Download as pdf or txt
Download as pdf or txt
You are on page 1of 55

See TARGETED THERAPY available online at

www.studentconsult.com C H A P T ER

The Heart
Frederick J. Schoen • Richard N. Mitchell
12
C H A P T E R CO N T E N T S

Cardiac Structure and Obstructive Lesions 537 Noninfected Vegetations 561


Specializations 523 Coarctation of the Aorta 537 Nonbacterial Thrombotic Endocarditis 561
Myocardium 524 Pulmonary Stenosis and Atresia 537 Endocarditis of Systemic Lupus Erythematosus
Valves 524 Aortic Stenosis and Atresia 537 (Libman-Sacks Disease) 562
Conduction System 524 Ischemic Heart Disease 538 Carcinoid Heart Disease 562
Blood Supply 525 Angina Pectoris 539 Complications of Prosthetic Valves 563
Cardiac Stem Cells 525 Myocardial Infarction 540 Cardiomyopathies 564
Effects of Aging on the Heart 525 Chronic Ischemic Heart Disease 550 Dilated Cardiomyopathy 565
Overview of Cardiac Arrhythmias 550 Arrhythmogenic Right Ventricular
Pathophysiology 526 Sudden Cardiac Death (SCD) 551 Cardiomyopathy 568
Heart Failure 526 Hypertrophic Cardiomyopathy 568
Hypertensive Heart Disease 552
Cardiac Hypertrophy: Pathophysiology Restrictive Cardiomyopathy 570
Systemic (Left-Sided) Hypertensive Heart
and Progression to Heart Disease 552 Myocarditis 570
Failure 527 Pulmonary (Right-Sided) Hypertensive Heart Other Causes of Myocardial Disease 571
Left-Sided Heart Failure 529 Disease (Cor Pulmonale) 553 Pericardial Disease 573
Right-Sided Heart Failure 530 Valvular Heart Disease 554 Pericardial Effusion and
Congenital Heart Disease 531 Calcific Valvular Degeneration 554 Hemopericardium 573
Left-to-Right Shunts 533 Calcific Aortic Stenosis 554 Pericarditis 573
Atrial Septal Defect 534 Calcific Stenosis of Congenitally Bicuspid Aortic Acute Pericarditis 573
Ventricular Septal Defect 535 Valve 555 Heart Disease Associated with
Patent Ductus Arteriosus 535 Mitral Annular Calcification 556 Rheumatologic Disorders 575
Right-to-Left Shunts 535 Mitral Valve Prolapse (Myxomatous Tumors of the Heart 575
Tetralogy of Fallot 535 Degeneration of the Mitral Valve) 556 Primary Cardiac Tumors 575
Transposition of the Great Arteries 536 Rheumatic Fever and Rheumatic Heart Cardiac Effects of Noncardiac
Tricuspid Atresia 537 Disease 557 Neoplasms 576
Infective Endocarditis 559 Cardiac Transplantation 577

The human heart is a remarkably efficient, durable, and described in this chapter include congenital heart abnor-
reliable pump, distributing more than 6000 liters of blood malities, ischemic heart disease, hypertensive heart disease,
through the body each day, and beating 30 to 40 million diseases of the cardiac valves, and primary myocardial
times a year—providing tissues with vital nutrients and disorders. A few comments about pericardial diseases and
facilitating waste excretion. Consequently, cardiac dys- cardiac neoplasms, as well as cardiac transplantation, are
function can have devastating physiologic consequences. also presented. This chapter begins with a brief review of
Cardiovascular disease (including coronary artery disease, the normal heart since most cardiac diseases manifest as
stroke, and peripheral vascular disease) is the number one structural and/or functional changes in one or more cardiac
cause of worldwide mortality, with about 80% of the components. General principles underlying cardiac hyper-
burden occurring in developing countries. In the United trophy and failure—common end points of several of the
States alone, cardiovascular disease accounts for roughly a different forms of heart disease—are also discussed.
third of all deaths, totaling about 800,000 individuals—or
nearly 1.5 times the number of deaths caused by all forms
of cancer combined. Cardiac Structure and Specializations
Disruption of any element of the heart—myocardium,
valves, conduction system, and coronary vasculature—can Heart weight varies with body habitus, averaging approxi-
adversely affect pumping efficiency, thus leading to mor- mately 0.4% to 0.5% of body weight (250 to 320 gm in
bidity and mortality. The major categories of cardiac disease females and 300 to 360 gm in males); the right ventricle
523
524 C H A P T E R 12 The Heart

wall thickness is usually 0.3 to 0.5 cm, while the left ven- Valves
tricle wall is 1.3 to 1.5 cm thick. Increases in heart weight
or ventricular thickness above these normal limits indi- The four cardiac valves—tricuspid, pulmonary, mitral, and
cates hypertrophy, and an enlarged chamber size implies aortic—maintain unidirectional blood flow. Valve function
dilation; both can represent compensatory changes in depends on the mobility, pliability, and structural integrity
response to heart disease and to volume and/or pressure of the leaflets of the atrioventricular valves (tricuspid and
overloads (see later). Increased cardiac weight or size (or mitral) or cusps of the semilunar valves (aortic and pulmo-
both)—resulting from hypertrophy and/or dilation—is nary). Cardiac valves are lined by endothelium and share
called cardiomegaly. a similar, tri-layered architecture:
• A dense collagenous core (fibrosa) at the outflow
Myocardium surface and connected to the valvular supporting
structures
The pumping function of the heart is accomplished via
the coordinated contraction (during systole) and relaxation • A central core of loose connective tissue (spongiosa)
(during diastole) of the cardiac myocytes that comprise the • A layer rich in elastin (ventricularis or atrialis depending
on which chamber it faces) on the inflow surface
myocardium. Left ventricular myocytes are arranged cir-
cumferentially in a spiral orientation that helps to generate The collagen of the ventricularis is largely responsible
a coordinated wave of contraction that spreads from the for the mechanical integrity of a valve, while the elastic
apex to the base of the heart. In contrast, right ventricular tissue of the atrialis/ventricularis imparts a rapid recoil to
myocytes have a somewhat less structured organization. achieve prompt valve closure. The proteoglycan-rich spon-
The contractile apparatus within myocytes is organized giosa facilitates the interactions of the collagenous (rela-
into a series of subunits called sarcomeres, composed of tively stiff) and elastic layers during the cardiac cycle.
highly ordered networks of thick filaments (primarily Crucial to function are the valvular interstitial cells, the
myosin in the center of the sarcomere) interlaced with thin most abundant cell type in the heart valves, and distrib-
filaments (largely actin, attached to the end of the sarco- uted throughout all of its layers. Valvular interstitial cells
mere) and regulatory proteins such as troponin and tropo- synthesize extracellular matrix (ECM) and express matrix
myosin. The actin and myosin filaments partially overlap degrading enzymes (including matrix metalloproteinases
with each other, giving rise to the striated appearance of [MMPs], along with inhibitors that remodel collagen and
cardiac myocytes (overlapping areas of actin and myosin other matrix components. Valvular interstitial cells com-
are dark while the intervening areas are light). prise a diverse and dynamic population of resident cells
Contraction results as myosin filaments ratchet adjacent that can alter their phenotypes and functions in response
actin filaments toward the center—shortening individual to changing hemodynamic stresses.
sarcomeres, and collectively leading to myocyte shorten- The function of the semilunar valves depends on the
ing. The amount of force generated is determined by the integrity and coordinated movements of the cuspal attach-
distance each sarcomere contracts. Thus, moderate ven- ments. Thus, dilation of the aortic root can hinder coapta-
tricular dilation during diastole creates a greater distance tion of the aortic valve cusps during closure and result in
over which the sarcomere can subsequently shorten and valvular regurgitation. In contrast, the competence of the
augment the force of systolic contraction. This compensa- atrioventricular valves depends on the proper function
tory mechanism serves to accommodate differing volume not only the leaflets but also the tendinous cords and the
and pressure demands. Unfortunately, there is an upper attached papillary muscles of the ventricular wall. Left
limit to the benefit of increased stretching during diastole. ventricular dilation, a ruptured tendinous cord, or papil-
With excessive dilation, the overlap of the actin and myosin lary muscle dysfunction can all interfere with valve closure,
filaments is reduced and the force of contraction decreases causing valvular insufficiency.
sharply, leading to heart failure. Because they are thin enough to be nourished by diffu-
Atrial myocytes are relatively haphazardly arranged, sion from the blood, normal leaflets and cusps have only
and thus generate weaker contractile forces than the ven- scant blood vessels limited to the proximal portion.
tricles. Some atrial cells have distinctive cytoplasmic Pathologic changes of valves are largely of three types:
electron-dense storage granules that contain atrial natri- damage to collagen that weakens the leaflets, exemplified
uretic peptide; this is a peptide hormone that promotes arte- by mitral valve prolapse; nodular calcification beginning
rial vasodilation and stimulates renal salt and water in interstitial cells, as in calcific aortic stenosis; and
elimination (natriuresis and diuresis), actions that are benefi- fibrotic thickening, the key feature in rheumatic heart
cial in the setting of hypertension and congestive heart disease (see later).
failure.
The coordinated beating of cardiac myocytes depends Conduction System
on intercalated discs—specialized intercellular junctions that
facilitate cell-to-cell mechanical and electrical (ionic) cou- Coordinated contraction of the cardiac muscle depends
pling. Within the intercalated discs (and at the lateral on propagation of electrical impulses—accomplished
borders of adjacent myocytes), gap junctions facilitate syn- through specialized excitatory and conducting myocytes
chronized waves of myocyte contraction by permitting within the cardiac conduction system that regulate heart
rapid movement of ions (e.g., sodium, potassium, calcium) rate and rhythm. The frequency of electrical impulses
between adjoining cells. Abnormalities in the spatial dis- that course through the conduction system is sensitive to
tribution of gap junctions in a variety of heart diseases can neural inputs (e.g., vagal stimulation), extrinsic adrenergic
cause electromechanical dysfunction (arrhythmia) and/or agents (e.g., adrenaline), hypoxia, and potassium concen-
heart failure. tration (i.e., hyperkalemia can block signal transmission
Effects of aging on the heart 525

altogether). Inputs from the autonomic nervous system can order animals is classically depicted as a permanent cell
increase the heart rate to twice normal within seconds, and population without replicative potential. However, increas-
are important for physiologic responses to exercise or other ing evidence points to the presence of bone marrow–
states associated with increased oxygen demand. derived precursors—as well as a small population of stem
The components of the conduction system include: cells within the myocardium—that are capable of repopu-
lating the mammalian heart. Besides self-renewal, these
• Sinoatrial (SA) node pacemaker (at the junction of the cardiac stem cells generate all cell lineages seen within the
right atrial appendage and superior vena cava)
myocardium. They constitute up to 5% to 10% of normal
• Atrioventricular (AV) node (located in the right atrium atrial cellularity, but represent only roughly 1 in 100,000
along the atrial septum)
cells in a normal ventricle.
• Bundle of His, connecting the right atrium to the ven- Cardiac stem cells have a very slow rate of proliferation,
tricular septum
which is greatest in neonates, and decreases with age. The
• Subsequent divisions into the right and left bundle human adult heart replaces roughly 1% of its total popula-
branches that stimulate their respective ventricles via tion each year, so that by the age of 50 years, almost 45%
further arborization into the Purkinje network of the total cardiomyocytes have been renewed. While
The cells of the cardiac conduction system depolarize stem cell numbers and progeny increase after myocardial
spontaneously, enabling them to function as cardiac pace- injury or hypertrophy, albeit to a limited extent, hearts that
makers. Because the normal rate of spontaneous depolar- suffer myocardial cell loss (e.g., due to infarction) do not
ization in the SA node (60 to 100 beats/minute) is faster recover any significant function in the necrotic zone (one
than the other components, it normally sets the pace. The of several features that distinguish humans from fish and
AV node has a gatekeeper function; by delaying the trans- newts). Nevertheless, the potential for stimulating prolif-
mission of signals from the atria to the ventricles, it ensures eration and differentiation of these cells in vivo is tantaliz-
that atrial contraction precedes ventricular systole. ing since it could facilitate recovery of myocardial function
following irreversible myocardial damage. Similarly, ex
Blood Supply vivo expansion and subsequent administration of stem
cells is another strategy being explored in the unfulfilled
Cardiac myocytes rely almost exclusively on oxidative quest to heal a broken heart. Until then a trip to the local
phosphorylation for their energy needs. Besides a high jewelry store will have to suffice!
density of mitochondria (20% to 30% of myocyte volume),
myocardial energy generation also requires a constant
supply of oxygenated blood—rendering myocardium Effects of Aging on the Heart
extremely vulnerable to ischemia. Nutrients and oxygen
are delivered via the coronary arteries, with takeoffs imme- The prevalence of most forms of heart disease increases
diately distal to the aortic valve. These initially course with each advancing decade. Consequently, as the average
along the external surface of the heart (epicardial coronary population in developed countries ages, changes in the
arteries) and then penetrate the myocardium (intramural cardiovascular system that accrue with aging become
arteries), subsequently branching into arterioles, and even- increasingly significant (Table 12-1).
tually forming a rich arborizing vascular network so that Epicardial fat increases, while the detritus of years of
each myocyte contacts roughly three capillaries. intracellular catabolism and oxidant stress accumulate in
There are three major epicardial coronary arteries the form of intracellular lipofuscin. Basophilic degeneration,
(so-called because they form a crown or corona at the base a gray-blue byproduct of glycogen metabolism within
of the heart): cardiac myocytes, is also increased. The size of the left
ventricular cavity, particularly in the base-to-apex dimen-
• Left anterior descending (LAD) and left circumflex sion, is reduced; this volume change is exacerbated by
(LCX) arteries arise from the left (main) coronary
artery systemic hypertension and bulging of the basal ventricular
septum into the left ventricular outflow tract (termed
• Right coronary artery sigmoid septum).
The divisions of the LAD are called diagonal branches, Valvular aging changes include calcification of the
and those of the LCX are marginal branches. The right and mitral annulus and aortic valve, the latter frequently
left coronary arteries function as end arteries, although leading to aortic stenosis. In addition, the valves can
anatomically most hearts have numerous intercoronary develop fibrous thickening, and the mitral leaflets tend
anastomoses (connections called collateral circulation). to buckle back toward the left atrium during ventricular
Blood flow to the myocardium occurs during ventricular systole, simulating a prolapsing (myxomatous) mitral
diastole, following closure of the aortic valve, and when valve. As this happens, increasing volume and pressure
the microcirculation is not compressed by cardiac contrac- overloads lead to left atrial dilation, and with it, an
tion. At rest, diastole comprises approximately two thirds increased incidence of atrial arrhythmias (e.g., fibrillation).
of the cardiac cycle; with tachycardia (increased heart rate), With time, small filiform processes (Lambl excrescences)
the relative duration of diastole also shortens, thus poten- form on the closure lines of aortic and mitral valves,
tially compromising cardiac perfusion. probably resulting from the organization of small
thrombi.
Cardiac Stem Cells The aorta becomes progressively stiffer, owing to the
fragmentation and loss of elastic tissue and increased col-
Although cardiac regeneration in metazoans (e.g., newts lagen deposition, along with the accumulation of athero-
and zebrafish) is well described, the myocardium of higher sclerotic plaque. The result is less elasticity, and increased
526 C H A P T E R 12 The Heart

Table 12-1 Changes in the Aging Heart


• Regurgitant flow. A portion of the output from each con-
Chambers traction flows backward through an incompetent valve,
Increased left atrial cavity size adding a volume overload to the affected atria or ven-
Decreased left ventricular cavity size tricles (e.g., left ventricle in aortic regurgitation; left
Sigmoid-shaped ventricular septum atrium and left ventricle in mitral regurgitation).
Valves • Shunted flow. Blood can be diverted from one part of the
heart to another (e.g., from the left ventricle to the right
Aortic valve calcific deposits
ventricle), through defects that can be congenital or
Mitral valve annular calcific deposits
Fibrous thickening of leaflets acquired (e.g., following myocardial infarction). Shunted
Buckling of mitral leaflets toward the left atrium flow can also occur between blood vessels, as in patent
Lambl excrescences ductus arteriosus (PDA).
Epicardial Coronary Arteries • Disorders of cardiac conduction. Conduction defects or
arrhythmias due to uncoordinated generation or trans-
Tortuosity
mission of impulses (e.g., atrial or ventricular fibrilla-
Diminished compliance
Calcific deposits tion) lead to nonuniform and inefficient myocardial
Atherosclerotic plaque contractions, and may in fact be lethal.
Myocardium • Rupture of the heart or a major vessel. In such circum-
stances (e.g., gunshot to the left ventricle, or aortic dis-
Decreased mass section and rupture), there is cataclysmic exsanguination,
Increased subepicardial fat
either into body cavities or externally.
Brown atrophy
Lipofuscin deposition Most cardiovascular disease results from a complex
Basophilic degeneration interplay of genetics and environmental factors; these
Amyloid deposits
may disrupt signaling pathways that control morphogen-
Aorta esis, impact myocyte survival after injury, or affect contrac-
Dilated ascending aorta with rightward shift tility or electrical conduction in the face of biomechanical
Elongated (tortuous) thoracic aorta stressors. Indeed, the pathogenesis of many congenital
Sinotubular junction calcific deposits heart defects involves an underlying genetic abnormality
Elastic fragmentation and collagen accumulation whose expression is modified by environmental or mater-
Atherosclerotic plaque nal factors (see later). Moreover, genes that control the
development of the heart may also regulate the response
to various forms of injury including aging. Subtle poly-
pressure spikes with each cardiac contraction that are morphisms can significantly impact the risk of many forms
transmitted to distal organs. of heart disease, and, as discussed later, a number of adult-
Compared with younger myocardium, “elderly” myo- onset heart disorders have a fundamentally genetic basis.
cardium has fewer myocytes, increased collagenized con- Thus, cardiovascular genetics provides an important
nective tissue and, often the deposition of extracellular window on the pathogenesis of heart disease and increas-
amyloid (most commonly due to poorly catabolized trans- ingly molecular diagnoses are becoming a critical part of
thyretin; see Chapter 6). its classification.
Most importantly, the progressive atherosclerosis
(Chapter 11)—over a period of 50 to 60 years—finally ends
up causing significant stenosis, or weakens the wall suffi- Heart Failure
ciently to give rise to catastrophic dissection of the aortic
wall (see later). Heart failure, often called congestive heart failure (CHF), is a
common, usually progressive condition with a poor prog-
nosis. Each year in the United States, CHF affects more
Overview of Cardiac Pathophysiology than 5 million individuals (approximately 2% of the popu-
lation), necessitating more than a million hospitalizations,
Cardiovascular dysfunction can be attributed to one (or and contributing to the death of nearly 300,000 people.
more) of six principal mechanisms: CHF occurs when the heart is unable to pump blood
at a rate sufficient to meet the metabolic demands of the
• Pump failure. In some conditions, the myocardium con- tissues or can do so only at an elevated filling pressure.
tracts weakly during systole and there is inadequate It is the common end stage of many forms of chronic heart
cardiac output. Conversely, myocardium may relax disease, often developing insidiously from the cumulative
insufficiently during diastole to permit adequate ven- effects of chronic work overload (e.g., in valve disease or
tricular filling. hypertension) or ischemic heart disease (e.g., following
• Flow obstruction. Lesions can obstruct blood flow through myocardial infarction with heart damage). However, acute
a vessel (e.g., atherosclerotic plaque) or prevent valve hemodynamic stresses, such as fluid overload, abrupt val-
opening or otherwise cause increased ventricular vular dysfunction, or myocardial infarction, can all precipi-
chamber pressure (e.g., aortic valvular stenosis, sys- tate sudden CHF.
temic hypertension, or aortic coarctation). In the case of When cardiac workload increases or cardiac function is
a valvular blockage, the increased pressure overloads compromised, several physiologic mechanisms maintain
the chamber that pumps against the obstruction. arterial pressure and organ perfusion:
Heart failure 527

• Frank-Starling mechanism, in which increased filling hypertrophy (e.g., due to hypertension or aortic stenosis),
volumes dilate the heart and thereby increase subse- new sarcomeres are predominantly assembled in parallel
quent actin-myosin cross-bridge formation, enhancing to the long axes of cells, expanding the cross-sectional area
contractility and stroke volume of myocytes in ventricles and causing a concentric increase
• Myocardial adaptations, including hypertrophy with or in wall thickness. In contrast, volume-overload hypertrophy is
without cardiac chamber dilation. In many pathologic characterized by new sarcomeres being assembled in series
states, heart failure is preceded by cardiac hypertrophy, within existing sarcomeres, leading primarily to ventricu-
the compensatory response of the myocardium to lar dilation. As a result, in dilation due to volume overload,
increased mechanical work. The collective molecular, or dilation that accompanies failure of a previously pres-
cellular, and structural changes that occur as a response sure overloaded heart, the wall thickness may be increased,
to injury or changes in loading conditions are called normal, or less than normal. Consequently, heart weight,
ventricular remodeling. rather than wall thickness, is the best measure of hypertro-
• Activation of neurohumoral systems to augment heart phy in dilated hearts.
function and/or regulate filling volumes and Cardiac hypertrophy can be substantial in clinical heart
pressures disease. Heart weights of two to three times normal are
common in patients with systemic hypertension, ischemic
• Release of norepinephrine by adrenergic cardiac heart disease, aortic stenosis, mitral regurgitation, or
nerves of the autonomic nervous system (which
dilated cardiomyopathy, and heart weights can be three- to
increases heart rate and augments myocardial con-
four-fold greater than normal in those with aortic regurgi-
tractility and vascular resistance)
tation or hypertrophic cardiomyopathy.
• Activation of the renin-angiotensin-aldosterone Important changes at the tissue and cell level occur
system with cardiac hypertrophy. Importantly, myocyte hyper-
• Release of atrial natriuretic peptide. The latter two trophy is not accompanied by a proportional increase in
factors act to adjust filling volumes and pressures. capillary numbers. As a result, the supply of oxygen and
nutrients to the hypertrophied heart, particularly one
These adaptive mechanisms may be adequate to undergoing pressure overload hypertrophy, is more
maintain normal cardiac output in the face of acute pertur- tenuous than in the normal heart. At the same time,
bations, but their capacity to do so may ultimately be over- oxygen consumption by the hypertrophied heart is ele-
whelmed. Moreover, superimposed pathologic changes, vated due to the increased workload that drives the
such as myocyte apoptosis, intracellular cytoskeletal process. Hypertrophy is also often accompanied by depo-
alterations, and extracellular matrix deposition, may cause sition of fibrous tissue (interstitial fibrosis). Molecular
further structural and functional disturbances. Heart changes include the expression of immediate-early genes
failure can result from progressive deterioration of myo- (e.g., FOS, JUN, MYC, and EGR1) (Chapter 2). With pro-
cardial contractile function (systolic dysfunction)—reflected longed hemodynamic overload, there may be a shift to a
as a decrease in ejection fraction (EF, the percentage of gene expression pattern resembling that seen during fetal
blood volume ejected from the ventricle during systole; cardiac development (including selective expression of
normal is approximately 45% to 65%). Reduction in EF can embryonic/fetal forms of β-myosin heavy chain, natri-
occur with ischemic injury, inadequate adaptation to pres- uretic peptides, and collagen).
sure or volume overload due to hypertension or valvular At a functional level, cardiac hypertrophy is associated
disease, or ventricular dilation. Increasingly, heart failure with heightened metabolic demands due to increases in
is recognized as resulting from an inability of the heart mass, heart rate, and contractility (inotropic state, or force
chamber to expand and fill sufficiently during diastole (dia- of contraction), all of which increase cardiac oxygen con-
stolic dysfunction), for example, due to left ventricular sumption. As a result of these changes, the hypertrophied heart
hypertrophy, myocardial fibrosis, constrictive pericarditis, is vulnerable to ischemia-related decompensation, which can
or amyloid deposition. evolve to cardiac failure and eventually lead to death.
The proposed sequence of initially beneficial—and later
harmful—events in response to increased cardiac work
Cardiac Hypertrophy: Pathophysiology and is summarized in Figure 12-2. The molecular and cellular
Progression to Heart Failure changes in hypertrophied hearts that initially mediate enhanced
function may themselves contribute to the development of heart
Sustained increase in mechanical work due to pressure failure. This can occur through:
or volume overload (e.g., systemic hypertension or aortic
stenosis) or trophic signals (e.g., those mediated through
• Abnormal myocardial metabolism
the activation of β-adrenergic receptors) cause myocytes • Alterations of intracellular handling of calcium ions
to increase in size (hypertrophy); cumulatively, this • Myocyte apoptosis
increases the size and weight of the heart (Fig. 12-1). • Reprogramming of gene expression, which may
Hypertrophy requires increased protein synthesis, thus occur in part through changes in expression of miRNAs,
enabling the assembly of additional sarcomeres, as well as small noncoding RNAs that inhibit gene expression
increasing the numbers of mitochondria. Hypertrophic (Chapter 1).
myocytes also have enlarged nuclei, attributable to The degree of structural abnormality of the heart in
increased DNA ploidy resulting from DNA replication in CHF does not always reflect the severity of dysfunction,
the absence of cell division. The pattern of hypertrophy and the structural, biochemical, and molecular basis for
reflects the nature of the stimulus. In pressure-overload myocardial contractile failure can be obscure.
528 C H A P T E R 12 The Heart

A B

C D
Figure 12-1 Left ventricular hypertrophy. A, Pressure hypertrophy due to left ventricular outflow obstruction. The left ventricle is on the lower right in this apical
four-chamber view of the heart. B, Left ventricular hypertrophy with and without dilation, viewed in transverse heart sections. Compared with a normal heart
(center), the pressure-hypertrophied hearts (left and in A) have increased mass and a thick left ventricular wall, while the hypertrophied, dilated heart (right)
has increased mass and a normal wall thickness. C, Normal myocardium. D, Hypertrophied myocardium (panels C and D are photomicrographs at the same
magnification). Note the increases in both cell size and nuclear size in the hypertrophied myocytes. (A,B, Reproduced with permission from Edwards WD:
Cardiac anatomy and examination of cardiac specimens. In Emmanouilides GC, et al [eds]: Moss and Adams Heart Disease in Infants, Children, and
Adolescents: Including the Fetus and Young Adults, 5th ed. Philadelphia, Williams & Wilkins, 1995, p 86.)

