Está en la página 1de 18

Congenital Heart Disease Compendium

Circulation Research Compendium on Congenital Heart Disease


Congenital Heart Disease: The Remarkable Journey From the “Post-Mortem Room” to Adult Clinics
Perspective on Congenital Heart Disease Research
Has the Congenitally Malformed Heart Changed Its Face? Journey From Understanding Morphology to Surgical Cure in
Congenital Heart Disease
Impact of Three-Dimensional Printing on the Study and Treatment of Congenital Heart Disease
Changing Landscape of Congenital Heart Disease
Genetics and Genomics of Congenital Heart Disease
Cardiac Regeneration: Lessons From Development
Neurodevelopmental Abnormalities and Congenital Heart Disease: Insights Into Altered Brain Maturation
Heart Failure in Pediatric Patients With Congenital Heart Disease
Noninvasive Imaging in Adult Congenital Heart Disease
Current Status and Future Potential of Transcatheter Interventions in Congenital Heart Disease
Current Interventional and Surgical Management of Congenital Heart Disease: Specific Focus on Valvular Disease and
Cardiac Arrhythmias
Downloaded from http://circres.ahajournals.org/ by guest on June 24, 2018

Ali J. Marian, Editor

Heart Failure in Pediatric Patients With Congenital


Heart Disease
Robert B. Hinton, Stephanie M. Ware

Abstract: Heart failure (HF) is a complex clinical syndrome resulting from diverse primary and secondary causes
and shared pathways of disease progression, correlating with substantial mortality, morbidity, and cost. HF in
children is most commonly attributable to coexistent congenital heart disease, with different risks depending on
the specific type of malformation. Current management and therapy for HF in children are extrapolated from
treatment approaches in adults. This review discusses the causes, epidemiology, and manifestations of HF in
children with congenital heart disease and presents the clinical, genetic, and molecular characteristics that are
similar or distinct from adult HF. The objective of this review is to provide a framework for understanding rapidly
increasing genetic and molecular information in the challenging context of detailed phenotyping. We review clinical
and translational research studies of HF in congenital heart disease including at the genome, transcriptome, and
epigenetic levels. Unresolved issues and directions for future study are presented.  (Circ Res. 2017;120:978-994.
DOI: 10.1161/CIRCRESAHA.116.308996.)
Key Words: cardiovascular malformation ■ genetics ■ mutation ■ single ventricle ■ stem cell ■ ventricular dysfunction

T he International Society for Heart and Lung Transplantation


defines pediatric heart failure (HF) as a clinical and patho-
physiologic syndrome that results from ventricular dysfunc-
heart disease (CHD) is frequently associated with ventricular
dysfunction, volume, or pressure overload. HF in pediatric pa-
tients with CHD has various causes, some of which overlap
tion, volume, or pressure overload, alone or in combination. with the causes of cardiomyopathy, resulting in both distinct
In children, it leads to characteristic signs and symptoms, such and shared mechanisms leading to ventricular dysfunction and
as poor growth, feeding difficulties, respiratory distress, exer- the clinical manifestation of HF (Figure 1). In this review, we
cise intolerance, and fatigue and is associated with circulato- focus on basic, translational, and clinical research as it applies
ry, neurohormonal, and molecular abnormalities.1 Congenital to HF in pediatric patients with CHD.

Original received October 17, 2016; revision received December 27, 2016; accepted December 28, 2016.
From the Department of Pediatrics and Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis.
Correspondence to Stephanie M. Ware, MD, PhD, Indiana University School of Medicine, Herman B Wells Center for Pediatric Research, 1044 W
Walnut St, R4-227, Indianapolis, IN 46202. E-mail stware@iu.edu
© 2017 American Heart Association, Inc.
Circulation Research is available at http://circres.ahajournals.org DOI: 10.1161/CIRCRESAHA.116.308996

978
Hinton and Ware   Heart Failure in Congenital Heart Disease   979

blood.2 The criteria for the diagnosis of HF are largely clini-


Nonstandard Abbreviations and Acronyms
cal, and several standardized diagnostic classification systems
AVSD atrioventricular septal defect have been proposed.3 Pediatric HF encompasses a broader
CHD congenital heart disease
range of associated findings than adult HF in part because of
the variety of ages of presentation. As a clinical entity, HF
DCM dilated cardiomyopathy
is further subdivided based on its onset, acuity, and severity.
ECM extracellular matrix
The well-known New York Heart Association Heart Failure
HCM hypertrophic cardiomyopathy
Classification does not apply well to children and on a practi-
HF heart failure
cal level is thought to lack the sensitivity needed to assess and
HLHS hypoplastic left heart syndrome
capture the progression of HF severity in children. For this
LV left ventricle
reason, the Ross Heart Failure Classification was developed
LVNC left ventricular noncompaction for the assessment of infants with HF, and a modified Ross
lncRNA long noncoding RNA Classification was developed to apply to additional age ranges
MFS Marfan syndrome of children.4 The New York University Pediatric Heart Failure
miRNA microRNA Index provides a weighted score (Table 1).5 In pediatric prac-
NSML Noonan syndrome with multiple lentigines tice, these classification systems are not widely used. As dis-
PA/IVS pulmonary atresia with intact ventricular septum cussed in subsequent sections, this creates specific challenges
PHN Pediatric Heart Network for research.
The clinical presentation of CHD is described primarily
Downloaded from http://circres.ahajournals.org/ by guest on June 24, 2018

RV right ventricle
SV single ventricle in physiological terms, for example, outflow tract obstruction
VSD ventricular septal defect (pressure overload) or pulmonary over-circulation (volume
overload). The treatment of CHD is typically surgical correc-
tion (anatomic), and because HF lacks specificity in this con-
text, the term ventricular dysfunction is often used. However,
Diagnosis and Epidemiology
ventricular dysfunction may be associated with poor contractil-
Defining HF in Children With CHD Presents ity (systolic dysfunction) or poor relaxation (diastolic dysfunc-
Significant Challenges tion), with or without the clinical presence of HF, thus creating
HF is defined by the American Heart Association and a semantic challenge that spans clinical and research efforts in
American College of Cardiology as a complex clinical syn- pediatric cardiology and cardiothoracic surgery. Considerable
drome that can result from any structural or functional cardiac practice variation across centers significantly confounds this
disorder that impairs the ability of the ventricle to fill or eject problem. Even large multicenter groups, including the Pediatric

