Diastolic murmur always pathologic
secondary concerns with mitral valve stenosis
Increase in mitral valve gradient – causes shortened diastolic phase
As atria expand, can get loss in atrial ventricular synchrony….get atrial fibrillation
Likely to get strokes
Valvular atrial fibrillation high risk of strokes

Fusion causes valve to become more spherical – less efficient

Tightening of valve – get turbulent flow in atrium
Tend to clot
Clots found in atrial appendage likely to embolize to brain
primary affects of mitral stenosis
Consequences:
Reduced ventricular filling
Reduced stroke volume
Decreased cardiac out put
Stroke volume low enough may result in a reduction in aortic pressure
mitral valve regurgitation

acute
Acute – infective endocarditis
Myxomatous disease
Ischemic heart disease

PE: loud S4, heard best at apex, radiates to axilla
mitral valve regurgitation

chronic
Chronic – myxomatous disease (prolapsed)
Rheumatic heart disease
Annulo-aortic ectasia – dilated aortic root
Ischemia – kill papillary muscle

Chronic:
Leak into left atrium
Left ventricle volume load
Compensation – left ventricle dilates and hypertrophies
Increase in stroke volume
Left atrium dilates
mitral valve stenosis PE findings
PE:
Jugular venous distension – normal until late
Carotids – normal until late
Apex impulse – laterally displaced and hyperdynamic
Pan systolic murmur in apex, axilla right precordium or back
mitral valve stenosis dx
Dx: systolic displace of mitral leaflet into atrium of at least 3mm, mitral regurgitation
Can have prolapsed but no leak can be of little concern
mitral valve stenosis tx
Tx. Surgery if see signs of atrium/ventricle dilation
normal changes in cardiovascular system with aging
increased stiffness of the left ventricle and arteries, and decreased maximal heart rate and cardiac output during exercise

3. These normal age related changes reduce cardiovascular reserve capacity, and thereby lower the threshold for development of common cardiovascular diseases, and reduce the ability to adapt to them
when does cardiovascular risk for women rise?
5. Cardiovascular risk for women rises after menopause and becomes similar to men by age by 70
cardiovascular disorders unique to elderly?
diastolic heart failure

calcific aortic stenosis
presentation of elderly with MI
Atypical presentations: 1/3 of elderly
Confusion
Dyspnea
Lassitude
New onset of CHF
Syncope
Stroke
Increases with age and women
gender differences in CHF
women more likely to have normal ejection fraction heart failure

Differences in preserved vs reduced
PEF – old, women, coronary artery disease reduced, morbidity
Reduced – middle aged, men, CAD common, morbidity,
venous insufficiency ulcer characteristics
gaiter distribution

larger with irregular border

shallow with moist, granulating base

surrounding pigmentation and inflammation

tx. compression and elevation
arterial insufficiency ulcer characteristics
painful with punched out appearance

dorsum of foot or toes

surrounding skin pale or molted

poorly developed granulation tissue at wound base

little bleeding seen with probing or debridment

tx. revascularization
neurotropic ulcer characteristics
completely painless

bleed with manipulation

appear over pressure points of toe or foot

surrounded by acut and chronic inflammatory reaction

associated with longstanding diabetes with neuropathy

tx. meticulous wound and foot care, well fitting shoes
arterial baroreflex vasconstriction
Receptors are sensitive to stretch…aortic arch and carotid sinus
Graded response
firing increases with increased stretch
sensitive to stretch, pulse stretch, rate of change
small deviations result in large change in firing because it responds to slope of curve (slope of change, not absolute amount of change)

drop in pressure = less firing

How it works:
From baroreceptor to nucleus tract soliatirus to either(1) CVLM – depressor or (2) RVLM – pressor centers in medulla
Barorecptor input into NTS…NTS sends excitatory to CVLM..CVLM projects inhibitory signal to RVLM…get less output from pressor centers….low BP

Baroreceptor input into NTS…NTS decreases signal to CVLM, CVLM has decreased firing on RVLM..RVLM increases vasoconstriction and BP
parasympathetic baroreflex of heart
Cell bodies originate in dorsal vagal/nucleus ambiguous of medulla
Excitatory connection to dorsal motor nucules and nucleus ambiguous with secondary projections to sympathetic
Get increased parasympathetic firing and thus decreased on heart
chemoreceptor control
Regulate respiratory activity
(a) Peripheral receptors
Carotid and aortic bodies
Increased CO2, acid, decreased O2, and hypoperfusion
Sends info via NTS to medulla

Humoral Control:
Catecholamines:
Released by adrenal medulla
80% Epi, 20% NE
NE also from end of sympathetic
NE stimulates release of Renin
receptor dependent vasodilators

local vascular control
histamine

sub P

SE

ACh
receptor independent vasodilators

local vascular control
sodium

nitroprusside

NO donor
vasocontrictors
PE

NE
NPY coreleased with NE augments actions of NE

Angiotensin II

arginine vasopressin

NE, Epi

Endothelin

Increased PO2
Special circulaitons

coronary
compensate increased demand with increased flow

local factors predominate over neural

use adenosine (vasodilator)
special circulation

cerebral
local factors predominate over neural
special circulation

skeletal muscle circulation
local factors and hypoxia regulate flow

metabolites out compete sympathetic vasoconstriction
special circulation

skin
control by neural and physical factors most important

tonic sympathetic tonic

hot vs cold temps
special circulation

splanchnic circulation
reservoir of blood

use to redistribute blood as needed

neural control predominates
special circulation

renal
two capillary segments in series

strong autoregulation - depends on sympathetic tone
special circulation

pulmonary
two criculations

(1) pulmonary - hypoxia causes vasoconstriction to maintain proper perfusion ratios