At autopsy, the hearts of patients with CHF are gener- sometimes referred to as physiologic hypertrophy. Static exer-
ally heavy and dilated, and may be relatively thin-walled; cise (e.g., weight lifting) is associated with pressure hyper-
they exhibit microscopic evidence of hypertrophy, but the trophy and appears more likely to be associated with
extent of these changes is extremely variable. In hearts that deleterious changes.
have suffered myocardial infarction, loss of pumping Whatever its basis, CHF is characterized by variable
capacity due to myocyte death leads to work-related degrees of decreased cardiac output and tissue perfusion
hypertrophy of the surrounding viable myocardium. In (sometimes called forward failure), as well as pooling of
valvular heart disease, the increased pressure or volume blood in the venous capacitance system (backward
overloads the myocardium globally. Increased heart mass failure); the latter may cause pulmonary edema, periph-
owing to disease is correlated with excess cardiac mortality eral edema, or both. As a result, many of the significant
and morbidity; indeed, cardiomegaly is an independent clinical features and morphologic changes noted in CHF
risk factor for sudden death. are actually secondary to injuries induced by hypoxia and
In contrast to pathologic hypertrophy (which is often congestion in tissues away from the heart.
associated with contractile impairment), hypertrophy The cardiovascular system is a closed circuit. Thus,
induced by regular strenuous exercise has varied effects on although left-sided and right-sided failure can occur
the heart depending on the type of exercise. Aerobic exer- independently, failure of one side (particularly the left)
cise (e.g., long distance running) tends to be associated often produces excessive strain on the other, terminat-
with volume-load hypertrophy that may be accompanied ing in global heart failure. Despite this interdepen-
by increases in capillary density (unlike other forms of dence, it is easiest to understand the pathology of heart
hypertrophy) and decreases in resting heart rate and blood failure by considering right- and left-sided heart failure
pressure—effects that are all beneficial. These changes are separately.
Heart failure 529

VALVULAR MYOCARDIAL
HYPERTENSION DISEASE INFARCTION perivascular and interstitial edema, particularly in the interlobular
septa, responsible for the characteristic Kerley B and C lines
Pressure Pressure and/or Regional dysfunction
overload volume overload with volume overload noted on chest X-ray study in CHF, (2) progressive edematous
widening of alveolar septa, and (3) accumulation of edema fluid
in the alveolar spaces. Some red cells and plasma proteins
Cardiac work
extravasate into the edema fluid within the alveolar spaces,
where they are phagocytosed and digested by macrophages,
Wall stress
which store the iron recovered from hemoglobin in the form of
hemosiderin. These hemosiderin-laden macrophages (also
Cell stretch
known as heart failure cells) are telltale signs of previous
episodes of pulmonary edema. Pleural effusions arise from
Hypertrophy and/or dilation elevated pleural capillary pressure and the resultant transuda-
Characterized by tion of fluid into the pleural cavities.
• heart size and mass
• protein synthesis
• induction of immediate-early genes
• induction of fetal gene program
Early left-sided heart failure symptoms are related to
• abnormal proteins pulmonary congestion and edema and may be subtle.
• fibrosis Initially, cough and dyspnea (breathlessness) may occur
• inadequate vasculature only with exertion. As CHF progresses, worsening pulmo-
nary edema may cause orthopnea (dyspnea when supine,
relieved by sitting or standing) or paroxysmal nocturnal
Cardiac dysfunction
dyspnea (dyspnea usually occurring at night that is so
Characterized by severe that it induces a feeling of suffocation). Dyspnea at
• heart failure (systolic/diastolic) rest may follow. Atrial fibrillation, an uncoordinated, chaotic
• arrhythmias contraction of the atrium, exacerbates CHF owing to the
• neurohumoral stimulation loss of the atrial “kick” and its 10% to 15% contribution to
ventricular filling.
Figure 12-2 Schematic representation of the causes and consequences of A reduced ejection fraction leads to diminished renal
cardiac hypertrophy. perfusion, causing activation of the renin-angiotensin-
aldosterone system as a compensatory mechanism to
Left-Sided Heart Failure correct the “perceived” hypotension. This leads to salt and
water retention, with expansion of the interstitial and intra-
Left-sided heart failure is most often caused by: vascular fluid volumes (Chapters 4 and 11) that then exac-
erbate the ongoing pulmonary edema. If the hypoperfusion
• Ischemic heart disease of the kidney becomes sufficiently severe, impaired excre-
• Hypertension tion of nitrogenous products may cause azotemia (called
• Aortic and mitral valvular diseases prerenal azotemia because of its vascular origin; Chapter 20).
• Primary myocardial diseases In far-advanced CHF, cerebral hypoperfusion can give rise
The clinical and morphologic effects of left-sided CHF to hypoxic encephalopathy (Chapter 28), with irritability, loss
are a consequence of passive congestion (blood backing of attention span, and restlessness that can progress to
up in the pulmonary circulation), stasis of blood in the stupor and coma with ischemic cerebral injury.
left-sided chambers, and inadequate perfusion of down- Left-sided heart failure can be divided into systolic and
stream tissues leading to organ dysfunction. diastolic failure:

• Systolic failure is defined by insufficient ejection fraction


MORPHOLOGY (pump failure), and can be caused by any of the many
disorders that damage or derange the contractile func-
Heart. The heart findings depend on the disease process, tion of the left ventricle.
ranging from myocardial infarcts, to stenotic or regurgitant
valves, to intrinsic myocardial pathology. Except for failure • In diastolic failure, the left ventricle is abnormally stiff
and cannot relax during diastole. Thus, although cardiac
caused by mitral valve stenosis or unusual restrictive cardiomy-
function is relatively preserved at rest, the heart is
opathies (described later), the left ventricle is usually hypertro-
unable to increase its output in response to increases in
phied and often dilated, sometimes massively. The microscopic
the metabolic demands of peripheral tissues (e.g., during
changes are nonspecific: primarily myocyte hypertrophy and
exercise). Moreover, because the left ventricle cannot
variable degrees of interstitial fibrosis. Impaired left ventricular
expand normally, any increase in filling pressure is
function usually causes secondary dilation of the left atrium,
immediately transferred back into the pulmonary circu-
which increases the risk of atrial fibrillation. This in turn results
lation, producing rapid onset pulmonary edema ( flash
in stasis of blood, particularly in the atrial appendage, which is
pulmonary edema). Diastolic failure predominantly
a common site of thrombus formation.
occurs in patients older than age 65 years and for unclear
Lungs. Pulmonary congestion and edema produce heavy,
reasons is more common in women. Hypertension is the
wet lungs, as described elsewhere (Chapters 4 and 15).
most common underlying etiology; diabetes mellitus,
Pulmonary changes—from mildest to most severe—include (1)
obesity, and bilateral renal artery stenosis may also
530 C H A P T E R 12 The Heart

contribute to risk. Reduced left ventricular relaxation


splenomegaly), and can also contribute to chronic congestion
may stem from myocardial fibrosis (e.g., in cardiomy-
and edema of the bowel wall. The latter may be sufficiently
opathies and ischemic heart disease), and infiltrative
severe as to interfere with nutrient (and/or drug) absorption.
disorders associated with restrictive cardiomyopathies
Pleural, Pericardial, and Peritoneal Spaces. Systemic
(e.g., cardiac amyloidosis). Diastolic failure may appear
venous congestion can lead to fluid accumulation in the pleural,
in older patients without any known predisposing
pericardial, or peritoneal spaces (effusions; peritoneal effu-
factors, possibly as an exaggeration of the normal stiff-
sions are also called ascites). Large pleural effusions can
ening of the heart with age. Constrictive pericarditis
impact lung inflation, causing atelectasis, and substantial
(discussed later) can also limit myocardial relaxation
ascites can also limit diaphragmatic excursion, causing dyspnea
and therefore mimics primary diastolic dysfunction.
on a purely mechanical basis.
Subcutaneous Tissues. Edema of the peripheral and
Right-Sided Heart Failure dependent portions of the body, especially ankle (pedal) and
pretibial edema, is a hallmark of right-sided heart failure. In
Right-sided heart failure is most commonly caused by
chronically bedridden patients presacral edema may predomi-
left-sided heart failure, as any increase in pressure in the
nate. Generalized massive edema (anasarca) may also occur.
pulmonary circulation from left-sided failure inevitably
burdens the right side of the heart. Consequently, the
causes of right-sided heart failure include all of those that
induce left-sided heart failure. Isolated right-sided heart The kidney and the brain are also prominently affected
failure is infrequent and typically occurs in patients with in right-sided heart failure. Renal congestion is more
one of a variety of disorders affecting the lungs; hence it marked with right-sided than left-sided heart failure,
is often referred to as cor pulmonale. Besides parenchymal leading to greater fluid retention and peripheral edema,
lung diseases, cor pulmonale can also arise secondary and more pronounced azotemia. Venous congestion and
to disorders that affect the pulmonary vasculature, for hypoxia of the central nervous system can also produce
example, primary pulmonary hypertension (Chapter 15), deficits of mental function akin to those seen in left-sided
recurrent pulmonary thromboembolism (Chapter 4), or heart failure with poor systemic perfusion.
conditions that cause pulmonary vasoconstriction (obstruc- Although we have discussed right and left heart failure
tive sleep apnea, altitude sickness). The common feature of separately, it is again worth emphasizing that in many
these disorders is pulmonary hypertension (discussed later), cases of chronic cardiac decompensation, patients present
which results in hypertrophy and dilation of the right side in biventricular CHF with symptoms reflecting both right-
of the heart. In extreme cases, leftward bulging of the inter- sided and left-sided heart failure.
ventricular septum can even cause left ventricular dysfunc- Standard therapy for CHF is mainly pharmacologic.
tion. The major morphologic and clinical effects of primary Drugs that relieve fluid overload (e.g., diuretics), that block
right-sided heart failure differ from those of left-sided the renin-angiotensin-aldosterone axis (e.g., angiotensin
heart failure in that pulmonary congestion is minimal converting enzyme inhibitors), and that lower adrenergic
while engorgement of the systemic and portal venous tone (e.g., beta-1 adrenergic blockers) are all particularly
systems is pronounced. beneficial. The efficacy of the latter two classes of drugs
supports the concept that neurohumoral changes in CHF
(e.g., renin and norepinephrine elevations) are maladap-
tive contributions to heart failure.
MORPHOLOGY Newer approaches to improving cardiac function
include mechanical assist devices, and resynchronization
Heart. As in left-heart failure, the cardiac morphology varies
of electrical impulses to maximize cardiac efficiency.
with cause. Rarely, structural defects such as tricuspid or pul-
Interestingly, some patients treated by mechanical assist
monary valvular abnormalities or endocardial fibrosis (as in car-
can recover sufficient function to be weaned from the
cinoid heart disease) may be present. However, since isolated
device. There is also considerable enthusiasm for novel
right heart failure is most often caused by lung disease, most
therapies, including cell-based approaches, although
cases exhibit only hypertrophy and dilation of the right atrium
several hurdles remain in their implementation.
and ventricle.
Liver and Portal System. Congestion of the hepatic and
portal vessels may produce pathologic changes in the liver, the
spleen, and the GI tract. The liver is usually increased in size KEY CONCEPTS
and weight (congestive hepatomegaly) due to prominent
passive congestion, greatest around the central veins Heart Failure
(Chapter 4). Grossly, this is reflected as congested red-brown ■ CHF occurs when the heart is unable to provide adequate
pericentral zones, with relatively normal-colored tan periportal perfusion to meet the metabolic requirements of peripheral
regions, producing the characteristic “nutmeg liver” appearance tissues; inadequate cardiac output is usually accompanied
(Chapter 4). In some instances, especially when left-sided by increased congestion of the venous circulation.
heart failure with hypoperfusion is also present, severe centri- ■ Left-sided heart failure is most commonly due to ischemic
lobular hypoxia produces centrilobular necrosis. With long- heart disease, systemic hypertension, mitral or aortic valve
standing severe right-sided heart failure, the central areas can disease, and primary diseases of the myocardium; symp-
become fibrotic, eventually culminating in cardiac cirrhosis toms are mainly a consequence of pulmonary congestion
(Chapter 18). Portal venous hypertension also causes enlarge- and edema, although systemic hypoperfusion can cause
ment of the spleen with platelet sequestration (congestive secondary renal and cerebral dysfunction.
Congenital heart disease 531

Table 12-2 Frequencies of Congenital Cardiac Malformations*


■ Right heart failure is most often due to left heart failure,
and less commonly to primary pulmonary disorders; symp- Incidence per
toms are chiefly related to peripheral edema and visceral Malformation Million Live Births %
congestion. Ventricular septal defect 4482 42
Atrial septal defect 1043 10
Pulmonary stenosis 836 8
Congenital Heart Disease Patent ductus arteriosus 781 7
Tetralogy of Fallot 577 5
Congenital heart disease (CHD) is a general term designat-
Coarctation of the aorta 492 5
ing abnormalities of the heart or great vessels that are
present at birth. Most congenital heart disease arises from Atrioventricular septal defect 396 4
faulty embryogenesis during gestational weeks 3 through Aortic stenosis 388 4
8, when major cardiovascular structures form and begin to Transposition of the great arteries 388 4
function. The most severe anomalies are incompatible with Truncus arteriosus 136 1
intrauterine survival and significant heart malformations
Total anomalous pulmonary 120 1
are common among premature infant and stillborns. On
venous connection
the other hand, defects that affect individual chambers or
discrete regions of the heart are often compatible with Tricuspid atresia 118 1
embryologic maturation and eventual live birth. In this Total 9757
category are septation defects, unilateral obstructions, and *Presented as upper quartile of 44 published studies. Percentages do not add up to 100%
outflow tract anomalies. Septal defects, or “holes in the because of rounding.
Source: Hoffman JIE, Kaplan S: The incidence of congenital heart disease. J Am Coll Cardiol
heart”, include atrial septal defects (ASDs) or ventricular 39:1890, 2002.
septal defects (VSDs). Stenotic lesion can be at the level of
the cardiac valve or entire cardiac chamber, as in hypoplas-
tic left heart syndrome. Outflow tract anomalies include multipotent progenitor cells that can produce all of the
inappropriate routing of the great vessels from the ven- major cell types of the heart: endocardium, myocardium,
tricular mass. These forms of congenital heart disease and smooth muscle cells. However, each heart field is dif-
usually produce clinically important manifestations only ferentially marked by the expression of distinct gene sets.
after birth—unveiled by the transition from fetal to perina- Thus, the first heart field expresses the transcription factor
tal circulation. Hand1, whereas the second heart field expresses the tran-
scription factor Hand2 and the secreted protein fibroblast
Incidence. With an incidence of up to 5%, congenital car- growth factor-10.
diovascular malformations are among the most prevalent Even at this very early stage of development, each heart
birth defects and are the most common type of pediatric field is destined to give rise to particular portions of the
heart disease. Approximately 1% of individuals have sig- heart. Thus, the left ventricle largely derives from cells
nificant forms of congenital heart disease that are diag- originating in the first heart field, whereas cells from the
nosed in the first year of life. However, milder forms of second heart field become the outflow tract, right ventricle,
congenital heart disease such as bicuspid semilunar valves, and most of the atria. By day 20, the initial cell crescent
with an incidence itself of 1-2%, may not become evident develops into a beating tube, which loops to the right and
until adulthood. Twelve disorders account for about 85% begins to form the basic heart chambers roughly 8 days
of cases; their frequencies are listed in Table 12-2. later. At the same time, two other critical events occur: (1)
The number of individuals who survive into adulthood neural crest–derived cells migrate into the outflow tract,
with congenital heart disease is increasing rapidly and is where they participate in the septation of the outflow tract
estimated at nearly 1 million individuals in the United and the formation of the aortic arches, and (2) interstitial
States. Many have benefited from advances in early post- connective tissue that will become the future atrioventricu-
natal (and even intrauterine) surgical repair of their struc- lar canal and outflow tract enlarges to produce swellings
tural defects. In some cases, however, surgical repairs fail known as endocardial cushions. By day 50, further septa-
to restore complete normalcy; patients may have already tion of the ventricles, atria, and atrioventricular valves pro-
sustained pulmonary or myocardial changes that are no duces a four-chambered heart.
longer reversible, or may suffer from arrhythmias due to Proper orchestration of these remarkable transforma-
surgical scarring. Other factors that impact the long-term tions depends on a network of transcription factors that are
outcome include risks associated with the use of prosthetic regulated by a number of signaling pathways, particularly
materials and devices (e.g., substitute valves or myocardial the Wnt, hedgehog, vascular endothelial growth factor
patches), and the cardiovascular stressors associated with (VEGF), bone morphogenetic factor, TGFβ, fibroblast
childbearing that may tip a repaired heart into failure. growth factor, and Notch pathways (Chapter 1). In addi-
tion, specific micro-RNAs play critical roles in cardiac
Cardiac Development. The diverse malformations seen in development by coordinating patterns and levels of tran-
congenital heart disease are caused by errors that occur scription factor expression. The heart is a mechanical organ
during cardiac morphogenesis (Fig. 12-3). The earliest that generates pulsatile blood from its earliest stages of
cardiac precursors originate in lateral mesoderm and move development. It is therefore likely that hemodynamic
to the midline in two migratory waves to create a crescent forces play an important role in cardiac development, just
of cells consisting of the first and second heart fields as they influence adaptations in the adult heart such as
by about day 15 of development. Both fields contain hypertrophy and dilation.
532 C H A P T E R 12 The Heart

RCA LCA
RSCA LSCA
First heart field
Second heart field
DA
Cardiac neural crest
CT AS Ao
PA
RA LA

CT LA
V
AVV
RA
V V
RV LV
LV
A A
RV
RA LA

A First heart field template B Second heart field migration C Neural crest migration D Completed formation of
four-chambered heart
Figure 12-3 Human cardiac development, emphasizing three main sources of cells. A, Day 15. First heart field (FHF) cells (shown in red) form a crescent shape
in the anterior embryo with second heart field (SHF) cells (shown in yellow) near the FHF. B, Day 21. SHF cells lie dorsal to the straight heart tube and begin
to migrate (arrows) into the anterior and posterior ends of the tube to form the right ventricle, conotruncus (CT), and part of the atria (A). C, Day 28. Following
rightward looping of the heart tube, cardiac neural crest cells (shown in blue) also migrate (arrow) into the outflow tract from the neural folds to septate the
outflow tract and pattern the bilaterally symmetric aortic arch arteries. D, Day 50. Septation of the ventricles, atria, and atrioventricular valves (AVV) results in
the appropriately configured four-chambered heart. Ao, Aorta; AS, aortic sac; DA, ductus arteriosus; LA, left atrium; LCA, left carotid artery; LSCA, left sub-
clavian artery; LV, left ventricle; PA, pulmonary artery; RA, right atrium; RCA, right carotid artery; RSCA, right subclavian artery; V, ventricle. (Modified with
permission from Srivastava D: Making or breaking the heart: from lineage determination to morphogenesis. Cell 126:1037, 2006.)

Many of the inherited defects that affect heart develop- Table 12-3 Selected Examples of Gene Defects Associated with
ment involve genes that encode transcription factors; Congenital Heart Disease*
these typically cause partial loss of function and are auto-
Gene Product
somal dominant (discussed later). Even relatively minor Disorder Gene(s) Function
decrements in activity can result in significant defects.
Nonsyndromic
Moreover, transient environmental stresses during the first
trimester of pregnancy that alter the activity of these same ASD or conduction defects NKX2.5 Transcription factor
genes could conceivably lead to acquired defects that ASD or VSD GATA4 Transcription factor
mimic those produced by heritable mutations. Tetralogy of Fallot ZFPM2 or NKX2.5 Transcription factors
Syndromic†
Etiology and Pathogenesis. Sporadic genetic abnormali-
Alagille syndrome—pulmonary JAG1 or NOTCH2 Signaling proteins or
ties are the major known causes of congenital heart
artery stenosis or tetralogy receptors
disease. They can take the form of single gene mutations, of Fallot
small chromosomal losses, and additions or deletions of
Char syndrome—PDA TFAP2B Transcription factor
whole chromosomes (trisomies and monosomies). In the
case of single gene mutations, the affected genes encode CHARGE syndrome—ASD, CHD7 Helicase-binding
proteins belonging to several different functional classes VSD, PDA, or hypoplastic protein
right side of the heart
(Table 12-3); as noted earlier, many of these involve tran-
scription factors. Since affected patients are heterozygous DiGeorge syndrome—ASD, TBX1 Transcription factor
for such mutations, it follows that a 50% reduction in the VSD, or outflow tract
obstruction
activity of these factors (or even less) may be sufficient to
derange cardiac development. Holt-Oram syndrome—ASD, TBX5 Transcription factor
Moreover, many of the transcription factors interact in VSD, or conduction defect
large protein complexes, providing a rationale for why Noonan syndrome—pulmonary PTPN11, KRAS, Signaling proteins
mutations in any one of several genes can produce similar valve stenosis, VSD, or SOS1
hypertrophic cardiomyopathy
defects. Thus, GATA4, TBX5, and NKX2-5, three transcrip-
tion factors that are mutated in some patients with atrial ASD, Atrial septal defect; CHARGE, posterior coloboma, heart defect, choanal atresia, retarda-
tion, genital and ear anomalies; PDA, patent ductus arteriosus; VSD, ventricular septal defect.
and ventricular septal defects, all bind to one another and *Different mutations can cause the same phenotype, and mutations in some genes can cause
co-regulate the expression of target genes required for multiple phenotypes (e.g., NKX2.5). Many of these congenital lesions also can occur sporadi-
cally, without specific genetic mutation.
proper cardiac development. Of further interest, GATA4 †
Only the cardiac manifestations of the syndrome are listed; the other skeletal, facial, neuro-
and TBX20 are also mutated in rare forms of adult-onset logic, and visceral changes are not.
cardiomyopathy (discussed later), indicating important
roles not only in development but also in maintaining
normal function of the postnatal heart.
Congenital heart disease 533

Other single gene mutations associated with congenital flow down pressure gradients from the left (systemic) side
heart disease can alter structural proteins or affect signal- to the right (pulmonary) side of the circulation or vice
ing pathway molecules. Thus, mutations in genes encoding versa. When blood from the right side of the circulation
various components of the Notch pathway (Chapter 1) flows directly into the left side (right-to-left shunt), hypox-
are associated with a variety of congenital heart defects, emia and cyanosis (a dusky blueness of the skin and mucous
including bicuspid aortic valve (NOTCH1, discussed membranes) result because the pulmonary circulation is
later) and tetralogy of Fallot (JAG1 and NOTCH2). As de- bypassed and poorly oxygenated venous blood shunts
scribed in Chapter 11, fibrillin mutations underlie Marfan directly into the systemic arterial supply. In addition, right-
syndrome—associated with valvular defects and aortic to-left shunts can allow emboli from the peripheral veins
aneurysms. Although fibrillin is an important structural to bypass the lungs and directly enter the systemic circula-
protein in the ECM, it is also an important negative regula- tion (paradoxical embolism). Severe, long-standing cyanosis
tor of TGF-β signaling, and hyperactive TGF-β signaling also causes a peculiar distal blunting and enlargement
contributes to the cardiovascular abnormalities in Marfan (“clubbing”) of the tips of the fingers and toes (called hyper-
syndrome and Loeys-Dietz syndrome. trophic osteoarthropathy), as well as polycythemia. The most
A notable example of a small chromosomal lesion important causes of right-to-left shunts are Tetralogy of
causing congenital heart disease is deletion of chromosome Fallot, transposition of the great arteries, persistent truncus
22q11.2, occurring in up to 50% of patients with DiGeorge arteriosus, tricuspid atresia, and total anomalous pulmo-
syndrome. In this syndrome, the fourth branchial arch and nary venous connection.
the derivatives of the third and fourth pharyngeal pouches In contrast, left-to-right shunts (e.g., ASD, VSD, and
(which contribute to the formation of the thymus, parathy- patent ductus arteriosus [PDA]) increase pulmonary blood
roids, and heart) develop abnormally. The syndrome is flow, but are not initially associated with cyanosis. However,
therefore associated with multiple deficits (memorable left-to-right shunts chronically elevate both volume and
through the mnemonic CATCH-22: cardiac abnormality, pressure in the normally low-pressure, low-resistance pul-
abnormal facies, thymic aplasia, cleft palate, and hypocal- monary circulation. To maintain relatively normal distal
cemia, all on chromosome 22). Of the 30 or so genes present pulmonary capillary and venous pressures, the muscular
on this chromosome segment, deletion specifically of the pulmonary arteries (<1 mm in diameter) initially respond
TBX1 transcription factor gene is probably the culprit by undergoing medial hypertrophy and vasoconstriction.
lesion. TBX1 regulates neural crest migration, as well as the However, prolonged pulmonary arterial vasoconstriction
expansion of cardiac progenitors in the second heart field. stimulates the development of irreversible obstructive
Interestingly, deletions in this region are also associated intimal lesions analogous to the arteriolar changes seen in
with mental illness, including schizophrenia. systemic hypertension; pulmonary arteries can even
Other important genetic causes of congenital heart develop frank atherosclerotic lesions (Chapter 11). The
disease include chromosomal aneuploidies, particularly right ventricle also responds to the pulmonary vascular
Turner syndrome (monosomy X) and trisomies 13, 18, and changes by undergoing progressive right ventricular
21. Indeed, the most common genetic cause of congenital heart hypertrophy. Eventually, pulmonary vascular resistance
disease is trisomy 21 (Down syndrome); roughly 40% of patients approaches systemic levels, and the original left-to-right
with Down syndrome have one or more heart defects, most shunt becomes a right-to-left shunt that introduces poorly
often affecting structures derived from the second heart oxygenated blood into the systemic circulation (Eisenmenger
field (e.g., the atrioventricular septae). The mechanisms by syndrome).
which aneuploidy causes congenital heart disease likely Once irreversible pulmonary hypertension develops,
involve the dysregulated expression of multiple genes. the structural defects of congenital heart disease are con-
Despite these genetic advances, our understanding sidered irreparable; subsequent right heart failure can lead
of the mechanisms underlying congenital heart disease to the patient’s death. This provides the rationale for early
remains rudimentary. Most affected patients have no iden- intervention to close significant left-to-right shunts.
tifiable genetic risk, and even in those that do, the nature Obstructive congenital heart disease occurs when there is
and severity of the defect are highly variable. Thus, envi- abnormal narrowing of chambers, valves, or blood vessels;
ronmental factors, alone or in combination with genetic these include coarctation of the aorta, aortic valvular
factors, are also increasingly implicated in congenital heart stenosis, and pulmonary valvular stenosis. A complete
disease. Examples include congenital rubella infection, ges- obstruction is called an atresia. In some disorders (e.g.,
tational diabetes, and teratogen exposure (including some Tetralogy of Fallot [TOF]), an obstruction (pulmonary ste-
therapeutic drugs). Nutritional factors may also influence nosis) and a shunt (right-to-left through a VSD) are both
risk. For instance, folate supplementation during early present.
pregnancy may reduce congenital heart disease risk. The altered hemodynamics of congenital heart disease
usually cause cardiac dilation or hypertrophy (or both).
Clinical Features. Most of the various structural anoma- However, some defects induce a decrease in the volume
lies in congenital heart disease can be organized into three and muscle mass of a cardiac chamber; this is called hypo-
major categories: plasia if it occurs before birth and atrophy if it develops
postnatally.
• Malformations causing a left-to-right shunt
• Malformations causing a right-to-left shunt
• Malformations causing an obstruction Left-to-Right Shunts
A shunt is an abnormal communication between cham- Left-to-right shunts are the most common congenital heart
bers or blood vessels. Abnormal channels permit blood disease; these include ASD, VSD, and PDA as shown in
534 C H A P T E R 12 The Heart

• Before the growing septum primum completely obliter-


Ao ates the ostium primum, it develops a second posterior
LA opening called the ostium secundum.
• The septum secundum is a subsequent membranous
PT ingrowth located to the right and anterior of the septum
primum.
LA
• As the septum secundum grows, it also leaves a small
RA opening called the foramen ovale that is continuous
with the ostium secundum—the foramen ovale/ostium
LV secundum permits continued right-to-left shunting of
RV
blood during intrauterine development.
• The septum secundum continues to enlarge until it
A forms a flap of tissue that covers the foramen ovale on
its left side.
ASD
This flap of tissue opens and closes in response to pres-
sure gradients between the left and right atria; the valve
Ao opens only when the pressure is greater in the right atrium.
Ao In fetal life, the lungs are nonfunctional, and the pressure
in the pulmonary circulation is greater than that of the sys-
temic circulation; thus, the right atrium is under higher pres-
PT PT sures than the left atrium, and the valve of the foramen
LA LA
ovale is normally open. At birth, with lung expansion, the
RA RA pulmonary vascular pressures drop, and the right atrial
pressures fall below those in the left atrium. As a result,
LV
LV the valve of the foramen ovale closes—and usually perma-
RV RV nently seals (see later).

B C MORPHOLOGY
VSD PDA ASDs are classified according to their location. Secundum
Figure 12-4 Common congenital left-to-right shunts (arrows indicate the ASD (90% of all ASD) result from a deficient septum secundum
direction of blood flow). A, Atrial septal defect (ASD). B, Ventricular septal formation near the center of the atrial septum. These are usually
defect (VSD). With VSD the shunt is left-to-right, and the pressures are the not associated with other anomalies, may be of any size, and
same in both ventricles. Pressure hypertrophy of the right ventricle and can be multiple or fenestrated. Primum anomalies (5% of
volume hypertrophy of the left ventricle are generally present. C, Patent
ASD) occur adjacent to the AV valves and are often associated
ductus arteriosus (PDA). Ao, Aorta; LA, left atrium; LV, left ventricle; PT,
pulmonary trunk; RA, right atrium; RV, right ventricle.
with AV valve abnormalities and/or a VSD. Sinus venosus
defects (5%) are located near the entrance of the superior vena
cava and can be associated with anomalous pulmonary venous
Figure 12-4. ASD typically increases only right ventricular return to the right atrium.
and pulmonary outflow volumes, while VSD and PDA
cause both increased pulmonary blood flow and pressure.
Depending on their size and location, manifestations of Clinical Features. ASDs result in a left-to-right shunt,
these shunts range in severity from no symptoms at all to largely because pulmonary vascular resistance is consider-
fulminant heart failure. ably less than systemic vascular resistance and because the
compliance (distensibility) of the right ventricle is much
Atrial Septal Defect greater than that of the left. Pulmonary blood flow may be
ASDs are abnormal, fixed openings in the atrial septum two to eight times normal. A murmur is often present as
caused by incomplete tissue formation that allows com- a result of excessive flow through the pulmonary valve
munication of blood between the left and right atria; ASDs and/or through the ASD. Despite the right-sided volume
are usually asymptomatic until adulthood (Table 12-2 and overload, ASDs are generally well tolerated and usually do
Fig. 12-4A). ASD should not be confused with patent foramen not become symptomatic before age 30; irreversible pulmo-
ovale (PFO; see later), which represents the failure to close nary hypertension is unusual. ASD closure (surgical or
a foramen (hole) that is part of normal development. Both catheter-based) reverses the hemodynamic abnormalities
ASD and PFO result from defects in the formation of the and prevents the complications, including heart failure,
interatrial septum; a brief summary of the developmental paradoxical embolization, and irreversible pulmonary vas-
stages of this structure follows: cular disease. Mortality is low, and long-term survival is
comparable to that of the normal population.
• The septum primum is a crescent-shaped membranous
ingrowth that sits posteriorly between the right and left Patent Foramen Ovale. The foramen ovale closes perma-
atria and partially separates them; the remaining ante- nently in approximately 80% of people by 2 years of age.
rior opening, called the ostium primum, allows move- However, in the remaining 20%, the unsealed flap can open
ment of blood from the right to left atrium during fetal if right-sided pressures become elevated. Thus, sustained
development. pulmonary hypertension or even transient increases in
Congenital heart disease 535

right-sided pressures, for example, during a bowel move- the development of irreversible obstructive pulmonary
ment, coughing, or sneezing, can produce brief periods of vascular disease.
right-to-left shunting, with the possibility of paradoxical
embolism. Patent Ductus Arteriosus
The ductus arteriosus arises from the pulmonary artery and
Ventricular Septal Defect joins the aorta just distal to the origin of the left subclavian
VSDs are incomplete closures of the ventricular septum, artery. During intrauterine life, it permits blood flow from
allowing free communication of blood between the left to the pulmonary artery to the aorta, thereby bypassing the
right ventricles; they are the most common form of con- unoxygenated lungs. Shortly after birth in healthy term
genital heart disease (Table 12-2 and Fig. 12-4B). infants, the ductus constricts and is functionally closed
after 1 to 2 days; this occurs in response to increased arte-
MORPHOLOGY rial oxygenation, decreased pulmonary vascular resistance,
and declining local levels of prostaglandin E2. Complete
VSDs are classified according to their size and location. Most structural obliteration occurs within the first few months
are about the size of the aortic valve orifice, and about 90% of extrauterine life to form the ligamentum arteriosum.
occur in the region of the membranous interventricular septum Ductal closure is often delayed (or even absent) in infants
(membranous VSD; Fig. 12-5). The remainder occur below with hypoxia (due to respiratory distress or heart disease),
the pulmonary valve (infundibular VSD) or within the muscular or when PDA occurs in association with other congenital
septum. Although most VSDs are single, those in the muscular defects, particularly VSDs that increase pulmonary vascu-
septum may be multiple. lar pressures. PDAs account for about 7% of cases of con-
genital heart disease (Table 12-2 and Fig. 12-4C), and 90%
Clinical Features. Most VSDs that clinically manifest in of these are isolated defects.
the pediatric age group are associated with other congeni- PDA produces a characteristic continuous harsh
tal cardiac anomalies such as Tetralogy of Fallot; only 20% “machinery-like” murmur. The clinical impact of a PDA
to 30% are isolated. Conversely, if a VSD is first detected depends on its diameter and the cardiovascular status of
only in an adult, it is usually an isolated defect. The func- the individual. PDA is usually asymptomatic at birth, and
tional consequences of a VSD depend on the size of the a narrow PDA may have no effect on the child’s growth
defect and whether there are associated right-sided malfor- and development. Because the shunt is initially left-to-
mations. Thus, large VSDs cause difficulties virtually from right, there is no cyanosis. However, with large shunts, the
birth; smaller lesions are generally well tolerated for years additional volume and pressure overloads eventually
and may not be recognized until much later in life. produce obstructive changes in small pulmonary arteries,
Moreover, approximately 50% of small muscular VSDs leading to reversal of flow and its associated consequences.
close spontaneously. Large defects are usually membra- In general, isolated PDA should be closed as early in life
nous or infundibular, and they generally cause significant as is feasible. Conversely, preservation of ductal patency
left-to-right shunting, leading to early right ventricular (by administering prostaglandin E) may be life saving for
hypertrophy and pulmonary hypertension. Over time, infants with various congenital malformations that obstruct
large unclosed VSDs almost universally lead to irreversible the pulmonary or systemic outflow tracts. In aortic valve
pulmonary vascular disease, ultimately resulting in shunt atresia, for example, a PDA may provide the entire sys-
reversal, cyanosis, and death. Surgical or catheter-based temic blood flow.
closure of asymptomatic VSD is generally delayed beyond
infancy, in hope of spontaneous closure. Early correction, Right-to-Left Shunts
however, must be performed for large defects to prevent
The diseases in this group cause cyanosis early in postnatal
life (cyanotic congenital heart disease). Tetralogy of Fallot,
the most common in this group, and transposition of the
great arteries are illustrated schematically in Figure 12-6.
The others include persistent truncus arteriosus, tricus-
pid atresia, and total anomalous pulmonary venous
connection.