Figure 1. Model of the relationship of


heart failure (HF) to congenital heart
disease (CHD) and cardiomyopathy.
Three phenotypes are shown: CHD
(yellow), CM (blue), and CHD with CM
(overlap). Cardiomyopathy by definition
results in ventricular dysfunction in 100%
of cases. CHD results in ventricular
dysfunction in ≈20% of cases, whereas
coexisting CHD and CM results in
ventricular dysfunction in 100% of cases.
HF occurs in ≈65%, 6%, and ≈40%,
respectively. Ventricular dysfunction,
either systolic or diastolic, precedes HF
and therefore offers opportunities for
early intervention. Causes of HF may
vary despite common clinical features
(different colored arrows). The clinical
time course (left) and associated causes
and mechanisms (right) suggest that
identifying distinct mechanisms in CM
vs CHD provides opportunities for early
intervention.
980  Circulation Research  March 17, 2017

Table 1.  Classification Approaches to Heart Failure


NYHA Modified Ross NYU PHFI*
I asymptomatic I asymptomatic 0–7 asymptomatic to minimal signs and
symptoms
II slight or moderate limitation of II mild tachypnea or diaphoresis with feeding in infants; dyspnea on 8–14 moderate signs and symptoms
physical activity exertion in older children
III marked limitation of physical activity III marked tachypnea or diaphoresis with feeding in infants; marked 15–21 advanced signs and symptoms and
dyspnea on exertion; prolonged feeding times with growth failure modest use of medication
IV symptoms at rest IV tachypnea, retractions, grunting, or diaphoresis at rest 22–30 advanced signs and symptoms and
liberal use of medicine
*HF severity is determined from signs and symptoms, medical regimen, and ventricular physiology. A total score can range from zero (no heart failure) to 30 (severe
heart failure) and is derived by adding 1 or 2 points for each of 13 individual criterion.5 NYHA indicates New York Heart Association; and NYU PHFI, New York University
Pediatric Heart Failure Index.

Heart Network (PHN), Society of Thoracic Surgeons, Pediatric in the United States, but 2 single-site studies in Europe indicate
Cardiomyopathy Registry, Pediatric Cardiac Genomics that more than half of the pediatric HF cases were in children
Consortium, and National Pediatric Cardiology Quality with CHD.14,15 In these studies, there were differences in the
Improvement Collaborative, use different approaches to iden- rate of HF in their CHD populations, with 1 study identifying
Downloaded from http://circres.ahajournals.org/ by guest on June 24, 2018

tify HF and classify severity, creating unintended inconsisten- HF in 10.4% of all patients with congenital and acquired heart
cies in the literature, ultimately resulting in studies that are disease and the other identifying HF in 34%, suggesting dif-
difficult to reconcile. For example, the PHN, the field’s premier ferences in study design or HF definitions.14,15 Within the CHD
clinical network, has used different approaches to classifying populations, the rate of HF was 6.2% and 39%, respectively.
cardiac dysfunction or HF in different studies, such as compar-
ing function between groups without distinguishing dysfunc- CHD Is the Most Common Cause of HF in Children
tion,6 using the Ross criteria,7 or focusing on imaging evidence HF has numerous causes that are a consequence of cardiac
of dysfunction without commenting on HF.8 Although there are and noncardiac disorders, either congenital or acquired.1 In
valid reasons for these specific differences in study design, the the pediatric population, rheumatic fever was the most com-
lack of a unified approach to ventricular dysfunction and HF mon cause of HF in children in the United States in the 1950s
confounds integrated analyses of the collective literature. The and continues to be a common cause of pediatric HF in devel-
small population sizes available for each specific type of CHD oping countries (Table 2). Traditionally, HF has been synony-
further hamper the ability to make generalizable observations. mous with cardiomyopathy in the literature of pediatric heart
Together, these observations illustrate that primary challenges disease, but over time, it became clear that cardiomyopathy
in the field are overcoming practice variation and standardizing is but one cause of HF. Although the proportion of CHD pa-
phenotyping. Nevertheless, it is important to recognize that it is tients with HF is lower than the proportion with rhythm dis-
feasible to work toward a solution using the multiple existing turbances or cardiomyopathy, CHD is a much more common
networks and consortia to optimize the use of data already col- disease and therefore contributes a greater number of cases
lected in these large cohorts. to the overall HF count. Nearly 60% of HF cases in pediatric
patients occurred within the first year of life, but in these stud-
Epidemiology of Pediatric HF Is Poorly Understood ies, the overall mortality was lower in the CHD population
Factors that have contributed to difficulties in compiling accu- than in patients with HF from other causes.14,15 The fact that
rate incidence and prevalence estimates for pediatric HF include the risk of HF varied depending on the underlying cause raises
the lack of standardized phenotyping for both CHD and HF and the fundamental question of whether HF is the same disease
the diversity of causes of HF in children with CHD. HF has process across the spectrum of age ranges and precipitating
been a major public health problem for decades. In the United causes. In addition, it suggests that there may be the potential
States, >550 000 new cases are diagnosed each year, and the for risk stratification and customization of therapy.
overall prevalence is >6 million individuals.2 Pediatric HF con- HF is a common morbidity identified in the adult CHD
tributes substantially to the economic impact of this disease as a population, an age range complicated by additional factors
result of the frequent need for procedure-based intervention and and not covered in many pediatric studies. HF is known to
the significant morbidity and mortality.9 Children whose hospi- occur in ≈25% of adult CHD patients by the age of 30, and the
talizations are complicated by HF have over a 20-fold increased incidence increases with age.16 Tracking the natural history of
risk of death.10 It is estimated that 11 000 to 14 000 children will specific CHD types or specific genetic causes, and comparing
be hospitalized with HF annually in the United States.10,11 These the similarities and differences of HF in children and adults,
numbers include children with cardiomyopathy, with an annual promises to provide insight into the risk of HF at the time
incidence estimated at 1.13 cases per 100 000 children,12 and of diagnosis in childhood. Taken together, these data indicate
children with CHD, with a traditionally cited incidence of 8 per that HF is an important cause of morbidity and mortality in
1000 live births and a need for cardiac intervention in 3 of ev- pediatric and adult CHD, and HF in this population is hetero-
ery 1000 newborns.13 There have been no comprehensive epi- geneous with regard to underlying cause and outcome. The
demiological studies addressing HF in the pediatric population issues emerging in the management of the growing adult CHD
Hinton and Ware   Heart Failure in Congenital Heart Disease   981

