You are on page 1of 24

5

Dysmorphology and Genetics


RONALD V. LACRO, MD

In 1966, Smith proposed that the term dysmorphology APPROACH TO THE CHILD WITH
be used to denote the study of abnormalities in morpho- STRUCTURAL DEFECTS
genesis, regardless of etiology, timing of origin, or severity.1
The field has expanded dramatically over the last several About 4% of all infants have at least one major defect in
decades, as great strides have been made in our under- structural development.3 A significant proportion of these
standing of the developmental pathogenesis of many struc- have congenital cardiovascular malformations, for which the
tural defects, including those affecting the cardiovascular incidence in the general population is between 0.4%
system. Congenital anomalies of the cardiovascular system and 1%.2 Furthermore, at least 25% of patients with a
are among the leading causes of morbidity and mortality in congenital heart defect have one or more extracardiac malfor-
infancy and childhood. With an overall incidence between mations.4 Cardiologists and cardiac surgeons frequently are
0.4% and 1% among live-born infants, congenital cardio- involved in the care of patients with multiple malforma-
vascular malformations (CCVMs) or congenital heart defects tions involving multiple organ systems. Consultations
(CHDs) constitute an etiologically heterogeneous group of between the cardiac team and a dysmorphologist or clini-
birth defects with a variety of known and unknown causes.2 cal geneticist, play an integral part in the management of
For most structural cardiovascular malformations, the affected patients and their families.
genetic and biochemical basis for the developmental error A developmental approach to the child with structural
is largely unknown. Vigorous research efforts are currently defects is depicted in Figure 5-1. The ultimate goal of such
directed at elucidating the genetic, biochemical, and cellular an approach is to make a specific diagnosis such that accu-
mechanisms involved in normal and abnormal cardiovas- rate prognosis can be predicted, the recurrence risk can be
cular development. An increasing number of specific determined, and an appropriate management plan may be
genes are now implicated in the pathogenesis of congeni- formulated. When evaluating an infant or child with a
tal cardiovascular malformations in humans and animals. structural defect, it must first be determined whether the
These developments in the dysmorphology and genetics of defect has a prenatal or postnatal onset. Usually this distinc-
pediatric heart disease have led to improvements in clinical tion can be deduced from a careful history and physical
diagnosis, management, and genetic counseling for indi- examination. The term prenatal onset is used to designate
viduals and families with congenital heart disease, whether structural abnormalities that are present at birth, whereas
isolated or associated with one or more extracardiac postnatal onset is used to designate structural abnormal-
malformations. In this chapter, the etiologic and genetic ities that are not present at birth, but rather develop post-
aspects of congenital cardiovascular malformations will natally. Some structural defects are categorized as postnatal
be reviewed, emphasizing those aspects of particular inter- in onset even though the genetic alteration responsible for
est to the pediatrician, pediatric cardiologist, and pediatric them was present prenatally. Once the distinction between
cardiac surgeon. prenatal and postnatal onset has been made, a rational

49
50 Dysmorphology

Dysmorphic features and extracardiac malformations are


commonly associated with congenital heart defects (25%)4
and should prompt an evaluation for a possible syndrome.
Multiple malformation syndromes arise from chromoso-
mal, genetic, teratogenic (environmental), and unknown
causes. For malformation syndromes, the prognosis and
recurrence risk for heart disease depend largely on the
underlying syndrome diagnosis. In the management of
fetuses, infants, children, and adults with congenital
cardiovascular malformations, the importance of a genetics
evaluation, including consultation with a clinical geneticist
and possible cytogenetics evaluation, cannot be overem-
phasized. When a major cardiovascular malformation is
FIGURE 5–1 A developmental approach to the child with struc- detected prenatally, amniocentesis should be strongly
tural defects. considered, particularly if there is a coexistent extracardiac
malformation. In particular, complete atrioventricular
canal defects are common in Trisomy 21 syndrome, and
differential diagnosis can be developed, since this determi- conotruncal malformations (truncus arteriosus, tetralogy
nation narrows the diagnostic possibilities considerably. of Fallot, type B interrupted aortic arch) are common in
The vast majority of structural cardiovascular defects the chromosome 22 deletion syndrome (velocardiofacial
have a prenatal rather than postnatal onset. Congenital syndrome, DiGeorge sequence), which is associated with
heart defects may be the result of malformation (abnormal genetic abnormalities on the long arm of chromosome 22.
development), disruption (interruption of a normal devel- In most patients with the chromosome 22 deletion
opmental process), or deformation (the effects of extrinsic syndrome, a deletion on the long arm of chromosome 22
mechanical forces on normal development). Although some can be detected by fluorescent in situ hybridization (FISH).
isolated defects are caused by single gene mutations Examples of multiple malformation syndromes of
(e.g., mutations of the elastin gene, ELN, in familial supraval- prenatal and postnatal onset are illustrated in Figures 5-2
var aortic stenosis) or known teratogens (e.g., retinoic acid), and 5-3. When structural malformations are present at the
most currently have no identifiable cause. In the past, most time of birth (prenatal onset), the diagnostic possibilities
isolated defects were thought to arise from multifactorial should include chromosomal abnormalities, genetically
influences—a combination of genetic and environmental determined syndromes, and environmental disorders due
causes. Recent research efforts have implicated an increas- to prenatal exposure to a teratogen. The child depicted in
ing number of specific genes in the pathogenesis of Figure 5-2 was born to a mother who used the acne medi-
congenital cardiovascular malformations. cation, cis-retinoic acid (Accutane) during her pregnancy.5

A B
Figure 5–2 Retinoic acid embryopathy in a 2-year-old boy. A, Triangular facies, down-slanting palpebral fissures, and widely
spaced eyes. B, Malformed ears. Brain malformations (e.g., hydrocephalus) and conotruncal abnormalities are common.
From Fyler DC (ed). Nadas’ Pediatric Cardiology. Philadephia: Hanley & Belfus, 1992.
Dysmorphology and Genetics 51

Other teratogens that are known to cause cardiac malfor-


mations in humans include alcohol, anticonvulsants
(hydantoins, trimethadione, valproic acid, carbamazepine),
lithium, thalidomide, warfarin (Coumadin), the rubella
virus, maternal diabetes, and maternal phenylketonuria (see
following discussions).
A patient with mucopolysaccharidosis type I (MPS I,
Hurler syndrome) is depicted in Figure 5-3. Although the
genetic alteration responsible for MPS I syndrome is pres-
ent prenatally, the structural abnormalities develop post-
natally. This child appeared to be normal at birth, having
thrived in utero. By 18 months of age, coarse facial features,
cloudy corneas, poor growth, mental deficiency, and multiple
skeletal anomalies became evident. Deposition of
mucopolysaccharides in cardiac valve tissue, myocardium,
and coronary arteries leads to progressive thickening and
stiffening of valve leaflets, valvar insufficiency, myocardial
dysfunction, sudden death from arrhythmia, and diffuse
coronary artery disease. A small subset of individuals with
severe MPS I have an early-onset fatal endocardiofibro-
elastosis.6 Postnatal onset of these structural defects
should focus the diagnostic evaluation on the etiologic
possibilities set forth in Figure 5-1, as they became mani-
fest after birth. They include genetically determined
inborn errors of metabolism, degenerative diseases of
Figure 5–3 Hurler syndrome (mucopolysaccharidosis I). After the central nervous system, and perinatal and postnatal
birth, the facial features become progressively more coarse. environmental factors such as anoxia, trauma, infection,
Deposition of mucopolysaccharides leads to cardiac complications and drugs.
including valvar insufficiency, myocardial dysfunction, sudden
death from arrhythmia, and diffuse coronary artery disease.
From Fyler DC (ed). Nadas’ Pediatric Cardiology. Philadephia: PATHOGENETIC CLASSIFICATION OF
Hanley & Belfus, 1992. CONGENITAL CARDIOVASCULAR
MALFORMATIONS

In these infants, malformations present at birth include The traditional nomenclature for classifying structural
craniofacial defects, especially anomalies of the ears and the heart defects is based on presumed embryologic events or
Robin sequence, defects of the central nervous system on anatomic characteristics and location. Although helpful
including hydrocephalus and microcephaly, thymic abnor- in naming complex cardiac defects, earlier classification
malities, and a characteristic spectrum of conotruncal systems may have obscured important pathogenetic rela-
malformations including tetralogy of Fallot, double-outlet tionships. More recently, congenital cardiovascular malfor-
right ventricle, supracristal ventricular septal defect, and mations have been classified by disordered mechanisms
type B interruption of the aortic arch. The retinoic acid (Table 5-1). This newer classification scheme, which is
embryopathy is an example of the effect of a teratogen on undergoing continuous reassessment and modification, is
the developing embryo or fetus. In particular, it demon- based on the assumption that there is a limited repertoire
strates how a specific teratogen can lead to a characteristic of developmental mechanisms and a relatively wide spec-
spectrum of cardiac lesions, presumably by acting via a trum of phenotypic expression. Clark proposed six major
common mechanism interfering with conotruncal devel- pathogenetic mechanisms that are likely to be involved in
opment. Disorders caused by environmental agents take the majority of cardiovascular malformations: I, ectomes-
on a special significance because prevention prior to concep- enchymal tissue migration or neural crest abnormalities
tion may be feasible. In general, recognizing that a terato- (conotruncal and aortic arch anomalies); II, abnormalities
genic agent was the cause of the structural defect means of intracardiac blood flow (flow defects); III, cell death
that the recurrence risk is negligible if the mother avoids abnormalities; IV, extracellular matrix abnormalities (endo-
the use of that agent during subsequent pregnancies. cardial cushion abnormalities); V, abnormal targeted growth;
52 Dysmorphology

TABLE 5–1. Classification of Cardiovascular Malformations by Pathogenetic Mechanisms


I. Ectomesenchymal tissue migration abnormalities (neural Pulmonary valve stenosis
crest defects) Pulmonary valve atresia with intact ventricular septum
Conotruncal septation defects Perimembranous ventricular septal defect
Increased mitral–aortic separation (a clinically silent III. Cell death abnormalities
forme fruste) Ebstein malformation of the tricuspid valve
Subarterial or malalignment ventricular septal defect Muscular ventricular septal defect
Double-outlet right ventricle IV. Extracellular matrix abnormalities
Tetralogy of Fallot with or without pulmonary atresia Endocardial cushion defects
Aorticopulmonary window Ostium primum atrial septal defect
Truncus arteriosus communis Atrioventricular canal type ventricular septal defect (inflow)
Branchial arch defects Complete atrioventricular canal defect
Interrupted aortic arch, type B Dysplastic pulmonary or aortic valve
Double aortic arch V. Abnormal targeted growth
Right aortic arch with mirror image branching Anomalous pulmonary venous connection
Aberrant subclavian artery Partially anomalous pulmonary venous connection
Abnormal conotruncal cushion position Totally anomalous pulmonary venous connection
D-transposition of the great arteries Cor triatriatum
II. Abnormalities of intracardiac blood flow VI. Abnormal situs and looping
Left heart defects Situs inversus totalis
Bicuspid aortic valve Isolated
Aortic valve stenosis Immotile cilia syndrome, Kartagener syndrome
Coarctation of the aorta Heterotaxy
Interrupted aortic arch, type A Asplenia syndrome, right isomerism, bilateral rightsidedness
Hypoplastic left heart, aortic atresia, and mitral atresia Polysplenia syndrome, left isomerism, bilateral leftsidedness
Right heart defects Looping abnormalities
Secundum atrial septal defect L-transposition of the great arteries
Bicuspid pulmonary valve Ventricular inversion

and VI, abnormal situs and looping (including but not Although Clark’s classification system is attractive, some
limited to heterotaxy).7–9 mechanistic groupings have proven more useful than
In the most simple situation (Fig. 5-4), one etiology acts others (e.g., conotruncal malformations, left-sided flow
through a single pathogenetic mechanism (A) to produce a defects, and heterotaxy). Recent studies suggest that the
single anatomic malformation. However, for many congenital mechanisms and pathways involved in the developmental
cardiovascular malformations, multiple etiologies may act pathogenesis of cardiovascular malformations are much
through a single mechanism (A) to produce a spectrum of
anatomic abnormalities.7 For example, chromosomal dele-
tions involving the long arm of chromosome 22 and pre-
natal exposure to retinoic acid (multiple etiologies) may act
through a single mechanism (disruption of neural crest cell
migration) to cause a spectrum of conotruncal malforma-
tions, including truncus arteriosus, type B interrupted
aortic arch, and tetralogy of Fallot. Although the specific
defects within each group are heterogeneous, they share a
common disordered mechanism.
Pathogenetic classification is currently being used in
some epidemiologic and family studies. Evaluation of cases
of congenital heart disease by mechanistic groups can help to
clarify relationships among malformations, suggest underly- Figure 5–4 Relationship of etiology, pathogenetic mechanism,
ing mechanisms, and elucidate familial patterns and recur- and anatomic abnormality (see text).
rence risks for relatives. Boughman and colleagues10 found From Clark EB. Growth, morphogenesis, and function: the
that the risk for congenital heart disease was higher among dynamics of cardiovascular development. In Moller JM, Neal WA
siblings of individuals with blood flow defects, as compared (eds). Fetal, Neonatal, and Infant Heart Disease. New York:
to other categories of disordered pathogenetic mechanism. Appleton-Century-Crofts, 1989, pp 1–22, with permission.
Dysmorphology and Genetics 53

more complex than had been predicted by the Clark model.