Tetralogy of Fallot
The four cardinal features of TOF are (1) VSD, (2) obstruc-
tion of the right ventricular outflow tract (subpulmonary
stenosis), (3) an aorta that overrides the VSD, and (4) right
ventricular hypertrophy (Fig. 12-6A). All of these features
result embryologically from anterosuperior displacement
of the infundibular septum.

MORPHOLOGY
The heart is typically enlarged and is classically “boot-shaped”
due to marked right ventricular hypertrophy. The VSD is usually
Figure 12-5 A ventricular septal defect (membranous type), denoted by the
large with the aortic valve at the superior border, thereby
arrow. (Courtesy William D. Edwards, MD, Mayo Clinic, Rochester, Minn.)
536 C H A P T E R 12 The Heart

soon thereafter. The more severe the subpulmonic stenosis,


Ao
the more hypoplastic are the pulmonary arteries (i.e.,
smaller and thinner-walled), and the larger is the overrid-
ing aorta. As the child grows and the heart increases in
size, the pulmonic orifice does not expand proportionally,
PT making the obstruction progressively worse. The subpul-
LA monary stenosis, however, protects the pulmonary vascu-
RA
lature from pressure overload, and right ventricular failure
is rare because the right ventricle is decompressed by the
shunting of blood into the left ventricle and aorta. Complete
LV
RV surgical repair is possible but becomes complicated for
individuals with pulmonary atresia and dilated bronchial
arteries.

Transposition of the Great Arteries


A Classic tetralogy of Fallot
TGA produces ventriculoarterial discordance. Thus, the
aorta lies anterior and arises from the right ventricle, while
the pulmonary artery is relatively posterior and emanates
Ao
Ao
from the left ventricle (Fig. 12-7; see also Fig. 12-6B). The
atrium-to-ventricle connections are normal (concordant),
with the right atrium joining the right ventricle and the left
PT PT
atrium emptying into the left ventricle. The embryologic
LA LA
defect in complete TGA stems from abnormal formation of
the truncal and aortopulmonary septa. The result is separa-
RA RA tion of the systemic and pulmonary circulations, a condi-
tion incompatible with postnatal life unless a shunt exists
LV LV for adequate mixing of blood.
RV RV The outlook for infants with TGA depends on the degree
of blood “mixing,” the magnitude of tissue hypoxia, and
the ability of the right ventricle to maintain the systemic
With VSD Without VSD
circulation. Patients with TGA and a VSD (approximately
35%) often have a stable shunt. However, dependence
B Complete transposition on a patent foramen ovale or ductus arteriosus for
Figure 12-6 Common congenital right-to-left shunts (cyanotic congenital blood mixing (approximately 65%) is problematic. These
heart disease). A, Tetralogy of Fallot. The direction of shunting across the systemic-to-pulmonary connections tend to close early and
ventricular septal defect (VSD) depends on the degree of the subpulmonary thus require intervention to create a new shunt within the
stenosis; when severe, a right-to-left shunt results (arrow). B, Transposition first few days of life (e.g., balloon atrial septostomy). With
of the great arteries with and without VSD. Ao, Aorta; LA, left atrium; LV, left
ventricle; PT, pulmonary trunk; RA, right atrium; RV, right ventricle.

overriding the defect and both ventricular chambers. The


obstruction to right ventricular outflow is most often due to
narrowing of the infundibulum (subpulmonic stenosis) but can
be accompanied by pulmonary valvular stenosis. Sometimes
there is complete atresia of the pulmonary valve and variable
portions of the pulmonary arteries, such that blood flow through
a PDA, dilated bronchial arteries, or both, is necessary for
survival. Aortic valve insufficiency or an ASD may also be
present; a right aortic arch is present in about 25% of cases.

Clinical Features. Even untreated, patients with TOF can


survive into adult life; 10% of untreated patients are alive
at 20 years and 3% survive for 40 years. The clinical conse-
quences depend primarily on the severity of the subpul-
monary stenosis, since this determines the direction of
blood flow. If the subpulmonary stenosis is mild, the
abnormality resembles an isolated VSD, and the shunt may
be left-to-right, without cyanosis (so-called “pink tetral-
ogy”). With more severe right ventricular outflow obstruction,
right-sided pressures approach or exceed left-sided pressures, and
right-to-left shunting develops, producing cyanosis (classic Figure 12-7 Transposition of the great arteries. (Courtesy William D. Edwards,
TOF). Most infants with TOF are cyanotic from birth or MD, Mayo Clinic, Rochester, Minn.)
Congenital heart disease 537

time, right ventricular hypertrophy becomes prominent, producing only minimal narrowing. Although coarctation
because this chamber functions as the systemic ventricle. of the aorta may occur as a solitary defect, in 50% of cases
Concurrently, the left ventricle becomes thin-walled (atro- it is accompanied by a bicuspid aortic valve and may also
phic) as it supports the low-resistance pulmonary circula- be associated with congenital aortic stenosis, ASD, VSD,
tion. Without surgery, most patients die within months. mitral regurgitation, or berry aneurysms of the circle of
However, improving surgical interventions allow many Willis.
patients with TGA to survive into adulthood. Clinical manifestations depend on the severity of the
narrowing and the patency of the ductus arteriosus.
Tricuspid Atresia Coarctation of the aorta with a PDA usually manifests early
Tricuspid atresia represents complete occlusion of the tri- in life; indeed, it may cause signs and symptoms immedi-
cuspid valve orifice. It results embryologically from ately after birth. In such cases, the delivery of unsaturated
unequal division of the AV canal; thus, the mitral valve is blood through the PDA produces cyanosis localized to the
larger than normal, and there is right ventricular underde- lower half of the body. Many such infants do not survive
velopment (hypoplasia). The circulation can be maintained the neonatal period without surgical or catheter-based
by right-to-left shunting through an interatrial communi- intervention to occlude the PDA.
cation (ASD or patent foramen ovale), in addition to a VSD The outlook is different with coarctation of the aorta
that affords communication between the left ventricle and without a PDA, unless the aortic constriction is severe. Most
the pulmonary artery arising from the hypoplastic right children are asymptomatic, and the disease may go unrec-
ventricle. Cyanosis is present virtually from birth, and ognized until well into adult life. Typically there is hyper-
there is a high early mortality. tension in the upper extremities with weak pulses and
hypotension in the lower extremities, associated with man-
Obstructive Lesions ifestations of arterial insufficiency (i.e., claudication and
coldness). Particularly characteristic is the development
Congenital obstruction to blood flow can occur at the level of collateral circulation between the pre-coarctation and
of the heart valves or within a great vessel. Common exam- post-coarctation arteries through enlarged intercostal and
ples include aortic or pulmonary valve stenosis or atresia, internal mammary arteries, often producing radiographi-
and coarctation of the aorta. Obstruction can also occur cally visible erosions (“notching”) of the undersurfaces of
within a chamber, as with subpulmonary stenosis in TOF. the ribs.
With significant coarctations, murmurs are present
Coarctation of the Aorta throughout systole; sometimes a vibratory “thrill” is also
Coarctation (narrowing, constriction) of the aorta ranks present. The long-standing pressure overload leads to con-
high in frequency among the common structural anoma- centric left ventricular hypertrophy. With uncomplicated
lies. It is twice as common in males as in females; interest- coarctation of the aorta, surgical resection and end-to-end
ingly, females with Turner syndrome are also frequently anastomosis or replacement of the affected aortic segment
affected (Chapter 5). There are two classic forms: (1) an by a prosthetic graft yields excellent results.
“infantile” form—often symptomatic in early childhood—
with tubular hypoplasia of the aortic arch proximal to a Pulmonary Stenosis and Atresia
PDA, and (2) an “adult” form with a discrete ridgelike Pulmonary stenosis or atresia is a relatively frequent mal-
infolding of the aorta just opposite the closed ductus arte- formation leading to obstruction at the level of the pulmo-
riosus (ligamentum arteriosum) distal to the arch vessels nary valve. This can be mild to severe; the lesion can also
(Fig. 12-8). Encroachment on the aortic lumen is variable, be isolated or part of a more complex anomaly—either TOF
sometimes leaving only a small channel and at other times or TGA. Right ventricular hypertrophy typically develops,
and there is sometimes poststenotic dilation of the pulmo-
nary artery due to injury of the wall by “jetting” blood.
With coexistent subpulmonary stenosis (as in TOF), the
Ao Ao pulmonary trunk is not dilated and may in fact be hypo-
plastic. When the valve is entirely atretic, there is no com-
munication between the right ventricle and lungs. In such
PT PT
cases the anomaly is associated with a hypoplastic right
LA LA
ventricle and an ASD; blood reaches the lungs through a
RA RA PDA. Mild stenosis may be asymptomatic and compatible
with long life, whereas symptomatic cases require surgical
LV LV correction.
RV RV
Aortic Stenosis and Atresia
Congenital narrowing and obstruction of the aortic valve
can occur at three locations: valvular, subvalvular, and
With PDA Without PDA supravalvular. Congenital aortic stenosis is an isolated
Coarctation of aorta lesion in 80% of cases. With valvular aortic stenosis the cusps
Figure 12-8 Schematic of aortic coarctation with and without patent ductus may be hypoplastic (small), dysplastic (thickened, nodular),
arteriosus (PDA). Ao, Aorta; LA, left atrium; LV, left ventricle; PT, pulmonary or abnormal in number (usually acommissural or unicom-
trunk; RA, right atrium; RV, right ventricle; PDA, patent ductus arteriosus. missural). In severe congenital aortic stenosis or atresia,
(Courtesy William D. Edwards, MD, Mayo Clinic, Rochester, Minn.) obstruction of the left ventricular outflow tract leads to
538 C H A P T E R 12 The Heart

hypoplasia of the left ventricle and ascending aorta, some- myocardial ischemia—an imbalance between myocardial
times accompanied by dense, porcelain-like left ventricular supply (perfusion) and cardiac demand for oxygenated
endocardial fibroelastosis. The ductus must be open to blood. Ischemia not only limits tissue oxygenation (and
allow blood flow to the aorta and coronary arteries, and thus ATP generation), but also reduces the availability of
the constellation of findings is called the hypoplastic left nutrients and the removal of metabolic wastes (Chapter 2).
heart syndrome. Unless a palliative procedure is done to Thus, cardiac ischemia is generally less well tolerated than
preserve PDA patency, duct closure in the first week of life hypoxia per se, such as may occur with severe anemia,
is generally lethal. However, less severe congenital aortic cyanotic heart disease, or advanced lung disease.
stenosis can be compatible with long survival. In more than 90% of cases, myocardial ischemia results
Subaortic stenosis is caused by a thickened ring or collar from reduced blood flow due to obstructive atherosclerotic
of dense endocardial fibrous tissue below the level of the lesions in the epicardial coronary arteries; consequently,
cusps. Supravalvular aortic stenosis is a congenital aortic IHD is frequently referred to as coronary artery disease
dysplasia with thickening of ascending aortic wall and con- (CAD). In most cases there is a long period (up to decades)
sequent luminal constriction. In some cases it is a compo- of silent, slow progression of coronary lesions before the
nent of a multiorgan developmental disorder resulting sudden onset of symptoms. Thus, IHD is often the late
from deletions on chromosome 7 that include the gene manifestation of coronary atherosclerosis that began during
for elastin. Other features of the syndrome include childhood or adolescence (Chapter 11).
hypercalcemia, cognitive abnormalities, and characteristic IHD can present as one or more of the following clinical
facial anomalies (Williams-Beuren syndrome). Elastin gene syndromes:
mutations may cause supravalvular stenosis by disrupting
elastin–smooth muscle cell interactions during aortic
• Myocardial infarction (MI), where ischemia causes frank
cardiac necrosis
morphogenesis.
Subaortic stenosis is usually associated with a promi- • Angina pectoris (literally “chest pain”), where ischemia
is not severe enough to cause infarction, but the symp-
nent systolic murmur and sometimes a thrill. Pressure toms nevertheless portend infarction risk
hypertrophy of the left ventricle develops as a consequence
of the obstruction to blood flow, but congenital stenoses • Chronic IHD with heart failure
are well tolerated unless very severe. Although mild ste- • Sudden cardiac death (SCD).
noses can often be managed conservatively with antibiotic In addition to coronary atherosclerosis, myocardial isch-
prophylaxis (to prevent endocarditis) and avoidance of emia can be caused by coronary emboli, myocardial vessel
strenuous activity, the resulting left ventricular hypertro- inflammation, or vascular spasm. Moreover, otherwise
phy still carries a risk of sudden death with exertion. modest vascular occlusions may become consequential in
the setting of increased cardiac energy demand (e.g., myo-
cardial hypertrophy or increased heart rate), hypoxemia,
KE Y CONCEPTS
or systemic hypotension (e.g., shock). Some conditions
Congenital Heart Disease can have multiple deleterious effects. Thus, tachycardia
■ Congenital heart disease represents defects of cardiac increases oxygen demand (because of more contractions
chambers or the great vessels; these either result in shunt- per unit time) while decreasing functional supply (by
ing of blood between the right and left circulation or cause decreasing the relative time spent in diastole, when cardiac
outflow obstructions. Lesions range from relatively asymp- perfusion occurs).
tomatic to rapidly fatal. Environmental (toxic or infectious)
and genetic causes both contribute, and the manifesta- Epidemiology. IHD is the leading cause of death in the
tions depend on the timing of the environmental insult or United States (one of every six deaths in 2008; more
which step in cardiac development is affected. than 400,000 individuals) and other developed nations
(approximately 7 million total per year). As high as these
■ Left-to-right shunts are most common and are typically
numbers are, they represent a substantial improvement
associated with ASDs, VSDs, or a PDA. These lesions
relative to just one to two generations ago. Since peaking
result in chronic right-sided pressure and volume over-
in 1963, the overall death rate from IHD has fallen in the
loads that eventually cause pulmonary hypertension with
United States by approximately 50%. This remarkable
reversal of flow and right-to-left shunts with cyanosis
improvement can be attributed to:
(Eisenmenger syndrome).
Right-to-left shunts are most commonly caused by TOF or

TGA. These are cyanotic lesions from the outset and are
• Prevention, achieved by modifying important risk fac-
tors, such as smoking, blood cholesterol, and hyperten-
associated with polycythemia, hypertrophic osteoarthrop- sion. Additional risk reduction and prevention may
athy, and paradoxical emboli. potentially be achieved by maintenance of normal blood
■ Obstructive lesions include aortic coarctation; the clinical glucose levels in diabetic patients, control of obesity,
severity of the lesion depends on the degree of stenosis and exercise.
and the patency of the ductus arteriosus.
• Diagnostic and therapeutic advances, allowing earlier
and more effective treatments. The latter include new
medications, including the use of cholesterol lowering
Ischemic Heart Disease drugs such as statins, coronary care units, thrombolysis
for MI, percutaneous transluminal coronary angio-
Ischemic heart disease (IHD) represents a group of patho- plasty, endovascular stents, coronary artery bypass
physiologically related syndromes resulting from graft (CABG) surgery, and improved control of heart
Ischemic heart disease 539

failure and arrhythmias via left ventricular assist of atheromatous plaques. However, the varied clinical
devices, implantable defibrillators, and cardiac resyn- manifestations of IHD cannot be explained by the anatomic
chronization with pacemakers. Even a simple daily pro- disease burden alone. This is particularly true for the
phylactic aspirin can have therapeutic benefit. so-called acute coronary syndromes, namely unstable
angina, acute MI, and sudden death. These acute coronary
Continuing this encouraging trend will be challenging, syndromes are typically initiated by an unpredictable and
particularly in view of the increased longevity of “baby abrupt conversion of a stable atherosclerotic plaque to an
boomers” (which will lead to a doubling of individuals unstable and potentially life-threatening atherothrombotic
older than age 65 by 2050), the “obesity epidemic,” and lesion through rupture, superficial erosion, ulceration,
other factors. Increasingly, new therapeutic advances will fissuring, or deep hemorrhage (Chapter 11). In most
depend on understanding the genetic determinants of cor- instances, plaque changes—typically associated with intra-
onary atherosclerosis and IHD. For example, the observa- lesional inflammation—precipitate the formation of a
tion that MIs occur in only a fraction of individuals with superimposed thrombus that partially or completely
coronary disease suggests that simple control of atheroscle- occludes the artery.
rotic risk factors is only part of the story. For example, MI
risk—but not coronary atherosclerosis—is associated with Consequences of Myocardial Ischemia. The common
genetic variants that modify leukotriene B4 metabolism. feature of the acute coronary syndromes is downstream
myocardial ischemia.
Pathogenesis. The dominant cause of IHD syndromes is
insufficient coronary perfusion relative to myocardial
• Stable angina results from increases in myocardial
oxygen demand that outstrip the ability of stenosed
demand; in the vast majority of cases, this is due to coronary arteries to increase oxygen delivery; it is
chronic, progressive atherosclerotic narrowing of the epi- usually not associated with plaque disruption.
cardial coronary arteries, and variable degrees of super-
imposed acute plaque change, thrombosis, and vasospasm. • Unstable angina is caused by plaque disruption that
results in thrombosis and vasoconstriction, and leads to
The individual elements and their interactions are dis- severe but transient reductions in coronary blood flow.
cussed next. In some cases, microinfarcts can occur distal to dis-
rupted plaques due to thromboemboli.
Chronic Vascular Occlusion. More than 90% of patients
with IHD have atherosclerosis involving one or more of the • Myocardial infarction (MI) is often the result of acute
plaque change that induces an abrupt thrombotic occlu-
epicardial coronary arteries (Chapter 11). The clinical man-
sion, resulting in myocardial necrosis.
ifestations of coronary atherosclerosis are generally due to
progressive narrowing of the lumen leading to stenosis • Sudden cardiac death may be caused by regional myo-
(“fixed” obstructions), or to acute plaque erosion or rupture cardial ischemia that induces a fatal ventricular
with thrombosis, all of which compromise blood flow. A arrhythmia.
fixed lesion obstructing greater than 75% of vascular cross- Each of these important syndromes is discussed in
sectional area defines significant coronary artery disease; detail next, followed by an examination of the important
this is generally the threshold for symptomatic ischemia myocardial consequences.
precipitated by exercise (typically manifesting as angina).
With this degree of obstruction, compensatory coronary Angina Pectoris
arterial vasodilation is no longer sufficient to meet even
moderate increases in myocardial demand. Obstruction of Angina pectoris is characterized by paroxysmal and
90% of the cross-sectional area of the lumen can lead to usually recurrent attacks of substernal or precordial chest
inadequate coronary blood flow even at rest. Progressive discomfort caused by transient (15 seconds to 15 minutes)
myocardial ischemia induced by slowly developing occlu- myocardial ischemia that is insufficient to induce myocyte
sions may stimulate the formation of collateral vessels over necrosis. The pain itself is likely a consequence of the
time, which can often protect against myocardial ischemia ischemia-induced release of adenosine, bradykinin, and
and infarction and mitigate the effects of high-grade other molecules that stimulate sympathetic and vagal
stenoses. afferent nerves. There are three overlapping patterns
Although only a single major coronary epicardial vessel of angina pectoris caused by varying combinations of
may be affected, two or all three—the LAD, LCX, and decreased perfusion, increased demand, and coronary
RCA—are often involved by obstructive atherosclerosis. arterial pathology. Importantly, not all ischemic events are
Clinically significant plaques can be located anywhere perceived by patients; silent ischemia is particularly
along the course of the vessels, particularly the RCA, common in the geriatric population and in the setting of
although they tend to predominate within the first several diabetic neuropathy.
centimeters of the LAD and LCX. Sometimes the major
epicardial branches are also involved (i.e., LAD diagonal • Stable (typical) angina is the most common form of
branches, LCX obtuse marginal branches, or posterior angina; it is caused by an imbalance in coronary perfusion
descending branch of the RCA), but atherosclerosis of the (due to chronic stenosing coronary atherosclerosis) relative to
intramural (penetrating) branches is rare. myocardial demand, such as that produced by physical
activity, emotional excitement or psychological stress.
Acute Plaque Change. The risk of an individual develop- Typical angina pectoris is variously described as a deep,
ing clinically important IHD depends in part on the poorly localized pressure, squeezing, or burning sensa-
number, distribution, structure, and degree of obstruction tion (like indigestion), but unusually as pain, and is
540 C H A P T E R 12 The Heart

usually relieved by rest (decreasing demand) or admin- Compelling evidence for this sequence derives from
istering vasodilators, such as nitroglycerin and calcium (1) autopsy studies of patients dying of acute MI; (2) angio-
channel blockers (increasing perfusion). graphic studies demonstrating a high frequency of throm-
• Prinzmetal variant angina is an uncommon form of episodic botic occlusion early after MI; (3) the high success rate of
myocardial ischemia; it is caused by coronary artery spasm. coronary revascularization following MI (i.e., thromboly-
Although individuals with Prinzmetal variant angina sis, angioplasty, stent placement, and surgery); and (4) the
may well have significant coronary atherosclerosis, the demonstration of residual disrupted atherosclerotic lesions
anginal attacks are unrelated to physical activity, heart by angiography after thrombolysis. Coronary angiography
rate, or blood pressure. Prinzmetal angina generally performed within 4 hours of MI onset shows coronary
responds promptly to vasodilators. thrombosis in almost 90% of cases. However, angiography
• Unstable or crescendo angina refers to a pattern of increas- after 12 to 24 hours reveals thrombosis only about 60% of
ingly frequent, prolonged (>20 min), or severe angina or chest the time, suggesting resolution due to fibrinolysis, relax-
discomfort that is described as frank pain, precipitated by ation of spasm, or both.
progressively lower levels of physical activity or even occur- In approximately 10% of cases, transmural MI occurs in
ring at rest. In most patients, unstable angina is caused the absence of the typical coronary atherothrombosis. In
by the disruption of an atherosclerotic plaque with such situations, other mechanisms may be responsible for
superimposed partial thrombosis and possibly emboli- the reduced coronary blood flow, including:
zation or vasospasm (or both). Approximately one-half • Vasospasm with or without coronary atherosclerosis,
of patients with unstable angina have evidence of perhaps in association with platelet aggregation or due
myocardial necrosis; for others, acute MI may be to drug ingestion (e.g., cocaine or ephedrine)
imminent. • Emboli from the left atrium in association with atrial
fibrillation, a left-sided mural thrombus, vegetations of
Myocardial Infarction infective endocarditis, intracardiac prosthetic material;
or paradoxical emboli from the right side of the heart or
Myocardial infarction, also commonly referred to as “heart the peripheral veins, traversing a patent foramen ovale
attack,” is the death of cardiac muscle due to prolonged and into the coronary arteries
severe ischemia. Roughly 1.5 million individuals in the
United States suffer an MI annually. • Ischemia without detectable or significant coronary athero-
sclerosis and thrombosis may be caused by disorders of
small intramural coronary vessels (e.g., vasculitis),
Incidence and Risk Factors. MI can occur at virtually any
hematologic abnormalities (e.g., sickle cell disease),
age; nearly 10% of myocardial infarcts occur in people
amyloid deposition in vascular walls, vascular dissec-
younger than age 40, and 45% occur in people younger
tion, marked hypertrophy (e.g., aortic stenosis), lowered
than age 65. Nevertheless, MI frequency rises progres-
systemic blood pressure (e.g., shock), or inadequate
sively with increasing age. The incidence of MI also strongly
myocardial “protection” during cardiac surgery.
correlates with genetic and behavioral predispositions to
atherosclerosis. Blacks and whites are equally affected. Myocardial Response. Coronary arterial obstruction
Through middle age, male gender increases the relative diminishes blood flow to a region of myocardium (Fig.
risk of MI; indeed, women are generally protected against 12-9), causing ischemia, rapid myocardial dysfunction,
MI during their reproductive years. However, postmeno- and eventually—with prolonged vascular compromise—
pausal decline in estrogen production is usually associated myocyte death. The anatomic region supplied by that
with accelerated CAD, and IHD is the most common cause artery is referred to as the area at risk. The outcome
of death in older women. Unfortunately, postmenopausal depends predominantly on the severity and duration of
hormonal replacement therapy has not been shown to be flow deprivation (Fig. 12-10).
protective, and in fact, in some cases, may be detrimental. The early biochemical consequence of myocardial isch-
emia is the cessation of aerobic metabolism within seconds,
Pathogenesis leading to inadequate production of high-energy phos-
Coronary Arterial Occlusion. The following sequence of phates (e.g., creatine phosphate and adenosine triphos-
events likely underlies most MIs (see Chapter 11 for addi- phate) and accumulation of potentially noxious metabolites
tional details): (e.g., lactic acid) (Fig. 12-10A). Because of the exquisite
dependence of myocardial function on oxygen and nutri-
• A coronary artery atheromatous plaque undergoes an
ents, myocardial contractility ceases within a minute or so
acute change consisting of intraplaque hemorrhage,
of the onset of severe ischemia. Such loss of function actu-
erosion or ulceration, or rupture or fissuring.
ally precipitates heart failure long before myocyte death
• When exposed to subendothelial collagen and necrotic occurs.
plaque contents, platelets adhere, become activated, As detailed in Chapter 2, ultrastructural changes (includ-
release their granule contents, and aggregate to form ing myofibrillar relaxation, glycogen depletion, cell and
microthrombi. mitochondrial swelling) also develop within a few minutes
• Vasospasm is stimulated by mediators released from of the onset of ischemia. Nevertheless, these early manifes-
platelets. tations of ischemic injury are potentially reversible. Indeed,
• Tissue factor activates the coagulation pathway, adding experimental and clinical evidence shows that only severe
to the bulk of the thrombus. ischemia (blood flow 10% or less of normal) lasting 20 to
• Within minutes, the thrombus can expand to completely 30 minutes or longer leads to irreversible damage (necro-
occlude the vessel lumen. sis) of cardiac myocytes. This delay in the onset of
Ischemic heart disease 541

Table 12-4 Approximate Time of Onset of Key Events in Ischemic


Cardiac Myocytes
Feature Time
Onset of ATP depletion Seconds
Loss of contractility <2 min
ATP reduced
to 50% of normal 10 min
to 10% of normal 40 min
Irreversible cell injury 20-40 min
Microvascular injury >1 hr
ATP, Adenosine triphosphate.

intracellular myocardial proteins into the circulation forms


the basis for blood tests that can sensitively detect irrevers-
ible myocyte damage, and are important for managing MI
(see later). With prolonged severe ischemia, injury to the
microvasculature follows injury to the cardiac myocytes.
Figure 12-9 Postmortem angiogram showing the posterior aspect of the The temporal progression of these events is summarized
heart of a patient who died during the evolution of acute myocardial infarction, in Table 12-4.
demonstrating total occlusion of the distal right coronary artery by an acute The progression of ischemic necrosis in the myocardium
thrombus (arrow) and a large zone of myocardial hypoperfusion involving the is summarized in Figure 12-11. Due to the myocardial per-
posterior left and right ventricles, as indicated by arrowheads, and having fusion pattern from epicardium to endocardium, ischemia
almost absent filling of capillaries. The heart has been fixed by coronary
is most pronounced in the subendocardium; thus, irrevers-
arterial perfusion with glutaraldehyde and cleared with methyl salicylate, fol-
lowed by intracoronary injection of silicone polymer (yellow). (Photograph
ible injury of ischemic myocytes occurs first in the suben-
courtesy Lewis L. Lainey. Reproduced with permission from Schoen FJ: docardial zone. With more extended ischemia, a wavefront
Interventional and Surgical Cardiovascular Pathology: Clinical Correlations of cell death moves through the myocardium to encompass
and Basic Principles. Philadelphia, WB Saunders, 1989, p. 60.) progressively more of the transmural thickness and breadth
of the ischemic zone. The precise location, size, and specific
morphologic features of an acute MI depend on:
permanent myocardial injury provides the rationale for
rapid diagnosis in acute MI—to permit early coronary • The location, severity, and rate of development of
intervention to establish reperfusion and salvage as much coronary obstructions due to atherosclerosis and
“at risk” myocardium as possible. thromboses
The earliest detectable feature of myocyte necrosis is the • The size of the vascular bed perfused by the obstructed
disruption of the integrity of the sarcolemmal membrane, vessels
allowing intracellular macromolecules to leak out of • The duration of the occlusion
necrotic cells into the cardiac interstitium and ultimately • The metabolic and oxygen needs of the myocardium
into the microvasculature and lymphatics. This escape of at risk

100
6 Onset of irreversible injury
ATP and Lactate (arbitrary units)

Fraction of at-risk myocardium

Ischemic myocardium
80 potentially salvageable
5
by timely intervention
Irreversible phase
Reversible phase

te
cta
4 La 60

3
40
2

20
1 ATP Cumulative
dead myocardium
0
0 5 10 15 20 30 40 50 0 1 2 3 4 5 6 12 18
A Minutes B Time Hours
Figure 12-10 Temporal sequence of early biochemical findings and progression of necrosis after onset of severe myocardial ischemia. A, Early changes include
loss of adenosine triphosphate (ATP) and accumulation of lactate. B, For approximately 30 minutes after the onset of even the most severe ischemia, myo-
cardial injury is potentially reversible. Thereafter, progressive loss of viability occurs that is complete by 6 to 12 hours. The benefits of reperfusion are greatest
when it is achieved early, and are progressively lost when reperfusion is delayed. (Modified with permission from Antman E: Acute myocardial infarction. In
Braunwald E, et al [eds]: Heart Disease: A Textbook of Cardiovascular Medicine, 6th ed. Philadelphia, WB Saunders, 2001, pp 1114-1231.)
542 C H A P T E R 12 The Heart

Aorta

Pulmonary
artery

Left circumflex coronary


artery
Right
coronary Left anterior descending
artery coronary artery
Acute coronary
arterial occlusion

Zone of perfusion
(area at risk)

Completed infarct
Cross-section involving nearly the
of myocardium entire area at risk

Obstructed
coronary
artery

Endocardium

Zone of Zone of
Zone of perfusion necrosis necrosis
(area at risk)

0 hr 2 hr 24 hr
Figure 12-11 Progression of myocardial necrosis after coronary artery occlusion. Necrosis begins in a small zone of the myocardium beneath the endocardial
surface in the center of the ischemic zone. The area that depends on the occluded vessel for perfusion is the “at risk” myocardium (shaded). Note that a very
narrow zone of myocardium immediately beneath the endocardium is spared from necrosis because it can be oxygenated by diffusion from the ventricle.