Table 2.  Causes of Heart Failure in Children mutations in developmental signaling pathways long known
to be associated with CHD such as the Notch pathway and
Congenital heart Left to right shunts (eg, VSD)
disease noncanonical Wnt signaling also cause LVNC in animal mod-
Conotruncal lesions (eg, TOF) els and humans.26–28 Given the developmental importance of
Single ventricle lesions (eg, HLHS) these signaling pathways for cardiogenesis, future research
Valve disease (eg, AS) investigating the shared developmental mechanisms leading
to both CHD and cardiomyopathy from disruption in these
Cardiomyopathy HCM
pathways will likely be informative.
(primary)
DCM In addition to the MYH7 gene, mutations in other sar-
RCM comeric genes can cause both CHD and cardiomyopathy.
Mutations in MHY6, encoding α-myosin heavy chain, cause
ARVC
hypertrophic cardiomyopathy (HCM), dilated cardiomyopa-
LVNC thy (DCM), familial atrial septal defect, and sick sinus syn-
Arrhythmia Tachycardias (eg, supraventricular tachycardia) drome.29 Similarly, mutations in ACTC1, encoding α-cardiac
Bradycardias (eg, complete AV block) actin, a component of the thin filament of the sarcomere, can
cause cardiomyopathy with or without septal defects.30,31
Infection Myocarditis
Mutations in MYBPC3 and TNNI3 have also been described
Acute rheumatic fever with both CHD and cardiomyopathy.32 Surprisingly, sarco-
Downloaded from http://circres.ahajournals.org/ by guest on June 24, 2018

HIV meric gene mutation analysis has not been applied to the CHD
population in a comprehensive manner to determine whether
Ischemia Coronary anomaly (eg, myocardial bridge)
rare variants or common polymorphisms in these genes might
Kawasaki disease be associated with ventricular function. Whether patients who
Toxin Chemotherapy have CHD with myocardial dysfunction that is out of propor-
Maternal drugs during pregnancy
tion to their heart defect might have a primary heart muscle
disease is unknown. Studies to analyze the natural history of
Other Diabetes mellitus
patients with mutations in sarcomeric genes and CHD are nec-
Essential hypertension essary to assess the penetrance of left ventricular (LV) dys-
ARVC indicates arrhythmogenic right ventricular cardiomyopathy; AS, aortic function or cardiomyopathy in these patients.
stenosis; AV, atrioventricular; DCM, dilated cardiomyopathy; HIV, human
immunodeficiency virus; HCM, hypertrophic cardiomyopathy; HLHS, hypoplastic Genetic syndromes associated With CHD Also
left heart syndrome; LVNC, left ventricular noncompaction; RCM, restrictive Cause Cardiomyopathy
cardiomyopathy; TOF, tetralogy of Fallot; and VSD, ventricular septal defect. The development of the heart is under genetic control, and
CHD is primarily a genetic disease, although teratogens may
population have been reviewed recently.17–20 Herein, we focus independently cause malformation, and maternal factors may
specifically on children with CHD and HF. contribute to disease manifestation.33,34 The genetic causes of
CHD include chromosome abnormalities, genomic disorders,
Overlapping Causes single gene causes, and multifactorial causes.34–36 Overall,
Mutations That Cause Cardiomyopathy Can Also the likelihood of identifying the genetic cause of CHD is less
Cause CHD than for cardiomyopathy (Table 3). In part, this stems from
Recent data indicate that mutations in sarcomeric genes the fact that cardiomyopathy most often illustrates Mendelian
are associated with CHD in addition to cardiomyopathy.21 inheritance in an autosomal dominant pattern. CHD, in con-
Mutations in specific protein domains of the MYH7 gene cause trast, is most often multifactorial with a complex interplay of
Ebstein anomaly in addition to causing cardiomyopathy.22 multiple genes and environment contributing a susceptibility
Interestingly, in the original description, 6% of a cohort of to the development of a structural defect.34,35 An exception to
patients with Ebstein anomaly, a defect of tricuspid valve for- this is syndromic CHD, in which the diagnostic rate is higher.
mation and position, had MYH7 mutations, of whom 75% had Genetic syndromes are often associated with specific classes
left ventricular noncompaction cardiomyopathy (LVNC). In of cardiovascular malformations, but phenotypic heteroge-
their families, the familial MYH7 mutation was also identified neity is common. In addition to this variability, these cases
in individuals with other types of CHD and in individuals with illustrate occasional nonpenetrance of cardiac defects that is
isolated LVNC but was not found in unaffected family mem- not well understood. Overall, genetic syndromes have been
bers. These cases illustrate that CHD and cardiomyopathy tremendously informative for identifying genes that cause
phenotypes occur with variable expressivity in family mem- CHD.37–41 The paradigm that understanding the genetic basis
bers carrying the same disease causing mutation, suggesting of syndromic CHD can identify important genes that cause or
that other factors, including genetic modifiers, can influence modify isolated CHD is conceptually important.
phenotypic expression. LVNC is known to be associated with Several genetic conditions with highly penetrant CHD are
specific types of CHD in a subset of cases.23–25 LVNC is char- also associated with HF and cardiomyopathy. For example,
acterized by abnormal and excessive ventricular trabeculation Noonan syndrome, an autosomal dominant genetic condi-
and a thin-walled myocardium. Whereas mutations in sarco- tion characterized by short stature, cardiac defects, and dys-
meric genes have been associated with the LVNC phenotype, morphic features, is caused by mutations in genes within the
982  Circulation Research  March 17, 2017

Table 3.  Comparison of Congenital Heart Disease and Cardiomyopathy Genetics in Children
Congenital Heart Disease Cardiomyopathy
Characteristics Syndromic and nonsyndromic (isolated) forms ≈30% and 70%, Syndromic and nonsyndromic forms ≈30% and 70%,
respectively respectively
Inheritance pattern Multifactorial>Mendelian; complex trait most common Mendelian>Multifactorial; autosomal dominant most
common
Decreased penetrance Common Less common
Variable expressivity Common Common
Diagnosis CMA, WES/WGS, NGS panels, single gene testing NGS panels
Diagnostic yield CMA: 3% to 25% in syndromic; 3% to 10% in isolated 20% to 70% depending on type of cardiomyopathy
WES/WGS: ≤10% de novo mutation rate
NGS panels: unknown
Genes Encode transcription factors, developmental signaling pathways, Encode structural proteins of the contractile apparatus, ion
chromatin remodeling, structural proteins of the contractile apparatus channels, signaling pathways, metabolic function
CMA indicates chromosome microarray; NGS, next-generation sequencing; WES, whole exome sequencing; and WGS, whole genome sequencing.
Downloaded from http://circres.ahajournals.org/ by guest on June 24, 2018