For example, complex interactions between genes and envi-
ronment and between multiple genes and proteins involved
in multiple developmental pathways have been shown to
be involved in conotruncal development and the determi-
nation of sidedness. Continuing genetic and developmental
investigations will lead inevitably to a better understanding
of the biologic basis of normal and abnormal morphogenesis.
An overview of Clark’s classification system is presented
here, with the realization that ongoing research will lead to
further refinements of the system.

Ectomesenchymal Tissue Migration


Abnormalities (Neural Crest Migration
Abnormalities/Chromosome 22q11
Deletion Syndrome)
Tissue from the neural crest and branchial arch
mesenchyme contributes to the formation and septation of
the outflow tract of the heart. Kirby and associates used
the quail-chick chimera to show that specific regions of
neural crest are major constituents of the conotruncal
septation process.11–13 Ectomesenchymal cells from neural
crest are essential for expression of tissue derivatives of
each branchial arch and pouch. In addition, neural crest
cells course through arches 4, 6, and probably 3 to partici-
pate in septation of the conotruncus and aortic sac.
Mechanical ablation of small amounts of preotic neural
crest produces a spectrum of conotruncal malformations
including truncus arteriosus and subarterial ventricular
septal defect (Fig. 5-5).12 Figure 5–5 Chromosome 22q11 deletion syndrome.
Conotruncal malformations, particularly type B inter-
ruption of the aortic arch, truncus arteriosus, and tetralogy
of Fallot, are overrepresented in patients with DiGeorge syndrome, CATCH 22 (an acronym for cardiac defect,
sequence, where they are seen in association with hypopla- abnormal facies, thymic hypoplasia or aplasia and T-cell
sia or aplasia of the thymus and parathyroid gland, which deficiency, cleft palate, hypoparathyroidism, and hypocal-
are derivatives of pharyngeal pouches III and IV. Tetralogy cemia), and some isolated cardiovascular malformations are
of Fallot is also one of the common cardiac defects in velo- now included in the chromosome 22 deletion syndrome
cardiofacial syndrome, which is characterized by congeni- spectrum.
tal heart defects, palatal abnormalities, learning disability, The first genetic model of the chromosome 22q11 dele-
and a characteristic facies. Previous studies have shown tion syndrome was a mouse mutant with an engineered
deletions and microdeletions of chromosome 22, within deletion of most of the mouse genomic region homologous
region 22q11, in the majority of patients with DiGeorge to the region deleted in human 22q11 deletion patients.21
sequence and velocardiofacial syndrome, and in some familial These mice, carrying a heterozygous deletion, have aortic
cases of congenital heart disease.14–18 More recently, arch defects and parathyroid, thymic, and neurobehavioral
microdeletions of chromosomal region 22q11 have been abnormalities reminiscent of those found in human patients.
identified in patients with nonsyndromic conotruncal Additional targeted mutations of the mouse genome
cardiac defects19–20 demonstrating a genetic contribution to allowed investigators to attribute most of these phenotypic
the development of some isolated conotruncal malforma- findings to haploinsufficiency of a single gene, Tbx1.22–24
tions and altering our understanding of the risk of heritability Extensive searches for mutations of the Tbx1 gene in
of these defects in certain families. DiGeorge sequence, human patients, eventually led to the identification of five
velocardiofacial syndrome, conotruncal anomaly face patients (three of whom were from the same family) carrying
54 Dysmorphology

a Tbx1 gene mutation.25 Most of these patients had a typical also found the chromosome 22q11 deletion in
chromosome 22q11 deletion phenotype including heart 16 (24%) of 66 patients with isolated anomalies of aortic
defects, but did not have learning disabilities. Hence, consis- arch laterality and branching, without associated intracar-
tent with mouse genetic results, human Tbx1 mutation diac defects.32 These arch anomalies include double aortic
(presumably leading to haploinsufficiency), is sufficient to arch, right aortic arch with or without aberrant left subcla-
cause most of the abnormalities observed in the chromo- vian artery, and left aortic arch with aberrant right subcla-
some 22q11 deletion syndrome. A thorough review of the vian artery.
role of Tbx1 in outflow tract and aortic arch development While abnormalities of ectomesenchymal migration
is beyond the scope of this chapter. Interested readers are account for many conotruncal septal defects and branchial
referred to a recent review by Baldini.26 arch defects, the pathogenesis of dextro (D)–transposition
The chromosome 22q11 deletion syndrome (Fig. 5-5) is of the great vessels is thought to involve failure of the
common, with an estimated incidence of at least one in conotruncal cushions to spiral.33 Other human neural crest/
4000.27 Rarely, an abnormality is detected by standard branchial arch syndromes which have an overrepresenta-
karyotype. In most cases (approximately 80%), a microdele- tion of conotruncal malformations and which are associated
tion can be demonstrated by FISH.14–17 In the remainder with abnormalities of branchial arch derivatives include the
of cases, there presumably is a mutation or small deletion facio-auriculo-vertebral spectrum (oculo-auriculo-vertebral
not detectable with currently available FISH probes. dysplasia, hemifacial microsomia, Goldenhar syndrome),
Deletions involving chromosome 10p also have been asso- CHARGE association, thalidomide embryopathy, and
ciated with the chromosome 22 deletion phenotype but retinoic acid embryopathy.
are less common.28 A broad spectrum of clinical manifes-
tations is associated with interstitial chromosome 22q11
deletions, and phenotypic expression is highly variable.29
Defects Associated with Abnormalities of
Cardiovascular anomalies are present in 75% to 80% of
Intracardiac Blood Flow
patients with a chromosome 22q11 deletion. When a
conotruncal malformation is detected either prenatally or Change in volume distribution of intracardiac blood flow
postnatally (tetralogy of Fallot, truncus arteriosus, type B may be a mechanism in the pathogenesis of right and left
interrupted aortic arch) FISH cytogenetic analysis is heart defects. Experimental manipulation of intracardiac
recommended to detect a 22q11 deletion. blood flow affects cardiac morphogenesis. Reduction in
McElhinney and colleagues30 have extensively studied the fetal mitral valve inflow produces a spectrum of left ventric-
frequency of chromosome 22q11 deletion among patients ular volume ranging from mild left ventricular hypoplasia
with various types of cardiovascular defects. They found a to hypoplastic left heart syndrome with aortic and mitral
chromosome 22q11 deletion in 20 (40%) of 50 patients atresia.34 Any event which results in decreased aortic blood
with truncus arteriosus. Anatomic features that were flow and increased pulmonary artery and ductal blood flow
significantly associated with a deletion included a right may lead to one of the defects in the spectrum of left-sided
aortic arch and/or an abnormal aortic arch branching pattern. flow defects. For example, in Turner syndrome, where
There was a trend toward the association of discontinuous there is deletion of part or all of one of the X chromosome
pulmonary arteries with a chromosome 22q11 deletion. in girls, there is an overrepresentation of left-sided flow
Interruption of the aortic arch and truncal valve morphol- defects such as bicommissural aortic valve, coarctation of
ogy and function did not correlate significantly with the the aorta, and hypoplastic left heart syndrome,35 but the
presence of a chromosome 22q11 deletion. In a separate link between deletion of the X chromosome and altered
study, they found the chromosome 22q11 deletion in blood flow has not been elucidated. The indirect sign of
12 (10%) of 125 patients with conoventricular, posterior discrepant ventricular size on fetal sonogram (right ventri-
malalignment, or conoseptal hypoplasia ventricular septal cle larger than left ventricle) can identify fetuses at risk for
defect.31 Anatomic features that were significantly associ- postnatal development of coarctation of the aorta.36
ated with a deletion included abnormal aortic arch sided- It appears likely that blood flow in the developing heart is
ness, an abnormal aortic arch branching pattern, a cervical controlled, at least in part, by genetic factors. During
aortic arch, and discontinuous pulmonary arteries. There mouse heart development, dHAND and eHAND, which
was no correlation between type of ventral-septal defect are related basic helix-loop-helix (bHLH) transcription
(VSD) and chromosome 22q11 deletion. Of 20 patients with factors, are expressed in a complementary fashion and are
an abnormal aortic arch and/or discontinuous pulmonary restricted to segments of the heart tube fated to form the
arteries, 45% had a deletion, compared with only 3% of those right and left ventricles, respectively. These factors repre-
with a left aortic arch, normal aortic arch branching, and sent the earliest cardiac chamber-specific transcription
continuous branch pulmonary arteries. These investigators factors yet identified.37
Dysmorphology and Genetics 55

atrioventricular canal defects in trisomy 21 syndrome.


Extracellular Matrix Abnormalities
The Col6A1,Col6A2 gene cluster, which encode the alpha-1
The endocardial cushions (atrioventricular and and alpha-2 chains for type VI collagen, respectively, fall
conotruncal) consist primarily of glycosaminoglycans sepa- within the congenital heart defect critical region on chro-
rating the endocardium and the myocardium, and consti- mosome 21. Differences in adhesion to type VI collagen
tute the largest proportion of extracellular matrix in the between cultured skin fibroblasts isolated from people with
embryo. Fusion of opposing cushions results in the forma- and without trisomy 21 suggest a potential mechanism for
tion of the tricuspid and mitral orifices at the atrioventric- developmental defects.41
ular level, and the pulmonary and aortic orifices in the Several families have been reported to have autosomal-
outflow tract. A thorough discussion of endocardial cush- dominant atrioventricular canal (AVC) defects with incom-
ion morphogenesis is beyond the scope of this chapter. plete penetrance,42 demonstrating that AVC defects can be
A recent excellent review details the advances in the devel- inherited as a single gene defect. There are two established
opmental biology of atrioventricular septation and genetic loci for isolated AVC defects, also known as atrio-
discusses the complex connection between extracellular ventricular septal defects (AVSDs), designated AVSD1
matrix and growth factor receptor signaling during endo- and AVSD2, which map to chromosomes 1p31-p21 and
cardial cushion morphogenesis38: 3p25, respectively. The CRELD family consists of two
matricellular proteins thought to be involved in cell adhe-
It is becoming clear that converging pathways sion processes. Identification and characterization of the
coordinate early heart valve development and CRELD1 gene on chromosome 3p25 recently led to the
remodeling into functional valve leaflets. The inte- establishment of CRELD1 as the first known genetic risk
gration of these pathways begins with macro and factor for isolated AVC defects.43 In a study of 52 people with
molecular interactions outside the cell in the extracel- non–trisomy 21–related AVC defects, approximately 6% of
lular matrix separating the myocardial and endocar- the subjects had missense mutations in the coding region
dial tissue components of the rudimentary heart. of CRELD1.44 The presence of unaffected family members
Such interactions regulate events at the cell surface carrying a CRELD1 mutation showed that CRELD1 muta-
through receptors, proteases, and other membrane tions are neither necessary nor sufficient to cause AVSD,
molecules, which in turn transduce signals into the indicating that mutation of CRELD1 increases susceptibil-
cell. These signals trigger intracellular cascades that ity for AVC defects rather than being a monogenic cause.
transduce cellular responses through both transcrip- Characterization of CRELD1 as a genetic risk factor is
tion factor and cofactor activation mediating gene consistent with sporadic occurrence of AVC defects in the
induction or suppression. Chamber septation and study population. CRELD1 has now been assigned to the
valve formation occur from these coordinated molecu- AVSD2 locus.
lar events within the endocardial cushions to sustain Bone morphogenic protein-4 (Bmp4) is expressed in the
unidirectional blood flow and embryo viability. endocardial cushions and adjacent tissues of the embryonic
mouse heart. Jiao and colleagues45 developed a transgenic
Clark classified atrioventricular canal defects as defects mouse strain using cardiac-specific conditional gene inac-
of extracellular matrix. However, given the complexity of tivation of a hypomorphic Bmp4 allele. Deficiency of
cushion morphogenesis, it can be assumed that defects in Bmp4 expression in cardiomyocytes resulted in atrioven-
many different classes of molecules could contribute to the tricular canal defects, demonstrating that Bmp4 provides a
pathogenesis of atrioventricular canal defects. Our discus- myocardial-derived signal essential for septation.
sion here will be limited to a few genes. The high frequency Bmp4-deficient mice have a spectrum of atrioventricular
of atrioventricular canal defects in Down (trisomy 21) canal defects that correlate with the level of Bmp4 expres-
syndrome is a clue to the genetic mechanism(s) involved in sion. The severity of defect correlated with the level of
cushion morphogenesis. Increased adhesion of trisomy 21 Bmp4 expression-the lower the level of Bmp4, the more
cells is the basis of a long-standing model for abnormal severe the heart defect.
atrioventricular canal development. Fetal trisomy 21 fibro- GATA4 is a transcription factor known to be essential
blasts explanted from endocardial-cushion-derived structures for cardiac development. Garg and colleagues46 identi-
appear more adhesive in vitro than those explanted from fied mutations in the GATA4 gene as the monogenic cause
normal control fetuses. If the fusion of the atrioventricular of cardiac septal defects in two families with autosomal
cushions is time and location dependent, and the migration dominant ostium secundum atrial septal defects. Clinical
of cells in the trisomic embryo is delayed, then there variability in some affected family members suggests
is a greater chance of the process not occurring.39,40 that GATA4 may be involved in the pathogenesis of
Type VI collagen may have a role in the pathogenesis of other types of congenital heart defects (AVSD, VSD,
56 Dysmorphology