• The extent of collateral blood vessels “dominant” (even though the LAD and LCX collectively
• The presence, site, and severity of coronary arterial perfuse the majority of the left ventricular myocardium).
spasm In a right dominant circulation (present in approximately
• Other factors, such as heart rate, cardiac rhythm, and 80% of individuals), the RCA supplies the entire right
blood oxygenation ventricular free wall, the posterobasal wall of the left
ventricle, and the posterior third of the ventricular septum,
Necrosis involves approximately half of the thickness while the LCX generally perfuses only the lateral wall of
of the myocardium in 2 to 3 hours of the onset of severe the left ventricle. Thus, RCA occlusions can potentially
myocardial ischemia, and is usually transmural within 6 lead to left ventricular damage. Although most hearts
hours. However, in instances where chronic sublethal isch- have numerous intercoronary anastomoses (collateral cir-
emia has induced a more well-developed coronary collat- culation), relatively little blood normally courses through
eral circulation, the progression of necrosis may follow a these. However, when a coronary artery is progressively
more protracted course (12 hours or longer). narrowed over time, blood flows via the collaterals
Knowledge of the areas of myocardium perfused from the high- to the low-pressure circulating causing
by the major coronary arteries allows correlation of the channels to enlarge. Through such progressive dilation
specific vascular obstructions with their corresponding and growth of collaterals, stimulated by ischemia, blood
areas of myocardial infarction. Typically, the LAD branch flow is provided to areas of myocardium that would
of the left coronary artery supplies most of the apex of otherwise be deprived of adequate perfusion. Indeed,
the heart, the anterior wall of the left ventricle, and the in the setting of extensive collateralization, the normal
anterior two thirds of the ventricular septum. By con- epicardial perfusion territories may be so expanded that
vention, the coronary artery—either RCA or LCX—that subsequent occlusion leads to infarction in paradoxical
perfuses the posterior third of the septum is called distributions.
Ischemic heart disease 543

TRANSMURAL INFARCTS NON-TRANSMURAL INFARCTS

Restoration of flow (reperfusion)


Permanent
Transient/partial
occlusion of
obstruction
left anterior
regional
descending
subendocardial
branch
infarct

Posterior

Permanent Global
occlusion of hypotension
left circumflex RV LV circumferential
branch subendocardial
infarct

Anterior
Permanent
occlusion of Small
right coronary intramural
artery (or its vessel
posterior occlusions
descending microinfarcts
branch)

Figure 12-12 Distribution of myocardial ischemic necrosis correlates with the location and nature of decreased perfusion. Left, The positions of transmural
acute infarcts resulting from occlusions of the major coronary arteries; top to bottom, left anterior descending, left circumflex, and right coronary arteries.
Right, The types of infarcts that result from a partial or transient occlusion, global hypotension, or intramural small vessel occlusions.

Patterns of Infarction. The distribution of myocardial endogenous catechols (epinephrine) or drugs (cocaine
necrosis correlates with the location and cause of the or ephedrine). Elevated levels of catechols also increase
decreased perfusion (Fig. 12-12). heart rate and myocardial contractility, exacerbating
ischemia caused by the vasospasm. The outcome of such
• Transmural infarction. Myocardial infarcts caused by vasospasm can be sudden cardiac death (usually caused
occlusion of an epicardial vessel (in the absence of any by a fatal arrhythmia) or an ischemic dilated cardiomy-
therapeutic intervention) are typically transmural—the opathy, so-called takotsubo cardiomyopathy (also called
necrosis involves virtually the full thickness of the ven- “broken heart syndrome” because of the association
tricular wall in the distribution of the affected coronary. with emotional duress).
This pattern of infarction is usually associated with a
combination of chronic coronary atherosclerosis, acute Owing to the characteristic electrocardiographic changes
plaque change, and superimposed thrombosis (dis- resulting from myocardial ischemia or necrosis in various
cussed earlier). distributions, a transmural infarct is sometimes referred to
• Subendocardial (nontransmural) infarction. As the suben- as an “ST elevation myocardial infarct” (STEMI) and a
docardial zone is normally the least perfused region subendocardial infarct as a “non–ST elevation infarct”
of myocardium, this area is most vulnerable to any (NSTEMI). Depending on the extent and location of the
reduction in coronary flow. A subendocardial infarct— vascular involvement, microinfarctions show nonspecific
typically involving roughly the inner third of the ven- changes or can even be electrocardiographically silent.
tricular wall—can occur as a result of a plaque disruption
followed by a coronary thrombus that becomes lysed
(therapeutically or spontaneously) before myocardial MORPHOLOGY
necrosis extends across the full thickness of the wall.
Subendocardial infarcts can also result from prolonged, The temporal evolution of the morphologic changes in acute MI
severe reduction in systemic blood pressure, as in shock and subsequent healing are summarized in Table 12-5.
superimposed on chronic, otherwise noncritical, coro- Nearly all transmural infarcts involve at least a portion of the
nary stenoses. In the subendocardial infarcts that occur left ventricle (comprising the free wall and ventricular septum)
as a result of global hypotension, myocardial damage is and encompass nearly the entire perfusion zone of the occluded
usually circumferential, rather than being limited to the coronary artery save for a narrow rim (approximately 0.1 mm)
distribution of a single major coronary artery. of preserved subendocardial myocardium that is preserved by
diffusion of oxygen and nutrients from the ventricular lumen.
• Multifocal microinfarction. This pattern is seen when
Of MIs caused by a right coronary obstruction, 15% to 30%
there is pathology involving only smaller intramural
extend from the posterior free wall of the septal portion of the
vessels. This may occur in the setting of microemboliza-
left ventricle into the adjacent right ventricular wall. Isolated
tion, vasculitis, or vascular spasm, for example, due to
544 C H A P T E R 12 The Heart

Table 12-5 Evolution of Morphologic Changes in Myocardial Infarction


Time Gross Features Light Microscope Electron Microscope
Reversible Injury
0- 12 hr None None Relaxation of myofibrils; glycogen
loss; mitochondrial swelling
Irreversible Injury
1
2 -4 hr None Usually none; variable waviness of fibers at border Sarcolemmal disruption;
mitochondrial amorphous densities
4-12 hr Dark mottling (occasional) Early coagulation necrosis; edema; hemorrhage
12-24 hr Dark mottling Ongoing coagulation necrosis; pyknosis of nuclei; myocyte
hypereosinophilia; marginal contraction band necrosis; early
neutrophilic infiltrate
1-3 days Mottling with yellow-tan infarct Coagulation necrosis, with loss of nuclei and striations; brisk
center interstitial infiltrate of neutrophils
3-7 days Hyperemic border; central Beginning disintegration of dead myofibers, with dying neutrophils;
yellow-tan softening early phagocytosis of dead cells by macrophages at infarct border
7-10 days Maximally yellow-tan and soft, Well-developed phagocytosis of dead cells; granulation tissue at
with depressed red-tan margins margins
10-14 days Red-gray depressed infarct Well-established granulation tissue with new blood vessels and
borders collagen deposition
2-8 wk Gray-white scar, progressive from Increased collagen deposition, with decreased cellularity
border toward core of infarct
>2 mo Scarring complete Dense collagenous scar

infarction of the right ventricle is unusual (only 1% to 3% of necrosis by immersion of tissue slices in a solution of triphenyl-
cases), as is infarction of the atria. tetrazolium chloride. This gross histochemical stain imparts
The frequencies of involvement of each of the three main a brick-red color to intact, noninfarcted myocardium where
arterial trunks and the corresponding sites of myocardial lesions lactate dehydrogenase activity is preserved. Because dehydro-
resulting in infarction (in the typical right dominant heart) are as genases leak out through the damaged membranes of dead
follows (Fig. 12-12A): cells, an infarct appears as an unstained pale zone (Fig. 12-13).
By 12 to 24 hours after infarction, an MI can usually be identified
• Left anterior descending coronary artery (40% to 50%):
grossly as a reddish-blue area of discoloration caused by
infarcts involving the anterior wall of left ventricle near the
stagnated, trapped blood. Thereafter, the infarct becomes
apex; the anterior portion of ventricular septum; and the apex
circumferentially
• Right coronary artery (30% to 40%): infarcts involving the
inferior/posterior wall of left ventricle; posterior portion of
ventricular septum; and the inferior/posterior right ventricular
free wall in some cases
• Left circumflex coronary artery (15% to 20%): infarcts involv-
ing the lateral wall of left ventricle except at the apex
Other locations of critical coronary arterial lesions causing
infarcts are sometimes encountered, such as the left main coro-
nary artery, the secondary (diagonal) branches of the left ante-
rior descending coronary artery, or the marginal branches of the
left circumflex coronary artery.
The gross and microscopic appearance of an infarct
depends on the duration of survival of the patient follow-
ing the MI. Areas of damage undergo a progressive sequence
of morphologic changes involving typical ischemic coagulative
necrosis (the predominant mechanism of cell death in MI,
although apoptosis may also occur), followed by inflammation
and repair that closely parallels responses to injury in other
tissues. Figure 12-13 Acute myocardial infarct, predominantly of the posterolateral
Early morphologic recognition of acute MI can be difficult, left ventricle, demonstrated histochemically by a lack of staining by triphen-
particularly when death occurs within a few hours of the onset yltetrazolium chloride in areas of necrosis (arrow). The staining defect is due
of symptoms. MIs less than 12 hours old are usually not appar- to the lactate dehydrogenase leakage that follows cell death. Note the myo-
ent on gross examination. However, if the infarct preceded cardial hemorrhage at one edge of the infarct that was associated with
cardiac rupture, and the anterior scar (arrowhead), indicative of old infarct.
death by 2 to 3 hours, it is possible to highlight the area of
Specimen is oriented with the posterior wall at the top.
Ischemic heart disease 545

progressively more sharply defined, yellow-tan, and soft. By 10 heal from the margins toward the center. Consequently, a large
days to 2 weeks, it is rimmed by a hyperemic zone of highly infarct may not heal as quickly or as completely as a small one.
vascularized granulation tissue. Over the succeeding weeks, A healing infarct can also appear nonuniform, with the most
the injured region evolves to a fibrous scar. advanced healing at the periphery. Once a lesion is completely
The histopathologic changes also proceed in a fairly predict- healed, it is impossible to determine its age (i.e., the dense
able sequence (Fig. 12-14). The typical changes of coagulative fibrous scar of 8-week-old and 10-year-old infarcts looks virtu-
necrosis become detectable in the first 6 to 12 hours. “Wavy ally identical).
fibers” may be present at the periphery of the infarct; these
changes probably result from the forceful systolic tugs of the
The following discussion considers the changes that
viable fibers on immediately adjacent, noncontractile dead
result from interventions that can limit infarct size by sal-
fibers, causing stretching and folding. An additional sublethal
vaging myocardium that is not yet necrotic.
ischemic change may be seen in the margins of infarcts:
so-called myocyte vacuolization or myocytolysis, which
Infarct Modification by Reperfusion. Reperfusion is the
reflects intracellular accumulations of salt and water within the
restoration of blood flow to ischemic myocardium threat-
sarcoplasmic reticulum. The necrotic muscle elicits acute
ened by infarction; the goal is to salvage cardiac muscle
inflammation (most prominent between 1 and 3 days).
at risk and limit infarct size. The cardiology adage that
Thereafter, macrophages remove the necrotic myocytes (most
“time is myocardium” succinctly captures the impetus to
noticeable by 3 to 7 days), and the damaged zone is progres-
intervene promptly once ongoing infarction is diagnosed;
sively replaced by the ingrowth of highly vascularized granu-
patient outcome materially worsens with progressively
lation tissue (most prominent at 1 to 2 weeks); as healing
larger infarcts. Not only does reperfusion improve short-
progresses, this is replaced by fibrous tissue. In most instances,
and long-term survival, but it also impacts short- and long-
scarring is well advanced by the end of the sixth week, but the
term myocardial function. Thus, prompt reperfusion is the
efficiency of repair depends on the size of the original lesion, as
preeminent objective for treatment of patients with MI.
well as the relative metabolic and inflammatory state of the host.
This can be accomplished by a host of coronary interven-
Since healing requires the participation of inflammatory cells,
tions, that is, thrombolysis, angioplasty, stent placement,
immune suppression (e.g., due to steroids) can impair the vigor
or coronary artery bypass graft [CABG] surgery. The goal
of the healing response. Moreover, delivering inflammatory cells
is to dissolve, mechanically alter, or bypass the lesion
to the site of necrosis requires intact vasculature; since blood
that precipitated the acute infarction. The benefits of reper-
vessels often survive only at the edges of an infarct, MIs typically
fusion correlate with (1) the rapidity of reestablishing

A B C

Figure 12-14 Microscopic features of myocardial


infarction and its repair. A, One-day-old infarct
showing coagulative necrosis and wavy fibers
(elongated and narrow, as compared with adjacent
normal fibers at right). Widened spaces between
the dead fibers contain edema fluid and scattered
neutrophils. B, Dense polymorphonuclear leuko-
cytic infiltrate in an acute myocardial infarction that
is 3 to 4 days old. C, Removal of necrotic myo-
cytes by phagocytosis (approximately 7 to 10
days). D, Granulation tissue characterized by loose
collagen and abundant capillaries. E, Healed myo-
cardial infarct, in which the necrotic tissue has
been replaced by a dense collagenous scar. The
D E residual cardiac muscle cells show evidence of
compensatory hypertrophy.
546 C H A P T E R 12 The Heart

A B
Figure 12-15 Consequences of myocardial ischemia followed by reperfusion. Gross (A) and microscopic (B) appearance of myocardium modified by reperfu-
sion. A, Large, densely hemorrhagic, anterior wall acute myocardial infarction in a patient with left anterior descending artery thrombus treated with strepto-
kinase, a fibrinolytic agent (triphenyl tetrazolium chloride-stained heart slice). Specimen oriented with posterior wall at top. B, Myocardial necrosis with
hemorrhage and contraction bands, visible as dark bands spanning some myofibers (arrow).

coronary blood flow (the first 3 to 4 hours following sarcomeres. These result from the exaggerated contraction
obstruction are critical) and (2) the extent of restoration of of sarcomeres when perfusion is reestablished, at which
blood flow and correction of the underlying causal lesion. time the interior of cells with damaged membranes is
Indeed, thrombolysis can remove a thrombus occluding a exposed to a high concentration of calcium ions from the
coronary artery, but does not alter the underlying athero- plasma. Thus, reperfusion not only salvages reversibly injured
sclerotic plaque that initiated it. In contrast, percutaneous cells but also alters the morphology of lethally injured cells.
transluminal coronary angioplasty (PTCA) with stent The effects of reperfusion on myocardial viability and
placement not only eliminates a thrombotic occlusion but function are discussed later and summarized in Figure
also relieves some of the original obstruction and instabil- 12-16. Although clearly beneficial, reperfusion can trigger
ity caused by the underlying disrupted plaque. CABG pro- deleterious complications, including arrhythmias as well
vides a new conduit for flow bypassing the area of vessel as damage superimposed on the original ischemia, so-called
blockage. reperfusion injury. This term encompasses various forms of
The typical appearance of reperfused myocardium damage that can occur after restoration of flow to “vulner-
is illustrated in Figure 12-15. Reperfused infarcts are able” myocardium that is ischemic but not yet irreversibly
usually hemorrhagic because the vasculature is injured damaged (Fig. 12-16B). As discussed in Chapter 2, reperfu-
during ischemia and there is bleeding after flow is re- sion injury may be mediated by oxidative stress, calcium
stored. Microscopic examination reveals that irreversibly overload, and inflammatory cells recruited after tissue
injured myocytes exhibit contraction bands, intensely eosin- reperfusion. Reperfusion-induced microvascular injury
ophilic intracellular “stripes” composed of closely packed not only results in hemorrhage but can also cause

PERMANENT OCCLUSION TEMPORARY OCCLUSION WITH REPERFUSION

100% 100%
Viability Restoration of flow
Function

Reperfusion injury
% of original

Salvage
Post-ischemic
ventricular
dysfunction

0% 0%
0 2 min 20 min 120 min 6 hr 0 2 min 20 min 120 min 6 hr Days
A Time B Time
Figure 12-16 Effects of reperfusion on myocardial viability and function. Following coronary occlusion, contractile function is lost within 2 minutes and viability
begins to diminish after approximately 20 minutes. If perfusion is not restored (A), then nearly all myocardium in the affected region suffers death. B, If flow is
restored, then some necrosis is prevented, myocardium is salvaged, and at least some function can return. The earlier reperfusion occurs, the greater the
degree of salvage. However, the process of reperfusion itself may induce some damage (reperfusion injury), and return of function of salvaged myocardium
may be delayed for hours to days (postischemic ventricular dysfunction or stunning).
Ischemic heart disease 547

endothelial swelling that occludes capillaries and may fraction of creatine kinase (CK-MB) (Fig. 12-17). The diag-
limit the reperfusion of critically injured myocardium nosis of myocardial injury is established when blood levels
(called no-reflow). Although the clinical significance of of these cardiac biomarkers are elevated. The rate of
myocardial reperfusion injury is debated, it has been esti- appearance of these markers in the peripheral circulation
mated that up to 50% (or more) of the ultimate infarct size depends on several factors, including their intracellular
can be attributed to its effects. location and molecular weight, the blood flow and lym-
Biochemical abnormalities (and their functional conse- phatic drainage in the area of the infarct, and the rate of
quences) may also persist for days to weeks in reperfused elimination of the marker from the blood.
myocytes. Such changes are thought to underlie a phenom- The most sensitive and specific biomarkers of myocar-
enon referred to as stunned myocardium, a state of pro- dial damage are cardiac-specific proteins, particularly
longed cardiac failure induced by short-term ischemia that cTnT and cTnI (proteins that regulate calcium-mediated
usually recovers after several days. Myocardium that is contraction of cardiac and skeletal muscle). Troponins I and
subjected to chronic, sublethal ischemia may also enter into T are not normally detectable in the circulation. Following
a state of lowered metabolism and function called hiberna- an MI, levels of both begin to rise at 3-12 hours; cTnT levels
tion. Subsequent revascularization (e.g., by CABG surgery, peak somewhere between 12-48 hours while cTnI levels are
angioplasty, or stenting) often restores normal function to maximal at 24 hours. Creatine kinase is an enzyme expressed
such hibernating myocardium. in brain, myocardium, and skeletal muscle; it is a dimer
composed of two isoforms designated “M” and “B.” While
Clinical Features. MI is diagnosed by clinical symptoms, MM homodimers are found predominantly in cardiac and
laboratory tests for the presence of myocardial proteins skeletal muscle, and BB homodimers in brain, lung, and
in the plasma, and characteristic electrocardiographic many other tissues, MB heterodimers are principally local-
changes. Patients with MI classically present with pro- ized to cardiac muscle (with considerably lesser amounts
longed (more than 30 minutes) chest pain described as found in skeletal muscle). Thus, the MB form of creatine
crushing, stabbing, or squeezing, associated with a rapid, kinase (CK-MB) is sensitive but not specific, since it can
weak pulse; profuse sweating (diaphoresis), and nausea also be elevated after skeletal muscle injury. CK-MB begins
and vomiting are common, and can suggest involvement to rise within 3 to 12 hours of the onset of MI, peaks at
of the posterior-inferior ventricle with secondary vagal about 24 hours, and returns to normal within approxi-
stimulation. Dyspnea due to impaired contractility of the mately 48 to 72 hours.
ischemic myocardium and the resultant pulmonary con- To summarize:
gestion and edema is a frequent symptom. However, in
as many as 25% of patients the onset is entirely asymptom-
• Time to elevation of CKMB, cTnT and cTnI is 3 to
12 hrs
atic (e.g., in the setting of diabetic neuropathy) and the
disease is discovered only by electrocardiographic changes • CK-MB and cTnI peak at 24 hours
or laboratory tests that show evidence of myocardial • CK-MB returns to normal in 48-72 hrs, cTnI in 5-10 days,
and cTnT in 5 to 14 days
damage (see later).
The laboratory evaluation of MI is based on measuring the Consequences and Complications of Myocardial Infarc-
blood levels of proteins that leak out of irreversibly damaged tion. Extraordinary progress has been made in the treat-
myocytes; the most useful of these molecules are cardiac- ment of patients with acute MI. Concurrent with the
specific troponins T and I (cTnT and cTnI), and the MB decrease in the overall mortality of IHD since the 1960s,

Myosin Actin Enzymes, such as CK-MB 40


(=multiples of upper reference limit)
Relative concentration

30
Troponin I
CK-MB
Troponin free
in cytoplasm 20 Myoglobin

10

Troponin
complex
released from 0
actin filament 4 20 40
Acute myocardial
Hours after onset of chest pains
infarction

1. Onset of myocardial 2. Plasma membrane of necrotic 3. Molecules leak 4. These molecules can be used
infarction myocytes becomes leaky out of cell into as biomarkers for diagnosis
circulation of myocardial infarction
Figure 12-17 Release of myocyte proteins in myocardial infarction. Some of these proteins, for example, troponin I or troponin T, and creatine kinase, MB
fraction (CK-MB) are routinely used as diagnostic biomarkers.
548 C H A P T E R 12 The Heart

the in-hospital death rate has declined from around 30% • Angiotensin-converting enzyme (ACE) inhibitors to
to approximately 5% in patients receiving timely therapy. limit ventricular dilation
Factors associated with a poorer prognosis include • Oxygen supplementation to improve blood oxygen
advanced age, female gender, diabetes mellitus, and—due saturation
to the cumulative loss of functional myocardium—previous
MI. Half of the deaths associated with acute MI occur Despite these interventions, many patients have one or
within 1 hour of onset, most commonly due to a fatal more complications following acute MI (Fig. 12-18):
arrhythmia; most of these individuals never reach the hos- • Contractile dysfunction. Myocardial infarcts produce
pital. MI therapeutic interventions include: abnormalities in left ventricular function roughly pro-
portional to their size. There is usually some degree of
• Morphine to relieve pain and improve dyspneic
left ventricular failure with hypotension, pulmonary
symptoms
vascular congestion, and interstitial pulmonary transu-
• Prompt reperfusion to salvage myocardium
dates, which can progress to pulmonary edema and
• Antiplatelet agents such as aspirin, P2Y12 receptor respiratory impairment. Severe “pump failure” (cardio-
inhibitors, and GPIIb/IIIa inhibitors genic shock) occurs in 10% to 15% of patients following
• Anticoagulant therapy with unfractionated heparin, acute MI, generally with large infarcts involving
low-molecular-weight heparin, direct thrombin inhibi- more than 40% of the left ventricle. Cardiogenic shock
tors, and/or factor Xa inhibitors to prevent coronary has a nearly 70% mortality rate; it accounts for two
artery clot propagation thirds of in-hospital deaths in those patients admitted
• Nitrates to induce vasodilation and reverse vasospasm for MI.
• Beta blockers to decrease myocardial oxygen demand Right ventricular infarcts can cause right-sided heart
and to reduce risk of arrhythmias failure associated with pooling of blood in the venous
• Antiarrhythmics to manage arrhythmias circulation and systemic hypotension.

A B C

D E F
Figure 12-18 Complications of myocardial infarction. A, Anterior myocardial rupture in an acute infarct (arrow). B, Rupture of the ventricular septum (arrow).
C, Complete rupture of a necrotic papillary muscle. D, Fibrinous pericarditis, showing a dark, roughened epicardial surface overlying an acute infarct. E, Early
expansion of anteroapical infarct with wall thinning (arrow) and mural thrombus. F, Large apical left ventricular aneurysm. The left ventricle is on the right in
this apical four-chamber view of the heart. (A-E, Reproduced with permission from Schoen FJ: Interventional and Surgical Cardiovascular Pathology: Clinical
Correlations and Basic Principles. Philadelphia, WB Saunders, 1989; F, Courtesy William D. Edwards, MD, Mayo Clinic, Rochester, Minn.)
Ischemic heart disease 549

• Arrhythmias. Many patients have myocardial irritability • Mural thrombus. With any infarct, the combination
and/or conduction disturbances following MI that lead of a local abnormality in contractility (causing stasis)
to potentially fatal arrhythmias. MI-associated arrhyth- and endocardial damage (creating a thrombogenic
mias include sinus bradycardia, atrial fibrillation, heart surface) can foster mural thrombosis and potentially
block, tachycardia, ventricular premature contractions, thromboembolism.
ventricular tachycardia, and ventricular fibrillation. • Papillary muscle dysfunction. Although papillary muscle
Because of the location of portions of the atrioventricu- rupture after an MI may certainly result in precipitous
lar conduction system (bundle of His) in the inferoseptal onset of mitral (or tricuspid) valve incompetence, most
myocardium, infarcts involving this site can also be post-infarct regurgitation results from ischemic dys-
associated with heart block (see also the discussion con- function of a papillary muscle (and underlying myocar-
cerning arrhythmias). dium), or later from ventricular dilation or from
• Myocardial rupture. The various forms of cardiac rupture papillary muscle fibrosis and shortening.
typically occur when there is transmural necrosis of a • Progressive late heart failure (chronic IHD) is discussed
ventricle. These include: later.
• Rupture of the ventricular free wall (most common),
with hemopericardium and cardiac tamponade The risk of postinfarct complications and the prognosis
(Fig. 12-18A) of the patient depend primarily on the infarct size, location,
and fraction of the wall thickness involved (subendocardial
• Rupture of the ventricular septum (less common), or transmural). Thus, large transmural infarcts yield a
leading to an acute VSD and left-to-right shunting
(Fig. 12-18B) higher probability of cardiogenic shock, arrhythmias, and
late CHF. Patients with anterior transmural infarcts are at
• Papillary muscle rupture (least common), resulting greatest risk for free-wall rupture, expansion, mural
in the acute onset of severe mitral regurgitation
thrombi, and aneurysm. In contrast, posterior transmural
(Fig. 12-18C)
infarcts are more likely to be complicated by conduction
Free-wall rupture occurs most frequently 2 to 4 days blocks, right ventricular involvement, or both; when acute
after MI, when coagulative necrosis, neutrophilic infil- VSDs occur in this area they are more difficult to manage.
tration, and lysis of the myocardial connective tissue Moreover, female gender, age older than 70 years, diabetes
have appreciably weakened the infarcted myocardium; mellitus and previous MI are poor prognostic factors in
the anterolateral wall at the mid-ventricular level is the patients with ST elevation myocardial infarcts. With sub-
most common site. Risk factors for free-wall rupture endocardial infarcts, only rarely do pericarditis, rupture,
include age older than 60, first MI, large, transmural and and aneurysms occur.
anterior MI, absence of left ventricular hypertrophy, In addition to the sequence of repair in the infarcted
and preexisting hypertension. Ventricular rupture tissues described earlier, the noninfarcted segments of the
occurs less frequently in patients with prior MI because ventricle undergo hypertrophy and dilation; collectively,
associated fibrotic scarring tends to inhibit myocardial these changes are termed ventricular remodeling. The com-
tearing. While acute free-wall ruptures are usually pensatory hypertrophy of noninfarcted myocardium is
rapidly fatal, a fortuitously located pericardial adhesion initially hemodynamically beneficial. However, this adap-
can abort a rupture and result in a false aneurysm (local- tive effect may be overwhelmed by ventricular dilation
ized hematoma communicating with the ventricular (with or without ventricular aneurysm) and increased
cavity). The wall of a false aneurysm consists only of oxygen demand, which can exacerbate ischemia and
epicardium and adherent parietal pericardium and thus depress cardiac function. There may also be changes in
many still ultimately rupture. ventricular shape and stiffening of the ventricle due to scar
• Ventricular aneurysm. In contrast to the false aneurysms formation and hypertrophy that further diminish cardiac
mentioned earlier, true aneurysms of the ventricular wall output. Some of these deleterious effects appear to be
are bounded by myocardium that has become scarred. reduced by ACE inhibitors, which lessen the ventricular
Aneurysms of the ventricular wall are a late complica- remodeling that can occurs after infarction.
tion of large transmural infarcts that experience early Long-term prognosis after MI depends on many factors,
expansion. The thin scar tissue wall of an aneurysm the most important of which are the residual left ventricu-
paradoxically bulges during systole (Fig. 12-18F). lar function and the extent of any vascular obstructions in
Complications of ventricular aneurysms include mural vessels that perfuse the remaining viable myocardium. The
thrombus, arrhythmias, and heart failure; rupture of the overall total mortality within the first year can be as high
tough fibrotic wall does not usually occur. as 30%; thereafter, each passing year is associated with an
• Pericarditis. A fibrinous or fibrinohemorrhagic peri- additional 3% to 4% mortality among survivors. Infarct
carditis usually develops about the second or third prevention (through control of risk factors) in individuals
day following a transmural infarct as a result of under- who have never experienced MI (primary prevention) and
lying myocardial inflammation (Dressler syndrome; prevention of reinfarction in MI survivors (secondary pre-
Fig. 12-18D). vention) are important strategies that have received much
• Infarct expansion. As a result of the weakening of necrotic attention and achieved considerable success.
muscle, there may be disproportionate stretching, thin- The relationship of the causes, pathophysiology, and
ning, and dilation of the infarct region (especially with consequences of MI are summarized in Figure 12-19,
anteroseptal infarcts), which is often associated with including the possible outcomes of chronic IHD and
mural thrombus (Fig. 12-18E). sudden death, discussed below.
550 C H A P T E R 12 The Heart

CORONARY ARTERY DISEASE KEY CONCEPTS


Ischemic Heart Disease
■ The vast majority of ischemic heart disease is due to coro-
Acute plaque Myocardial ischemia nary artery atherosclerosis; vasospasm, vasculitis, or
Myocardial embolism are less common causes.
change; coronary of increased severity
ischemia artery thrombosis and duration ■ Cardiac ischemia results from a mismatch in coronary
supply and myocardial demand, and presents as different,
albeit overlapping syndromes:
■ Angina pectoris is chest pain due to inadequate perfu-
MYOCARDIAL INFARCTION sion, and is typically due to atherosclerotic disease with
with muscle loss
and arrhythmias greater than 70% fixed stenosis (so-called critical
stenosis).
■ Unstable angina results from a small fissure or rupture

Hypertrophy, of atherosclerotic plaque triggering platelet aggregation,


Infarct Ventricular
healing remodeling dilation of vasoconstriction, and formation of a mural thrombus
viable muscle that need not necessarily be occlusive.
■ Acute myocardial infarction typically results from acute

Chronic ischemic heart disease (IHD) thromboses after plaque disruption; most occur in
plaques that did not previously exhibit critical stenosis.
■ Sudden cardiac death usually results from a fatal

Congestive heart failure arrhythmia, typically without significant acute myocar-


dial damage.
■ Chronic ischemic heart disease is progressive heart

failure due to ischemic injury, either from prior infarc-


SUDDEN CARDIAC DEATH tions or chronic low-grade ischemia.