RAS/MAPK pathway.42 These patients may show evidence of I receptor signaling and loss of focal adhesion kinase activ-
HCM, CHD (classically pulmonary valve stenosis), or both.43 ity in the absence of aortic or valvular disease. An additional
There is a lifelong risk for the development of HCM with study identified 2 distinct phenotypes in aged Marfan mice,
Noonan syndrome, necessitating ongoing cardiac surveillance one of which was LV enlargement associated with elevated
even in affected individuals without CHD. Other RASopathies ERK1/2 and p38 MAPK phosphorylation and higher BNP
include Cardiofaciocutaneous syndrome, Costello syndrome, expression.48 The variable phenotypic responses noted in this
and Noonan syndrome with multiple lentigines (NSML; for- study parallel the small subset of MFS patients who develop
merly LEOPARD) syndrome. NSML is usually caused by DCM. Combined approaches using genomics, mouse models,
mutations in PTPN11, a protein tyrosine phosphatase 2 that and patient-oriented research will be required to dissect the
regulates the RAS/MAPK cascade. Mutations in PTPN11 are complex interactions that underlie this phenotypic variability.
also the most common cause of Noonan syndrome, where
they result in constitutive activation of the protein. In con- Surveillance
trast, in NSML, mutations in PTPN11 render the protein cata-
lytically impaired. Greater than 80% of patients with NSML The Development of HF in CHD Is Dependent on
have HCM that is caused by the hyperactivation of the AKT/
Age and Lesion Type
mTOR pathway. Using a mouse model of NSML, the ability Although genetic causes of CHD or cardiomyopathy can
to prevent HCM was investigated by early treatment with the identify children at high risk of developing HF, this occurs
mTOR inhibitor rapamycin. Mice treated early did not devel- in a minority of cases. Clinical factors related to phenotype
op HCM, and those treated at later stages demonstrated rever- and presentation remain the most common means for evaluat-
sal of disease.44 Recently, the first trial of an mTOR inhibitor ing risk and instituting surveillance for HF. Age is an impor-
was reported in an infant with NSML and rapidly progressive tant factor in assessing clinical features of HF. In the fetus,
HCM with a goal of halting progression of hypertrophy and HF is characterized by decreased fetal movement, pericar-
outflow tract obstruction until the time of transplant.45 An un- dial effusion, and ascites.49 In the preterm low birth weight
derstanding of the genetic basis of HF in this case led to trial newborn, HF is characterized by acidemia, anemia, and hy-
of a pathway-specific inhibitor for treatment. poxemia. In the term newborn, tachypnea, fatigue with feed-
Marfan syndrome (MFS), a connective tissue disorder ings, and decreased urine output are common symptoms of
caused by mutations in an extracellular matrix (ECM) protein HF. Decompensation at this age is often rapid, with the in-
encoded by FBN1,46 is another syndrome for which new data fant moving from asymptomatic to cardiac shock quickly in
implicate a propensity to HF in a subset of affected individu- part because of the decreased cardiac reserve at this age.50,51
als. Mutations in fibrillin-1 lead to an upregulation of trans- Accordingly, more classic signs of HF, such as edema and
forming growth factor-β signaling in individuals with MFS. pathological pulses, are less common and often not present.
The structural network of the microfibril controls the release The timing of HF is a clue to the underlying malformation.
of signaling molecules important for morphogenesis and tis- For example, HF in hypoplastic left heart syndrome (HLHS)
sue homeostasis. Case reports documenting the development develops on days 3 to 7 of life, whereas HF in severe coarcta-
of LV dysfunction or DCM in patients with MFS suggest a tion of the aorta tends to develop on days 7 to 10. Some CHD
potential overlap in molecular networks controlling cardiac types do not manifest HF until after the pulmonary vascular
function. Using a mouse model of MFS, it was determined resistance decreases, for example, a large ventricular septal
that DCM in fibrillin-1–deficient mice results from abnor- defect (VSD) or an atrioventricular septal defect (AVSD), in
mal mechanosignaling by cardiomyocytes.47 Specifically, the which case HF develops in 1 to 3 months. But other lesions,
mice spontaneously developed increased angiotensin II type such as an atrial septal defect, do not lead to symptoms until
Hinton and Ware   Heart Failure in Congenital Heart Disease   983

3 to 5 years of life, if at all. Although some of these observa- whereas in other lesions, such as HLHS, the right ventricle
tions simply reflect the underlying anatomy and physiology, (RV) is the systemic ventricle. Differences in left- and right-
it is increasingly recognized that additional factors, including sided ventricles, as well as ventricular–ventricular interactions,
both genes and environment, contribute to the timing of the are critical to understand cardiac function and performance. In
onset of disease. HLHS, the RV is dilated and hypertrophied and managing 3×
Lesion type is another factor useful for risk stratification the normal cardiac output. The morphology of the RV is not
for HF. HF in patients with CHD is typically attributed to con- built for the demands of systemic circulation. The size of the
comitant pressure or volume overload. Therefore, infants with RV is known to be important in PA/IVS, and the shape has been
specific types of CHD will universally experience HF with- shown to result in different outcomes in HLHS.8,52 It is possible
out surgical correction, for example, critical aortic stenosis that each lesion may be further distinguished by the pattern of
(pressure overload) or Ebstein anomaly with severe tricuspid specific findings, for example, whether the aortic valve is atretic
regurgitation (volume overload). However, genetic factors, or stenotic in HLHS. Careful phenotyping is important to deter-
as well as various environmental triggers, are implicated in mine whether specific subphenotypes have unique genetic and
the complex process leading to HF. Accordingly, identifying molecular causes and characteristics.
new-onset ventricular dysfunction is important for emerging Although functional SV defects have high rates of ventricu-
early intervention strategies. In general, children beyond the lar dysfunction or HF, many lesions are more difficult to gauge.
first year of life have better overall health and greater cardiac For example, predicting the need for and timing of surgery for
reserve than adults and can remain in a compensated state for valve diseases (stenosis and regurgitation) can be challenging.
Downloaded from http://circres.ahajournals.org/ by guest on June 24, 2018