thickening of the pulmonary valve, or insufficiency of


Situs and Looping Abnormalities
cardiac valves).
One of the earliest events in cardiac morphogenesis is the
formation of the normal d (dextral)-cardiac loop from the
CELL DEATH ABNORMALITIES previously symmetric, midline cardiac tube. Abnormalities
at this stage of cardiac development frequently lead to
Controlled cell death is an important molding process in complex congenital heart disease and associated visceral
the embryonic heart.47–48 Ebstein malformation and muscu- malformations. In humans, situs abnormalities (heterotaxy
lar ventricular septal defect have been postulated to arise syndrome) have been observed in pedigrees consistent
from abnormalities in cell death. The tricuspid valve cusps with autosomal dominant, autosomal recessive, and
are almost exclusively derived from the interior of the X-linked recessive inheritance, suggesting that multiple
embryonic right ventricular myocardium by a process of genes regulate cardiac looping and determine cardiac and
undermining of the right ventricular wall.49 Abnormalities visceral situs. Heterotaxy may also be caused by terato-
of this process of reabsorption of ventricular myocardium genic exposures, especially maternal diabetes. Isolated
may lead to the Ebstein malformation with displacement congenital heart defects resulting from isomerisms and
of the functional tricuspid valve annulus into the right ventri- disturbed looping may be caused by mutations in genes
cle. The muscular ventricular septum forms early in devel- that control early left–right patterning and the earliest
opment as trabeculations at the apex of the heart coalesce steps in cardiogenesis. Belmont and colleagues recently
and the margins of the cardiac tube grow toward the endo- reviewed the complex molecular genetics of heterotaxy
cardial cushions and atrioventricular orifice.50 Muscular syndromes. Genes currently implicated in human hetero-
ventricular septal defects may arise from abnormal trabecu- taxy include ZIC3, LEFTYA, CRYPTIC, and ACVR2B.
lar organization or secondarily from foci of cellular death Roles for NKX2.5 and CRELDA are also suggested by
that occur during active cardiac remodeling.7 recent case reports.55
Genes expressed in dorsal midline cells, specifically
notochord cells or adjacent cells induced by the notochord
Abnormal Targeted Growth
(such as neural tube floorplate), are essential for cardiac
Anomalous pulmonary venous connections are believed to left–right development. Several mammalian genes, includ-
arise from abnormalities in targeted growth. The pulmonary ing transcription factors, dyneins, and cell-cell signaling
veins form as an outpouching of endothelial-lined mesenchy- factors, have been implicated in left–right development
mal tissue from the lung buds, and coalesce into the common either by mutations or by asymmetric expression patterns.56
pulmonary vein, which bridges across the splanchnic space Mutations in the gene ZIC3, which maps to human chro-
and fuses with the posterior wall of the primitive left mosome Xq24-27.1, have been identified in humans with
atrium at 5 weeks’ gestation.51 By further remodeling, the X-linked familial situs ambiguous.57–58 ZIC3 is a member
common pulmonary vein is gradually absorbed into the of the ZIC (zinc finger protein of the cerebellum) tran-
posterior wall of the left atrium such that the four pulmonary scription factor family. In mice and Xenopus, ZIC3 is
veins enter the heart individually by eight weeks’ gestation. expressed in the primitive streak and developing neural
The mechanism is undefined but likely involves an attrac- tissues, suggesting that it is part of the midline pathway for
tion between the pulmonary veins and the left atrium. In left–right development. In addition, humans with ZIC3
the chick embryo, following experimental excision and mutations have midline defects in addition to cardiac
reimplantation of the lung bud in an inverted orientation, defects.56–58
venous connection from lung bud to the left atrium is Experiments in the iv/iv mouse (inversus viscerum) are
established in 80% of cases.52 If connection of the common consistent with a control gene, the presence of which defines
pulmonary vein to the left atrium does not occur, primitive the normal relationship of situs solitus. In the absence of
venous drainage to systemic veins persists (totally anom- the control gene (recessive mutation), there is a random
alous pulmonary venous connection). If absorption of the chance of situs inversus or situs solitus.59 Cardiac abnor-
common pulmonary vein into the left atrium is incomplete, malities are found in 40% of developing iv vivo and iv vitro
a membrane may persist between the pulmonary veins and embryos, and the spectrum of abnormalities is strikingly
the left atrium (cor triatriatum). Bleyl and colleagues53 have similar to that seen in humans with heterotaxy syndrome.60
mapped a gene for familial totally anomalous pulmonary The IV gene, which has been mapped to the subtelomeric
venous connection to a locus at chromosome 4q12 in large region of mouse chromosome 12, which is syntenic (homol-
Utah kindreds, and they have preliminary evidence implicat- ogous) with distal human chromosome 14,61 encodes a
ing the gene encoding platelet-derived growth factor recep- novel axonemal dynein, called a left–right dynein, that is
tor alpha (PDGFRA), which is localized in this region.54 expressed in the node and midline. Recent research suggests
Dysmorphology and Genetics 57

that dynein, a microtubule-based motor, is involved in the and familial studies suggest that specific genetic influences
determination of left–right asymmetry and provides insight may be more important than previously recognized, and
into the early molecular mechanisms of this process.62 that certain malformations are more likely to have a
Humans with immotile cilia syndrome (Kartagener stronger genetic component.10,18–20,56–58,66–77
syndrome, primary ciliary dyskinesia), an autosomal reces- Clinical and echocardiographic studies of first-degree
sive disorder, have ciliary dynein defects and laterality relatives of patients with complete common atrioventricu-
defects, as well as respiratory problems and male infertility.63 lar canal have detected previously unsuspected congenital
Although situs inversus totalis can be seen in immotile cilia heart defects that were clinically less significant than the
syndrome, situs inversus totalis is generally considered proband’s congenital heart defect but were part of the same
separate from the heterotaxy syndromes. Yet the coexis- mechanistic spectrum (e.g., atrioventricular type of septal
tence of ciliary abnormalities and polysplenia has been defects and left-axis deviation).78 Brenner and colleagues
reported.64,65 The precise role of ciliary abnormalities in performed echocardiograms on relatives of 11 infants with
situs determination in humans has not been fully HLHS, and observed 5 of 41 first-degree relatives with
delineated. unrecognized bicommissural aortic valve.79 In a study
designed to define the incidence of cardiac anomalies in
first-degree relatives of apparently nonsyndromic children
GENETIC COUNSELING FOR with congenital aortic valve stenosis (AVS), coarctation of the
CARDIOVASCULAR MALFORMATIONS aorta (CoA), and hypoplastic left heart syndrome (HLHS),
Lewin and colleagues80 found bicommissural aortic valve
When possible, identification of a specific diagnosis or (BAV) in 4.68% of first-degree relatives. Additional rela-
etiology improves accuracy in the prediction of prognosis tives had anomalies of the aorta, aortic valve, left ventricle,
and the estimation of recurrence risk. The requirements or mitral valve. Overall, anomalies were found in 20.2%
for optimal genetic counseling for cardiovascular malforma- (21 of 104) of mothers, 14.7% (14 of 95) of fathers, 21.6%
tions include (a) a thorough understanding of the anatomy, (8 of 37) of brothers, and 4.8% (2 of 42) of sisters.
management, and outcome of the particular defect, Echocardiographic anomalies were noted in 10 (35.7%) of
(b) identification of other affected family members and care- 28 AVS families, 22 (42.3%) of 52 CoA families, and 11
ful pedigree analysis for prediction of familial risks, (c) (29.1%) of 32 families of HLHS probands, for a total of 33
identification of associated malformations or syndromes, (29.5%) of 113 families. They concluded that the parents
and (d) options for prenatal diagnosis. Ideally, genetic and siblings of affected patients with left-sided obstructive
counseling should be provided both by a clinical geneticist defects should be screened by echocardiography, as the
or dysmorphologist knowledgeable about cardiac defects presence of asymptomatic BAV may carry a significant
and outcome and by a pediatric cardiologist with a keen long-term health risk.
awareness and interest in genetic issues.66 The techniques of molecular genetics are becoming
The etiology of congenital heart disease is currently the increasingly useful for the study of familial congenital
focus of intense research. Based on the results of this heart disease. An increasing number of specific genes are
research, our concepts of the causes of congenital heart now implicated in the pathogenesis of congenital cardio-
disease have evolved over the years.67–69 In the past, genetic vascular malformations in humans. These genes are
counseling for isolated congenital cardiovascular malforma- summarized in Table 5-2 and discussed in the following
tions (i.e., without extracardiac malformations or syndromic sections. Although the general principles of multifactorial
diagnosis) was transmitted as generalized advice, with the inheritance may still apply in many situations, the recur-
use of an overall recurrence risk for first-degree relatives rence risk may be underestimated in other situations.
of 2% to 5%. These malformations were said to be multi- Therefore, risk projections should be based on the specific
factorial, which refers to defects caused by the combined congenital heart defect involved as well as the genetic and
effects of one or more alleles at a number of loci interact- teratogenic history in an individual family or pregnancy.
ing with stochastic and/or environmental factors. However, The recurrence risks for siblings (Table 5-3) and offspring
the familial (apparent mendelian or single gene) occur- (Table 5-4) are for isolated, nonsyndromic malforma-
rence of virtually all forms of congenital heart disease has tions and are based on combined data published during
been noted. In the past, the study of familial congenital two decades from European and North American
heart disease in humans has been hindered by a number of populations.75
factors including reduced penetrance, variable expressiv- Recent research, accelerated through the Human
ity, genetic heterogeneity, small family size, and decreased Genome Project has resulted in the rapid identification of
survival and reproductive capability especially in those disease genes causing congenital heart defects. Gelb80
individuals with complex defects. Recent epidemiologic recently reviewed genes relevant for atrial (GATA4, TBX5,
58 Dysmorphology