Figure 12-19 Schematic for the causes and outcomes of ischemic heart
■ Myocardial ischemia leads to loss of function within 1 to 2
disease (IHD), showing the interrelationships among coronary artery disease, minutes, but causes necrosis only after 20 to 40 minutes.
acute plaque change, myocardial ischemia, myocardial infarction, chronic Myocardial infarction is diagnosed based on symptoms,
IHD, congestive heart failure, and sudden cardiac death. electrocardiographic changes, and measurement of serum
CK-MB and troponins. Gross and histologic changes of
infarction require hours to days to develop.
■ Infarction can be modified by therapeutic intervention (e.g.,
Chronic Ischemic Heart Disease thrombolysis or stenting), which salvages myocardium at
risk, but potentially induces reperfusion-related injury.
The designation chronic IHD (often called ischemic cardio-
■ Complications of infarction include: ventricular rupture,
myopathy by clinicians) is used here to describe progressive
papillary muscle rupture, aneurysm formation, mural
congestive heart failure as a consequence of accumulated
thrombus, arrhythmia, pericarditis, and CHF.
ischemic myocardial damage and/or inadequate compen-
satory responses. In most instances there has been prior MI
and sometimes previous coronary arterial interventions
and/or bypass surgery. Chronic IHD usually appears Arrhythmias
postinfarction due to the functional decompensation of
hypertrophied noninfarcted myocardium (see earlier dis- Abnormalities in myocardial conduction can be sustained
cussion of cardiac hypertrophy). However, in other cases or sporadic (paroxysmal). Aberrant rhythms can be initiated
severe obstructive coronary artery disease may present as anywhere in the conduction system, from SA node down
chronic congestive heart failure in the absence of prior to the level of an individual myocyte; they are typically
infarction. Patients with chronic IHD account for almost designated as originating from the atrium (supraventricu-
50% of cardiac transplant recipients. lar) or within the ventricular myocardium. Arrhythmias
can manifest as tachycardia (fast heart rate), bradycardia
(slow heart rate), an irregular rhythm with normal ven-
tricular contraction, chaotic depolarization without func-
MORPHOLOGY tional ventricular contraction (ventricular fibrillation), or no
electrical activity at all (asystole). Patients may be unaware
Hearts from patients with chronic IHD have cardiomegaly, with
of a rhythm disorder, or may note a “racing heart” or pal-
left ventricular hypertrophy and dilation. Invariably there is some
pitations (irregular rhythm); loss of adequate cardiac output
degree of stenotic coronary atherosclerosis. Discrete scars rep-
due to sustained arrhythmia can produce light-headedness
resenting healed infarcts are usually present. The mural endo-
(near syncope), loss of consciousness (syncope), or sudden
cardium often has patchy fibrous thickenings (due to abnormal
cardiac death (see later)
wall shear forces), and mural thrombi may be present.
Ischemic injury is the most common cause of rhythm disor-
Microscopic findings include myocardial hypertrophy, diffuse
ders, either through direct damage, or through the dilation
subendocardial vacuolization, and fibrosis.
of heart chambers that alters conduction system firing.
Arrhythmias 551

Table 12-6 Selected Examples of Causal Genes in Inherited


• If the SA node is damaged (e.g., sick sinus syndrome),
Arrhythmogenic Diseases*
other fibers or even the AV node can take over pace-
maker function, albeit at a much slower intrinsic rate Disorder Gene Function
(causing bradycardia). Long QT KCNQ1 K+ channel (LOF)
• If the atrial myocytes become “irritable” and depolarize syndrome† KCNH2 K+ channel (LOF)
independently and sporadically (as occurs with atrial SCN5A Na+ channel (GOF)
dilation), the signals are variably transmitted through CAV3 Caveolin, Na+ current (GOF)
the AV node leading to the random “irregularly irregu- Short QT KCNQ1 K+ channel (GOF)
lar” heart rate of atrial fibrillation. syndrome† KCNH2 K+ channel (GOF)
• If the AV node is dysfunctional, varying degrees of Brugada SCN5A Na+ channel (LOF)
heart block occur, ranging from simple prolongation syndrome† CACNB2b Ca++ channel (LOF)
of the P-R interval on the electrocardiogram (ECG; first SCN1b Na+ channel (LOF)*
degree heart block), to intermittent transmission of the CPVT RYR2 Diastolic Ca++ release (GOF)
signal (second degree heart block), to complete failure syndrome† CASQ2 Diastolic Ca++ release (LOF)
(third degree heart block). *Different mutations can cause the same general syndrome and mutations in some genes can
cause multiple different phenotypes; thus, loss of function (LOF) mutations may cause long QT
As already discussed, coordinated cardiac contraction intervals, whereas gain of function (GOF) mutations result in short repolarization intervals.
depends on the orderly transmission of electrical currents †
Long QT syndrome manifests as arrhythmias associated with excessive prolongation of the
cardiac repolarization; patients often present with stress-induced syncope or sudden cardiac
from myocyte to myocyte via gap junctions. Thus, abnor- death (SCD), and some forms are associated with swimming. Short QT syndrome patients
malities in the structure or spatial distribution of gap junc- have arrhythmias associated with abbreviated repolarization intervals; they can present with
tions, which are seen in a variety of disorders (e.g., IHD palpitations, syncope, and SCD. Brugada syndrome manifests as ECG abnormalities (ST
segment elevations and right bundle branch block) in the absence of structural heart disease;
and dilated cardiomyopathies), can cause arrhythmias. patients classically present with syncope or SCD during rest or sleep, or after large meals.
Ischemia, myocyte hypertrophy, and inflammation (e.g., CPVT does not have characteristic ECG changes; patients often present in childhood with
myocarditis or sarcoidosis) also promote increased “irrita- life-threatening arrhythmias due to adrenergic stimulation (stress-related).
LOF, Loss of function mutations; GOF, gain of function mutations; CPVT, catecholaminergic
bility” that leads to spontaneous aberrant myocyte depo- polymorphic ventricular tachycardia.
larization; because of the electrical interconnection of Modified from Cerrone M , Priori SG: Genetics of sudden death: focus on inherited channelopa-
myocytes, such random events can cause inappropriate thies. Eur Heart J, 2011:32, 2109-2118.
firing of adjacent cells and create abnormal electrical cir-
cuits (so-called reentry circuits) that lead to ventricular
tachycardia, which may progress to fatal ventricular fibril-
lation. Likewise, deposition of nonconducting material needed for the normal function of many tissues, and certain
(e.g., amyloid), and even small areas of fibrosis, can disrupt channelopathies are also associated with skeletal muscle
myocyte-to-myocyte signaling, again sowing the seeds for disorders and diabetes. Nevertheless, the most common
development of reentry circuits that can give rise to poten- channelopathies are isolated disorders of the heart, and
tially fatal arrythmias. their most feared consequence is sudden cardiac death
Heritable conditions associated with arrhythmias are (discussed below).
important to recognize, since they may alert physicians to
the need for intervention to prevent sudden cardiac death Sudden Cardiac Death (SCD)
(discussed later) in the proband and their family members.
Some of these disorders are associated with recognizable SCD is most commonly defined as unexpected death from
anatomic abnormalities (e.g., congenital anomalies, hyper- cardiac causes either without symptoms, or within 1 to 24
trophic cardiomyopathy, mitral valve prolapse). However, hours of symptom onset (different authors use different
other heritable disorders precipitate arrythmias and criteria); this happens in some 300,000 to 400,000 individu-
sudden death in the absence of structural cardiac pathol- als each year in the United States alone. Coronary artery
ogy (so-called primary electrical disorders). These syn- disease is the leading cause of SCD, responsible for 80% to
dromes can only be diagnosed by genetic testing, which is 90% of cases; unfortunately, SCD is often the first manifes-
performed in those with a positive family history or an tation of IHD. Interestingly, there is typically only chronic
unexplained nonlethal arrhythmia. severe atherosclerotic disease; acute plaque disruption is
The primary electrical abnormalities of the heart that found in only 10% to 20% of cases. Healed remote MIs are
predispose to arrhythmias are listed in Table 12-6. The present in about 40%.
most important of these are the so-called channelopathies, With younger victims, other nonatherosclerotic causes
which are caused by mutations in genes that are required are more common etiologies for SCD:
for normal ion channel function. These disorders (mostly
with autosomal dominant inheritance) either involve genes
• Hereditary or acquired abnormalities of the cardiac con-
duction system
that encode the structural components of ion channels
(including Na+, K+, and Ca+ channels), or accessory proteins • Congenital coronary arterial abnormalities
that are essential for normal channel function. Ion channels • Mitral valve prolapse
are responsible for conducting the electrical currents that • Myocarditis or sarcoidosis
mediate contraction of the heart, and it is thus not surpris- • Dilated or hypertrophic cardiomyopathy
ing that defects in these channels may provoke arrythmias. • Pulmonary hypertension
The prototype is the long QT syndrome, characterized by • Myocardial hypertrophy. Increased cardiac mass is an
prolongation of the QT segment in ECGs and susceptibility independent risk factor for SCD; thus, some young indi-
to malignant ventricular arrhythmias. Ion channels are viduals who die suddenly—including athletes—have
552 C H A P T E R 12 The Heart

hypertensive hypertrophy or unexplained increased


cardiac mass as the only finding. Hypertensive Heart Disease
• Other miscellaneous causes, such as pericardial tam-
ponade, pulmonary embolism, systemic metabolic and Hypertensive heart disease (HHD) is a consequence of the
hemodynamic alterations, catecholamines, and drugs of increased demands placed on the heart by hypertension,
abuse, particularly cocaine and methamphetamine. causing pressure overload and ventricular hypertrophy.
Although most commonly seen in the left heart as the
The mechanism of SCD is most often a lethal arrhythmia result of systemic hypertension, pulmonary hypertension
(e.g., asystole or ventricular fibrillation). Notably, infarc- can cause right-sided HHD, or cor pulmonale.
tion need not occur; 80% to 90% of patients who suffer SCD
but are successfully resuscitated do not show any enzy- Systemic (Left-Sided) Hypertensive Heart Disease
matic or ECG evidence of myocardial necrosis—even when
the original cause was ischemic heart disease. Although Hypertrophy of the heart is an adaptive response to the
ischemic injury (and other pathologies) can directly affect pressure overload of chronic hypertension. However, such
the major components of the conduction system, most compensatory changes may be ultimately maladaptive and
cases of fatal arrhythmia are triggered by electrical irritabil- can lead to myocardial dysfunction, cardiac dilation, CHF,
ity of myocardium distant from the major elements of the and in some cases sudden death. The minimal pathologic
conduction system. criteria for the diagnosis of systemic HHD are the following:
The prognosis of patients vulnerable to SCD is (1) left ventricular hypertrophy (usually concentric) in the
markedly improved by pharmaceutical intervention, and absence of other cardiovascular pathology and (2) a clinical
particularly by implantation of automatic cardioverter history or pathologic evidence of hypertension in other organs
defibrillators that can sense and electrically counteract epi- (e.g., kidney). The Framingham Study established unequiv-
sodes of ventricular fibrillation. ocally that even mild hypertension (levels only slightly
above 140/90 mm Hg)—if sufficiently prolonged—induces
left ventricular hypertrophy. Approximately 30% of the
population of the United States suffers from hypertension
MORPHOLOGY of at least this degree. The pathogenesis of hypertension is
discussed in Chapter 11.
Marked coronary atherosclerosis with a critical (>75% cross-
sectional area) stenosis involving one or more of the three major
vessels is present in 80% to 90% of SCD victims; only 10% to
20% of cases are of nonatherosclerotic origin. Usually there are MORPHOLOGY
high-grade stenoses (>90% of area); in approximately one half,
Hypertension induces left ventricular pressure overload hyper-
acute plaque disruption is observed, and in approximately 25%
trophy, initially without ventricular dilation. As a result, the left
diagnostic changes of acute MI are seen. This suggests that
ventricular wall thickening increases the weight of the heart
many patients who die suddenly are suffering an MI, but the
disproportionately to the increase in overall cardiac size (Fig.
short interval from onset to death precludes the development
12-20A). The thickness of the left ventricular wall may exceed
of diagnostic myocardial changes. However, in one study of
2.0 cm, and the heart weight may exceed 500 gm. In time the
those who had been successfully resuscitated from a sudden
increased thickness of the left ventricular wall, often associated
cardiac arrest, a new MI occurred in only 39% of the patients.
with increased interstitial connective tissue, imparts a stiffness
Thus, most SCD is not associated with acute MI; most of these
that impairs diastolic filling, frequently with consequent left atrial
deaths are thought to result from myocardial ischemia-induced
enlargement.
irritability that initiates malignant ventricular arrhythmias. Scars
Microscopically, the earliest change of systemic HHD is an
of previous infarcts and subendocardial myocyte vacuolization
increase in the transverse diameter of myocytes, which may be
indicative of severe chronic ischemia are common in such
difficult to appreciate on routine microscopy. At a more
patients.
advanced stage variable degrees of cellular and nuclear enlarge-
ment become apparent, often accompanied by interstitial
fibrosis.

KE Y CONCEPTS
Arrhythmias Compensated systemic HHD may be asymptomatic,
■ Arrhythmias can be caused by ischemic or structural producing only electrocardiographic or echocardiographic
changes in the conduction system or by intrinsic myocyte evidence of left ventricular enlargement. In many patients,
electrical instability. In structurally normal hearts, arrhyth- systemic HHD comes to attention due to new atrial fibril-
mias are more often due to mutations in ion channels that lation induced by left atrial enlargement, or by progressive
cause aberrant repolarization or depolarization. CHF. Depending on the severity, duration, and underlying
basis of the hypertension, and on the adequacy of thera-
■ SCD typically results from ventricular fibrillation, and is
peutic control, the patient may (1) enjoy normal longevity
most frequently a consequence of coronary artery disease.
and die of unrelated causes, (2) develop IHD due to both
Myocardial irritability typically results from nonlethal isch-
the potentiating effects of hypertension on coronary ath-
emia or from preexisting fibrosis from previous myocardial
erosclerosis and the ischemia induced by increased oxygen
injury. SCD is less often due to acute plaque rupture with
demand from the hypertrophic muscle, (3) suffer renal
thrombosis that induces a rapidly fatal arrhythmia.
damage or cerebrovascular stroke as direct effects of
Hypertensive heart disease 553

Figure 12-20 Hypertensive heart disease, systemic


and pulmonary. A, Systemic (left-sided) hypertensive
heart disease. There is marked concentric thicken-
ing of the left ventricular wall causing reduction in
lumen size. The left ventricle and left atrium (asterisk)
are on the right in this apical four-chamber view of
the heart. A pacemaker is present in the right ven-
tricle (arrow). B, Pulmonary (right-sided) hyperten-
sive heart disease (cor pulmonale). The right ventricle
is markedly dilated and has a thickened free wall and
hypertrophied trabeculae (apical four-chamber view
of heart, right ventricle on left). The shape of the left
ventricle (to the right) has been distorted by the A B
enlarged right ventricle.

hypertension, or (4) experience progressive heart failure or Table 12-7 Disorders Predisposing to Cor Pulmonale
SCD. Effective control of hypertension can prevent cardiac Diseases of the Pulmonary Parenchyma
hypertrophy, or can lead to its regression; with normaliza-
Chronic obstructive pulmonary disease
tion of the blood pressure, the associated risks of HHD are Diffuse pulmonary interstitial fibrosis
diminished. Pneumoconioses
Cystic fibrosis
Bronchiectasis
Pulmonary (Right-Sided) Hypertensive Heart Diseases of the Pulmonary Vessels
Disease (Cor Pulmonale) Recurrent pulmonary thromboembolism
Primary pulmonary hypertension
Normally, because the pulmonary vasculature is the low Extensive pulmonary arteritis (e.g., granulomatosis with polyangiitis)
pressure side of the circulation, the right ventricle has a Drug-, toxin-, or radiation-induced vascular obstruction
thinner and more compliant wall than the left ventricle. Extensive pulmonary tumor microembolism
Isolated pulmonary HHD, or cor pulmonale, stems from
right ventricular pressure overload. Chronic cor pulmo- Disorders Affecting Chest Movement
nale is characterized by right ventricular hypertrophy, Kyphoscoliosis
dilation, and potentially right-sided failure. Typical causes Marked obesity (sleep apnea, pickwickian syndrome)
Neuromuscular diseases
of chronic cor pulmonale are disorders of the lungs, espe-
cially chronic parenchymal diseases such as emphysema, Disorders Inducing Pulmonary Arterial Constriction
and primary pulmonary hypertension (Table 12-7; see also Metabolic acidosis
Chapter 15). Acute cor pulmonale can follow massive pul- Hypoxemia
monary embolism. Nevertheless, it should also be remem- Chronic altitude sickness
bered that pulmonary hypertension most commonly occurs Obstruction of major airways
Idiopathic alveolar hypoventilation
as a complication of left-sided heart disease.

KEY CONCEPTS
Hypertensive Heart Disease
MORPHOLOGY ■ Hypertensive heart disease can affect either the left ven-
In acute cor pulmonale there is marked dilation of the right tricle or the right ventricle; the latter is called cor pulmo-
ventricle without hypertrophy. On cross-section the normal nale. Elevated pressures induce myocyte hypertrophy and
crescent shape of the right ventricle is transformed to a dilated interstitial fibrosis that increases wall thickness and myo-
ovoid. In chronic cor pulmonale the right ventricular wall thick- cardial stiffness.
ens, sometimes up to 1.0 cm or more (Fig. 12-20B). More ■ The chronic pressure overload of systemic hypertension
subtle right ventricular hypertrophy may take the form of thick- causes left ventricular concentric hypertrophy, often asso-
ening of the muscle bundles in the outflow tract, immediately ciated with left atrial dilation due to impaired diastolic filling
below the pulmonary valve, or thickening of the moderator of the ventricle. Persistently elevated pressure overload
band, the muscle bundle that connects the ventricular septum can cause ventricular failure with dilation.
to the anterior right ventricular papillary muscle. Sometimes, the ■ Cor pulmonale results from pulmonary hypertension due
hypertrophied right ventricle compresses the left ventricular to primary lung parenchymal or vascular disorders. There
chamber, or leads to regurgitation and fibrous thickening of the is commonly right ventricular and right atrial hypertrophy;
tricuspid valve. right ventricular and atrial dilation can occur.
554 C H A P T E R 12 The Heart

Table 12-8 Major Etiologies of Acquired Heart Valve Disease


Valvular Heart Disease Mitral Valve Disease Aortic Valve Disease
Valvular disease can come to clinical attention due to Mitral Stenosis Aortic Stenosis
stenosis, insufficiency (synonyms: regurgitation or incom- Postinflammatory scarring (rheumatic heart Postinflammatory scarring
petence), or both. Stenosis is the failure of a valve to open disease) (rheumatic heart disease)
Senile calcific aortic stenosis
completely, which impedes forward flow. Insufficiency results Calcification of congenitally
from failure of a valve to close completely, thereby allowing deformed valve
reversed flow. These abnormalities can be present alone
or coexist, and may involve only a single valve, or more Mitral Regurgitation Aortic Regurgitation
than one valve. Functional regurgitation is used to describe Abnormalities of Leaflets and Commissures
the incompetence of a valve stemming from an abnor- Postinflammatory scarring Postinflammatory scarring
Infective endocarditis (rheumatic heart disease)
mality in one of its support structures, as opposed to a Mitral valve prolapse
primary valve defect. For example, dilation of the right or Drugs (e.g., fen-phen)
left ventricle can pull the ventricular papillary muscles
Abnormalities of Tensor Apparatus
down and outward, thereby preventing proper closure Rupture of papillary muscle Degenerative aortic dilatation
of otherwise normal mitral or tricuspid leaflets. Functional Papillary muscle dysfunction (fibrosis) Syphilitic aortitis
mitral valve regurgitation is particularly common Rupture of chordae tendineae Ankylosing spondylitis
and clinically important in IHD, as well as in dilated Rheumatoid arthritis
cardiomyopathy. Marfan syndrome
The clinical consequences of valve dysfunction vary Abnormalities of Left Ventricle and/or
depending on the valve involved, the degree of impair- Annulus
ment, the tempo of disease onset, and the rate and quality LV enlargement (myocarditis, dilated
of compensatory mechanisms. For example, sudden cardiomyopathy)
destruction of an aortic valve cusp by infection (infective Calcification of mitral ring
endocarditis; see later) can cause acute, massive, and LV, Left ventricular.
Modified from Schoen FJ: Surgical pathology of removed natural and prosthetic valves. Hum
rapidly fatal regurgitation. In contrast, rheumatic mitral Pathol 18:558, 1987.
stenosis typically develops indolently over years, and its
clinical effects can be well tolerated for extended periods.
Certain conditions can complicate valvular heart disease
by increasing the demands on the heart; for example, the • Mitral insufficiency: myxomatous degeneration (mitral
increased output demands of pregnancy can exacerbate valve prolapse)
valve disease and lead to unfavorable maternal or fetal
outcomes. Valvular stenosis or insufficiency often pro- Calcific Valvular Degeneration
duces secondary changes, both proximal and distal to the
affected valve, particularly in the myocardium. Generally, Heart valves are subjected to high levels of repetitive
valvular stenosis leads to pressure overload cardiac hyper- mechanical stress, particularly at the hinge points of the
trophy, whereas mitral or aortic valvular insufficiency cusps and leaflets; this is a consequence of (1) 30 to 40
leads to volume overload; both situations can culminate in million or more cardiac contractions per year, (2) substan-
heart failure. In addition, the ejection of blood through tial tissue deformations during each contraction, and (3)
narrowed stenotic valves can produce high speed “jets” of transvalvular pressure gradients in the closed phase of
blood that injure the endocardium where they impact. each contraction of approximately 120 mm Hg for the
Valvular abnormalities can be congenital (discussed mitral and 80 mm Hg for the aortic valve. It is therefore
earlier) or acquired. Acquired valvular stenosis has relatively not surprising that these delicate structures can suffer
few causes; it is almost always a consequence of a remote cumulative damage and calcification that lead to clinically
or chronic injury of the valve cusps that declares itself clini- important dysfunction.
cally only after many years. In contrast, acquired valvular
insufficiency can result from intrinsic disease of the valve Calcific Aortic Stenosis
cusps or damage to or distortion of the supporting struc- The most common of all valvular abnormalities, calcific
tures (e.g., the aorta, mitral annulus, tendinous cords, aortic stenosis is usually the consequence of age-associated
papillary muscles, ventricular free wall). Thus, valvular “wear and tear” of either anatomically normal valves or
insufficiency has many causes and may appear acutely, as congenitally bicuspid valves (in approximately 1% of the
with rupture of the cords, or chronically in disorders asso- population). The prevalence of aortic stenosis is estimated
ciated with leaflet scarring and retraction. at 2% and is increasing as the general population ages.
The causes of acquired heart valve diseases are sum- Aortic stenosis of previously normal valves (termed senile
marized in Table 12-8. The most frequent causes of the calcific aortic stenosis) usually comes to clinical attention
major functional valvular lesions are: in the seventh to ninth decades of life, whereas stenotic
bicuspid valves tend to become clinically significant 1 to 2
• Aortic stenosis: calcification and sclerosis of anatomically decades earlier.
normal or congenitally bicuspid aortic valves Aortic valve calcification is likely a consequence of
• Aortic insufficiency: dilation of the ascending aorta, often recurrent chronic injury due to hyperlipidemia, hyperten-
secondary to hypertension and/or aging sion, inflammation, and other factors similar to those
• Mitral stenosis: rheumatic heart disease implicated in atherosclerosis. Bicuspid valves incur greater
Valvular heart disease 555

mechanical stress than normal tricuspid valves, which may narrowing of the valve orifice (valve area approximately
explain their accelerated stenosis. The chronic progressive 0.5 to 1 cm2 in severe aortic stenosis; normal approximately
injury leads to valvular degeneration and incites the depo- 4 cm2) and an increasing pressure gradient across the calci-
sition of hydroxyapatite (the same calcium salt found in fied valve, reaching 75 to 100 mm Hg in severe cases. Left
bone). Although this model provides a good starting point ventricular pressures rise to 200 mm Hg or more in such
for understanding calcific degeneration, it is increasingly instances, producing concentric left ventricular (pressure
clear that the valve injury of calcific aortic stenosis differs overload) hypertrophy. The hypertrophied myocardium
in some important respects from atherosclerosis. Most tends to be ischemic (as a result of diminished microcircu-
notably, the abnormal valves contain cells resembling latory perfusion, often complicated by coronary atheroscle-
osteoblasts that synthesize bone matrix proteins and rosis), and angina pectoris may occur. Both systolic and
promote the deposition of calcium salts. Moreover, inter- diastolic myocardial function may be impaired; eventually,
ventions that improve atherosclerotic risk (e.g., statins), cardiac decompensation and CHF can ensue. The onset of
do not appear to significantly impact valvular calcific symptoms (angina, CHF, or syncope) in aortic stenosis
degeneration. heralds cardiac decompensation and carries an extremely
poor prognosis. If untreated, most patients with aortic ste-
nosis will die within 5 years of developing angina, within
MORPHOLOGY 3 years of developing syncope, and within 2 years of CHF
onset. Treatment requires surgical valve replacement, as
The gross morphologic hallmark of nonrheumatic, calcific aortic medical therapy is ineffective in severe symptomatic aortic
stenosis (involving either tricuspid or bicuspid valves) is mounded stenosis.
calcified masses within the aortic cusps that ultimately protrude
through the outflow surfaces into the sinuses of Valsalva, and Calcific Stenosis of Congenitally Bicuspid Aortic Valve
prevent cuspal opening. The free edges of the cusps are usually Bicuspid aortic valve (BAV) is a developmental abnormal-
not involved (Fig. 12-21A). Microscopically, the layered archi- ity with prevalence in the population of approximately 1%.
tecture of the valve is largely preserved. The calcific process Some cases of BAV show familial clustering, often with
begins in the valvular fibrosa on the outflow surface of the associated aorta or left ventricular outflow tract malforma-
valve, at the points of maximal cusp flexion (near the margins tions. While the heritability of BAV is well-established, and
of attachment). Inflammation is variable, and metaplastic bone three loci on chromosomes 18q, 5q, and 13q have been
(and even bone marrow) may be seen. In aortic stenosis, the identified in kindred studies, the specific genes that are
functional valve area is decreased by large nodular calcific responsible for the disorder remain largely unknown. Thus
deposits that can eventually cause measurable outflow obstruc- far, only loss-of-function mutations in NOTCH1 (mapping
tion; this subjects the left ventricular myocardium to progres- to chromosome 9q34.3) have been specifically associated
sively increasing pressure overload. with BAV in a few families; tantalizingly, modulation of
In contrast to rheumatic (and congenital) aortic stenosis (see Notch activity in animal models also impacts valvular
Fig. 12-23E), commissural fusion is not usually seen. The mitral calcification.
valve is generally normal, although some patients may have In a congenitally bicuspid aortic valve, there are only
direct extension of aortic valve calcific deposits onto the anterior two functional cusps, usually of unequal size, with the
mitral leaflet. In contrast, virtually all patients with rheumatic larger cusp having a midline raphe, resulting from incom-
aortic stenosis also have concomitant and characteristic struc- plete commissural separation during development; less
tural abnormalities of the mitral valve (see later discussion). frequently the cusps are of equal size and the raphe is
absent. The raphe is frequently a major site of calcific
deposits. Once stenosis is present, the clinical course is
Clinical Features. In calcific aortic stenosis (superimposed similar to that described earlier for calcific aortic stenosis.
on a previously normal or bicuspid aortic valve), the Valves that become bicuspid because of an acquired
obstruction to left ventricular outflow leads to gradual deformity (e.g., rheumatic valve disease) have a