longer periods, but tend to transition rapidly to acute HF. The Aortic stenosis may be an emergency in the newborn period but
proportion of cases of specific types of CHD that have ven- typically is asymptomatic in childhood until progression results
tricular dysfunction or HF at the time of diagnosis varies. For in HF. HF is also common, but not universal, in the context of
example, pulmonary insufficiency in a patient with repaired chronic severe pulmonary insufficiency after repair later in life.
tetralogy of Fallot may or may not lead to ventricular dysfunc- The manifestation of ventricular dysfunction or HF by lesion
tion and HF, and the ability to predict these events is limited. is also variable. Left to right shunts include complete AVSDs,
The higher the rate of HF at diagnosis, the higher the risk for which uniformly develop refractory HF within months of birth
HF later in life.16 Coordinated lesion-specific approaches to without surgical correction. In contrast, conotruncal lesions,
HF may provide enhanced surveillance algorithms. such as tetralogy of Fallot, may have HF at the time of diagnosis
Standardized and Detailed Phenotyping Is Critical if there is little to no RV outflow tract obstruction, but typically
Specific types of CHD universally result in HF that can be cyanosis dictates the clinical course of these children before re-
addressed by surgical repair. Functional single ventricle (SV) pair. Postsurgical scarring or rhythm abnormalities can lead to
lesions (a variety of malformations grouped for their com- HF over time. However, in the absence of these injuries, the
mon physiology, whereby 1 ventricle is under-developed, varying susceptibility of patients to HF, either in the immediate
and the other ventricle supports both pulmonary and sys- postsurgical period or as long-term sequelae, suggests that ad-
temic circulations) have the highest incidence of ventricular ditional factors such as genetics may impact the relative risk for
dysfunction and HF at the time of diagnosis, followed by HF and ultimately the long-term outcome. The ability to predict
conotruncal lesions (eg, tetralogy of Fallot and transposition ventricular dysfunction and HF using genetic and molecular in-
of the great arteries), left to right shunts (eg, VSD and AVSD), formation such as predictive biomarkers may inform care and
and valve disease (eg, aortic stenosis and pulmonary insuf- improve outcomes in the future.
ficiency). In some cases, decisions about the timing of sur- Numerous classification schemes for CHD have been de-
gery are based on balancing the need for the infant to age and veloped, the first being Maude Abbott’s atlas.53 Fyler et al54
grow and the morbidities associated with HF, including poor subsequently developed a refined system emphasizing anato-
growth. Postsurgically, identifying patients at risk for persis- my and physiology, and this classification system remains in
tent ventricular dysfunction or HF is confounded by many wide use given its practical application to surgical intervention.
factors, including the heterogeneity within groups of lesions, More recently, the National Birth Defects Prevention Network
surgery-specific factors, and differences in medical manage- has developed a new scheme that incorporates developmental
ment. Approaches to improve our ability to identify patients and etiologic considerations into a system that organizes de-
at risk for HF pre- and postsurgically are needed. Analyzing tailed lesions in groups.55 This type of organization will be nec-
outcomes within subgroups of large multicenter cohorts will essary to incorporate genetic and molecular information into
elucidate the common clinical characteristics and molecular the phenotype. In addition, the Society of Thoracic Surgeons
features of patients whose ventricular dysfunction does not has developed a classification system used by their Registry in
resolve and progresses to HF. North America, and the International Nomenclature Committee
Functional SV defects are complex malformations that con- has recently produced the International Pediatric Cardiac Code
tribute disproportionately to the prevalence of HF in part be- for cross-mapping.56 However, none of these systems identify
cause they may have both pressure and volume overload issues ventricular dysfunction or HF as an independent phenotype,
affecting both systemic and pulmonary circulations, and in part but emerging evidence showing the genetic basis of these traits
because cyanotic lesions are also at risk for subendocardial isch- suggests it may be prudent to reconsider.
emia. In some SV lesions, such as pulmonary atresia with intact In many ways, our ability to standardize detailed and accu-
ventricular septum (PA/IVS), the LV is the systemic ventricle, rate phenotype data has lagged behind our ability to database
984  Circulation Research  March 17, 2017

genomic findings.57 The field of phenomics results from the this mechanism.66–68 Interestingly, mutations in histone-mod-
need to obtain structured, comprehensive phenotype data, ifying proteins were commonly observed to occur as de novo
termed deep phenotyping, as well as computational phenomic mutations, suggesting that abnormal heart patterning resulting
analysis.58–60 These big data analysis methods provide major op- from epigenetic alterations may be relatively common.
portunities for progress. As whole exome and whole genome The distinction between monogenic and complex traits
sequencing become more common both in research and clini- can be overly simplistic, as is drawing a distinct boundary be-
cal care, a wealth of data is accumulating that can be iteratively tween syndromic and nonsyndromic causes given that variants
interrogated. However, to exploit this resource to begin to un- in genes known to cause syndromic forms of CHD are now
derstand complex clinical phenotypes such as HF in the CHD identified in nonsyndromic cases. In addition, traits that seem
population, it needs to be properly combined with accurate, stan- to be monogenic can be influenced by variation in multiple
dardized phenotyping and longitudinal patient follow-up.55,56,60,61 modifier genes, as the above example of cardiac dysfunction
in MFS illustrates. The reverse is also true: complex traits can
Distinct Mechanisms be strongly influenced by variation in a single gene. These
findings may explain the decreased penetrance and variable
Genome and Transcriptome in CHD and HF
expressivity that are so common in CHD.
Because the transcription factors, signaling pathways, and
In an effort to better understand genetic causes of CHD,
structural proteins that are important for cardiogenesis are
systems biology approaches have been used to assess func-
delineated, an overlapping network with genes required for
tional convergence of causative CHD genes, effectively
both cardiac structure and cardiac function is emerging. As
Downloaded from http://circres.ahajournals.org/ by guest on June 24, 2018