TABLE 5–2. Known Cardiovascular Gene Defects in Humans


Structural Defects CRYPTIC, CFC1, intercellular signaling molecule, chromosome
Thoracic aortic aneurysms and aortic dissection: 2q21.1
11q23.3-q24, 5q13-q14, 3p24.2-p25, genes unknown ACVR2B, activin A receptor, type IIB, chromosome 3p22
Marfan syndrome and related connective tissue disorders: Cardiomyopathies and Arrhythmias
fibrillin gene mutations, chromosome 15q15.21 (FBN1), Hypertrophic cardiomyopathy
5q23-q31 (FBN2) Cardiac troponin T2, TNNT2, on chromosome 1q32
Congenital contractural arachnodactyly, severe form (ventral- Cardiac troponin T, TNNI3, 19q13.4
septal defect [VSD], atrial-septal defect [ASD], IAA): Beta-myosin heavy chain, MYH7, chromosome 14q12
Fibrillin 2 gene mutations (FBN2), chromosome 5q23-q31 Alpha-tropomyosin, TPM1, chromosome 15q22.1
Familial supravalvar aortic stenosis: Cardiac myosin binding protein C, MYBPC3, chromosome 11p11.2
elastin gene point mutations (ELN), chromosome 7q11.23 Essential myosin light chain 3, MYL3, chromosome 3p
Williams syndrome (contiguous gene deletion syndrome): Regulatory myosin light chain 2, MYL2, chromosome 12q23-24.3
1- to 2-Mb deletion including elastin (ELN) and other genes, Alpha-actin cardiac, ACTC, chromosome 15q14
chromosome 7q11.23 Dilated cardiomyopathy
Alagille syndrome, peripheral pulmonary stenosis, tetralogy of Dystrophin (XLDC), DMD, Xp21; Duchenne and Becker
Fallot: muscular dystrophy and familial X-linked cardiomyopathy
JAG1, Jagged1, a ligand for Notch1 receptor, chromosome 20p12 Myotonic dystrophy gene, DMPK, a protein kinase,
Rendu-Osler-Weber (hereditary hemorrhagic telangiectasia) chromosome 19q13.2-13.3
syndrome, telangiectasia and pulmonary arteriovenous fistulas: Autosomal dominant gene loci on chromosomes 9q13q22
Endoglin, endothelial cell transforming growth factor-beta (FDC1), 1q31-q32 (FDC2), 10q21-q23 (FDC3)
binding protein, chromosome 9q33-4 Tafazzin, TAZ, Xq28, also neutropenia and short stature
Ehlers-Danlos syndrome, type IV, aortic dilation and aneurysms: Conduction defects with late development of dilated
COL3A1 gene, Type III collagen, chromosome 2q31-2 cardiomyopathy:
Holt-Oram syndrome/atrial septal defects (heterogeneous): Autosomal recessive gene loci on 1q-p1 (CDDC) and 3p22-p25
Chromosome 12q24.1, TBX5, a transcription factor (CDDC2)
Noonan syndrome (heterogeneous): Isolated dilated cardiomyopathy:
Chromosome 12q24.1, PTPN11, a tyrosine phosphatase Lamin A/C, LMNA, 1q21.1
LEOPARD syndrome (heterogeneous): Myosin binding protein C, Cardiac Type, MYBPC3, 11p11.2
Chromosome 12q24.1, PTPN11, a tyrosine phosphatase Cardiac beta myosin heavy chain, MYH7, 14q11
(allelic with Noonan syndrome) Cardiac troponin T, TNNI2, 1q32
Ellis-van Creveld syndrome: Tropomyosin 1, alpha chain, TPM1, 15q22.1
Chromosome 4p16, EVC and EVC2 Myocardial mitochondrial DNA deletions or mutations
Cornelia de Lange syndrome(heterogeneous): Arrhythmogenic right ventricular dysplasia (ARVD)
Chromosome 5p13.1, NIPBL, nped-B-like protein Gene loci at chromosomes 14q23-q24, 1q42-q43, 14q12-q22
CHARGE syndrome (heterogeneous): chromosome 8q12, CHD7 Long QT syndrome
Kabuki syndrome (heterogeneous): LQT1, KCNQ1, potassium channel gene, chromosome 11p15.5
Chromosome 8p22-p23.1 duplication LQT2, KCNH2, potassium channel gene, chromosome 7q35-36
Familial atrial septal defects (heterogeneous): LQT3, SCN5A, cardiac sodium channel gene, chromosome 3
CSX gene (NKX2.5), transcription factor, chromosome 5q34-35 p21-24
Chromosome 8p23.1-p22 (GATA4), transcription factor LQT4, ANK2, Ankyrin 2, 4q25-q27
Chromosome 5p, gene unknown LQT5, KCNE1, potassium channel gene, chromosome 21q22.2
Familial totally anomalous pulmonary venous connection: LQT6, KCNE2, potassium channel gene, chromosome 21q22.1
Chromosome 4p13-q12, gene unknown, possibly PDGFRA LQT7, KCNJ2, potassium channel gene, chromosome
Atrioventricular canal defects: 17q23.1-q24.2
Chromosome 21q22.2-21q22.3, genes unknown (trisomy 21) Other conduction defects
Chromosome 1p31-p21, (AVSD1), gene unknown Familial heart block, chromosome 19q13.3
Chromosome 3p25, (CRELD1, AVSD2) Catecholaminergic polymorphic ventricular tachycardia,
Chromosome 8p23.1-p22 (GATA4) AD form: Cardiac ryanodine receptor channel, RYR2, 1q42.1-q43
Conotruncal defects/CATCH 22 syndrome: AR form: Calsequestrin, CASQ2, 1p13.3-p11
Deletion on chromosome 22q11,TBX1 and other genes Wolff-Parkinson-White syndrome, linkage of 3 kindreds to 7q3
Deletion on chromosome 10p13, genes unknown Mitochondrial cardiomyopathy, associated with skeletal and
Heterotaxy neuromuscular defects
ZIC3, transcription factor, chromosome Xq24-27.1 Missense mutations
LEFTYA, TGF-beta family of cell-signaling molecules, chromo- Point mutations
some 1q42 Deletion mutations
Dysmorphology and Genetics 59

Table 5–3. Recurrence Risks for Congenital Heart recurrence risk for families of children with congenital
Defects in Siblings heart defects.
Heterozygous mutations in the NKX2.5 gene, which
Percent at Risk
encodes a DNA transcription factor, were among the first
One Two evidence of a genetic cause for congenital heart defects
Sibling Siblings and were initially found in pedigrees with autosomal domi-
Defect Affected Affected nant transmission of cardiac septal defects and atrioven-
Ventricular septal defect 3 10 tricular conduction abnormalities. NKX2.5 mutations have
Patent ductus arteriosus 3 10 subsequently been identified in individuals with ASD,
Atrial septal defect 2.5 8 VSD, Ebstein malformation, tetralogy of Fallot, truncus
Tetralogy of Fallot 2.5 8 arteriosus, double-outlet right ventricle, L-transposition of
Pulmonary stenosis 2 6
Coarctation of aorta 2 6 the great arteries, interrupted aortic arch, subvalvar aortic
Aortic stenosis 2 6 stenosis, hypoplastic left heart syndrome, and coarctation
Transposition 1.5 5 of the aorta.69,70 NKX2.5 mutations are a rare cause of
Endocardial cushion defects 3 10 secundum ASD. Elliot and colleagues71 found only one
Fibroelastosis 4 12 NKX2.5 mutation among 102 individuals with ASD.
Hypoplastic left heart 2 6
Tricuspid atresia 1 3 GATA4 encodes a transcription factor that was known to
Ebstein anomaly 1 3 play a critical role in cardiogenesis. Garg and colleagues46
Truncus arteriosus 1 3 studied a large kindred inheriting a fully penetrant autosomal
Pulmonary atresia 1 3 dominant disorder in which all affected individuals had
From Nora JJ, Nora AH. Update on counseling the family with a first- secundum atrial septal defects. Several also had additional
degree relative with a congenital heart defect. Am J Med Genet 29:137, CHDs, including ventricular septal defects, atrioventricu-
1988. Reprinted with permission of John Wiley & Sons, Inc. lar canal defects, and pulmonary valve stenosis. They
subsequently identified a second family inheriting secun-
dum atrial septal defects, with a 1–base-pair (bp) deletion
NKX2-5) and atrioventricular septal defects (CRELD1), in GATA4, which results in premature truncation of the
patent ductus arteriosus (TFAP2B), bicuspid aortic valve protein. These investigators also showed that three genes,
and coarctation of the aorta (KCNJ2), valvar pulmonary GATA4, TBX5, and NKX2-5, which have been associated
stenosis (PTPN11), and branch pulmonary artery stenosis with secundum atrial septal defects, are now known to
(JAG1). These studies provide insights into the molecular produce proteins that form complexes with one another.
genetic causes of congenital heart defects, and suggest that See further discussion of CRELD1 under Extracellular
DNA testing may become standard for many forms of Matrix Abnormalities above, and TBX5, PTPN11, and
congenital heart defects, improving clinicians’ ability to JAG1, in the sections on Holt Oram, Noonan, and Alagille
anticipate complications for their patients and predict syndromes.
A number of general conclusions can be made based on
this exciting research: (a) Whereas in the past it was
believed that most cardiovascular malformations were due
Table 5–4. Recurrence Risks for Congenital Heart to multifactorial influences, it is now believed that human
Defects in Offspring Given One Affected Parent congenital heart disease is frequently due to single gene
Percent at Risk defects and that even sporadic defects may arise from a
single gene abnormality; (b) a common genetic defect or
Mother Father pathogenetic mechanism may cause several apparently
Defect Affected Affected different forms of congenital cardiovascular malformations.
Aortic stenosis 13–18 3 For example, chromosome 22q deletions cause a variety of
Atrial septal defect 4–4.5 1.5 conotruncal malformations and aortic arch anomalies;
Atrioventricular canal 14 1 (c) the same cardiac malformation can be caused by mutant
Coarctation of aorta 4 2
genes at different loci, indicating that several different, but
Patent ductus arteriosus 3.5–4 2.5
Pulmonary stenosis 4–6.5 2 single, gene defects may cause the same apparent pheno-
Tetralogy of Fallot 2.5 1.5 type; (d) the elucidation of the genetic basis of congenital
Ventricular septal defect 6–10 2 heart disease provides clues to normal cardiovascular
From Nora JJ, Nora AH. Update on counseling the family with a first-
developmental biology. As causative or candidate genes are
degree relative with a congenital heart defect. Am J Med Genet 29:137, isolated, ablation studies in mice will be essential to define
1988. Reprinted with permission of John Wiley & Sons, Inc. interactions with other genes and to understand the
60 Dysmorphology

mechanism by which congenital heart disease becomes severity of expression). For example, many different muta-
manifest; (e) interactions of clinical investigators (cardiol- tions of the fibrillin gene have been identified in Marfan
ogists, geneticists, and cardiothoracic surgeons) with basic syndrome and related connective tissue disorders (allelic
scientists should allow more rapid progress in defining the heterogeneity).87 In contrast, mutations for several differ-
genetic basis of congenital cardiovascular malformations. ent genes encoding myocardial structural proteins
Such research will improve our ability to provide gene- (e.g., β-myosin heavy chain, α-tropomyosin, cardiac
specific treatment for cardiovascular disease with the hope troponin T) are associated with hypertrophic cardiomyopa-
of improved overall prognosis.67–69 thy (nonallelic heterogeneity).81–83 Similarly, mutations for
several different genes encoding cardiac ion channels are
associated with long QT syndrome.82,86 For hypertrophic
Known or Presumed Cardiovascular Gene
cardiomyopathy and long QT syndrome, the severity of
Defects in Humans
clinical manifestations and risk for complications such as
The genes implicated in the pathogenesis of cardiovascu- sudden death depend on the nature of the genetic muta-
lar malformations in humans are summarized in Table 5-2. tion. Some mutations are associated with mild disease,
Such a list is constantly evolving and never complete. whereas others are associated with a severe clinical pheno-
Some of the genes listed are relatively well characterized, type. Genetic testing is now available for some families with
whereas others have been mapped to a chromosomal loca- hypertrophic cardiomyopathy and long QT syndrome, and
tion by familial linkage studies but the precise genetic defect gene-specific therapies are available for some families with
remains unknown. Additional studies in animal models, long QT syndrome.81,86
particularly in the mouse, zebrafish, and Xenopus, have Mitochondrial Disorders: Cardiomyopathy is a
added to our understanding of cardiovascular development, common clinical feature of several well-known mitochon-
but a comprehensive discussion of this research is beyond drial syndromes (MELAS [mitochondrial myopathy,
the scope of this chapter. encephalopathy, lacticacidosis, stroke] syndrome, MERRF
Malformation Syndromes Versus Nonsyndromic [myoclonic epilepsy and ragged-red fibers] syndrome,
Defects: A number of the genetic defects listed in NADH-coenzyme Q reductase deficiency, Kearns-Sayre
Table 5-2 cause malformation syndromes with cardiovascu- syndrome, MIMyCA [maternally inherited myopathy and
lar involvement (e.g., fibrillin mutations cause Marfan cardiomyopathy]). Mitochondrial cardiomyopathy, which
syndrome), whereas others cause isolated familial cardio- is usually accompanied by skeletal and neuromuscular
vascular defects (e.g., elastin mutations cause familial abnormalities, has been attributed to missense mutations,
supravalvar aortic stenosis). Still others cause malforma- point mutations, and deletion mutations. In addition,
tion syndromes in some individuals and nonsyndromic isolated dilated cardiomyopathy has been reported associ-
cardiovascular malformations in other individuals (e.g., ated with myocardial mitochondrial DNA deletions or
chromosome 22q11 deletions). mutations.81,84
Familial Cardiomyopathy and Arrhythmias:
Genetic mutations in several genes encoding sarcomeric
proteins have been associated with hypertrophic MULTIPLE MALFORMATION SYNDROMES
cardiomyopathy.81–83 Mutations in the gene encoding WITH CARDIOVASCULAR INVOLVEMENT
dystrophin not only cause Duchenne and Becker muscular
dystrophy, but also cause familial X-linked dilated Congenital heart defects are common in malformation
cardiomyopathy.84 Other genes for familial cardiomyopa- syndromes. Approximately 70% of spontaneous abortuses
thy and familial arrhythmias have been mapped to chro- and stillborn fetuses with a congenital heart defect and 25%
mosomal positions but the genes are still unknown.81–86 of children with a congenital heart defect have associated
Abnormalities in genes encoding cardiac potassium and extracardiac malformations, often as part of a multiple
sodium channels have been linked to long QT syndrome malformation syndrome. Such syndromes are caused by
(see Chapter 61).68,82,86 chromosomal aberrations, teratogens, genetic abnormalities,
Allelic and Nonallelic Heterogeneity: Genetic and unknown causes. For multiple malformation syndromes,
heterogeneity has been documented or is suspected for a the prognosis and recurrence risk for congenital heart
number of conditions. In other words, there can be differ- disease depend largely on the underlying syndrome.
ent causes for the same disease or phenotype. Genetic Detection of a congenital heart defect should prompt a
heterogeneity can be allelic (different mutations at the search for associated extracardiac malformations and iden-
same gene locus) or nonallelic (mutations at different gene tification of a syndrome. Similarly, individuals with malfor-
loci causing similar phenotypes), and can explain, at least mation syndromes should be evaluated for the presence of
in part, the variability in clinical phenotype (pattern and a cardiovascular defect. This discussion will be limited to
Dysmorphology and Genetics 61