A B C D
Figure 12-21 Calcific valvular degeneration. A, Calcific aortic stenosis of a previously normal valve (viewed from aortic aspect). Nodular masses of calcium
are heaped up within the sinuses of Valsalva (arrow). Note that the commissures are not fused, as occurs with postrheumatic aortic valve stenosis (see
Fig. 12-23E). B, Calcific aortic stenosis of a congenitally bicuspid valve. One cusp has a partial fusion at its center, called a raphe (arrow). C and D, Mitral
annular calcification, with calcific nodules at the base (attachment margin) of the anterior mitral leaflet (arrows). C, Left atrial view. D, Cut section of myocardium
showing the lateral wall with dense calcification that extends into the underlying myocardium (arrow).
556 C H A P T E R 12 The Heart

fused commissure that produces a conjoined cusp that is interactions and dysregulate TGF-β signaling. Interestingly,
generally twice the size of the nonconjoined cusp. BAVs mice with mutated FBN-1 develop a form of mitral
may also become incompetent as a result of aortic dilation, valve prolapse that is prevented by TGF-β inhibitors,
cusp prolapse, or infective endocarditis. The mitral valve indicating that excess TGF-β activity can cause the
is generally normal in patients with a congenitally bicuspid characteristic structural laxity and myxomatous changes.
aortic valve. Whether similar mechanisms contribute to sporadic MVP
Although BAV is usually asymptomatic early in life, late is unknown. Genetic linkage analyses have also mapped
complications include aortic stenosis or regurgitation, inherited forms of MVP to loci involved in the remodeling
infective endocarditis, and aortic dilation and/or dissec- of valvular extracellular matrix and cell : cell adhesion.
tion. In particular, BAVs are predisposed to progressive
calcification, similar to that occurring in aortic valves with
initially normal anatomy (Fig. 12-21B); calcified BAV com-
prise approximately 50% of cases of aortic stenosis in MORPHOLOGY
adults. Structural abnormalities of the aortic wall also com- The characteristic anatomic change in MVP is interchordal bal-
monly accompany BAV, even when the valve is hemody- looning (hooding) of the mitral leaflets or portions thereof (Fig.
namically normal, and this may potentiate aortic dilation 12-22A-C). The affected leaflets are often enlarged, redundant,
or aortic dissection (see later). thick, and rubbery. The associated tendinous cords may be
elongated, thinned, or even ruptured, and the annulus may be
Mitral Annular Calcification dilated. The tricuspid, aortic, or pulmonary valves may also
As opposed to the predominantly cuspal involvement in be affected. The key histologic change in the tissue is marked
aortic valve calcification, degenerative calcific deposits in thickening of the spongiosa layer with deposition of mucoid
the mitral valve typically develop in the fibrous annulus. (myxomatous) material, called myxomatous degeneration;
Grossly, these appear as irregular, stony hard, occasionally there is also attenuation of the collagenous fibrosa layer of the
ulcerated nodules (2 to 5 mm in thickness) at the base of valve, on which the structural integrity of the leaflet depends
the leaflets (Fig. 12-21C, D). Mitral annular calcification (Fig. 12-22E). Secondary changes reflect the stresses and
usually does not affect valvular function. However, in tissue injury incident to the billowing leaflets: (1) fibrous thicken-
exceptional cases it can lead to: ing of the valve leaflets, particularly where they rub against each
other; (2) linear fibrous thickening of the left ventricular endo-
• Regurgitation by interfering with physiologic contrac- cardial surface where the abnormally long cords snap or rub
tion of the valve ring against it; (3) thickening of the mural endocardium of the left
• Stenosis by impairing opening of the mitral leaflets ventricle or atrium as a consequence of friction-induced injury
• Arrhythmias and occasionally sudden death by penetra- induced by the prolapsing, hypermobile leaflets; (4) thrombi on
tion of calcium deposits to a depth sufficient to impinge the atrial surfaces of the leaflets or the atrial walls (Fig. 12-22B);
on the atrioventricular conduction system. and (5) focal calcifications at the base of the posterior mitral
leaflet (Fig. 12-22C). Notably, mitral valve myxomatous degen-
Because calcific nodules may also provide a site for eration can also occur as a secondary consequence of regur-
thrombus formation, patients with mitral annular calcifica- gitation of other etiologies (e.g., ischemic dysfunction).
tion have an increased risk of embolic stroke, and the cal-
cific nodules can become a nidus for infective endocarditis.
Heavy calcific deposits are sometimes visualized on echo-
cardiography or seen as distinctive, ringlike opacities on Clinical Features. Most individuals diagnosed with MVP
chest radiographs. Mitral annular calcification is most are asymptomatic; in such cases, the condition is discov-
common in women older than age 60 and individuals with ered incidentally by auscultation of mid-systolic clicks,
mitral valve prolapse (see later). sometimes followed by a mid to late systolic murmur. The
diagnosis is confirmed by echocardiography. A minority
of patients have chest pain mimicking angina (although
Mitral Valve Prolapse (Myxomatous not exertional in nature), and a subset has dyspnea, pre-
Degeneration of the Mitral Valve) sumably related to valvular insufficiency. Although the
great majority of persons with MVP have no untoward
In mitral valve prolapse (MVP), one or both mitral valve effects, approximately 3% develop one of four serious com-
leaflets are “floppy” and prolapse, or balloon back, into plications: (1) infective endocarditis; (2) mitral insuffi-
the left atrium during systole. MVP affects approximately ciency, sometimes with chordal rupture; (3) stroke or other
2-3% of adults in the United States with an approximate systemic infarct, resulting from embolism of leaflet thrombi;
7 : 1 female-to-male ratio; it is most often an incidental or (4) arrhythmias, both ventricular and atrial. Rarely,
finding on physical examination, but in a small minority of MVP is the only finding in sudden cardiac death.
affected individuals may lead to serious complications. The risk of serious complications is very low in MVP
discovered incidentally in young asymptomatic patients;
Pathogenesis. The etiologic basis for the changes that the risk is higher for men, older patients, and those with
weaken the valve leaflets and associated structures is arrhythmias or mitral regurgitation. Valve repair or
unknown in most cases. Uncommonly, MVP is associated replacement surgery can be done for symptomatic patients
with heritable disorders of connective tissue including or those with increased risk for significant complications;
Marfan syndrome, caused by fibrillin-1 (FBN-1) muta- indeed, in the United States, MVP is the most common
tions (Chapter 5). Fibrillin-1 defects alter cell-matrix cause for mitral valve surgery.
Valvular heart disease 557

D E
Figure 12-22 Myxomatous degeneration of the mitral valve. A, Long axis view (left ventricle is on the right) demonstrating hooding with prolapse of the posterior
mitral leaflet into the left atrium (arrow). B, Opened valve, showing pronounced hooding of the posterior mitral leaflet with thrombotic plaques at sites of leaflet-
left atrium contact (arrows). C, Opened valve with pronounced hooding (double arrows) in a patient who died suddenly. Note also mitral annular calcification
(arrowhead). Normal heart valve (D) and myxomatous mitral valve (E). In myxomatous valves, collagen in the fibrosa is loose and disorganized, proteoglycan
deposition (asterisk) in the spongiosa is markedly expanded, and elastin in the atrialis is disorganized. (A, Courtesy William D. Edwards, MD, Mayo Clinic,
Rochester, Minn; D, E, Movat pentachrome stain, in which collagen is yellow, elastin is black, and proteoglycans are blue). From Rabkin E, et al: Activated
interstitial myofibroblasts express catabolic enzymes and mediate matrix remodeling in myxomatous heart valves. Circulation 104:2525-2532, 2001.)

RHD is characterized principally by deforming fibrotic


Rheumatic Fever and Rheumatic Heart Disease valvular disease, particularly involving the mitral valve;
indeed, RHD is virtually the only cause of mitral stenosis. The
Rheumatic fever (RF) is an acute, immunologically medi- incidence and mortality rate of RF and RHD have declined
ated, multisystem inflammatory disease classically occur- remarkably in many parts of the world over the past
ring a few weeks after an episode of group A streptococcal century, as a result of improved sanitation, and rapid
pharyngitis; occasionally, RF can follow streptococcal diagnosis and treatment of streptococcal pharyngitis.
infections at other sites, such as the skin. Acute rheumatic Nevertheless, in developing countries, and in many
carditis is a common manifestation of active RF and may crowded, economically depressed urban areas, RHD
progress over time to chronic rheumatic heart disease remains an important public health problem, affecting an
(RHD), mainly manifesting as valvular abnormalities. estimated 15 million people.
558 C H A P T E R 12 The Heart

Pathogenesis. Acute rheumatic fever results from host


round-to-ovoid nuclei (occasionally binucleate) in which the
immune responses to group A streptococcal antigens that
chromatin condenses into a central, slender, wavy ribbon
cross-react with host proteins. In particular, antibodies
(hence the designation “caterpillar cells”).
and CD4+ T cells directed against streptococcal M proteins
During acute RF, diffuse inflammation and Aschoff bodies
can also in some cases recognize cardiac self-antigens.
may be found in any of the three layers of the heart, resulting
Antibody binding can activate complement, as well as
in pericarditis, myocarditis, or endocarditis (pancarditis).
recruit Fc-receptor bearing cells (neutrophils and macro-
Inflammation of the endocardium and the left-sided valves
phages); cytokine production by the stimulated T cells
typically results in fibrinoid necrosis within the cusps or tendi-
leads to macrophage activation (e.g., within Aschoff
nous cords. Overlying these necrotic foci and along the lines of
bodies). Damage to heart tissue may thus be caused by a
closure are small (1 to 2 mm) vegetations, called verrucae.
combination of antibody- and T cell–mediated reactions
Thus, RHD is one of the forms of vegetative valve disease, each
(Chapter 6).
of which exhibit their own characteristic morphologic features
(Fig. 12-24). Subendocardial lesions, perhaps exacerbated by
MORPHOLOGY regurgitant jets, can induce irregular thickenings called
MacCallum plaques, usually in the left atrium.
Key pathologic features of acute RF and chronic RHD are The cardinal anatomic changes of the mitral valve in chronic
shown in Figure 12-23. During acute RF, focal inflammatory RHD are leaflet thickening, commissural fusion and short-
lesions are found in various tissues. Distinctive lesions occur in ening, and thickening and fusion of the tendinous cords
the heart, called Aschoff bodies, consisting of foci of T lym- (Fig. 12-23D). In chronic disease the mitral valve is virtually
phocytes, occasional plasma cells, and plump activated mac- always involved. The mitral valve is affected in isolation in
rophages called Anitschkow cells (pathognomonic for RF). roughly two thirds of RHD, and along with the aortic valve in
These macrophages have abundant cytoplasm and central another 25% of cases. Tricuspid valve involvement is

C D E
Figure 12-23 Acute and chronic rheumatic heart disease. A, Acute rheumatic mitral valvulitis superimposed on chronic rheumatic heart disease. Small vegeta-
tions (verrucae) are visible along the line of closure of the mitral valve leaflet (arrows). Previous episodes of rheumatic valvulitis have caused fibrous thickening
and fusion of the chordae tendineae. B, Microscopic appearance of an Aschoff body in a patient with acute rheumatic carditis. The myocardium exhibits a
circumscribed nodule of mixed mononuclear inflammatory cells with associated necrosis; within the inflammation, large activated macrophages show prominent
nucleoli, as well as chromatin condensed into long, wavy ribbons (caterpillar cells; arrows). C and D, Mitral stenosis with diffuse fibrous thickening and distor-
tion of the valve leaflets and commissural fusion (arrows, C), and thickening of the chordae tendineae (D). Note neovascularization of anterior mitral leaflet
(arrow, D). E, Surgically resected specimen of rheumatic aortic stenosis, demonstrating thickening and distortion of the cusps with commissural fusion.
(E, Reproduced from Schoen FJ, St. John-Sutton M: Contemporary issues in the pathology of valvular heart disease. Hum Pathol 18:568, 1967.)
Valvular heart disease 559

heart failure, individuals with chronic RHD may suffer


infrequent, and the pulmonary valve is only rarely affected.
from arrhythmias (particularly atrial fibrillation in the
Because of the increase in calcific aortic stenosis (see earlier)
setting of mitral stenosis), thromboembolic complications,
and the reduced frequency of RHD, rheumatic aortic stenosis
and infective endocarditis (see later). The long-term prog-
now accounts for a small fraction of cases of acquired aortic
nosis is highly variable. Surgical repair or prosthetic
stenosis.
replacement of diseased valves has greatly improved the
In rheumatic mitral stenosis, calcification and fibrous bridging
outlook for persons with RHD.
across the valvular commissures create “fish mouth” or “but-
tonhole” stenoses. With tight mitral stenosis, the left atrium
progressively dilates and may harbor mural thrombi that can Infective Endocarditis
embolize. Long-standing congestive changes in the lungs may
Infective endocarditis (IE) is a microbial infection of the
induce pulmonary vascular and parenchymal changes; over
heart valves or the mural endocardium that leads to the
time, these can lead to right ventricular hypertrophy. The left
formation of vegetations composed of thrombotic debris
ventricle is largely unaffected by isolated pure mitral stenosis.
and organisms, often associated with destruction of the
Microscopically, valves show organization of the acute inflam-
underlying cardiac tissues. The aorta, aneurysms, other
mation, with post-inflammatory neovascularization and trans-
blood vessels, and prosthetic devices can also become
mural fibrosis that obliterate the leaflet architecture. Aschoff
infected. Although fungi and other classes of microorgan-
bodies are rarely seen in surgical specimens or autopsy tissue
isms can be responsible, most infections are bacterial (bacte-
from patients with chronic RHD, as a result of the long intervals
rial endocarditis). Prompt diagnosis, identification of the
between the initial insult and the development of the chronic
offending agent, and effective treatment of IE is important
deformity.
in limiting morbidity and mortality.
Traditionally, IE has been classified on clinical grounds
Clinical Features. RF is characterized by a constellation into acute and subacute forms. This subdivision reflects the
of findings: (1) migratory polyarthritis of the large joints, range of the disease severity and tempo, which are deter-
(2) pancarditis, (3) subcutaneous nodules, (4) erythema mined in large part by the virulence of the infecting micro-
marginatum of the skin, and (5) Sydenham chorea, a neu- organism and whether underlying cardiac disease is
rologic disorder with involuntary rapid, purposeless present. Acute infective endocarditis is typically caused by
movements. The diagnosis is established by the so-called infection of a previously normal heart valve by a highly
Jones criteria: evidence of a preceding group A strepto- virulent organism (e.g., Staphylococcus aureus) that rapidly
coccal infection, with the presence of two of the major produces necrotizing and destructive lesions. These infec-
manifestations listed earlier or one major and two minor tions may be difficult to cure with antibiotics alone, and
manifestations (nonspecific signs and symptoms that usually require surgery. Despite appropriate treatment,
include fever, arthralgia, or elevated blood levels of acute- death can ensue within days to weeks. In contrast, subacute
phase reactants). IE is characterized by organisms with lower virulence
Acute RF typically appears 10 days to 6 weeks after a (e.g., viridans streptococci) that cause insidious infections
group A streptococcal infection in about 3% of patients. It of deformed valves with overall less destruction. In such
occurs most often in children between ages 5 and 15, but cases the disease may pursue a protracted course of weeks
first attacks can occur in middle to later life. Although to months, and cures can be achieved with antibiotics.
pharyngeal cultures for streptococci are negative by the
time the illness begins, antibodies to one or more strepto- Pathogenesis. Although highly virulent organisms can
coccal enzymes, such as streptolysin O and DNase B, can infect previously normal valves, a variety of cardiac and
be detected in the sera of most patients with RF. The pre- vascular abnormalities increase the risk of developing IE.
dominant clinical manifestations are carditis and arthritis, Rheumatic heart disease with valvular scarring has histori-
the latter more common in adults than in children. Arthritis cally been the major antecedent disorder; as RHD becomes
typically begins with migratory polyarthritis (accompa- less common, it has been supplanted by mitral valve pro-
nied by fever) in which one large joint after another lapse, degenerative calcific valvular stenosis, bicuspid
becomes painful and swollen for a period of days and then aortic valve (whether calcified or not), artificial (prosthetic)
subsides spontaneously, leaving no residual disability. valves, and unrepaired and repaired congenital defects.
Clinical features related to acute carditis include pericardial The causal organisms differ among the major high-risk
friction rubs, tachycardia, and arrhythmias. Myocarditis groups. Endocarditis of native but previously damaged or
can cause cardiac dilation that may culminate in func- otherwise abnormal valves is caused most commonly (50%
tional mitral valve insufficiency or even heart failure. to 60% of cases) by Streptococcus viridans, a normal compo-
Approximately 1% of affected individuals die of fulminant nent of the oral cavity flora. In contrast, more virulent S.
RF involvement of the heart. aureus organisms commonly found on the skin can infect
After an initial attack there is increased vulnerability to reac- either healthy or deformed valves and are responsible for
tivation of the disease with subsequent pharyngeal infections, 20% to 30% of cases overall; notably, S. aureus is the major
and the same manifestations are likely to appear with each recur- offender in IE among intravenous drug abusers. Other
rent attack. Damage to the valves is cumulative. Turbulence bacterial causes include enterococci and the so-called
induced by ongoing valvular deformities leads to addi- HACEK group (Haemophilus, Actinobacillus, Cardiobacterium,
tional fibrosis. Clinical manifestations appear years or even Eikenella, and Kingella), all commensals in the oral cavity.
decades after the initial episode of RF and depend on Prosthetic valve endocarditis is caused most commonly
which cardiac valves are involved. In addition to various by coagulase-negative staphylococci (e.g., S. epidermidis).
cardiac murmurs, cardiac hypertrophy and dilation, and Other agents causing endocarditis include gram-negative
560 C H A P T E R 12 The Heart

RHD IE NBTE LSE


Figure 12-24 Comparison of the four major forms of vegetative endocarditis. The rheumatic fever phase of rheumatic heart disease (RHD) is marked by small,
warty vegetations along the lines of closure of the valve leaflets. Infective endocarditis (IE) is characterized by large, irregular masses on the valve cusps that
can extend onto the chordae (see Fig. 12-25A). Nonbacterial thrombotic endocarditis (NBTE) typically exhibits small, bland vegetations, usually attached at
the line of closure. One or many may be present (see Figure 12-26). Libman-Sacks endocarditis (LSE) has small or medium-sized vegetations on either or
both sides of the valve leaflets.

bacilli and fungi. In about 10% of all cases of endocarditis,


common sites of infection, although the valves of the right heart
no organism can be isolated from the blood (“culture-
may also be involved, particularly in intravenous drug abusers.
negative” endocarditis); reasons include prior antibiotic
Vegetations can be single or multiple and may involve more
therapy, difficulties in isolating the offending agent, or
than one valve; they can occasionally erode into the underlying
because deeply embedded organisms within the enlarging
myocardium and produce an abscess (ring abscess; Fig.
vegetation are not released into the blood.
12-25B). Vegetations are prone to embolization; because the
Foremost among the factors predisposing to endocardi-
embolic fragments often contain virulent organisms, abscesses
tis are those that cause microorganism seeding into the
frequently develop where they lodge, leading to sequelae such
blood stream (bacteremia or fungemia). The source may be
as septic infarcts or mycotic aneurysms.
an obvious infection elsewhere, a dental or surgical proce-
The vegetations of subacute endocarditis are associated
dure, a contaminated needle shared by intravenous drug
with less valvular destruction than those of acute endocarditis,
users, or seemingly trivial breaks in the epithelial barriers
although the distinction can be subtle. Microscopically, the
of the gut, oral cavity, or skin. In patients with valve abnor-
vegetations of subacute IE typically exhibit granulation tissue at
malities, or with known bacteremia, IE risk can be lowered
their bases indicative of healing. With time, fibrosis, calcification,
by antibiotic prophylaxis.
and a chronic inflammatory infiltrate can develop.

MORPHOLOGY
Clinical Features. Acute endocarditis has a stormy onset
Vegetations on heart valves are the classic hallmark of IE;
with rapidly developing fever, chills, weakness, and las-
these are friable, bulky, potentially destructive lesions containing
situde. Although fever is the most consistent sign of IE, it
fibrin, inflammatory cells, and bacteria or other organisms (Figs.
can be slight or absent, particularly in older adults, and the
12-24 and 12-25). The aortic and mitral valves are the most
only manifestations may be nonspecific fatigue, loss of

A B
Figure 12-25 Infective (bacterial) endocarditis. A, Endocarditis of mitral valve (subacute, caused by Streptococcus viridans). The large, friable vegetations are
denoted by arrows. B, Acute endocarditis of congenitally bicuspid aortic valve (caused by Staphylococcus aureus) with extensive cuspal destruction and ring
abscess (arrow).
Valvular heart disease 561

Table 12-9 Diagnostic Criteria for Infective Endocarditis* culture results, echocardiographic findings, and laboratory
Pathologic Criteria information.
Complications of IE generally begin within the first few
Microorganisms, demonstrated by culture or histologic examination, in a
vegetation, embolus from a vegetation, or intracardiac abscess
weeks of onset, and can include glomerular antigen-
Histologic confirmation of active endocarditis in vegetation or intracardiac antibody complex deposition causing glomerulonephritis
abscess (Chapter 20). Earlier diagnosis and effective treatment has
nearly eliminated some previously common clinical mani-
Clinical Criteria festations of long-standing IE—for example, microthrom-
Major boemboli (manifest as splinter or subungual hemorrhages),
Blood culture(s) positive for a characteristic organism or persistently positive erythematous or hemorrhagic nontender lesions on the
for an unusual organism palms or soles (Janeway lesions), subcutaneous nodules in
Echocardiographic identification of a valve-related or implant-related mass or the pulp of the digits (Osler nodes), and retinal hemor-
abscess, or partial separation of artificial valve rhages in the eyes (Roth spots).
New valvular regurgitation
Minor Noninfected Vegetations
Predisposing heart lesion or intravenous drug use
Fever Noninfected (sterile) vegetations occur in nonbacterial
Vascular lesions, including arterial petechiae, subungual/splinter thrombotic endocarditis and the endocarditis of systemic
hemorrhages, emboli, septic infarcts, mycotic aneurysm, intracranial lupus erythematosus (SLE), called Libman-Sacks endocar-
hemorrhage, Janeway lesions†
ditis (see later).
Immunological phenomena, including glomerulonephritis, Osler nodes,‡ Roth
spots,§ rheumatoid factor
Microbiologic evidence, including a single culture positive for an unusual
Nonbacterial Thrombotic Endocarditis
organism Nonbacterial thrombotic endocarditis (NBTE) is charac-
Echocardiographic findings consistent with but not diagnostic of endocarditis, terized by the deposition of small sterile thrombi on the
including worsening or changing of a preexistent murmur leaflets of the cardiac valves (Figs. 12-24 and 12-26). The
*Diagnosis by these guidelines, often called the Duke Criteria, requires either pathologic or lesions are 1 to 5 mm in size, and occur as single or mul-
clinical criteria; if clinical criteria are used, 2 major, 1 major + 3 minor, or 5 minor criteria are tiple vegetations along the line of closure of the leaflets or
required for diagnosis.

Janeway lesions are small erythematous or hemorrhagic, macular, nontender lesions on the cusps. Histologically, they comprise bland thrombi that are
palms and soles and are the consequence of septic embolic events.

loosely attached to the underlying valve; the vegetations
Osler nodes are small, tender subcutaneous nodules that develop in the pulp of the digits or
occasionally more proximally in the fingers and persist for hours to several days.
are not invasive and do not elicit any inflammatory reac-
§
Roth spots are oval retinal hemorrhages with pale centers. tion. Thus, although the local effect of the vegetations is
Modified from Durack DT, et al: New criteria for diagnosis of infective endocarditis: utilization usually trivial, they can be the source of systemic emboli
of specific echocardiographic findings. Am J Med, 96:200, 1994, and Karchmer AW: Infective
Endocarditis. In Braunwald E, Zipes DP, Libby P (eds): Heart Disease. A Textbook of
that produce significant infarcts in the brain, heart, or
Cardiovascular Medicine, 6th ed. Philadelphia, WB Saunders, 2001, p 1723. elsewhere.
NBTE is often encountered in debilitated patients, such
as those with cancer or sepsis—hence the previous term
marantic endocarditis (root word marasmus, relating to mal-
weight, and a flulike syndrome. Murmurs are present nutrition). It frequently occurs concomitantly with deep
in 90% of patients with left-sided IE, either from a new venous thromboses, pulmonary emboli, or other findings
valvular defect or from a preexisting abnormality. The suggesting an underlying systemic hypercoagulable
so-called modified Duke criteria (Table 12-9) facilitate eval- state (Chapter 4). Indeed, there is a striking association
uation of individuals with suspected IE that takes into with mucinous adenocarcinomas, potentially relating to
account predisposing factors, physical findings, blood the procoagulant effects of tumor-derived mucin or tissue

A
Figure 12-26 Nonbacterial thrombotic endocarditis (NBTE). A, Nearly complete row of thrombotic vegetations along the line of closure of the mitral valve leaflets
(arrows). B, Photomicrograph of NBTE, showing bland thrombus, with virtually no inflammation in the valve cusp (c) or the thrombotic deposit (t). The thrombus
is only loosely attached to the cusp (arrow).
562 C H A P T E R 12 The Heart

factor that can also cause migratory thrombophlebitis massive hepatic metastatic burden, since the liver normally
(Trousseau syndrome, Chapter 4). Endocardial trauma, as catabolizes circulating mediators before they can affect the
from an indwelling catheter, is another well-recognized heart. Classically, endocardium and valves of the right
predisposing condition, and right-sided valvular and heart are primarily affected since they are the first cardiac
endocardial thrombotic lesions frequently track along the tissues bathed by the mediators released by gastrointesti-
course of pulmonary artery catheters. nal carcinoid tumors. The left side of the heart is afforded
some measure of protection because the pulmonary vascu-
lar bed degrades the mediators. However, left heart carci-
Endocarditis of Systemic Lupus Erythematosus noid lesions can occur in the setting of atrial or septal
(Libman-Sacks Disease) defects and right-to-left flow, or can be elicited by primary
Mitral and tricuspid valvulitis with small, sterile vegeta- pulmonary carcinoid tumors.
tions, called Libman-Sacks endocarditis, is occasionally
encountered in systemic lupus erythematosus. Due to the Pathogenesis. The mediators elaborated by carcinoid
use of steriods, the incidence of this complication has been tumors include serotonin (5-hydroxytryptamine), kalli-
greatly reduced. The lesions are small (1 to 4 mm in diam- krein, bradykinin, histamine, prostaglandins, and tachyki-
eter), single or multiple, sterile, pink vegetations with a nins. Although it is not clear which of these is causal,
warty (verrucous) appearance. They may be located on the plasma levels of serotonin and urinary excretion of the
undersurfaces of the atrioventricular valves, on the valvu- serotonin metabolite 5-hydroxyindoleacetic acid correlate
lar endocardium, on the chords, or on the mural endocar- with the severity of the cardiac lesions. The valvular
dium of atria or ventricles. Histologically the vegetations plaques in carcinoid syndrome are also similar to lesions
consist of a finely granular, fibrinous eosinophilic material that occur in patients taking fenfluramine (an appetite sup-
containing cellular debris including nuclear remnants. pressant) or ergot alkaloids (for migraine headaches); inter-
Vegetations are often associated with an intense valvulitis, estingly, these agents affect systemic serotonin metabolism.
characterized by fibrinoid necrosis of the valve substance Similarly, left-sided plaques have been reported following
and reflecting the activation of complement and recruit- methysergide or ergotamine therapy for migraines;
ment of Fc-receptor-bearing cells. notably, these drugs are metabolized to serotonin as they
Thrombotic heart valve lesions with sterile vegetations pass through the pulmonary vasculature. Despite this tan-
or rarely fibrous thickening can occur in the setting of the talizing evidence, however, it is not known how serotonin
antiphospholipid syndrome, which can also induce a might induce the observed cardiac changes, nor has it been
hypercoagulable state (Chapter 4). The mitral valve is more proven that treatment with serotonin inhibitors has any
frequently involved than the aortic valve, and regurgita- effect on the development or progression of heart lesions.
tion is the usual functional abnormality.

Carcinoid Heart Disease MORPHOLOGY


The cardiovascular lesions associated with the carcinoid syn-
The carcinoid syndrome refers to a systemic disorder
drome are distinctive, glistening white intimal plaquelike thicken-
marked by flushing, diarrhea, dermatitis, and broncho-
ings of the endocardial surfaces of the cardiac chambers and
constriction that is caused by bioactive compounds such
valve leaflets (Fig. 12-27). The lesions are composed of smooth
as serotonin released by carcinoid tumors (Chapter 17).
muscle cells and sparse collagen fibers embedded in an acid
Carcinoid heart disease refers to the cardiac manifestations
mucopolysaccharide-rich matrix material. Underlying structures
caused by the bioactive compounds and occurs in roughly
are intact. With right-sided involvement, typical findings are
half of the patients in whom the systemic syndrome devel-
tricuspid insufficiency and pulmonary stenosis.
ops. Cardiac lesions do not typically occur until there is a

A B
Figure 12-27 Carcinoid heart disease. A, Characteristic endocardial fibrotic lesion involving the right ventricle and tricuspid valve. B, Microscopic appearance
of carcinoid heart disease with intimal thickening. Movat stain shows myocardial elastic tissue (black) underlying the acid mucopolysaccharide-rich lesion
(blue-green). The underlying myocardium is unaffected.
Valvular heart disease 563

Complications of Prosthetic Valves Table 12-10 Complications of Cardiac Valve Prostheses


Thrombosis/thromboembolism
Replacement of damaged cardiac valves with prostheses is Anticoagulant-related hemorrhage
a common and often lifesaving mode of therapy. There are Prosthetic valve endocarditis
two types of valvular prostheses: Structural deterioration (intrinsic)
Wear, fracture, poppet failure in ball valves, cuspal tear, calcification
• Mechanical valves. These consist of different configura- Other forms of dysfunction
tions of rigid nonphysiologic material, such as caged Inadequate healing (paravalvular leak), exuberant healing (obstruction),
balls, tilting disks, or hinged semicircular flaps (bi-leaflet hemolysis
tilting disk valves).
• Tissue valves (bioprostheses). Porcine aortic valves or
bovine pericardium are preserved in a dilute glu-
taraldehyde solution and then mounted on a pros-
thetic frame. Alternatively, frozen human valves from with the attendant risk of hemorrhagic stroke or other
deceased donors (called cryopreserved “homografts”) forms of serious bleeding.
can also be used. Tissue valves are flexible and function • Structural deterioration rarely causes failure of mechani-
similarly to natural semilunar valves. However, the cal valves in current use. However, virtually all bio-
chemical treatment of the animal valves cross-links prostheses eventually become incompetent due to
the valvular proteins, especially collagen, and renders calcification and/or tearing (Fig. 12-28B).
the tissue nonviable. Similarly, the freezing and thawing • Infective endocarditis is a potentially serious complication
of human homografts may also render them largely of any valve replacement. The vegetations of prosthetic
nonviable. valve endocarditis are usually located at the prosthesis-
tissue interface, and often cause the formation of a ring
Approximately 60% of substitute valve recipients
abscess, which can eventually lead to a paravalvular
develop a serious prosthesis-related problem within 10
regurgitant blood leak. In addition, vegetations may
years after the surgery. The complications that occur
directly involve the tissue of bioprosthetic valvular
depend on which type of valve has been implanted (Table
cusps. The major organisms causing such infections are
12-10 and Fig. 12-28).
staphylococcal skin contaminants (e.g., S. epidermidis),
• Thromboembolism is the major consideration with S. aureus, streptococci, and fungi.
mechanical valves (Fig. 12-28A); this may take the form • Other complications include paravalvular leak due to
of either thrombotic occlusion of the prosthesis or inadequate healing, obstruction due to overgrowth of
emboli released from thrombi formed on the valve. fibrous tissue during healing, valve-orifice dispropor-
Because blood flow in all mechanical devices is non- tion (where the effective valve area is too small for
laminar, foci of turbulence and stasis are produced by the needs of the patient, leading to a relative stenosis),
prostheses that predispose to thrombus formation. The intravascular hemolysis due to high shear forces,
risk of such complications necessitates long-term anti- or excessive noise owing to hard contacts of moving
coagulation in all individuals with mechanical valves, rigid parts.