combining knowledge from genetics and developmental


discussed above, there are now clear examples of CHD result-
biology.69,70 Developmental pathways acting independently
ing from mutations in genes that cause cardiomyopathy and
or coordinately contribute to heart development.71,72 These
HF, providing evidence that abnormal expression of a gene re-
pathways often exhibit extensive cross-talk, suggesting a
sponsible for cardiac contractile function can also cause struc-
highly complex milieu in which individual or multiple ge-
tural heart defects. Likewise, there are examples of genetic
netic variants could potentially act to disrupt normal heart
defects causing CHD that can also lead to cardiomyopathy or
morphogenesis. The integration of genetic analysis with
HF in some proportion of patients. These examples imply that
developmental biology knowledge provides an ability to
the redundant gene networks responsible for cardiac structure
begin to unravel the additive effects of multiple susceptibil-
and function can lead to divergent phenotypes when abnor-
ity alleles in combination. Interestingly, these approaches
mal. As reviewed in Fahed et al,62 there is increasing evidence
have suggested that different CHD risk factors are more
that at least some cases of LV dysfunction in CHD result from
likely to act on distinct components of a common func-
a genetic predisposition and that combinatorial interactions
tional network than to directly converge on a single genetic
of genes and environment (eg, hemodynamic stressors) result
or molecular target.69,70
in HF in this patient population. Studying the genetic back-
HF is a phenotypic result of multiple heterogeneous
ground in specific CHD types may further develop this idea
causes, both environmentally acquired and genetic. Like
and help identify at risk individuals.
CHD, systems biology approaches have been applied to HF
Overlapping HF Molecular Pathways in CHD and in an effort to integrate complex interactions.73 However,
Cardiomyopathy patients with CHD and HF represent a combination that
There are shared genetic causes between CHD and cardio- most studies have not considered. The molecular adaptive
myopathy that increase the likelihood of HF in the context responses to the mismatch of energetic demands on the heart
of CHD. However, given the frequency with which ventric- involve the reinduction of the fetal gene program and mul-
ular dysfunction and HF occurs in individuals with CHD, a tiple transcriptional and signaling pathways. Preexisting
shared single genetic cause is unlikely to explain most cases. genetic abnormalities in these pathways could potentially
The development of HF in subsets of patients with specific compromise the adaptive response, thereby leading to HF
genetic syndromes hints at overlapping gene networks. What (Figure 1). To illustrate how pathogenic mutations leading
about the more common nonsyndromic CHD? Here, the ge- to either cardiomyopathy or CHD might intersect, we per-
netic architecture suggests that a majority of cases result from formed a basic network analysis using genes available on
multifactorial causes and behave as a complex trait, although current clinical genetic testing panels (Figure 2). Although
Mendelian inheritance does occur, albeit less frequently. there are some categories associated with only CHD or car-
Recent studies in multiplex families with isolated CHD diomyopathy, Figure 2A illustrates that a variety of catego-
have identified mutations in TBX5, GATA4, TFAP2B, ELN, ries are associated with both cardiomyopathy and CHD. For
MYH6, and NOTCH1.63–65 Of note, 2 of these genes, MYH6 example, the cardiac chamber morphogenesis feature maps
and NOTCH1, are known to overlap with cardiomyopathy. to both, whereas striated muscle contraction maps only to
Second, TBX5 and TFAP2B cause Holt–Oram syndrome and cardiomyopathy. The line darkness denotes the strength of
Char syndrome, respectively, 2 genetic syndromic conditions association, such that arrhythmia is more strongly associated
that can have very subtle extracardiac findings, highlighting with cardiomyopathy than CHD. Figure 2B shows similar
the importance of careful phenotyping. De novo mutations are findings at the gene level.
another important cause of isolated CHD. Recent studies in- In addition to genetic factors contributing to HF, there
dicate that ≤10% of nonsyndromic CHD may be explained by is substantial interest in the role of epigenetics in CHD
Hinton and Ware   Heart Failure in Congenital Heart Disease   985
Downloaded from http://circres.ahajournals.org/ by guest on June 24, 2018

Figure 2. Genes causing congenital heart disease (CHD) and cardiomyopathy form a complex network. The network diagrams were
generated using ToppCluster (www.toppcluster.cchmc.org) software. Gene lists were derived from clinically available next-generation
sequencing panels for cardiomyopathy genes (n=50) and CHD genes (n=44). A, Abstracted cluster network showing a selected subset
of features from the following categories: Gene ontology (GO): molecular function; GO: biological processes; GO: cellular component;
human phenotype; mouse phenotype; pathway; disease (cardiomyopathy and CHD). The features are color coded by category and
connected to cardiomyopathy (white circle) and CHD gene nodes. B, Gene level network with nodes (blue) selected from features in GO:
biological processes category. The network illustrates that regulation of cellular component of movement, actin filament-based processes,
and cardiac chamber morphogenesis are shared in common between cardiomyopathy and CHD genes, whereas regulation of force of
heart contraction and tube development are unshared.

and HF. MicroRNAs (miRNAs) are noncoding RNAs that lncRNAs are >200 base pair RNAs that function in the
consist of 18 to 22 nucleotides. They have been identified regulation of transcriptional and post-transcriptional events.
as important regulators of gene expression at the post-tran- Although less well studied in HF than miRNAs, lncRNAs
scriptional level and act as important modulators of cardiac are emerging as important in this disease process. For exam-
hypertrophy, HF, and fibrosis.74 miRNAs show promise as ple, the mitochondrial lncRNA LIPCAR identified patients
circulating biomarkers and their relative stability in the undergoing cardiac remodeling who were independently at
blood when compared with mRNA enhances their suitability risk for future cardiovascular deaths.78 CHAST is another
for translation to clinical care. Study of miRNAs in mouse lncRNA that has recently been shown to be deregulated in
models has shown that overexpression can result in cardiac pressure overload–induced cardiac hypertrophy in mouse
hypertrophy and HF and that deletion can be protective. and significantly upregulated in hypertrophic heart tissue
Profibrotic miRNAs can be blocked by antagomirs result- from aortic stenosis patients.79 It will be important to de-
ing in decreased interstitial fibrosis and improved cardiac termine the degree to which pediatric miRNA and lncRNA
function in a mouse model of hypertrophy caused by pres- profiles change in a developmental stage–specific manner.
sure overload. Upregulation of specific miRNAs can also be In summary, it seems likely that genetic and epigenetic
seen in patients with HF.75 The development of antagomirs factors may combinatorially affect the susceptibility to both
for therapeutic use in HF is reportedly in preclinical devel- CHD and HF, and some variants will increase or decrease risk
opment.76 Interestingly, pediatric HF patients seem to have for either CHD or HF or both. Identifying these molecular pat-
unique miRNA profiles,77 suggesting that further investiga- terns requires systems biology approaches, bioinformatics ex-
tion that is specific to the pediatric population is required pertise, and statistical genetic methods. In addition, improved
for the application of antagomir-based therapy. Long non- attention to and methodology for accurate phenotyping is es-
coding RNAs (lncRNAs) are another group of RNA mol- sential. The integration of genetic and epigenetic findings with
ecules that play important roles in development and disease. deep phenotyping will improve our understanding of disease
986  Circulation Research  March 17, 2017

cause, in part by elaborating distinct and shared genetic fac- maladaptation, the failing heart loses the ability to function
tors and ultimately advance medical care. efficiently.84