the more common multiple malformation syndromes and from familial SVAS (see discussion on Williams syndrome
those less common ones in which congenital heart defects and familial SVAS).
are either frequent or distinctive. Defect Required for Diagnosis of Syndrome:
Finally, in some syndromes, the cardiovascular malforma-
Patterns of Syndromic Congenital tion is required for diagnosis of the syndrome. In other
words, the diagnostic criteria for the syndrome include the
Cardiovascular Malformations
presence of a cardiovascular abnormality. For example,
Since there is a very large number of malformation aortic root dilation or lens dislocation is required to make
syndromes with cardiovascular involvement, it is helpful to a diagnosis of Marfan syndrome. Similarly, the diagnosis of
discuss these syndromes by the patterns of cardiovascular Holt-Oram syndrome, an autosomal dominant syndrome
defects associated with them. of upper limb and cardiac malformations, requires a cardiac
Nonspecific Increased Risk: For many malformation defect in at least one member of an affected family.
syndromes, there is a nonspecific increased risk for
congenital cardiovascular malformations (i.e., the overall
risk for cardiac defects is higher than the general popula- CHROMOSOMAL SYNDROMES
tion risk, but the distribution of types of defects is similar
to the general population). In these syndromes, defects Given the large number of chromosomal syndromes,
that are common are those that are also common in the this review will be limited to the most common disorders.
general population, such as atrial septal defect, ventricular The incidence of chromosomal abnormalities is about one
septal defect, and patent ductus arteriosus. in 200 at birth and much higher among spontaneous abor-
Increased Risk for Specific Defect(s): For some tions and stillborn fetuses. In a series of spontaneous abor-
syndromes, there is an increased risk for a specific congen- tuses and stillborn fetuses with congenital heart defects, a
ital heart defect. For example, in Noonan syndrome, chromosomal abnormality was confirmed in 19% and
valvar pulmonary stenosis due to a dysplastic pulmonary suspected in up to 36%.87 Recent studies using high-
valve is common, whereas aortic stenosis is very rare. resolution chromosomal analysis have shown that as many
Hypertrophic cardiomyopathy is seen in approximately as 13% of all infants with congenital cardiovascular malfor-
20% to 30% of individuals with Noonan syndrome. In such mations have chromosomal abnormalities.88 The vast
syndromes, the nature of the cardiovascular malforma- majority of these infants have trisomy 21 syndrome (Down
tion can support or contradict the proposed syndrome syndrome), which accounts for about 10% of all infants
diagnosis. with congenital cardiovascular malformations.
Increased Risk for a Specific Family or Spectrum
of Defects: For some syndromes, there is an increased
Trisomy 21 Syndrome (Down Syndrome)
risk for a pathogenetic family or spectrum of related defects.
For example, in Turner syndrome, the entire spectrum of Although the classic description of Down syndrome
left heart obstructive/hypoplastic lesions is observed. (Fig. 5-6) was published in 1866, the cytogenetic confirma-
Although coarctation is most common, the spectrum of tion of its trisomic etiology was not reported until 1959.89–91
defects associated with Turner syndrome ranges from a Trisomy 21 syndrome is the most frequent chromosomal
bicommissural aortic valve without stenosis to hypoplastic aberration affecting live born infants, with an incidence of
left heart syndrome. Similarly, the full spectrum of atrio- 1 in 660 live births: 94% are due to nondisjunction, which
ventricular canal defects is observed in individuals with results in three full copies of chromosome 21 in each cell,
trisomy 21 syndrome, and conotruncal and aortic arch 3% are due to parental translocation, and 3% are due to
malformations are overrepresented in the chromosome 22 mosaicism.
deletion syndrome. The phenotype of trisomy 21 syndrome is well recog-
Defect Pathognomonic for a Specific Syndrome: nized. Cardiovascular malformations are found in 40%
Occasionally, a specific cardiovascular malformation is to 50% of individuals with Down syndrome. There is a distinc-
virtually pathognomonic for the syndrome. For example, tive spectrum of cardiac defects with an overrepresenta-
supravalvar aortic stenosis (SVAS) and the associated tion of endocardial cushion defects (atrioventricular canal
elastin arteriopathy are relatively uncommon in the general defects) compared to the general population. The most
population but are very common in Williams syndrome. common abnormalities (in decreasing order of frequency)
When SVAS is diagnosed in a child with dysmorphic include common atrioventricular canal, ventricular septal
features, growth delay, and/or developmental delay, the diag- defect, tetralogy of Fallot, and patent ductus arteriosus.
nosis is likely to be Williams syndrome. Fluorescent in situ Left-sided obstructive defects such as coarctation and
hybridization (FISH) can differentiate Williams syndrome valvar aortic stenosis are rare. Transposition of the great
62 Dysmorphology

Figure 5–6 Trisomy 21 (Down) syndrome. Note the flat, expressionless face, small nose, low nasal bridge, bilateral epicanthal folds, and
protrusion of the tongue.
From Fyler DC (ed). Nadas’ Pediatric Cardiology. Philadephia: Hanley & Belfus, 1992.

arteries has not been reported in Down syndrome or any with an incidence of one in 3500 newborns.89–91 The major
other autosomal trisomy. features found in virtually all affected individuals include
Prenatally, trisomy 21 is most often identified when intrauterine growth retardation, microcephaly, characteris-
there is advanced maternal age or when maternal serum tic craniofacial features (prominent occiput, short palpe-
factors or fetal sonographic findings indicate an increased bral fissures, malformed and low-set ears, small mouth,
risk. Prenatal detection of a common atrioventricular canal narrow palate, and micrognathia), clenched hands, a short
defect should raise suspicion for Down syndrome, particu- sternum, a low arch pattern of the dermal ridges on the
larly when seen with other sonographic findings suggestive fingertips, and severe cardiac malformations.
of the syndrome, such as increased nuchal thickness and Cardiovascular defects are found in virtually all liveborn
echogenic foci. Postnatally, the clinical phenotype is so infants with trisomy 18 syndrome. Van Praagh et al.92
distinctive that cytogenetic analysis is performed primarily reported on the cardiac malformations in 41 karyotyped
to identify cases due to translocation or mosaicism. The and autopsied cases of trisomy 18 syndrome. The salient
risk of recurrence for Down syndrome is 1%, except in findings included a ventricular septal defect in all cases,
rare cases of the translocation carrier parent, when the risk polyvalvular disease (malformations of more than one
for recurrence will depend on the type of translocation and valve) in 93%, a subpulmonary infundibulum in 98%, and
the sex of the parent that carries it. a striking absence of transposition of the great arteries and
inversion at any level (cardiac or visceral), findings which
appear to be characteristic of all autosomal trisomies. The
Trisomy 18 Syndrome
ventricular septal defect was associated with anterosuperior
Trisomy 18 syndrome (Fig. 5-7) is the second most conal septal malalignment in 61% of cases. The malforma-
common autosomal chromosomal aberration in humans, tions of the atrioventricular and semilunar valves were
Dysmorphology and Genetics 63

trisomy 18 syndrome. The risk of recurrence for trisomy


18 syndrome is estimated to be less than 1%.

Trisomy 13 Syndrome
Trisomy 13 syndrome has an incidence of about one in
5000 live births.89–91 The major features include cleft lip
and palate; holoprosencephalic defects of the eye, nose,
lip, and forebrain; postaxial polydactyly; and localized skin
defects of the scalp at the vertex (cutis aplasia congenita).
Microphthalmia, colobomata of the iris, and retinal dysplasia
are common. Apneic spells, seizures, and severe mental
deficiency are hallmarks.
The incidence of cardiovascular malformations is about
80%. The most frequent defects include patent ductus arte-
riosus (63%), ventricular septal defect (48%), atrial septal
defect (40%), abnormal valves (22%), coarctation of the
aorta (10%), and dextrocardia (6%). The majority of
affected infants (75%) have complex defects.
Infants with trisomy 13 syndrome have a dismally poor
prognosis, as in trisomy 18 syndrome. Although long-term
survival has been reported occasionally, all survivors are
profoundly impaired. Prenatal detection of facial abnor-
malities, such as holoprosencephaly, cleft lip with or with-
out cleft palate, hand and foot malformations, particularly
postaxial polydactyly, and cardiovascular malformations
suggests trisomy 13 syndrome. The risk of recurrence is
presumed to be less than 1%.

Trisomy or Tetrasomy 22p (Cat-Eye


Syndrome)
Figure 5–7 Trisomy 18 syndrome in a newborn infant with
complex cardiac defects. Note petite facial features, clenched The small marker chromosome in the cat-eye syndrome
hands, and short sternum. is either a tandem duplication of proximal 22q or an isodi-
From Fyler DC (ed). Nadas’ Pediatric Cardiology. Philadephia: centric (22)(pter→q11).89–91 The classic pattern of malfor-
Hanley & Belfus, 1992. mations includes mild mental deficiency, hypertelorism,
down-slanting palpebral fissures, iris coloboma, preauricu-
lar pits or tags, and anal and renal malformations, but there
characterized by redundant or thick myxomatous leaflets, is wide variability of phenotypic expression. Cardiovascular
long chordae tendineae, and hypoplastic or absent papillary malformations have been reported in about 40%, and the
muscles. Other defects included double-outlet right occurrence of totally anomalous pulmonary venous connec-
ventricle (10%), all with mitral atresia, and tetralogy of tion has been observed in several studies. Additional defects
Fallot (15%). include tetralogy of Fallot, ventricular septal defect,
Most infants with trisomy 18 syndrome have severe peri- persistent left superior vena cava, interruption of the inferior
natal difficulties attributable to severe brain dysfunction and vena cava, and tricuspid atresia.98
severe cardiac defects. Most die within a week of birth,
most commonly because of apneic spells. Cardiovascular
failure and aspiration pneumonia are common. Survival into
Turner Syndrome
the teens and adult age has been reported, but all have been The major features of Turner syndrome (Fig. 5-8)
profoundly mentally retarded and completely dependent include short stature, primary amenorrhea due to ovarian
on care. dysgenesis, webbed neck, congenital lymphedema, and
Prenatal detection of intrauterine growth retardation, cubitus valgus.89–91 The incidence of Turner syndrome
decreased fetal activity, and clenched fists, accompanied is one in 5000 liveborn female infants. Although common
by visceral, cardiac, and limb malformation suggests in the first trimester, most 45,X conceptuses are
64 Dysmorphology

Figure 5–8 Turner syndrome. Turner syndrome in a young woman with coarctation of the aorta. Note the webbed neck, broad chest,
prominent ears, and multiple pigmented nevi.
From Fyler DC (ed). Nadas’ Pediatric Cardiology. Philadephia: Hanley & Belfus, 1992.

spontaneously aborted. They constitute 25% of sponta- are unclear.93,94 Systemic hypertension is more common in
neous abortions during the first trimester of pregnancy. Turner syndrome than the general population, and should
More than half of liveborn individuals has a 45,X karyotype be treated aggressively.
without evidence of mosaicism. The remainder show Prenatal recognition of coarctation of the aorta, left-
mosaicism and/or more complex rearrangements involving sided hypoplasia, or hypoplastic left heart syndrome in a
the X chromosome (ring, deletion, isochromosome Xq, female infant with hydrops fetalis or cystic hygroma and
isodicentric X, etc.). renal malformation suggests Turner syndrome.
Between 20% and 40% of girls with Turner syndrome
have significant cardiovascular malformations, most
commonly coarctation of the aorta (70%), often bicommis- TERATOGENS
sural aortic valve, and aortic stenosis—defects typically not
common in girls. In fact, girls with Turner syndrome are Teratogens are chemical, physical, and biologic agents
prone to a spectrum of left-sided obstructive/hypoplastic capable of inducing congenital anomalies. Table 5-5
defects ranging in severity from asymptomatic bicommis- summarizes the known or potential teratogens that involve
sural aortic valve to hypoplastic left heart syndrome. Aortic the cardiovascular system. Some teratogens produce distinct
dilation, dissection, and rupture also have been reported, clinical syndromes or recognizable patterns of malforma-
but the incidence and risk factors for these complications tion, the most common of which is the fetal alcohol
Dysmorphology and Genetics 65