Figure 12-28 Complications of artificial heart valves. A, Thrombosis of a


mechanical prosthetic valve. B, Calcification with secondary tearing of a
porcine bioprosthetic heart valve, viewed from the inflow aspect.
564 C H A P T E R 12 The Heart

KE Y CONCEPTS contraction or relaxation, or to dysregulated ion transport


across cell membranes. Although chronic myocardial dys-
Valvular Heart Disease function secondary to ischemia, valvular abnormalities, or
■ Valve pathology can lead to occlusion (stenosis) and/or to hypertension can cause significant ventricular dysfunction
regurgitation (insufficiency); acquired aortic and mitral (as described previously), these conditions should not be
valves stenoses account for approximately two thirds of all denoted as cardiomyopathies.
valve disease. Cardiomyopathies can be classified according to a
■ Valve calcification is a degenerative process that typically
variety of criteria, including the underlying genetic basis
results in stenosis; abnormal matrix synthesis and turnover
of dysfunction; indeed, we have already discussed a
result in myxomatous degeneration and insufficiency.
number of the arrhythmia-inducing channelopathies,
which may be included in cardiomyopathies. However, we
■ Inflammatory valve diseases lead to post-inflammatory
will confine our list of cardiomyopathies to disorders that
neovascularization and scarring. Rheumatic heart disease
produce anatomic abnormalities in the heart. These fall
results from anti-streptococcal antibodies that cross-react
into three pathologic patterns (Fig. 12-29 and Table 12-11):
with cardiac tissues; it most commonly affects the mitral
valve and is responsible for 99% of acquired mitral • Dilated cardiomyopathy (including arrhythmogenic
stenoses. right ventricular cardiomyopathy)
■ Infective endocarditis can be aggressive and rapidly • Hypertrophic cardiomyopathy
destroy normal valves (acute IE), or can be indolent and • Restrictive cardiomyopathy
minimally destructive of previously abnormal valves (sub- Among the three major patterns, dilated cardiomyopa-
acute IE). Systemic embolization can produce septic thy is most common (90% of cases), and restrictive cardio-
infarcts. myopathy is the least frequent. Within each pattern, there
■ Nonbacterial thrombotic endocarditis occurs on previously is a spectrum of clinical severity, and in some cases clinical
normal valves due to hypercoagulable states; embolization features overlap among the groups. In addition, each of
is an important complication. these patterns can be caused by a specific identifiable
■ Mechanical prosthetic valves have thrombotic or hemor- cause, or can be idiopathic (Tables 12-11 and 12-12).
rhagic complications related to the nonlaminar flow
of blood and the need for chronic anti-coagulation.
Bioprosthetic valves are nonviable and are therefore sus-
ceptible to long-term calcification and/or degeneration
with tearing. Both types of valves have an increased risk
of developing endocarditis relative to native valves. LA LA
Ao Ao

Cardiomyopathies LV LV
Although the term cardiomyopathy (literally, heart muscle
disease) has been historically applied to any cardiac dys-
function resulting from a myocardial abnormality, a more
nuanced definition is probably appropriate. Thus—
stimulated by the recognition of new phenotypes and the Normal Dilated
advent of more sophisticated molecular characterization— cardiomyopathy
an expert panel has suggested: “[C]ardiomyopathies are a
heterogeneous group of diseases of the myocardium associated
with mechanical and/or electrical dysfunction that usually (but
not invariably) exhibit inappropriate ventricular hypertrophy or
dilatation and are due to a variety of causes that frequently are Ao LA Ao LA
genetic. Cardiomyopathies either are confined to the heart or are
part of generalized systemic disorders, often leading to cardio-
vascular death or progressive heart failure-related disability.” LV
Thus, cardiomyopathies manifest as failure of myocar- LV
dial performance; this can be mechanical (e.g., diastolic or
systolic dysfunction) leading to CHF, or can culminate in
life-threatening arrhythmias. Primary cardiomyopathies
can be genetic or acquired diseases of myocardium,
whereas secondary cardiomyopathies have myocardial Hypertrophic Restrictive
involvement as a component of a systemic or multiorgan cardiomyopathy cardiomyopathy
disorder. A major advance in our understanding of cardio-
Figure 12-29 The three major morphologic patterns of cardiomyopathy.
myopathies stems from the frequent identification of Dilated cardiomyopathy leads primarily to systolic dysfunction, whereas
underlying genetic causes, including mutations in myocar- restrictive and hypertrophic cardiomyopathies result in diastolic dysfunction.
dial proteins involved in contraction, cell-cell contacts, Note the changes in atrial and/or ventricular wall thickness. Ao, Aorta;
and the cytoskeleton. These, in turn, lead to abnormal LA, left atrium; LV, left ventricle.
Cardiomyopathies 565

Table 12-11 Cardiomyopathy and Indirect Myocardial Dysfunction: Functional Patterns and Causes
Functional Left Ventricular Mechanisms of Indirect Myocardial Dysfunction
Pattern Ejection Fraction* Heart Failure Causes of Phenotype (Mimicking Cardiomyopathy)
Dilated <40% Impairment of contractility Genetic; alcohol; peripartum; myocarditis; Ischemic heart disease; valvular
(systolic dysfunction) hemochromatosis; chronic anemia; doxorubicin heart disease; hypertensive heart
(Adriamycin) toxicity; sarcoidosis; idiopathic disease; congenital heart disease
Hypertrophic 50% to 80% Impairment of compliance Genetic; Friedreich ataxia; storage diseases; Hypertensive heart disease; aortic
(diastolic dysfunction) infants of diabetic mother stenosis
Restrictive 45% to 90% Impairment of compliance Amyloidosis; radiation-induced fibrosis; idiopathic Pericardial constriction
(diastolic dysfunction)
*Normal, approximately 50% to 65%.

Dilated Cardiomyopathy DCM phenotype can result from diverse causes, both
primary and secondary.
Dilated cardiomyopathy (DCM) is characterized morpho-
logically and functionally by progressive cardiac dilation Pathogenesis. By the time of diagnosis, DCM has typically
and contractile (systolic) dysfunction, usually with con- progressed to end-stage disease; the heart is dilated and
comitant hypertrophy. Many cases are familial, but the poorly contractile. Unfortunately, at that point, even an
exhaustive evaluation frequently fails to suggest a specific
Table 12-12 Conditions Associated with Heart Muscle Diseases etiology. Increasingly, familial (genetic) forms of DCM are
Cardiac Infections recognized, but the final pathology can also result from
Viruses various acquired myocardial insults; as this implies, several
Chlamydia different pathways can lead to DCM (Fig. 12-31).
Rickettsia
Bacteria • Genetic Influences. DCM is familial in at least 30% to 50%
Fungi of cases, in which it is caused by mutations in a diverse
Protozoa group of more than 20 genes encoding proteins involved
in the cytoskeleton, sarcolemma, and nuclear envelope
Toxins
(laminin A/C). In particular, mutations in TTN, a gene
Alcohol that encodes titin (so-called because it is the largest
Cobalt
Catecholamines
protein expressed in humans), may account for approxi-
Carbon monoxide mately 20% of all cases of DCM (Fig. 12-30).
Lithium In the genetic forms of DCM, autosomal dominant
Hydrocarbons inheritance is the predominant pattern; X-linked, auto-
Arsenic somal recessive, and mitochondrial inheritance are less
Cyclophosphamide common. In some families there are deletions in mito-
Doxorubicin (Adriamycin) and daunorubicin
chondrial genes that result in defects in oxidative phos-
Metabolic phorylation; in others there are mutations in genes
Hyperthyroidism encoding enzymes involved in β-oxidation of fatty
Hypothyroidism acids. Mitochondrial defects typically manifest in the
Hyperkalemia pediatric population, while X-linked DCM typically
Hypokalemia presents after puberty and into early adulthood.
Nutritional deficiency (protein, thiamine, other avitaminoses) X-linked cardiomyopathy can also be associated with
Hemochromatosis
mutations affecting the membrane-associated dystro-
Neuromuscular Disease phin protein that couples cytoskeleton to the extracel-
Friedreich ataxia lular matrix; recall that dystrophin is mutated in the
Muscular dystrophy most common skeletal myopathies (i.e., Duchenne and
Congenital atrophies Becker muscular dystrophies; Chapter 27). Some patients
Storage Disorders and Other Depositions and families with dystrophin gene mutations have
DCM as the primary clinical feature. Interestingly, and
Hunter-Hurler syndrome
Glycogen storage disease probably resulting from the common developmental
Fabry disease origin of contractile myocytes and conduction elements,
Amyloidosis congenital abnormalities of conduction may also be
associated with DCM.
Infiltrative
Leukemia
• Myocarditis. Sequential endomyocardial biopsies have
documented progression from myocarditis to DCM. In
Carcinomatosis
Sarcoidosis other studies, the detection of the genetic fingerprints of
Radiation-induced fibrosis coxsackie B and other viruses within myocardium of
patients with DCM suggests that viral myocarditis can
Immunologic be causal (see later discussion).
Myocarditis (several forms)
Posttransplant rejection
• Alcohol and other toxins. Alcohol abuse is strongly asso-
ciated with the development of DCM, raising the
566 C H A P T E R 12 The Heart

Laminin α-2
Dystrophin-associated
glycoproteins Extracellular matrix
Dystroglycans
Plasma membrane
δ
δ

Cytoplasm
δ-Sarcoglycan Emerin

Lamin A/C

Dystrophin Chromatin

Nucleus β-Myosin
Actin heavy chain
Desmin

Z-disc Titin Myosin


binding
protein C Myosin light
Mitochondrial chains
proteins
Troponin I/T α-Tropomyosin

Figure 12-30 Schematic of a myocyte, showing key proteins mutated in dilated cardiomyopathy (red labels), hypertrophic cardiomyopathy (blue labels), or both
(green labels). Mutations in titin (the largest known human protein at approximately 30,000 amino acids) account for approximately 20% of all dilated cardio-
myopathy. Titin spans the sarcomere and connects the Z and M bands thereby limiting the passive range of motion of the sarcomere as it is stretched. Titin
also functions like a molecular spring, with domains that unfold when the protein is stretched and refold when the tension is removed, thereby impacting the
passive elasticity of striated muscle.

possibility that ethanol toxicity (Chapter 9) or a second- cardiomyopathy can be elicited in mouse models
ary nutritional disturbance can underlie myocardial by increased levels of circulating antiangiogenic media-
injury. Alcohol or its metabolites (especially acet- tors including vascular endothelial growth factor inhibi-
aldehyde) have a direct toxic effect on the myocar- tors (e.g., sFLT1, as occurs with preeclampsia) or
dium. Moreover, chronic alcoholism may be associated antiangiogenic cleavage products of the hormone pro-
with thiamine deficiency, which can lead to beriberi lactin (which rises late in pregnancy). Proangiogenic
heart disease (also indistinguishable from DCM). approaches, including the blockade of prolactin secre-
Nevertheless, no morphologic features serve to distin- tion by bromocriptine, represent new therapeutic strate-
guish alcoholic cardiomyopathy from DCM of other causes. gies for treating this disease.
In other cases, some other toxic insult can progress • Iron overload in the heart can result from either heredi-
to eventual myocardial failure. Particularly important is tary hemochromatosis (Chapter 18) or from multiple
myocardial injury caused by certain chemotherapeutic transfusions. DCM is the most common manifestation
agents, including doxorubicin (Adriamycin), and even of such iron excess, and may be caused by interference
targeted cancer therapeutics (e.g., tyrosine kinase inhib- with metal-dependent enzyme systems or to injury
itors). Cobalt is an example of a heavy metal with car- from iron-mediated production of reactive oxygen
diotoxicity and has caused DCM in the setting of species.
inadvertent tainting (e.g., in beer production). • Supraphysiologic stress can also result in DCM. This can
• Childbirth. A special form of DCM, termed peripartum happen with persistent tachycardia, hyperthyroidism,
cardiomyopathy, can occur late in pregnancy or up or even during development, as in the fetuses of insulin-
to months postpartum. The mechanism underlying dependent diabetic mothers. Excess catecholamines, in
this entity is poorly understood but is probably multi- particular, may result in multifocal myocardial con-
factorial. Pregnancy-associated hypertension, volume traction band necrosis that can eventually progress to
overload, nutritional deficiency, other metabolic DCM. This can happen in individuals with pheochromo-
derangements, or an as yet poorly characterized immu- cytomas, tumors that elaborate epinephrine (Chapter 24);
nological reaction have been proposed as causes. Recent use of cocaine or vasopressor agents such as dopamine
work suggests that the primary defect is a microvascu- can have similar consequences. Such “catecholamine
lar angiogenic imbalance within the myocardium effect” also occurs in the setting of intense autonomic
leading to functional ischemic injury. Thus, peripartum stimulation, for example, secondary to intracranial
Cardiomyopathies 567

DILATED CARDIOMYOPATHY HYPERTROPHIC CARDIOMYOPATHY

Non-genetic causes 20-50% genetic causes 100% genetic causes


• Myocarditis Various proteins, Sarcomeric proteins
• Peri partum predominantly related
• Toxic (e.g., alcohol) to cytoskeleton (from
• Idiopathic nucleus to sarcomere
to cell membrane to
adjacent myocytes) Defect in energy transfer from
or mitochondria mitochondria to sarcomere and/or
direct sarcomeric dysfunction
Defect in force generation,
force transmission, and/or
myocyte signaling Hypertrophic cardiomyopathy
phenotype
• Hypertrophy, marked
Dilated cardiomyopathy • Asymmetrical septal hypertrophy
phenotype • Myofiber disarray
• Hypertrophy • Fibrosis, interstitial and
• Dilation replacement
• Fibrosis, interstitial • LV outflow tract plaque
• Intracardiac thrombi • Thickened septal vessels

Clinical
• Heart failure
• Sudden death
• Atrial fibrilation
• Stroke

Figure 12-31 Causes and consequences of dilated and hypertrophic cardiomyopathy. Some dilated cardiomyopathies and virtually all hypertrophic cardiomy-
opathies are genetic in origin. The genetic causes of dilated cardiomyopathy involve mutations in any of a wide range of genes. They encode proteins pre-
dominantly of the cytoskeleton, but also the sarcomere, mitochondria, and nuclear envelope. In contrast, all of the mutated genes that cause hypertrophic
cardiomyopathy encode proteins of the sarcomere. Although these two forms of cardiomyopathy differ greatly in subcellular basis and morphologic phenotypes,
they share a common set of clinical complications. LV, left ventricle.

lesions or emotional duress. Thus, takotsubo cardiomyopa-


regurgitation). Either the coronary arteries are free of significant
thy is an entity characterized by left ventricular contrac-
narrowing or the obstructions present are insufficient to explain
tile dysfunction following extreme psychological stress;
the degree of cardiac dysfunction.
affected myocardium may be stunned or show multi-
The histologic abnormalities in DCM are nonspecific
focal contraction band necrosis. For unclear reasons,
and usually do not point to a specific etiology. Most
the left ventricular apex is most often affected leading
muscle cells are hypertrophied with enlarged nuclei, but some
to “apical ballooning” that resembles a “takotsubo,”
are attenuated, stretched, and irregular. Interstitial and endo-
Japanese for “fishing pot for trapping octopus” (hence,
cardial fibrosis of variable degree is present, and small suben-
the name).
docardial scars may replace individual cells or groups of cells,
The mechanism of catecholamine cardiotoxicity is
probably reflecting healing of previous ischemic necrosis of
uncertain, but likely relates either to direct myocyte
myocytes caused by hypertrophy-induced imbalance between
toxicity due to calcium overload or to focal vasoconstric-
perfusion and demand. Moreover, the severity of morphologic
tion in the coronary arterial macro- or microcirculation
changes may not reflect either the degree of dysfunction or the
in the face of an increased heart rate. Similar changes
patient’s prognosis.
may be encountered in individuals who have recovered
from hypotensive episodes or have been resuscitated
from a cardiac arrest; in such cases, the damage is a
result of ischemia-reperfusion (see earlier) with subse- Clinical Features. DCM can occur at any age, including
quent inflammation. in childhood, but it most commonly affects individuals
between the ages of 20 and 50. It presents with slowly
progressive signs and symptoms of CHF including
dyspnea, easy fatigability, and poor exertional capacity.
MORPHOLOGY At the end stage, ejection fractions are typically less than
25% (normal = 50% to 65%). Secondary mitral regurgitation
In DCM the heart is usually enlarged, heavy (often weighing two
and abnormal cardiac rhythms are common, and embolism
to three times normal), and flabby, due to dilation of all cham-
from intracardiac thrombi can occur. Death usually results
bers (Fig. 12-32). Mural thrombi are common and may be a
from progressive cardiac failure or arrhythmia, and can
source of thromboemboli. There are no primary valvular altera-
occur suddenly. Although the annual mortality is high
tions; if mitral (or tricuspid) regurgitation is present, it results
(10% to 50%), some severely affected patients respond well
from left (or right) ventricular chamber dilation (functional
to pharmacologic therapy. Cardiac transplantation is also
568 C H A P T E R 12 The Heart

A B
Figure 12-32 Dilated cardiomyopathy. A, Four-chamber dilatation and hypertrophy are evident. There is a mural thrombus (arrow) at the apex of the left ventricle
(on the right in this apical four-chamber view). The coronary arteries were patent. B, Histologic section demonstrating variable myocyte hypertrophy and
interstitial fibrosis (collagen is highlighted as blue in this Masson trichrome stain).

increasingly performed, and long-term ventricular assist fatty infiltration and fibrosis (Fig. 12-33). Although myo-
can be beneficial. Interestingly, in some patients, relatively cardial inflammation may be present, ARVC is not consid-
short-term mechanical cardiac support can induce durable ered an inflammatory cardiomyopathy. Classical ARVC
improvement of cardiac function. has autosomal dominant inheritance with a variable pen-
etrance. The disease has been attributed to defective cell
adhesion proteins in the desmosomes that link adjacent
Arrhythmogenic Right cardiac myocytes. Naxos syndrome is a disorder character-
Ventricular Cardiomyopathy ized by arrhythmogenic right ventricular cardiomyopathy
and hyperkeratosis of plantar palmar skin surfaces specifi-
Arrhythmogenic right ventricular cardiomyopathy (ARVC) is cally associated with mutations in the gene encoding the
an inherited disease of myocardium causing right ventricu- desmosome-associated protein plakoglobin.
lar failure and rhythm disturbances (particularly ventricu-
lar tachycardia or fibrillation) with sudden death. Left-sided Hypertrophic Cardiomyopathy
involvement with left-sided heart failure may also occur.
Morphologically, the right ventricular wall is severely Hypertrophic cardiomyopathy (HCM) is a common
thinned due to loss of myocytes, accompanied by extensive (incidence, 1 in 500), clinically heterogeneous, genetic

A B
Figure 12-33 Arrhythmogenic right ventricular cardiomyopathy. A, Gross photograph, showing dilation of the right ventricle and near-transmural replacement
of the right ventricular free-wall by fat and fibrosis. The left ventricle has a virtually normal configuration in this case, but can also be involved by the disease
process. B, Histologic section of the right ventricular free wall, demonstrating replacement of myocardium (red) by fibrosis (blue, arrow) and fat (Masson tri-
chrome stain).
Cardiomyopathies 569

disorder characterized by myocardial hypertrophy, from its source of generation (mitochondria) to its site of
poorly compliant left ventricular myocardium leading to use (sarcomeres). In addition, the interstitial fibrosis in
abnormal diastolic filling, and (in about one third of HCM probably occurs secondary to exaggerated responses
cases) intermittent ventricular outflow obstruction. It is of the myocardial fibroblasts to the primary myocardial
the leading cause of left ventricular hypertrophy unex- dysfunction. In contrast, DCM is mostly associated with
plained by other clinical or pathologic causes. The heart is abnormalities of cytoskeletal proteins (Fig. 12-30), and can
thick-walled, heavy, and hypercontracting, in striking con- be conceptualized as a disease of abnormal force genera-
trast to the flabby, hypocontracting heart of DCM. HCM tion, force transmission, or myocyte signaling. To compli-
causes primarily diastolic dysfunction; systolic function is cate matters, mutations in certain genes, depicted in Figure
usually preserved. The two most common diseases that 12-30, can give rise to either HCM or DCM, depending on
must be distinguished clinically from HCM are deposition the site and nature of the mutation.
diseases (e.g., amyloidosis, Fabry disease) and hyperten-
sive heart disease coupled with age-related subaortic septal
hypertrophy (see earlier discussion under Hypertensive MORPHOLOGY
Heart Disease). Occasionally, valvular or congenital sub- The essential feature of HCM is massive myocardial hyper-
valvular aortic stenosis can also mimic HCM. trophy, usually without ventricular dilation (Fig. 12-34). The
classic pattern involves disproportionate thickening of the ven-
Pathogenesis. In most cases, the pattern of transmission is tricular septum relative to the left ventricle free wall (with a ratio
autosomal dominant with variable penetrance. HCM is of septum to free wall greater than 3 : 1), termed asymmetric
caused by mutations in any one of several genes that septal hypertrophy. In about 10% of cases, the hypertrophy
encode sarcomeric proteins; there are more than 400 differ- is concentric and symmetrical. On longitudinal sectioning, the
ent known mutations in nine different genes, most being normally round-to-ovoid left ventricular cavity may be com-
missense mutations. Mutations causing HCM are found pressed into a “banana-like” configuration by bulging of the
most commonly in the gene encoding β-myosin heavy ventricular septum into the lumen (Fig. 12-34A). Although
chain (β-MHC), followed by the genes coding for cardiac marked hypertrophy can involve the entire septum, it is usually
TnT, α-tropomyosin, and myosin-binding protein C most prominent in the subaortic region. The left ventricular
(MYBP-C); overall, these account for 70% to 80% of all outflow tract often exhibits a fibrous endocardial plaque associ-
cases. Different affected families may have distinct muta- ated with thickening of the anterior mitral leaflet. Both findings
tions involving the same protein. For example, approxi- result from contact of the anterior mitral leaflet with the septum
mately 50 different mutations of β-MHC are known to during ventricular systole; they correlate with the echocardio-
cause HCM. The prognosis of HCM varies widely and cor- graphic “systolic anterior motion” of the anterior leaflet, with
relates strongly with specific mutations. Although it is functional left ventricular outflow tract obstruction during
clear that these genetic defects are critical to the etiology of mid-systole.
HCM, the sequence of events leading from mutations to The most important histologic features of HCM myocardium
disease is still poorly understood. are (1) massive myocyte hypertrophy, with transverse myocyte
As discussed above, HCM is a disease caused by muta- diameters frequently greater than 40 μm (normal, approximately
tions in proteins of the sarcomere. Although such sarco- 15 μm); (2) haphazard disarray of bundles of myocytes, indi-
meric alterations have been thought to be pathologic on the vidual myocytes, and contractile elements in sarcomeres within
basis of abnormal cardiac contraction causing a secondary cells (termed myofiber disarray); and (3) interstitial and
compensatory hypertrophy, newer evidence suggests that replacement fibrosis (Fig. 12-34B).
HCM may instead arise from defective energy transfer

Figure 12-34 Hypertrophic cardiomyopathy with


asymmetric septal hypertrophy. A, The septal
muscle bulges into the left ventricular outflow
tract, and the left atrium is enlarged. The anterior
mitral leaflet has been reflected away from the
septum to reveal a fibrous endocardial plaque
(arrow) (see text). B, Histologic appearance dem-
onstrating myocyte disarray, extreme hypertro-
phy, and exaggerated myocyte branching, as
well as the characteristic interstitial fibrosis (col- A B
lagen is blue in this Masson trichrome stain).
570 C H A P T E R 12 The Heart

Clinical Features. The central abnormality in HCM is causes a restrictive functional defect. Ventricular mural
reduced stroke volume due to impaired diastolic filling. thrombi sometimes develop, and indeed the endocar-
This is a consequence of a reduced chamber size, as well dial fibrosis may result from thrombus organization.
as the reduced compliance of the massively hypertrophied The etiology is unknown.
left ventricle. In addition, approximately 25% of patients • Loeffler endomyocarditis also results in endomyocardial
with HCM have dynamic obstruction to the left ventricular fibrosis, typically with large mural thrombi, with an
outflow. The compromised cardiac output in conjunction overall morphology similar to the tropical disease.
with a secondary increase in pulmonary venous pressure However, in addition to the cardiac changes, there is
explains the exertional dyspnea seen in these patients. often a peripheral eosinophilia and eosinophilic infil-
Auscultation discloses a harsh systolic ejection murmur, trates in multiple organs, including the heart. The
caused by the ventricular outflow obstruction as the ante- release of toxic products of eosinophils, especially major
rior mitral leaflet moves toward the ventricular septum basic protein, is postulated to initiate endomyocardial
during systole. Because of the massive hypertrophy, high necrosis, followed by scarring of the necrotic area, layer-
left ventricular chamber pressure, and frequently thick- ing of the endocardium by thrombus, and finally orga-
walled intramural arteries, focal myocardial ischemia com- nization of the thrombus. Many patients with Loeffler
monly results, even in the absence of concomitant coronary endomyocarditis have a myeloproliferative disorder
artery disease. Major clinical problems in HCM are atrial associated with chromosomal rearrangements involv-
fibrillation, mural thrombus formation leading to emboli- ing either the platelet-derived growth factor receptor
zation and possible stroke, intractable cardiac failure, ven- (PDGFR)-α or -β genes (Chapter 13). These rearrange-
tricular arrhythmias, and, not infrequently, sudden death, ments produce fusion genes that encode constitutively
especially with certain specific mutations. Indeed, HCM is active PDGFR tyrosine kinases. Treatment of such
one of the most common causes of sudden, otherwise patients with the tyrosine kinase inhibitor imatinib has
unexplained death in young athletes. resulted in hematologic remissions associated with
The natural history of HCM is highly variable. Most reversal of the endomyocarditis, which is otherwise
patients can be helped by pharmacologic intervention (e.g., often rapidly fatal.
β-adrenergic blockade) to decrease heart rate and contrac- • Endocardial fibroelastosis is an uncommon heart disease
tility. Some benefit can also be gained by reducing the characterized by fibroelastic thickening that typically
septal myocardial mass, thus relieving the outflow tract involves the left ventricular endocardium. It is most
obstruction. This can be achieved either by surgical exci- common in the first 2 years of life; in a third of cases, it
sion of muscle or by carefully controlled septal infarction is accompanied by aortic valve obstruction or other con-
through a catheter-based infusion of alcohol. genital cardiac anomalies. Endocardial fibroelastosis
may actually represent a common morphologic end-
Restrictive Cardiomyopathy point of several different insults including viral infec-
tions (e.g., intrauterine exposure to mumps) or mutations
Restrictive cardiomyopathy is characterized by a primary in the gene for tafazzin, which affects mitochondrial
decrease in ventricular compliance, resulting in impaired inner membrane integrity. Diffuse involvement may be
ventricular filling during diastole. Because the contractile responsible for rapid and progressive cardiac decom-
(systolic) function of the left ventricle is usually unaffected, pensation and death.
the functional abnormality can be confused with that of
constrictive pericarditis or HCM. Restrictive cardiomyopa-
thy can be idiopathic or associated with distinct diseases Myocarditis
or processes that affect the myocardium, principally radia-
tion fibrosis, amyloidosis, sarcoidosis, metastatic tumors, Myocarditis is a diverse group of pathologic entities
or the deposition of metabolites that accumulate due to in which infectious microorganisms and/or a primary
inborn errors of metabolism. inflammatory process cause myocardial injury. Myocarditis
The morphologic features are not distinctive. The ven- should be distinguished from conditions such as ischemic
tricles are of approximately normal size or slightly enlarged, heart disease, where myocardial inflammation is second-
the cavities are not dilated, and the myocardium is firm ary to other causes.
and noncompliant. Biatrial dilation is commonly observed.
Microscopically, there may be only patchy or diffuse inter- Pathogenesis. In the United States, viral infections are the
stitial fibrosis, which can vary from minimal to extensive. most common cause of myocarditis. Coxsackie viruses A
Endomyocardial biopsy can often reveal a specific etiology. and B and other enteroviruses probably account for most
An important specific subgroup is amyloidosis (described of the cases. Other less common etiologic agents include
later). cytomegalovirus, HIV, and influenza (Table 12-13). In
Several other restrictive conditions merit brief mention. some (but not all) cases, the responsible virus can be ascer-
tained by serologic studies or by identifying viral nucleic
• Endomyocardial fibrosis is principally a disease of chil- acid sequences in myocardial biopsies. Depending on
dren and young adults in Africa and other tropical the pathogen and the host, viruses can potentially cause
areas, characterized by fibrosis of the ventricular endo- myocardial injury either as a direct cytopathic effect, or
cardium and subendocardium that extends from the by eliciting a destructive immune response. Inflammatory
apex upward, often involving the tricuspid and mitral cytokines produced in response to myocardial injury can
valves. The fibrous tissue markedly diminishes the also cause myocardial dysfunction that is out of proportion
volume and compliance of affected chambers and so to the degree of actual myocyte damage.
Cardiomyopathies 571