Shared Mechanisms Regulation of Cardiac Metabolism Impacts the


Course of HF
HF Is Modified by Metabolic, Molecular, and At the metabolic level, HF represents a state of inefficient
Neurohormonal Factors energy expenditure. The heart uses fatty acids and glucose
Most of what we know about HF results from studies in adults. as energy sources with the former as the preferred substrate.
These metabolic, molecular, and neurohormonal abnormali- Mitochondrial dysfunction is a key characteristic of HF and
ties that occur in HF have been the subject of recent reviews, is both a cause and a consequence of abnormal energetics.85
including a previous Compendium issue on HF, and the reader A few of the consequences of mitochondrial dysfunction in-
is referred there for in-depth information on these complex clude impaired fatty acid oxidation, increased DNA damage
topics.80 These findings have been applied to the pediatric and apoptosis, and alterations in mitochondrial calcium lev-
population. At the molecular level, HF is characterized by els. Prenatally, the heart uses glucose as its primary energy
transcriptional, translational, and epigenetic alterations that substrate. Postnatally, there is a transition from a low oxygen
are a consequence of the adaptive and compensatory mecha- intrauterine environment to a high oxygen environment that is
nisms used by the heart in an attempt to respond to its func- accompanied by a switch from anaerobic glycolysis to fatty
tional demands (Figure 3). In both adult and pediatric HF, the acid oxidation and oxidative phosphorylation as a means of
progressive deterioration of myocardial function leads to an ATP production, a metabolically more efficient method of en-
Downloaded from http://circres.ahajournals.org/ by guest on June 24, 2018

inability of the heart to meet body requirements and is, essen- ergy generation.86,87 This transition in substrate utilization is
tially, a mismatch of supply and demand. Energy starvation is accompanied by mitochondrial proliferation and is a critical
proposed as a unifying mechanism underlying cardiac con- transition window. For example, infants with rare inborn er-
tractile failure.81,82 This is frequently the result of a downward rors of metabolism such as fatty acid oxidation disorders or
spiral of events in which decreases in oxygen and substrate mitochondrial disorders often experience cardiac decompen-
availability trigger adaptive mechanisms, including neuro- sation during this transition. There are important differences
endocrine overdrive, activation of signaling pathways, ECM in the expression of nuclear-encoded mitochondrial genes
remodeling, and alterations in mechanical load, among oth- during development, suggesting that stage-specific control
ers. Although these adaptive mechanisms stabilize contractile of mitochondrial biogenesis exists.88 The temporal span over
function in the short term, over the long term they may result in which this occurs in humans has not been well defined. Mouse
further compromise because of a vicious cycle characterized models have demonstrated that there is a strong activation of
in part by an increased mismatch of the supply:demand ratio.83 mitochondrial biogenesis in the myocardium during the peri-
Because metabolic remodeling progresses from adaptation to natal period. Ablation of transcription factors critical for this

Figure 3. Schematic of the interactions between causal, predisposing, and contributing factors that result in congenital heart
disease (CHD) and heart failure (HF). The figure depicts that CHD has a genetic cause and additional genetic components that
predispose the heart to maladaptive responses to environmental factors and may, individually or synergistically, lead to a mismatch
between cardiac output and demand with subsequent decompensation and ventricular dysfunction (yellow arrow). This in turn triggers
epigenetic modifications and many metabolic, molecular, and neurohormonal compensatory responses, which are also under primary
genetic regulation and therefore may predispose to disease. The effectiveness of these responses is a critical determinant of the
progression to HF. The signs and symptoms of HF are also subject to individual genetic variation; individual genetic and epigenetic
variation can provide protective or deleterious effects that influence severity. HF leads to the institution of medical therapies and surgical
interventions, including transplantation, which provide an additional layer of individual variation because of differential pharmacogenomics
and molecular responses to the stresses of surgery. ECM indicates extracellular matrix; lncRNA, long noncoding RNA; miRNA, microRNA;
and RAAS, renin–angiotensin–aldosterone system.
Hinton and Ware   Heart Failure in Congenital Heart Disease   987

metabolic switch, such as TFAM or PGC-1 isoforms, results it is logical to infer that additional differences and ongoing
in upregulation of glycolytic genes and rapidly fatal neona- changes impact HF in older children.
tal HF.89,90 Interestingly, there is now evidence that the transi-
tion from a low oxygen to a high oxygen environment after New Findings in Regulation of the Adrenergic
birth leads to mitochondrial damage which results in a block
Signaling Cascade in Infants and Children
There is a vast literature describing adrenergic signaling dur-
of cell cycle progression and withdrawal from the cell cycle.
ing HF, the deleterious effects of chronic stimulation and in-
In contrast, hypoxic cardiomyocytes retain a fetal or neonatal
creased sympathetic drive, the distinct functions of α1, α2, and
phenotype characterized by smaller size, fewer mitochondria,
β receptors, and the importance of the adrenergic system as a
and less evidence of oxidative damage, mononucleation, and,
therapeutic target.80,105 In pediatric patients with HF, therapy is
importantly, increased proliferative capacity.91,92 These studies
directed at mitigating this increased sympathetic drive, analo-
have implications for approaches to cardiac regeneration and
gous to treatment in the adult population. However, clinical
management of energetic demands during HF.
and translational research studies are beginning to identify im-
Sarcomeric Architecture Can Revert to Fetal State portant differences in the pediatric HF patient. Animal models
Through Molecular Switching have previously shown that β-adrenergic receptor-adenylyl
Sarcomere isoform switching and reinduction of the fetal gene cyclase cAMP pathway–mediated contractility responsive-
program occurs in HF.93–97 More recent evidence indicates that ness is less robust in the fetal/newborn heart versus the adult
these switches apply to miRNAs as well.74,98 The contractile heart. Furthermore, phosphodiesterase inhibition has less ef-
apparatus is responsive to ions, particularly calcium, and the fect on contractility alone, but enhanced contractility when
Downloaded from http://circres.ahajournals.org/ by guest on June 24, 2018