Table 5–5. Cardiovascular Abnormalities Caused by Teratogens


Recognizable
Phenotypes
(Syndromes) Cardiac Abnormalities

CHEMICAL TERATOGENS
Fetal alcohol syndrome Ventricular septal defect, atrial septal defect, tetralogy of Fallot, coarctation of the aorta

Fetal hydantoin syndrome Ventricular septal defect, tetralogy of Fallot, pulmonary stenosis, patent ductus arteriosus, atrial
septal defect, coarctation of the aorta
Fetal trimethadione Combined defects
syndrome
Fetal valproate syndrome Nonspecific
Fetal carbamazepine Ventricular septal defect, tetralogy of Fallot
syndrome
Retinoic acid Conotruncal malformations
embryopathy
Thalidomide embryopathy Conotruncal malformations
Fetal warfarin syndrome Patent ductus arteriosus, peripheral pulmonary stenosis
BIOLOGIC TERATOGENS
Maternal PKU fetal Tetralogy of Fallot, ventricular septal defect, coarctation
effects of the aorta
Maternal lupus fetal Complete heart block, cardiomyopathy, L-transposition of the
effects great arteries
Fetal rubella syndrome Patent ductus arteriosus, peripheral pulmonary stenosis, fibromuscular and intimal proliferation
of medium and large arteries, ventricular septal defect, atrial septal defect
Nonsyndromic increased risk for
malformations
Lithium Ebstein anomaly, tricuspid atresia, atrial septal defect
Maternal diabetes Transposition of the great arteries, ventricular septal defect, coarctation of the aorta, hyper-
trophic cardiomyopathy

syndrome, whereas other agents cause an increased inci- SINGLE GENE DISORDERS
dence of single or multiple malformations without a
specific syndrome pattern (e.g., maternal ingestion of
Marfan Syndrome
lithium or maternal diabetes). Biologic teratogens include
maternal illnesses (e.g., maternal phenylketonuria or Marfan syndrome is the most common and best character-
maternal lupus erythematosus) or maternal-fetal infections ized of the connective tissue disorders with serious cardio-
(e.g., rubella) that can cause birth defects. vascular manifestations.96–98 This condition is an autosomal
dominant, multisystem disorder with a wide variability of
phenotypic expression involving the skeletal, ocular, cardio-
Fetal Alcohol Syndrome
vascular, and other systems. Mutations involving the gene,
One of the most common malformation syndromes, FBN1, for the connective tissue protein fibrillin-1, have been
with an estimated frequency greater than one in 1000 live documented.96 Nearly all patients have some cardiovascular
births, the fetal alcohol syndrome (Fig. 5-9) is also one of involvement, most commonly mitral valve prolapse and
the most common identifiable causes of mental retarda- aortic root dilation. The risk for aortic regurgitation, dissec-
tion. The most serious consequence of prenatal alcohol tion, and rupture increases with progressive enlargement
exposure is its effects on brain development and function. of the aortic root. Beta-blockers may retard the rate of dila-
About half of all individuals with fetal alcohol syndrome tion of the aortic root and ascending aorta.97 In adults,
have congenital cardiovascular malformations.95 By far the replacement of the root and ascending aorta is recommended
most common defect is ventricular septal defect, followed when the aortic root diameter is greater than 5.5 cm or
by atrial septal defect and tetralogy of Fallot. Less greater than 5.0 cm if there is a family history of dissection.
commonly reported lesions include atrioventricular canal Worldwide, surgery for patients with Marfan syndrome
defect, hypoplasia or absence of one pulmonary artery, is performed by experienced surgeons at a very low risk,
subaortic stenosis, and complex defects. with excellent outcomes. Many experienced centers are now
66 Dysmorphology

Figure 5–10 Noonan syndrome.

families were mapped to chromosome 12q,103 and in 2001,


Tartaglia and colleagues104,105 reported on their discovery
that mutations of the PTPN11 gene account for about half
of the cases with a clinical diagnosis of Noonan syndrome,
Figure 5–9 Fetal alcohol syndrome. Typical facies in a child with consistent with genetic heterogeneity. The major features
severe manifestations of prenatal alcohol exposure. Note the include short stature, seen in about half of affected individu-
short, downslanting palpebral fissures, short upturned nose, long als; mental retardation (usually mild) seen in about 25% of
smooth philtrum, and thin upper lip. cases; characteristic facial appearance including widely
From Fyler DC (ed). Nadas’ Pediatric Cardiology. Philadephia:
spaced eyes, epicanthal folds, ptosis, and lowset, malformed
Hanley & Belfus, 1992.
ears; prominent trapezius muscle with a short webbed neck
and low posterior hairline; a shield chest deformity with
routinely offering a valve-sparing aortic root replacement pectus carinatum superiorly and pectus excavatum inferi-
for people with Marfan syndrome, instead of a composite orly; cubitus valgus; and cryptorchidism among males.
valve graft which involves a prosthetic aortic valve. Because At least 50% of individuals with Noonan syndrome have
of the low risk of prophylactic aortic root replacement in congenital heart defects. About 75% of those with congen-
Marfan syndrome and the devastating effects on short- and ital heart defects have valvar pulmonary stenosis secondary
long-term survival after dissection, some are now advocating to a dysplastic pulmonary valve with thickened valve leaflets.
surgery when the aortic root diameter exceeds 45 mm, espe- Particularly unusual is the high incidence of leftward or
cially if there is a family history of aortic dissection, when superior frontal QRS vector, even in the face of severe right
there is a rapid growth of the aorta (i.e., > 5–10 mm/year), ventricular hypertrophy, presumably due to a conduction
or when significant aortic insufficiency is present.99 abnormality. Other defects reported include atrial septal
defect (30%, usually associated with pulmonary stenosis),
patent ductus arteriosus (10%), and ventricular septal defect
Noonan Syndrome
(10%). Rare lesions include tetralogy of Fallot, coarctation
Noonan syndrome (Fig. 5-10) is a well-known autosomal of the aorta, subaortic stenosis, Ebstein malformation, and
dominant, multisystem disorder associated with congenital complex defects. Hypertrophic cardiomyopathy, which can
heart defects.100–102 Based on genetic linkage studies, some involve both ventricles, is observed in 10% to 20% of cases.
Dysmorphology and Genetics 67

The PTPN11 gene encodes SHP-2, a nonmembranous mapped to 12q by family linkage studies, encodes a
protein tyrosine phosphatase with diverse roles in signal transcription factor, TBX5.107–109 Other families have not
transduction. Particularly relevant to the cardiac involvement mapped to this locus, consistent with genetic heterogeneity.
in Noonan syndrome, SHP-2 has been shown to act in epider- An affected individual may have isolated upper limb defects,
mal growth factor signaling in semilunar valvulogenesis.69 isolated heart defects, or a combination. All gradations of
skeletal defects involving the upper limb and shoulder
girdle, ranging from mild hypoplasia of the thumb to
Holt-Oram Syndrome
phocomelia, can occur even within the same family. All
This syndrome of upper limb and cardiovascular malfor- patients with involvement of the upper limbs have defects
mations (Fig. 5-11) was first described in 1960 by Holt and of the thumb. There is no correlation between the severity
Oram,106 who noted the association of radial anomalies of the defect of the limb and the cardiac defect.
with atrial septal defect. It is an autosomal dominant disor- Congenital heart defects are observed in at least half of
der with variable expression, but complete or near- affected individuals, although there may be an ascertain-
complete penetrance. The gene for Holt-Oram syndrome, ment bias toward cardiac defects. Atrial septal defect is the
most frequent lesion, but a number of cardiac phenotypes
have been reported including normal, first-degree atrio-
ventricular block, ostium primum atrial septal defects,
isolated ventricular septal defects, and hypoplastic left heart
syndrome. Prognosis is dependent on the degree of
skeletal deformity and the nature of the cardiac anomaly.
Intelligence is normal, and there are no associated visceral
malformations.

Alagille Syndrome
The typical manifestations of Alagille syndrome (arterio-
hepatic dysplasia), an autosomal dominant disorder, include
intrahepatic cholestasis due to bile duct paucity, distinctive
facies (prominent forehead and chin, deep-set eyes, long
nose), anterior chamber abnormalities of the eye, especially
posterior embryotoxon, and butterfly hemivertebrae.
Peripheral pulmonary artery stenosis is most common,
although other defects including tetralogy of Fallot have also
been observed.110 This disorder is caused by mutations or
deletions of the JAG1 gene, which encodes for Jagged 1, a
ligand in the Notch intercellular signaling pathway.69,111–113
McElhinney and colleagues114 examined a large cohort
of 200 individuals with JAG1 mutations and/or Alagille
syndrome with respect to their cardiac phenotype. They
determined that 94% had cardiac involvement. Aside from
peripheral pulmonary artery stenosis, which was the most
prevalent lesion, other defects included tetralogy of Fallot,
pulmonary valve stenosis, atrial septal defect, ventricular
septal defect, and assorted other defects at low frequency.
A noteworthy finding was that the clinical outcome among
those with tetralogy of Fallot was poor, with 43% mortality.
Analysis of genotype–phenotype correlation failed to
disclose a relationship between the type or location of the
Figure 5–11 Holt-Oram syndrome in a newborn infant with an JAG1 mutation and the cardiac phenotype. Overall, this
atrial septal defect. Note the bilateral deficiency of the upper limbs. study revealed a relatively high percentage of left-sided
From Fyler DC (ed). Nadas’ Pediatric Cardiology. Philadephia: defects (11%) and tetralogy of Fallot with apparent excess of
Hanley & Belfus, 1992. those with pulmonary atresia and none with right aortic arch.
68 Dysmorphology

Ellis–Van Creveld Syndrome


(Chondroectodermal Dysplasia)
Ellis–van Creveld syndrome is an autosomal recessive
disorder with the following major features: short stature of
prenatal onset (short limbs), ectodermal dysplasia mani-
fested by hypoplastic nails and dental anomalies (neonatal
teeth, partial anodontia, small teeth and/or delayed erup-
tion), postaxial polydactyly, narrow thorax, and cardiac
defects.115 About 50% of patients have congenital cardio-
vascular malformations. Atrial septal defect or common
atrium occurs in about half of patients with congenital heart
disease. Other less frequent defects include patent ductus
arteriosus, persistent left superior vena cava, hypoplastic
left heart syndrome, coarctation of the aorta, totally anom-
alous pulmonary venous connection, and transposition of
the great arteries. Most survivors have normal intelligence.
Adult stature is in the range of 43 to 60 inches. Dental
problems are frequent.
The syndrome has been mapped to chromosome
4p16116 and mutations of two nonhomologous genes at this
locus (EVC and EVC2) have been identified in affected
families.117,118 For couples with at least one affected child,
the recurrence risk is 25% for each subsequent pregnancy.