(2) bronchial - maintain flow to trachea and bronchial structures
MI of LAD
anteroseptal LV
MI of LCX
lateral LV
MI of RCA
posteroseptal LV
stages of MI
early acute: 6-24 hrs

acute 1-6 days

organizing 1-3wks

Remote 3mos or more
Early Acute MI
6-24 hrs

thin wavy fibers, eosinophilia

pallor, patchy, hyperemia
Acute MI
1-6 days

obvious pale, yellow

necrotic myocytes
organizing MI
1-3wks

red-brown edge around pale center

granulation tissue, acute and or chronic inflammation
remote MI
3 mos or longer


firm, white, scar

collagen
Transmural infarcts
infarction of full thickness of ventricular wall

caused by severe CAD with acute plaque disruption and occlusive thrombosis
subendocardial infarct
limited to inner 1/3 to 1/2 of ventricular wall

diminished perfusion
transmural infarct pathogenesis
90% due to CA stenosis and disrupted plaques

significant plaques mainly in proximal CA

less common: vasospasm, PLT aggregation, emboli

complete vessel occlusion may not cause MI if sufficient collateral flow

Almost all IN LV

initial event - plaque disruption with thrombosis following
subendocardial infarct pathogenesis
diffuse CAD and global perfusion made transiently critical by increased demand, vasospasm, hypotension

disrupted plaque with overlying thrombus that lyses , thus limiting extent of MI
Rupture of MI which stage?
Acute more likely
LV aneurysm of MI in which stage?
organizing
CHF of MI in which phase?
early acute
dysrhythmia of MI in which stage?
early acute
pericarditis of MI in which stage?
organizing fibrous in remote
chronic ischemic heart disease
due to left heart failure

Pure RV failure tdue to intrinsic lung disease

major manifestations - portal, systemic and dependent peripheral congestion and edema and effusions
Idiopathic dilated cardiomyopathy
results from viral myocarditis or toxic myocarditis

male>female

african americans>caucasains

genetic predisposition

heart biopsy is not diagnostic

treat like CHF - beta blocker, angiotensin, aldosterone antagonist
Viral myocarditis
probably cause of idiopathic cardiomyopathy

coxsackie B most common agent

immunologic mechanism - develops weeks after original infection

enhanced susceptibility - animal studies...radiation, malnutrition, steriods, exercise, previous MI

tends to be more aggressive in infants and pregnant women

myocytes infiltrate heart

other: HIV, CMV, hepatitis
ricketsia
bacterial
spirochetal
protozal
peripartum cardiomyopathy
distinct type of cardiomyopathy related to pregnancy

clear relationship to recent pregnancy with high risk of relapse on subsequent pregnancies

embolic phenomena common

better prognosis

evidance of inflammation good prognosticator
hypertrophic cardiomyopathy
cardiac muscle disease characterized by abnormal hypertrophy, mainly involving ventricular septum

histology: myofibrillar disarray

functional abnormalities

outflow tract obstruction

diastolic dysfunction

atrial and ventricular arrhythmias

50% autodominant

thickening of septum impingnes on outflow tract. plus systolic anterior movement of mitral valve causing mitral regurgitation.

have decreased pressure in outflow tract and aorta
Infectious pericarditis causative agents
coxsackie, echo virus, and HIV

TB outside US
non-infectious pericarditis causative agents
idiopathic

neoplasm - metastatic lung, breast or primary mesothelioma

renal failure
hypersensitive pericarditis causes
collagen vascular disease - LUPUS, rheumatoid arthitis

drug induced

post MI
Dx of pericarditis
diffuse ST segment elevation in early stage

depresison of PR segment and T wave inversion in late stages

or eletrical alternans
Dx of pericardial effusions
CXR

ECG - low voltage

echo
cardiac tamponade
accumulation of fluid in the pericardial space leads to an increased pericardial pressure

increased pressure = cardiac compression and impaired diastolic ventricular filling....means decreased cardiac output
clinical presentation of cardiac tamponade
mimics heart failure

dyspnea on exertion and orthopnea

trachycardia, tachypnea

jugular venous distention

pulsus paradoxus - typically 10mm variation in systole exhale/inhale...greater than 10 then cardiac tamponade
constrictive paricarditis
results from dense fibrosis and adhesion of the parietal and visceral layers

creates rigid cause around heart

early ventricular filling is unimpeded but diastolic filling is subsequently abruptly reduced as a result of the ventricles fo fill secondary to the restraints imposed by a rigid, thickened, and calcified pericardium
presentation of constrictive pericarditis
right heart failure

weakness, dyspnea, orthopnea, anorexia

peripheral edema, hepatomegaly, splenomegaloy, ascites

prominent Y decent with jugular venous pressures

exaggerated filling of jugular with inspiration

effects end diastole when ventricle volume is higher
non penetrating trauma and heart
most commonly causes cardiac contusion - new arrhythmias and ECG changes

can have transaction of descending thoracic aorta