Table 12-13 Major Causes of Myocarditis


biopsies are diagnostic in some cases, they can be spuriously
Infections negative because inflammatory involvement of the myocardium
Viruses (e.g., coxsackievirus, ECHO, influenza, HIV, cytomegalovirus) may be focal or patchy. If the patient survives the acute phase
Chlamydiae (e.g., Chlamydophyla psittaci) of myocarditis, the inflammatory lesions either resolve, leaving
Rickettsiae (e.g., Rickettsia typhi, typhus fever) no residual changes, or heal by progressive fibrosis.
Bacteria (e.g., Corynebacterium diphtheriae, Neisseria meningococcus, Hypersensitivity myocarditis has interstitial infiltrates, prin-
Borrelia (Lyme disease)
cipally perivascular, composed of lymphocytes, macrophages,
Fungi (e.g., Candida)
Protozoa (e.g., Trypanosoma cruzi [Chagas disease], toxoplasmosis) and a high proportion of eosinophils (Fig. 12-35B). A morpho-
Helminths (e.g., trichinosis) logically distinctive form of myocarditis, called giant-cell myo-
carditis, is characterized by a widespread inflammatory cellular
Immune-Mediated Reactions infiltrate containing multinucleate giant cells (fused macro-
Postviral phages) interspersed with lymphocytes, eosinophils, plasma
Poststreptococcal (rheumatic fever) cells, and macrophages. Focal to frequently extensive necrosis
Systemic lupus erythematosus
is present (Fig. 12-35C). This variant likely represents the fulmi-
Drug hypersensitivity (e.g., methyldopa, sulfonamides)
Transplant rejection nant end of the myocarditis spectrum and carries a poor
prognosis.
Unknown The myocarditis of Chagas disease is distinctive by virtue
Sarcoidosis of the parasitization of scattered myofibers by trypanosomes
Giant cell myocarditis accompanied by a mixed inflammatory infiltrate of neutrophils,
HIV, Human immunodeficiency virus. lymphocytes, macrophages, and occasional eosinophils
(Fig. 12-35D).
Nonviral agents are also important causes of infectious
myocarditis, particularly the protozoan Trypanosoma cruzi, Clinical Features. The clinical spectrum of myocarditis is
the agent of Chagas disease. Chagas disease is endemic in broad. At one end, the disease is entirely asymptomatic,
some regions of South America, with myocardial involve- and patients can expect a complete recovery without
ment in most infected individuals. About 10% of patients sequelae; at the other extreme is the precipitous onset
die during an acute attack; others develop a chronic of heart failure or arrhythmias, occasionally with sudden
immune-mediated myocarditis that may progress to death. Between these extremes are the many levels of
cardiac insufficiency in 10 to 20 years. Trichinosis (Trichinella involvement associated with symptoms such as fatigue,
spiralis) is the most common helminthic disease associated dyspnea, palpitations, precordial discomfort, and fever.
with myocarditis. Parasitic diseases, including toxoplas- The clinical features of myocarditis can mimic those of
mosis, and bacterial infections such as Lyme disease acute MI. As noted previously, patients can develop dilated
and diphtheria, can also cause myocarditis. In the case of cardiomyopathy as a late complication of myocarditis.
diphtheritic myocarditis, the myocardial injury is a conse-
quence of diphtheria toxin release by the causal organism,
Corynebacterium diphtheriae (Chapter 8). Myocarditis occurs Other Causes of Myocardial Disease
in approximately 5% of patients with Lyme disease, a sys-
temic illness caused by the bacterial spirochete Borrelia Cardiotoxic Drugs. Cardiac complications of cancer
burgdorferi (Chapter 8); it manifests primarily as a self- therapy are an important clinical problem; cardiotoxicity
limited conduction system disorder that may require a has been associated with conventional chemotherapeutic
temporary pacemaker. AIDS-associated myocarditis may agents, tyrosine kinase inhibitors, and certain forms of
reflect inflammation and myocyte damage without a clear immunotherapy. The anthracyclines doxorubicin and dau-
etiologic agent, or a myocarditis attributable directly to norubicin are the chemotherapeutic agents most often
HIV or to an opportunistic pathogen. associated with toxic myocardial injury; they cause dilated
There are also noninfectious causes of myocarditis. Broadly cardiomyopathy with heart failure attributed primarily
speaking they are either immunologically mediated (hyper- to peroxidation of lipids in myocyte membranes. Anthra
sensitivity myocarditis) or idiopathic conditions with distinc- cycline toxicity is dose-dependent, with the cardiotoxicity
tive morphology (giant cell myocarditis) suspected to be of risk increasing when cumulative life-time doses exceed
immunologic origin (Table 12-13). 500 mg/m2.
Many other therapeutic agents, including lithium,
phenothiazines, and chloroquine can idiosyncratically
MORPHOLOGY induce myocardial injury and sometimes sudden death.
Common findings in affected myocardium include myofi-
Grossly, the heart in myocarditis may appear normal or dilated;
ber swelling, cytoplasmic vacuolization, and fatty change.
some hypertrophy may be present depending on disease dura-
Discontinuing the offending agent often leads to prompt
tion. In advanced stages the ventricular myocardium is flabby
resolution, without apparent sequelae. Occasionally, how-
and often mottled by either pale foci or minute hemorrhagic
ever, more extensive damage produces myocyte necrosis
lesions. Mural thrombi may be present.
that can evolve to a dilated cardiomyopathy.
Active myocarditis is characterized by an interstitial inflamma-
tory infiltrate associated with focal myocyte necrosis (Fig.
Amyloidosis. Amyloidosis results from the extracellular
12-35). A diffuse, mononuclear, predominantly lymphocytic infil-
accumulation of protein fibrils that are prone to
trate is most common (Fig. 12-35A). Although endomyocardial
forming insoluble β-pleated sheets (Chapter 6). Cardiac
572 C H A P T E R 12 The Heart

A B

C D
Figure 12-35 Myocarditis. A, Lymphocytic myocarditis, associated with myocyte injury. B, Hypersensitivity myocarditis, characterized by interstitial inflammatory
infiltrate composed largely of eosinophils and mononuclear inflammatory cells, predominantly localized to perivascular and expanded interstitial spaces.
C, Giant-cell myocarditis, with mononuclear inflammatory infiltrate containing lymphocytes and macrophages, extensive loss of muscle, and multinucleated
giant cells (fused macrophages). D, The myocarditis of Chagas disease. A myofiber distended with trypanosomes (arrow) is present along with individual
myofiber necrosis, and modest amounts of inflammation.

amyloidosis can appear as a consequence of systemic amy-


loidosis (e.g., due to myeloma or inflammation-associated MORPHOLOGY
amyloid) or can be restricted to the heart, particularly in In cardiac amyloidosis the heart varies in consistency from
the aged (senile cardiac amyloidosis). Senile cardiac amyloi- normal to firm and rubbery. The chambers are usually of
dosis characteristically occurs in individuals 70 years and normal size, but can be dilated and have thickened walls.
older, and has a far better prognosis than systemic amyloi- Small, semitranslucent nodules resembling drips of wax may
dosis. Senile cardiac amyloid deposits are largely com- be seen on the atrial endocardial surface, particularly on the
posed of transthyretin, a normal serum protein synthesized left. Histologically, hyaline eosinophilic deposits of amyloid
in the liver that transports thyroxine and retinol-binding may be found in the interstitium, conduction tissue, valves,
protein. Mutant forms of transthyretin can accelerate the endocardium, pericardium, and small intramural coronary arter-
cardiac (and associated systemic) amyloid deposition; 4% ies (Fig. 12-36); they can be distinguished from other deposits
of African-Americans have a transthyretin mutation sub- by special stains such as Congo red, which produces classic
stituting isoleucine for valine at position 122 that produces apple-green birefringence when viewed under polarized light
a particularly amyloidogenic protein, responsible for (Fig. 12-36B). Intramural arteries and arterioles may have suf-
autosomal dominant familial transthyretin amyloidosis. ficient amyloid in their walls to compress and occlude their
Isolated atrial amyloidosis can also occur secondary to lumens, inducing myocardial ischemia (“small-vessel disease”).
deposition of atrial natriuretic peptide, but its clinical sig-
nificance is uncertain.
Cardiac amyloidosis most frequently produces a restric-
KEY CONCEPTS
tive cardiomyopathy, but it can also be asymptomatic,
manifest as dilation or arrhythmias, or mimic ischemic or Cardiomyopathy
valvular disease. The varied presentations depend on the ■ Cardiomyopathy is intrinsic cardiac muscle disease; there
predominant location of the deposits, for example, intersti- may be specific causes, or it can be idiopathic.
tium, conduction system, vasculature, or valves.
Pericardial disease 573

A B
Figure 12-36 Cardiac amyloidosis. A, Hematoxylin and eosin stain, showing amyloid appearing as amorphous pink material around myocytes. B, Congo red
stain viewed under polarized light, in which amyloid shows characteristic apple-green birefringence (compared with collagen, which appears white).

or pus (purulent pericarditis). With long-standing cardiac


■ There are three general pathophysiologic categories of enlargement or with slowly accumulating fluid, the peri-
cardiomyopathy: dilated (90%), hypertrophic, and restric- cardium has time to dilate. This permits a slowly accumu-
tive (least common). lating pericardial effusion to become quite large without
■ Dilated cardiomyopathy results in systolic (contractile) dys- interfering with cardiac function. Thus, with chronic effu-
function. Causes include myocarditis, toxic exposures sions of less than 500 mL in volume, the only clinical sig-
(e.g., alcohol), and pregnancy. In 20% to 50% of cases, nificance is a characteristic globular enlargement of the
genetic cytoskeletal protein defects are causal, with titin heart shadow on chest radiographs. In contrast, rapidly
mutations representing up to 20% of cases of dilated developing fluid collections of as little as 200 to 300 mL—e.g.,
cardiomyopathy. due to hemopericardium caused by a ruptured MI or aortic
■ Hypertrophic cardiomyopathy results in diastolic (relax- dissection—can produce clinically devastating compres-
ation) dysfunction. Virtually all cases are due to autosomal sion of the thin-walled atria and venae cavae, or the ven-
dominant mutations in the proteins comprising the con- tricles themselves; cardiac filling is thereby restricted,
tractile apparatus, in particular β-myosin heavy chain. producing potentially fatal cardiac tamponade.
■ Restrictive cardiomyopathy results in a stiff, noncompliant
myocardium and can be due to deposition (e.g., amyloid), Pericarditis
increased interstitial fibrosis (e.g., due to radiation), or
endomyocardial scarring. Pericardial inflammation can occur secondary to a variety
■ Myocarditis is myocardial damage caused by inflammatory of cardiac, thoracic, or systemic disorders, metastases from
infiltrates secondary to infections or immune reactions. remote neoplasms, or cardiac surgical procedures. Primary
Coxsackie A and B viruses are the most common causes pericarditis is unusual and almost always of viral origin.
in the United States. Clinically, myocarditis can be asymp- The major causes of pericarditis are listed in Table 12-14.
tomatic, give rise to acute heart failure, or evolve into Most evoke an acute pericarditis, but a few, such as tuber-
dilated cardiomyopathy. culosis and fungi, produce chronic reactions.

Acute Pericarditis
Serous pericarditis is characteristically produced by non-
Pericardial Disease infectious inflammatory diseases, including rheumatic
fever, SLE, and scleroderma, as well as tumors and uremia.
The most important pericardial disorders cause fluid accu- An infection in the tissues contiguous to the pericardium—
mulation, inflammation, fibrous constriction, or some com- for example, a bacterial pleuritis—may incite sufficient
bination of these processes, usually in association with irritation of the parietal pericardial serosa to cause a sterile
other cardiac pathology or a systemic disease; isolated serous effusion that can progress to serofibrinous pericar-
pericardial disease is unusual. ditis and ultimately to a frank suppurative reaction. In
some instances a well-defined viral infection elsewhere—
Pericardial Effusion and Hemopericardium upper respiratory tract infection, pneumonia, parotitis—
antedates the pericarditis and serves as the primary focus
Normally, the pericardial sac contains less than 50 mL of of infection. Infrequently, usually in young adults, a viral
thin, clear, straw-colored fluid. Under various circum- pericarditis occurs as an apparent primary infection
stances the parietal pericardium may be distended by that may be accompanied by myocarditis (myopericardi-
serous fluid (pericardial effusion), blood (hemopericardium), tis). Tumors can cause a serous pericarditis by lymphatic
574 C H A P T E R 12 The Heart

Table 12-14 Causes of Pericarditis


• Seeding from the blood
Infectious Agents • Lymphatic extension
Viruses • Direct introduction during cardiotomy
Pyogenic bacteria
Tuberculosis The exudate ranges from a thin cloudy fluid to frank pus
Fungi up to 400 to 500 mL in volume. The serosal surfaces are
Other parasites reddened, granular, and coated with the exudate (Fig.
12-37). Microscopically there is an acute inflammatory
Presumably Immunologically Mediated
reaction, which sometimes extends into surrounding struc-
Rheumatic fever tures to induce mediastinopericarditis. Complete resolution
Systemic lupus erythematosus
Scleroderma
is infrequent, and organization by scarring is the usual
Postcardiotomy outcome. The intense inflammatory response and the sub-
Postmyocardial infarction (Dressler) syndrome sequent scarring frequently produce constrictive pericarditis,
Drug hypersensitivity reaction a serious consequence (see later). Clinical findings in the
active phase resemble those seen in fibrinous pericarditis,
Miscellaneous
although the frank infection leads to more marked sys-
Myocardial infarction
temic symptoms including spiking fevers and rigors.
Uremia
Following cardiac surgery
Hemorrhagic pericarditis has an exudate composed of
Neoplasia blood mixed with a fibrinous or suppurative effusion; it is
Trauma most commonly caused by the spread of a malignant neo-
Radiation plasm to the pericardial space. In such cases, cytologic
examination of fluid removed through a pericardial tap
often reveals neoplastic cells. Hemorrhagic pericarditis
invasion or direct contiguous extension into the pericar- can also be found in bacterial infections, in persons with
dium. Histologically, serous pericarditis elicits a mild an underlying bleeding diathesis, and in tuberculosis.
inflammatory infiltrate in the epipericardial fat consisting Hemorrhagic pericarditis often follows cardiac surgery
predominantly of lymphocytes; tumor-associated pericar- and is occasionally responsible for significant blood loss or
ditis may also exhibit neoplastic cells. Organization into even tamponade, requiring re-operation. The clinical sig-
fibrous adhesions rarely occurs. nificance is similar to that of fibrinous or suppurative
Fibrinous and serofibrinous pericarditis are the most pericarditis.
frequent types of pericarditis; these are composed of serous Caseous pericarditis is, until proved otherwise, tuber-
fluid variably admixed with a fibrinous exudate. Common culous in origin; infrequently, fungal infections evoke a
causes include acute MI (Fig. 12-18D), postinfarction similar reaction. Pericardial involvement occurs by direct
(Dressler) syndrome (an autoimmune response appearing spread from tuberculous foci within the tracheobronchial
days-weeks after an MI), uremia, chest radiation, rheu- nodes. Caseous pericarditis is a common antecedent of
matic fever, SLE, and trauma. A fibrinous reaction also disabling, fibrocalcific, chronic constrictive pericarditis.
follows routine cardiac surgery.

MORPHOLOGY
In fibrinous pericarditis the surface is dry, with a fine granular
roughening. In serofibrinous pericarditis a more intense inflam-
matory process induces the accumulation of larger amounts of
yellow to brown turbid fluid, containing leukocytes, erythro-
cytes, and fibrin. As with all inflammatory exudates, fibrin may
be lysed with resolution of the exudate, or can become orga-
nized (Chapter 3).

Symptoms of fibrinous pericarditis characteristically


include pain (sharp, pleuritic, and position dependent)
and fever; congestive failure may also be present. A loud
pericardial friction rub is the most striking clinical finding.
However, the collection of serous fluid can actually prevent
rubbing by separating the two layers of the pericardium.
Purulent or suppurative pericarditis reflects an active
infection caused by microbial invasion of the pericardial
space; this can occur through:

• Direct extension from neighboring infections, such as an


empyema of the pleural cavity, lobar pneumonia, medi-
astinal infections, or extension of a ring abscess through Figure 12-37 Acute suppurative pericarditis arising from direct extension of
the myocardium or aortic root an adjacent pneumonia. Extensive purulent exudate is evident.
Tumors of the heart 575

Chronic or Healed Pericarditis. In some cases organiza- resembling those of chronic rheumatic valvular disease.
tion merely produces plaque-like fibrous thickenings of The Libman-Sacks valvular lesions associated with SLE
the serosal membranes (“soldier’s plaque”) or thin, delicate were discussed previously.
adhesions that rarely cause impairment of cardiac function.
In other cases, fibrosis in the form of mesh-like stringy
adhesions completely obliterates the pericardial sac. In Tumors of the Heart
most instances, this adhesive pericarditis has no effect on
cardiac function. Primary tumors of the heart are rare; in contrast, metastatic
Adhesive mediastinopericarditis may follow infectious tumors to the heart occur in about 5% of persons dying of
pericarditis, previous cardiac surgery, or mediastinal irra- cancer. The most common primary cardiac tumors, in
diation. The pericardial sac is obliterated, and adherence descending order of frequency (overall, including adults
of the external aspect of the parietal layer to surrounding and children) are myxomas, fibromas, lipomas, papillary
structures strains cardiac function. With each systolic fibroelastomas, rhabdomyomas, and angiosarcomas. The
contraction, the heart pulls not only against the parietal five most common tumors are all benign and collectively
pericardium but also against the attached surrounding account for 80% to 90% of primary tumors of the heart.
structures. Systolic retraction of the rib cage and dia-
phragm, pulsus paradoxus, and a variety of other charac- Primary Cardiac Tumors
teristic clinical findings may be observed. The increased
workload causes occasionally severe cardiac hypertrophy Myxomas are the most common primary tumor of the
and dilation. adult heart (Fig. 12-38). These are benign neoplasms
In constrictive pericarditis the heart is encased in a dense, thought to arise from primitive multipotent mesenchymal
fibrous or fibrocalcific scar that limits diastolic expansion cells. Although sporadic myxomas do not show consistent
and cardiac output, features that mimic a restrictive car- genetic alterations, familial syndromes associated with
diomyopathy. A prior history of pericarditis may or may myxomas have activating mutations in the GNAS1 gene,
not be present. The fibrous scar can be up to a centimeter encoding a subunit of G protein (Gsα) (in association
in thickness, obliterating the pericardial space and some- with McCune-Albright syndrome) or null mutations in
times calcifying; in extreme cases it can resemble a plaster PRKAR1A, encoding a regulatory subunit of a cyclic-AMP-
mold (concretio cordis). Because of the dense enclosing scar, dependent protein kinase (Carney complex). About 90% of
cardiac hypertrophy and dilation cannot occur. Cardiac myxomas arise in the atria, with a left-to-right ratio of
output may be reduced at rest, but more importantly the approximately 4 : 1.
heart has little if any capacity to increase its output in
response to increased systemic demands. Signs of constric-
MORPHOLOGY
tive pericarditis include distant or muffled heart sounds,
elevated jugular venous pressure, and peripheral edema. The tumors are usually single, but can rarely be multiple. The
Treatment consists of surgical resection of the shell of con- region of the fossa ovalis in the atrial septum is the favored site
stricting fibrous tissue (pericardiectomy). of origin. Myxomas range from small (<1 cm) to large (≥10 cm),
and can be sessile or pedunculated lesions (Fig. 12-38A). They
vary from globular hard masses mottled with hemorrhage
to soft, translucent, papillary, or villous lesions having a gelati-
Heart Disease Associated with nous appearance. The pedunculated form is often sufficiently
Rheumatologic Disorders mobile to move during systole into the atrioventricular valve

The heart (vessels, myocardium, valves, or pericardium)


can be significantly impacted by chronic rheumatologic
diseases (e.g., rheumatoid arthritis, SLE, systemic sclerosis,
ankylosing spondylitis, and psoriatic arthritis). Indeed, as
improved therapies lead to longer life expectancies, the
cardiovascular manifestations of such systemic inflamma-
tion are increasingly recognized. In addition, ischemic
heart disease can be accelerated in the setting of systemic
inflammation.
Although rheumatoid arthritis is primarily a joint dis-
order, it also has several extra-articular manifestations,
including subcutaneous rheumatoid nodules, vasculitis,
and neutropenia (Chapter 26). The heart is also involved
in 20-40% of severe cases. The most common finding is a
fibrinous pericarditis that may progress to fibrous thickening
of the visceral and parietal pericardium and dense adhe- A B
sions. Granulomatous rheumatoid nodules resembling the
Figure 12-38 Atrial myxoma. A, A large pedunculated lesion arises from the
subcutaneous nodules may also occur in the myocardium, region of the fossa ovalis and extends into the mitral valve orifice. B, Abundant
endocardium, valves, and aortic root. Rheumatoid valvulitis amorphous extracellular matrix contains scattered multinucleate myxoma
can lead to marked fibrous thickening and secondary cal- cells (arrowheads) in various groupings, including abnormal vessel-like forma-
cification of the aortic valve cusps, producing changes tions (arrow).
576 C H A P T E R 12 The Heart

(hamartin and tuberin, respectively) function in a complex


opening, causing intermittent obstruction that may be position-
that inhibits the activity of the mammalian target of rapa-
dependent. Sometimes mobile tumors exert a “wrecking-ball”
mycin (mTOR), a kinase that stimulates cell growth and
effect, causing damage to the valve leaflets.
regulates cell size. TSC1 or TSC2 expression is often absent
Histologically, myxomas are composed of stellate or globular
in tuberous sclerosis-associated rhabdomyomas, providing
myxoma cells embedded within an abundant acid mucopoly-
a mechanism for myocyte overgrowth. Because rhabdo-
saccharide ground substance (Fig. 12-38B). Peculiar vessel-like
myomas often regress spontaneously, they may be consid-
or gland-like structures are characteristic. Hemorrhage and
ered as hamartomas rather than true neoplasms.
mononuclear inflammation are usually present.

MORPHOLOGY
The major clinical manifestations are due to valvular
“ball-valve” obstruction, embolization, or a syndrome Rhabdomyomas are gray-white myocardial masses that can be
of constitutional symptoms, such as fever and malaise. small or up to several centimeters in diameter. They are usually
Sometimes fragmentation and systemic embolization calls multiple and involve the ventricles preferentially, protruding into
attention to these lesions. Constitutional symptoms are the lumen. Microscopically, they are composed of bizarre,
probably due to the elaboration by some myxomas of the markedly enlarged myocytes. Routine histologic processing
cytokine interleukin-6, a major mediator of the acute-phase often artifactually reduces the abundant cytoplasm to thin
response. Echocardiography provides the opportunity to strands that stretch from the nucleus to the surface membrane,
identify these masses noninvasively. Surgical removal is an appearance referred to as “spider” cells.
usually curative; rarely, presumably with incomplete exci-
sion, the neoplasm can recurs months to years later.
Sarcoma. Cardiac angiosarcomas and other sarcomas are
Lipoma. Lipomas are localized, well-circumscribed, not clinically or morphologically distinctive from their
benign tumors composed of mature fat cells that can occur counterparts in other locations, and so require no further
in the subendocardium, subepicardium, or myocardium. comment here.
They may be asymptomatic, or produce ball-valve obstruc-
tions or arrhythmias. Lipomas are most often located in the Cardiac Effects of Noncardiac Neoplasms
left ventricle, right atrium, or atrial septum. In the atrial
septum, nonneoplastic depositions of fat sometimes occur With enhanced patient survival due to diagnostic and ther-
that are called “lipomatous hypertrophy.” These lesions apeutic advances, significant cardiovascular effects of non-
include white and brown adipose tissue, as well as small cardiac neoplasms and their therapy are increasingly
interspersed areas of myocardium. encountered (Table 12-15). The pathologic consequences
include direct tumor infiltration, effects of circulating
Papillary Fibroelastoma. Papillary fibroelastomas are mediators, and therapeutic complications.
curious, usually incidental, sea-anemone-like lesions, most The most frequent metastatic tumors involving the heart
often identified at autopsy. They may embolize and thereby are carcinomas of the lung and breast, melanomas, leuke-
become clinically important. Clonal cytogenetic abnormali- mias, and lymphomas. Metastases can reach the heart
ties have been reported, suggesting that fibroelastomas and pericardium by retrograde lymphatic extension (most
are unusual benign neoplasms. They resemble the much carcinomas), by hematogenous seeding (many tumors), by
smaller, usually trivial, Lambl excrescences that may repre- direct contiguous extension (primary carcinoma of the
sent remotely organized thrombus on the aortic valves of lung, breast, or esophagus), or by venous extension (tumors
older individuals. of the kidney or liver). Clinical symptoms are most often
associated with pericardial spread, which can cause
MORPHOLOGY
Papillary fibroelastomas are usually (>80%) located on valves,
particularly the ventricular surfaces of semilunar valves and the Table 12-15 Cardiovascular Effects of Noncardiac Neoplasms
atrial surfaces of atrioventricular valves. Each lesion, typically 1 Direct Consequences of Tumor
to 2 cm in diameter, consists of a distinctive cluster of hairlike Pericardial and myocardial metastases
projections up to 1 cm in length. Histologically, the projections Large vessel obstruction
are covered by a surface endothelium surrounding a core of Pulmonary tumor emboli
myxoid connective tissue with abundant mucopolysaccharide
Indirect Consequences of Tumor (Complications of Circulating
matrix and elastic fibers. Mediators)
Nonbacterial thrombotic endocarditis
Carcinoid heart disease
Rhabdomyoma. Rhabdomyomas are the most frequent
Pheochromocytoma-associated heart disease
primary tumor of the pediatric heart, and are commonly Myeloma-associated amyloidosis
discovered in the first years of life because of obstruction
of a valvular orifice or cardiac chamber. Approximately Effects of Tumor Therapy
half of cardiac rhabdomyomas are due to sporadic muta- Chemotherapy
tions; the other 50% of cases are associated with tuberous Radiation therapy
sclerosis (Chapter 28), with mutations in the TSC1 or TSC2 Modified from Schoen FJ, et al: Cardiac effects of non-cardiac neoplasms. Cardiol Clin 2:657,
1984.
tumor suppressor gene. The TSC1 and TSC2 proteins
Cardiac transplantation 577

A B
Figure 12-39 Complications of heart transplantation. A, Cardiac allograft rejection typified by lymphocytic infiltrate associated with cardiac myocyte damage.
B, Allograft arteriopathy, with severe diffuse concentric intimal thickening producing critical stenosis. The internal elastic lamina (arrow) and media are intact
(Movat pentachrome stain, elastin black). (B, Reproduced with permission from Salomon RN, et al: Human coronary transplantation-associated arteriosclerosis.
Evidence for chronic immune reaction to activated graft endothelial cells. Am J Pathol 138:791, 1991.)

symptomatic pericardial effusions or a mass-effect that is Of the major complications, allograft rejection is the
sufficient to restrict cardiac filling. Myocardial metastases primary problem requiring surveillance; routine endomyo-
are usually clinically silent or have nonspecific features, cardial biopsy is the only reliable means of diagnosing
such as a generalized defect in ventricular contractility or acute cardiac rejection before substantial myocardial
compliance. Bronchogenic carcinoma or malignant lym- damage has occurred and at a stage that is reversible in the
phoma may infiltrate the mediastinum extensively, causing majority of instances. Classic cellular rejection is character-
encasement, compression, or invasion of the superior vena ized by interstitial lymphocytic inflammation with associ-
cava with resultant obstruction to blood coming from the ated myocyte damage; the histology resembles myocarditis
head and upper extremities (superior vena cava syndrome). (Fig. 12-39A). There may also be interstitial edema due to
Renal cell carcinoma often invades the renal vein, and may vascular injury, and local cytokine elaboration can impact
grow as a continuous column of tumor up the inferior vena myocardial contractility without necessarily eliciting
cava and into the right atrium, blocking venous return to myocyte damage. Increasingly, antibody-mediated rejection is
the heart. also recognized as a pathologic mechanism of injury;
Noncardiac tumors may also affect cardiac function donor-specific antibodies directed against major histocom-
indirectly, sometimes via circulating tumor-derived sub- patibility complex proteins lead to complement activation
stances. The consequences include nonbacterial thrombotic and the recruitment of Fc-receptor-bearing cells. Mild rejec-
endocarditis, carcinoid heart disease, pheochromocytoma- tion may resolve spontaneously, while prompt recognition
associated myocardial damage and myeloma-associated of more severe episodes allows successful treatment by
AL-type amyloidosis. augmenting baseline levels of immunosuppression; occa-
Complications of chemotherapy were discussed earlier sionally aggressive anti-T cell or anti-B cell immunotherapy,
in this chapter. Radiation used to treat breast, lung, or with or without plasmapheresis may be necessary.
mediastinal neoplasms can cause pericarditis, pericardial Allograft arteriopathy is the single most important long-
effusion, myocardial fibrosis, and chronic pericardial dis- term limitation for cardiac transplantation. It is a late, pro-
orders. Other cardiac effects of radiation therapy include gressive, diffusely stenosing intimal proliferation in the
accelerated coronary artery disease and mural and valvu- coronary arteries (Fig. 12-39B), leading to ischemic injury.
lar endocardial fibrosis. Within 5 years of transplantation, 50% of patients develop
significant allograft arteriopathy, and virtually all patients
have lesions within 10 years. The pathogenesis of allograft
Cardiac Transplantation arteriopathy involves immunologic responses that induce
local production of growth factors that promote intimal
Transplantation of cardiac allografts is now frequently per- smooth muscle cell recruitment and proliferation with
formed (approximately 3000 per year worldwide) for ECM synthesis. Allograft arteriopathy is a particularly
severe, intractable heart failure of diverse causes—most vexing problem because it can lead to silent MI (transplant
commonly DCM and IHD. Three major factors have con- patients have denervated hearts and do not experience
tributed to the improved outcome of cardiac transplanta- angina), progressive CHF, or sudden cardiac death.
tion since the first human to human transplant in 1967: (1) Other postoperative problems include infection and
more effective immunosuppressive therapy (including the malignancies, particularly Epstein-Barr virus-associated
use of cyclosporin A, glucocorticoids, and other agents), (2) B-cell lymphomas that arise in the setting of chronic T-cell
careful selection of candidates, and (3) early histopatho- immunosuppression. Despite these problems, the overall
logic diagnosis of acute allograft rejection by endomyocar- outlook is good; the 1-year survival is 90% and 5-year sur-
dial biopsy. vival is greater than 60%.

You might also like