CONTIGUOUS GENE DELETION


SYNDROMES

Williams Syndrome
Williams and colleagues119,120 described this condition in Figure 5–12 Williams syndrome in a 2-year-old boy with
four unrelated children with mental retardation, an supravalvar aortic stenosis. Note the typical facies including a
unusual facial appearance, and supravalvar aortic stenosis stellate pattern of the iris, short anteverted nose, long philtrum,
prominent lips, and large, open mouth.
(Fig. 5-12). Hypocalcemia has been an infrequent finding,
From Fyler DC (ed). Nadas’ Pediatric Cardiology. Philadephia:
particularly beyond the neonatal period. At least half of Hanley & Belfus, 1992.
these individuals have cardiovascular defects. Supravalvar
aortic stenosis is the most frequent single defect, but any
of the systemic and pulmonary arteries can be affected.
Narrowing at the sinotubular ridge can be detected by Williams syndrome is a contiguous gene deletion
echocardiography and angiography postnatally, but may be syndrome associated with a 1- to 2-megabase deletion on
difficult to detect prenatally. the long arm of chromosome 7, including the entire elastin
The SVAS complex or elastin arteriopathy can occur as gene and 20 additional genes. The loss of these additional
an isolated autosomal dominant disorder (nonsyndromic genes presumably account for the nonelastin-related mani-
SVAS) or as part of Williams syndrome.121 The SVAS festations of Williams syndrome. The vast majority of
complex is a diffuse arteriopathy which can affect all patients have a deletion involving one copy of the elastin
segments of the aorta; any of its branches including the gene, detectable by FISH.121–123
coronary arteries, the brachiocephalic vessels, the mesen- In contrast, familial (nonsyndromic) SVAS is caused by
teric and the renal arteries, as well as the pulmonary and mutations of the elastin gene. A translocation disrupting
cerebral arteries. Abnormalities in elastin production are the elastin gene as well as intragenic deletions and muta-
thought to be responsible for the cardiovascular phenotype tions involving the elastin gene have been identified in
in both Williams syndrome and nonsyndromic SVAS, so nonsyndromic SVAS.121,124–126 Individuals with familial SVAS
the similarity in vascular abnormalities is predictable. Both typically do not show evidence of a complete deletion of
conditions link to the elastin gene on chromosome.7 elastin by FISH.
Dysmorphology and Genetics 69

Chromosome 22q Deletions (DiGeorge and/or defects of the central nervous system. In some
instances, the severity of the malformations has led to death
Sequence, Velocardiofacial Syndrome,
in the perinatal period. Recently Vissers and colleagues127
CATCH 22)
detected a microdeletion on chromosome 8q12 in two
The DiGeorge sequence, velocardiofacial syndrome, individuals with CHARGE and mutations in the
conotruncal face anomaly syndrome, CATCH 22, and CHD7 gene in 10 of 17 individuals with CHARGE.
some isolated conotruncal malformations are associated
with deletions or mutations involving the long arm of chro-
mosome 22 (critical region 22q11.2). The clinical and cardio- REFERENCES
vascular manifestations are discussed earlier in this chapter
under Ectomesenchymal tissue migration abnormalities 1. Smith DW. Dysmorphology (teratology). J Pediatr 69:1150,
(neural crest migration abnormalities). 1966.
2. Ferencz C, Rubin JD, McCarter RJ, et al. Congenital heart
disease prevalence at live birth—the Baltimore-Washington
Infant Study. Am J Epidemiol 121:31, 1985.
ASSOCIATIONS 3. Marden PM, Smith DW, McDonald MJ. Congenital anom-
alies in the newborn infant including minor variants.
An association is a nonrandom occurrence of multiple J Pediatr 64:357, 1965.
anomalies not known to be a developmental field defect, 4. Greenwood RD, Rosenthal A, Parisi L, et al. Extracardiac
sequence, or syndrome. The definition refers solely to statis- abnormalities in infants with congenital heart disease.
tically (not pathogenetically or causally) related anomalies. Pediatrics 55:485, 1975.
A given association, such as the VATER association, can 5. Lammer EJ, Chen DT, Hoar RM, et al. Retinoic acid embry-
occur in isolation or as part of a broader pattern of malfor- opathy. N Engl J Med 313:837, 1985.
mation, such as in trisomy 18 syndrome or infants born to 6. Clarke LA (Updated 6 August 2004). Mucopolysaccharidosis
diabetic mothers. type I. In GeneReviews at GeneTests: Medical Genetic
Information Resource (database online). Seattle: University
VATER Association. The VATER association refers to
of Washington, 1997-2005. Available at http://www.
the nonrandom occurrence of defects, including vertebral genetests.org. Accessed 13 March 2005.
defects, anal atresia, tracheoesophageal fistula with 7. Clark EB. Growth, morphogenesis, and function: the
esophageal atresia, and radial and renal dysplasia. The dynamics of cardiovascular development. In Moller JM,
spectrum has been extended to include cardiac defects, Neal WA (eds.). Fetal, Neonatal, and Infant Heart Disease.
single umbilical artery, and growth deficiency of prenatal New York, Appleton-Century-Crofts, 1989, pp 1–22.
onset. Although ventricular septal defects are most 8. Rose V, Clark E. Etiology of congenital heart disease.
common, a wide variety of cardiac lesions has been seen. In Freedom RM, Benson LN, Smallhorn JF (eds). Neonatal
The etiology of this pattern of malformation is not known. Heart Disease. London: Springer-Verlag, 1992, pp 3–17.
Generally, it has occurred sporadically in an otherwise 9. Clark EB. Pathogenetic mechanisms of congenital cardiovas-
normal family. Identification of VATER association defects cular malformations revisited. Semin Perinatol 20:465, 1996.
10. Boughman JA, Berg KA, Astemborski JA, et al. Familial risks
in a particular patient does not, in itself, imply a diagnosis.
of congenital heart defect assessed in a population-based
Rather, the presence of one or more VATER-associated epidemiologic study. Am J Med Genet 26:839, 1987.
malformations should alert the clinician to the presence of 11. Kirby ML, Gale TF, Stewart DE. Neural crest cells
other VATER defects. contribute to normal aorticopulmonary septation. Science
CHARGE Association. The CHARGE association is a 220:1059, 1983.
nonrandom association of congenital anomalies including 12. Kirby ML, Turnage KL, Hays BM. Characterization of
coloboma, heart disease, atresia choanae, retarded growth conotruncal malformations following ablation of “cardiac”
and development and/or central nervous system anom- neural crest. Anat Rec 213:87, 1985.
alies, genital anomalies and/or hypogonadism, and ear 13. Kirby ML, Bockman DR. Neural crest and normal develop-
anomalies and/or deafness. Heart defects have been ment. A new perspective. Anat Rec 209:1, 1984.
described in 50% to 70% of the patients. Of those with 14. Driscoll DA, Budarf ML, Emanuel BS. A genetic etiology
for DiGeorge syndrome: consistent deletions and microdele-
congenital heart disease, 42% had conotruncal anomalies
tions of 22q11. Am J Hum Genet 50:924, 1992.
(tetralogy of Fallot, double-outlet right ventricle, truncus 15. Driscoll DA, Spinner NB, Budarf ML, et al. Deletions and
arteriosus) and 36% had aortic arch anomalies (vascular ring, microdeletions of 22q11.2 in velo-cardio-facial syndrome.
aberrant subclavian artery, interrupted aortic arch). Other Am J Med Genet 44:261, 1992.
defects include patent ductus arteriosus, atrioventricular 16. Scambler PJ, Carey AH, Wyse RKH, et al. Microdeletions
canal, ventricular septal defect, and atrial septal defect. within 22q11 associated with sporadic and familial DiGeorge
Most patients have some degree of mental deficiency syndrome. Genomics 10:201, 1991.
70 Dysmorphology

17. Scambler PJ, Kelly D, Lindsay E, et al. Velo-cardio-facial of fetuses with nuchal cystic hygromas, hydrops fetalis, and
syndrome associated with chromosome 22 deletions encom- female genitalia. Pediatrics 81:445, 1988.
passing the DiGeorge locus. Lancet 339:1138, 1992. 36. Benacerraf BR, Saltzman DH, Sanders SP. Sonographic sign
18. Wilson DI, Goodship JA, Burn J, et al. Deletions within suggesting the prenatal diagnosis of coarctation of the aorta.
chromosome 22q11 in familial congenital heart disease. J Ultrasound Med 8:65, 1989.
Lancet 340:573, 1992. 37. Thomas T, Yamagishi H, Overbeek PA, et al. The bHLH
19. Goldmuntz E, Driscoll D, Budarf ML, et al. Microdeletions factors, dHAND and eHAND, specify pulmonary and
of chromosomal region 22q11 in patients with congenital systemic cardiac ventricles independent of left-right sided-
conotruncal cardiac defects. J Med Genet 30:807, 1993. ness. Dev Biol 196:228, 1998.
20. Goldmuntz E, Clark BJ, Mitchell LE, et al. Frequency of 38. Schroeder JA, Jackson LF, Lee DC, et al. Form and function
22q11 deletions in patients with conotruncal defects. J Am of developing heart valves: coordination by extracellular
Coll Cardiol 32:499, 1998. matrix and growth factor signaling. J Mol Med 81:392,
21. Lindsay EA, Botta A, Jurecic V, et al. Congenital heart 2003.
disease in mice deficient for the DiGeorge syndrome region. 39. Kurnit DM, Aldridge JF, Matsuoka R, et al. Increased adhe-
Nature 401:379, 1999. siveness of trisomy 21 cells and atrioventricular canal malfor-
22. Jerome LA, Papaioannou VE. DiGeorge syndrome pheno- mations in Down syndrome: a stochastic model. Am J Med
type in mice mutant for the T-box gene, Tbx1. Nat Genet Genet 20:385, 1985.
27:286, 2001. 40. Kurnit DM, Layton WM, Matthysse S. Genetics, chance,
23. Lindsay EA, Vitelli F, Su H, et al. Tbx1 haploinsufficiency in and morphogenesis. Am J Hum Genet 41:979, 1987.
the DiGeorge syndrome region causes aortic arch defects in 41. Jongewaard IN, Lauer RM, Behrendt DA, et al. Beta 1 inte-
mice. Nature 410:97, 2001. grin activation mediates adhesive differences between
24. Merscher S, Funke B, Epstein JA, et al. TBX1 is responsible trisomy 21 and non-trisomic fibroblasts on type VI collagen.
for cardiovascular defects in velo-cardio-facial/DiGeorge Am J Med Genet 109:298, 2002.
syndrome. Cell 104:619, 2001. 42. Maslen CL. Molecular genetics of atrioventricular septal
25. Yagi H, Furutani Y, Hamada H, et al. Role of TBX1 in human defects. Curr Opin Cardiol 19:205, 2004.
del22q11.2 syndrome. Lancet 362:1366, 2003. 43. Rupp PA, Fouad GT, Egelston CA, et al. Identification,
26. Baldini A. DiGeorge syndrome: an update. Curr Opin genomic organization and mRNA expression of CRELD1,
Cardiol 19:201, 2004. the founding member of a unique family of matricellular
27. Burn J, Wilson DI, Cross I, et al. The clinical significance of proteins. Gene 293:47, 2002.
22q11 deletion. In Clark EB, Markwald RR, Takao A (eds). 44. Robinson SW, Morris CD, Goldmuntz E, et al. Missense
Developmental Mechanisms of Heart Disease. Armonk, NY: mutations in CRELD1 are associated with cardiac atrioven-
Futura Publishing, 1995, pp 559–567. tricular septal defects. Am J Hum Genet 72:1047, 2003.
28. Daw SCM, Taylor C, Kraman M, et al. A common region of 45. Jiao K, Kulessa H, Tompkins K, et al. An essential role of
10p deleted in DiGeorge and velocardiofacial syndromes. Bmp4 in the atrioventricular septation of the mouse heart.
Nat Genet 13:458, 1996. Genes Dev 15:1, 2003.
29. Ryan AK, Goodship JA, Wilson DI, et al. Spectrum of 46. Garg V, Kathiriya IS, Barnes R, et al. GATA4 mutations cause
clinical features associated with interstitial chromosome human congenital heart defects and reveal an interaction
22q11 deletions: a European collaborative study. J Med with TBX5. Nature 424:443, 2003.
Genet 34:798, 1997. 47. Pexieder T. Cell death in the morphogenesis and teratogen-
30. McElhinney DB, Driscoll DA, Emanuel BS, et al. esis of the heart. Adv Anat Embryol Cell Biol 51:1, 1975.
Chromosome 22q11 deletion in patients with truncus arte- 48. James TN. Normal and abnormal consequences of apoptosis
riosus. Pediatr Cardiol 24:569, 2003. in the human heart. Annu Rev Physiol 60:309, 1998.
31. McElhinney DB, Driscoll DA, Levin ER, et al. 49. Van Mierop LHS, Gessner IH. Pathogenetic mechanisms in
Chromosome 22q11 deletion in patients with ventricular congenital cardiovascular malformations. Prog Cardiovasc Dis
septal defect: frequency and associated cardiovascular anom- 15:67, 1972.
alies. Pediatrics 112:472, 2003. 50. Ben-Shachar G, Arcilla R, Lucas RV, et al. Ventricular
32. McElhinney DB, Clark BJ, Weinberg PM, et al. Association trabeculations in the chick embryo and their contribution to
of chromosome 22q11 deletion with isolated anomalies of the ventricular and muscular septal development. Circ Res
aortic arch laterality and branching. J Am Coll Cardiol 57:759, 1985.
37:114, 2001. 51. Neill CA. Development of the pulmonary veins with refer-
33. Van Mierop LHS, Alley RD, Kausel HW, et al. Pathogenesis ence to embryology of anomalies of pulmonary venous
of transposition complexes, I: embryology of the ventricles return. Pediatrics 18:880, 1956.
and great arteries. Am J Cardiol 12:216, 1963. 52. Clark EB, Martini DR, Rosenquist GC. Spectrum of
34. Harh JY, Paul MH, Gallen WJ, et al. Experimental produc- pulmonary venous connections following lung bud inversion
tion of hypoplastic left heart syndrome in the chick embryo. in the chick embryo. In Pexieder T (ed). Perspectives in
Am J Cardiol 10:127, 1986. Cardiovascular Research, Mechanisms of Cardiac
35. Lacro RV, Jones KL, Benirschke K. Pathogenesis of coarcta- Morphogenesis and Teratogenesis, Vol 5. New York, Raven
tion of the aorta in the Turner syndrome: a pathologic study Press, 1981, pp 419–430.
Dysmorphology and Genetics 71
53. Bleyl S, Nelson L, Odelberg SJ, et al. A gene for familial total 73. Whittemore R, Hobbins JC, Engle MA. Pregnancy and its
anomalous pulmonary venous return maps to chromosome outcome in women with and without surgical treatment of
4p13-q12. Am J Hum Genet 56:408, 1995. congenital heart disease. Am J Cardiol 50:641, 1982.
54. Bleyl SB, Saijoh Y, Klewer SE, et al. Evidence that PDGF 74. Rose V, Gold RJ, Lindsay G, et al. A possible increase in the
signaling is disrupted in human anomalies of the pulmonary incidence of congenital heart defects among the offspring of
veins. (Abstract 1046, presented at American Society for affected parents. J Am Coll Cardiol 6:376, 1985.
Human Genetics meeting, October 2004, Toronto). 75. Nora JJ, Nora AH. Update on counseling the family with a
55. Belmont JW, Mohapatra B, Towbin JA, et al. Molecular first-degree relative with a congenital heart defect. Am J
genetics of heterotaxy syndromes. Curr Opin Cardiol 19:216, Med Genet 29:137, 1988.
2004. 76. Nora JJ. Causes of congenital heart diseases: old and
56. Yost HJ. The genetics of midline and cardiac laterality new modes, mechanisms, and models. Am Heart J 125:1409,
defects. Curr Opin Cardiol 13:185, 1998. 1993.
57. Casey B, Devoto M, Jones KL, et al. Mapping a gene for 77. Whittemore R, Wells JA, Castellsague X. A second-genera-
familial situs abnormalities to human chromosome Xq24-q27.1. tion study of 427 probands with congenital heart defects and
Nat Genet 5:403, 1993. their 837 children. J Am Coll Cardiol 23:1459, 1994.
58. Gebbia M, Ferrero GB, Pilia G, et al. X-linked situs abnor- 78. Disegni E, Pierpont ME, Bass JL, et al. Two-dimensional
malities result from mutations in ZIC3. Nat Genet 17:305, echocardiographic identification of endocardial cushion
1997. defect in families. Am J Cardiol 55:1649, 1985.
59. Brueckner M, McGrath J, D’Eustachio P, et al. Establishment 79. Brenner JI, Berg KA, Schneider DS, et al. Cardiac malfor-
of left-right asymmetry in vertebrates: genetically distinct mations in relatives of infants with hypoplastic left-heart
steps are involved in biological asymmetry and handedness. syndrome. Am J Dis Child 140:41, 1986.
Ciba Found Symp 162:202, 1991. 80. Lewin MB, McBride KL, Pignatelli R, et al. Echocardiographic
60. Icardo JM, de Vega S. Spectrum of heart malformations in evaluation of asymptomatic parental and sibling cardiovascu-
mice with situs solitus, situs inversus, and associated visceral lar anomalies associated with congenital left ventricular
heterotaxy. Circulation 84:2547, 1991. outflow tract lesions. Pediatrics 114:691, 2004.
61. Brueckner M, D’Eustachio P, Horwich AL. Linkage 81. Malik MSA, Watkins H. The molecular genetics of hyper-
mapping of a mouse gene, iv, that controls left-right asymme- trophic cardiomyopathy. Curr Opin Cardiol 12:295, 1997.
try of the heart and viscera. Proc Natl Acad Sci U S A 82. Vincent GM. Role of DNA testing for diagnosis, manage-
86:5035, 1989. ment, and genetic screening in long QT syndrome, hyper-
62. Supp DM, Witte DP, Potter SS, et al. Mutation of an axone- trophic cardiomyopathy, and Marfan syndrome. Heart 86:12,
mal dynein affects left-right asymmetry in inversus viscerum 2002.
mice. Nature 389:963, 1997. 83. Seidman JG, Seidman C. The genetic basis for cardiomyopa-
63. Afzelius BA. A human syndrome caused by immotile cilia. thy. Cell 104:557, 2001.
Science 193:317, 1976. 84. Mestroni L, Giacca M. Molecular genetics of dilated
64. Teichberg S, Markowitz J, Silverberg M, et al. Abnormal cilia cardiomyopathy. Curr Opin Cardiol 12:303, 1997.
in a child with the polysplenia syndrome and extrahepatic 85. Familial Dilated Cardiomyopathy Project Group. Available
biliary atresia. J Pediatr 100:399, 1982. at http://www.fdc.to.
65. Schidlow DV, Moriber Katz S, Turtz MG, et al. Polysplenia 86. Wang Q, Chen Q, Li H, Towbin JA. Molecular genetics of
and Kartagener syndromes in a sibship: association with long QT syndrome from genes to patients. Curr Opin
abnormal respiratory cilia. J Pediatr 100:401, 1982. Cardiol 12:310, 1997.
66. Lin AE, Garver KL. Genetic counseling for congenital heart 87. Lin AE. Congenital heart defects in malformation
defects. J Pediatr 113:1105, 1988. syndromes. Clin Perinatol 17:641, 1990.
67. Strauss AW, Johnson MC. The genetic basis of pediatric 88. Ferencz C, Neill CA, Boughman JA, et al. Congenital
cardiovascular disease. Semin Perinatol 20:564, 1996. cardiovascular malformations associated with chromosome
68. Lewin MB, Glass IA, Power P. Genotype-phenotype correlation abnormalities: an epidemiologic study. J Pediatr 114:79,
in congenital heart disease. Curr Opin Cardiol 19:221, 2004. 1989.
69. Gelb BD. Genetic basis of congenital heart disease. Curr 89. Pierpont MEM, Moller JH (eds). Genetics of Cardiovascular
Opin Cardiol 19:110, 2004. Disease. Boston: Martinus Nijhoff, 1987.
70. McElhinney DB, Geiger E, Blinder J, et al. NKX2.5 muta- 90. Borgaonkar DS. Chromosomal Variation in Man: A Catalog
tions in patients with congenital heart disease. J Am Coll of Chromosomal Variants and Anomalies. New York: Wiley-
Cardiol 42:1650, 2003. Liss, 1997.
71. Elliot DA, Kirk EP, Yeoh T, et al. Cardiac homeobox gene 91. Jones KL. Smith’s Recognizable Patterns of Human
NKX2-5 mutations and congenital heart disease: associations Malformation. Philadelphia: WB Saunders, 1997.
with atrial septal defect and hypoplastic left heart syndrome. 92. Van Praagh S, Truman T, Firpo A, et al. Cardiac malforma-
J Am Coll Cardiol 41:2072, 2003. tions in trisomy 18: a study of 41 postmortem cases. J Am
72. Brenner JI, Berg KA, Schneider DS, et al. Cardiac malfor- Coll Cardiol 13:1586, 1989.
mations in relatives of infants with hypoplastic left-heart 93. Allen DB, Hendricks SA, Levy JM. Aortic dilation in Turner
syndrome. Am J Dis Child 143:1492, 1989. syndrome. J Pediatr 109:302, 1986.
72 Dysmorphology

94. Lin AE, Lippe BM, Geffner ME, et al. Aortic dilation, 113. Krantz ID, Colliton RP, Genin A, et al. Spectrum and
dissection, and rupture in patients with Turner syndrome. frequency of jagged1 (JAG1) mutations in Alagille
J Pediatr 109:820, 1986. syndrome patients and their families. Am J Hum Genet
95. Jones KL, Smith DW, Ulleland CN, et al. Pattern of malfor- 62:1361, 1998.
mation of offspring of chronic alcoholic mothers. Lancet 114. McElhinney DB, Krantz ID, Bason L. Analysis of cardio-
1:1267, 1973. vascular phenotype and genotype-phenotype correlation in
96. Milewicz DM. Molecular genetics of Marfan syndrome individuals with JAG1 mutation and/or Alagille syndrome.
and Ehlers-Danlos type IV. Curr Opin Cardiol 13:198, Circulation 106:2567, 2002.
1998. 115. Ellis RWB, van Creveld S. A syndrome characterized by
97. McKusick VA. The defect in Marfan syndrome. Nature ectodermal dysplasia, polydactyly, chondro-dysplasia and
352:279, 1991. congenital morbus cordis: report of three cases. Arch Dis
98. Gray JR, Davies SJ. Marfan syndrome. J Med Genet 33:403, Child 15:65, 1940.
1996. 116. Polymeropoulos MH, Ide SE, Wright M, et al. The gene for
99. Braverman AC. Timing of aortic surgery in the Marfan the Ellis-van Creveld syndrome is located on chromosome
syndrome. Curr Opin Cardiol 19:549, 2004. 4p16. Genomics 35:1, 1996.
100. Mendez HMM, Opitz JM. Noonan syndrome: a review. 117. Ruiz-Perez VL, Ide SE, Strom TM, et al. Mutations in a
Am J Med Genet 21:493, 1985. new gene in Ellis-van Creveld syndrome and Weyers acro-
101. Allanson JE. Noonan syndrome. J Med Genet 24:9, 1987. dental dysostosis. Nat Genet 24:283, 2000.
102. Allanson JE, Hall JG, Hughes HE, et al. Noonan syndrome: 118. Ruiz-Perez VL, Tompson SW, Blair HJ, et al. Mutations in
the changing phenotype. Am J Med Genet 21:507, 1985. two nonhomologous genes in a head-to-head configuration
103. Jamieson CR, van der Burgt I, Brady AF, et al. Mapping a gene cause Ellis-van Creveld syndrome. Am J Hum Genet
for Noonan syndrome to the long arm of chromosome 12. Nat 72:728, 2003.
Genet 8:357, 1994. 119. Williams JCP, Barratt-Boyes BG, Lowe JB. Supravalvular
104. Tartaglia M, Mehler EL, Goldberg R, et al. Mutations in aortic stenosis. Circulation 24:1311, 1961.
PTPN11 encoding the protein tyrosine phosphatase SHP-2, 120. Burn J: Williams syndrome. J Med Genet 23:389, 1986.
cause Noonan syndrome. Nat Genet 29:465, 2001. 121. Morris CA: Genetic aspects of supravalvular aortic stenosis.
105. Tartaglia M, Kalidas K, Shaw A, et al. PTPN11 mutations in Curr Opin Cardiol 13:214, 1998.
Noonan syndrome: molecular spectrum, genotype pheno- 122. Ewart AK, Morris CA, Atkinson D, et al. Hemizygosity at
type correlation, and phenotypic heterogeneity. Am J Hum the elastin locus in a developmental disorder, Williams
Genet 70:1555, 2002. syndrome. Nat Genet 5:11, 1993.
106. Basson CT, Cowley GS, Solomon SD, et al. The clinical and 123. Lowery MC, Morris CA, Ewart A, et al. Strong correlation
genetic spectrum of the Holt-Oram syndrome (heart-hand of elastin deletions, detected by FISH, with Williams
syndrome). N Engl J Med 330:885, 1994. syndrome: evaluation of 235 patients. Am J Hum Genet
107. Li QY, Newbury-Ecob RA, Terrett JA, et al. Holt-Oram 57:49, 1995.
syndrome is caused by mutations in TBX5, a member of the 124. Curran ME, Atkinson DL, Ewart AK, et al. The elastin
Brachyury (T) gene family. Nat Genet 15:21, 1997. gene is disrupted by a translocation associated with
108. Basson CT, Bachinsky DR, Lin RC, et al. Mutations in supravalvular aortic stenosis. Cell 73:159, 1993.
human cause limb and cardiac malformation in Holt-Oram 125. Ewart AK, Jin W, Atkinson D, et al. Supravalvular aortic
syndrome. Nat Genet 15:30, 1997. stenosis associated with a deletion disrupting the elastin
109. Mori AD, Bruneau BG. TBX5 mutations and congenital gene. J Clin Invest 93:1071, 1994.
heart disease: Holt-Oram syndrome revealed. Curr Opin 126. Urban Z, Michels VV, Thibodeau SN, et al. Supravalvular
Cardiol 19:211, 2004. aortic stenosis (SVAS): predominance of truncating point
110. Krantz ID, Piccoli DA, Spinner NB. Alagille syndrome. mutations within the elastin gene. Am J Hum Genet
J Med Genet 34:152, 1997. 63:A390, 1998.
111. Oda T, Elkahloun AG, Pike BL, et al. Mutations in the 127. Vissers LE, van Ravenswaaij CM, Admiraal R, et al.
human Jagged1 gene are responsible for Alagille syndrome. Mutations in a new member of the chromodomain gene
Nat Genet 16:235, 1997. family cause CHARGE syndrome. Nat Genet 36:955, 2004.
112. Li L, Krantz ID, Deng Y, et al. Alagille syndrome is caused
by mutations in human Jagged1, which encodes a ligand for
Notch1. Nat Genet 16:243, 1997.

You might also like