autonomic nervous system. The excitation–contraction cou- combined with isoproterenol.101 Several recent studies investi-
pling that occurs as a result of calcium influx to the myocyte gate the molecular findings in explanted hearts from children
and subsequent calcium release from the sarcoplasmic re- with HF and demonstrate age-related differences similar to
ticulum is fundamentally abnormal in HF, typically resulting those seen in mouse. Pediatric patients with DCM showed a
in decreased contractile force and delayed or incomplete re- differential adaptation of the β-adrenergic signaling pathway
laxation. Although pediatric HF recapitulates many of these when compared with adults with DCM or nonfailing con-
changes, the sarcomere of a fetus and neonate is not identical trols.106 Specifically, downregulation of β1- and β2-adrenergic
to that of an adult.99 The sarcomeric proteins, myosin, actin, receptors is identified in children, whereas β2-AR expression
troponin, tropomyosin, and titin, show isoform switching is maintained in adults. Differences in the phosphorylation
during development as a result of distinct gene expression or status of phospholamban are also noted in children versus
alternative splicing.100 During HF, isoform transitions occur, adults.106 Investigation of phosphodiesterase isoform expres-
often with reinduction of the fetal gene program. In addition, sion and responsiveness to phosphodiesterase inhibition also
calcium transients are different in neonates, with increased differed in pediatric versus adult samples.107,108 Although the
utilization of trans-sarcolemmal calcium flux. Taken together approach to medical therapy is similar, there is a lack of data
with the differences in contractility because of sarcomere iso- on the adaptive responses triggered in patients with structur-
form differences, the neonatal heart is stiffer, with less con- ally abnormal hearts when compared with those with primary
tractile reserve than the adult heart.101 heart muscle disease. In the investigation of phosphodiesterase
expression, intriguing differences were noted in samples from
The Matrix Has a Structural and Functional Impact DCM explants when compared with single RV CHD hearts
In addition to the components of the sarcomere, the ECM in the myocardial response to milrinone.108,109 Additional stud-
plays an important structural and signaling role during HF pro- ies are required to understand the characteristic adaptive pro-
gression. Increasing ECM deposition results in interstitial and grams invoked by ventricular dysfunction of different causes.
perivascular fibrosis. Elevated levels of serum markers of col- These findings suggest a differential responsiveness to HF
lagen turnover may be useful for risk stratification of those at medical therapy depending on age, a conclusion that requires
high risk of death and HF hospitalization in adults.102 In addi- more investigation.
tion, studies on ECM deposition and difference in microfibril
content of neonatal cardiomyocytes suggest that the likeli- Clinical Research for Pediatric Ventricular
hood of fibrosis differs depending on the age of the cardio- Dysfunction and HF Is Limited
myocyte. The ultrastructure of fetal cardiomyocytes is distinct HF has been extensively studied in adults but has only re-
from adult cardiomyocytes, containing fewer microfibrils and cently begun to be evaluated in the pediatric population. To
mitochondria. Furthermore, the numbers of cardiomyocytes assess the current clinical research in this area, we queried
differ by age, with cardiomyocyte numbers increasing imme- the clinical trials website (www.clinicaltrials.gov) and deter-
diately after birth but subsequently permanently withdrawing mined that 1.3% of HF studies currently open for enrollment
from the cell cycle leaving the heart with limited regenerative include children with CHD (n=29). Fundamental questions
capacity.103,104 Clearly, an understanding of the developmen- about whether HF represents the same disease in children re-
tal differences in cardiomyocyte response to injury has im- main to be answered, but small numbers and heterogeneity are
portant therapeutic implications, but these studies also show problematic. Below, we give examples of clinical trials within
that treatment of infants with HF may require substantially the SV population, which has been studied rigorously in re-
different approaches than adult HF patients. Because the hu- cent years. One limitation to current studies is the difficulty
man heart continues to grow in size in a developing organism, with comparing results between different trials. For example,
988  Circulation Research  March 17, 2017

the Pediatric Cardiac Genomics Consortium has not reported high risk or low risk of myocardial dysfunction could lead to
the presence of ventricular dysfunction or HF as a distinct or more tailored approaches and protocols to monitor patients
modifying phenotype, and the National Pediatric Cardiology and medically intervene.
Quality Improvement Collaborative has analyzed only cases The anatomic and morphological features of the ventricle
that are at least moderately abnormal.68,110 A clear overarch- are also important determinants of outcome. The RV does not
ing need is to leverage existing cohorts by combining them to adapt well to the demands of the systemic ventricle. Previous
facilitate trials where both primary and secondary questions work has shown that at least 50% of patients with CHDs where
can be answered. Adopting a learning health system, such the RV is the systemic ventricle, such as l-TGA (congenitally
as the National Pediatric Cardiology Quality Improvement corrected transposition of great arteries) and HLHS, develop
Collaborative, may improve the results of trials performed us- RV dysfunction. In addition, there is a 15% incidence of death
ing existing cohorts because this approach has shown signifi- or heart transplantation in early adulthood caused by myocar-
cant increases in patient participation, a common limitation in dial dysfunction.125,126 Transcriptional profiling has shown that
pediatric heart disease trials.111,112 Clinical trials using learn- the inability of the RV to respond to chronic pressure over-
ing health systems may improve outcomes through quality load is correlated with its inability to assume a LV expression
improvement efforts and accelerate findings and subsequent pattern of genes such as angiotensin, adrenergic receptors,
dissemination. Partnerships with nonprofit organizations fa- G-proteins, cytoskeletal, and contractile components.127–129
cilitate this type of research in part by assuming some of the Just as variation in sarcomeric genes might explain a propor-
cost, but more importantly by mobilizing patients and orga- tion of HF, or predisposition to HF in CHD cases, it is possible
Downloaded from http://circres.ahajournals.org/ by guest on June 24, 2018

nizing patients’ collective experience to identify and advance that genetic variation in these pathways predisposes some in-
clinically relevant patient-oriented research questions.113 dividuals to myocardial dysfunction when superimposed on
In patients with CHD, complex defects have a higher rate the background of CHD (Figure 1). Thus, variation that might
of associated HF. Functional SV heart defects account for be silent in the normal population may strongly predispose to
a disproportionate share of the morbidity and mortality in (or protect from) poor outcome in the CHD population. This
CHD, and mortality in the first year of life ranges from 10% level of genetic variation has not yet been explored. Although
to 35%.114,115 In addition, management of these patients is as- expression analyses have not been performed in SV patients,
sociated with a significant economic burden and high levels it is tempting to speculate that the maladaptive responses that
of resource requirements. Clinical research, including stud- the RV exhibits under pressure overload would be similar
ies performed by the PHN, has reported detailed clinical out- under chronic volume overload. Thus, patients with LV SVs
comes in this population of patients. traditionally have had better survival than those with RV SVs.
The clinical predictors of poor outcome in functional Better understanding the developmental and genetic basis of
SV defects are based on the type of CHD and complications these differences and the progression to HF should not only
developed during staged palliative repairs. Although the sur- provide insight into the relative contribution of pressure and
gical approach is similar among SV lesions, the underlying volume overload but also provide new important data for nov-
genetic causes are different, and well-characterized genetic el therapeutics.
syndromic conditions predict worse outcome. The PHN SV Clinical trials are ongoing to evaluate stem cell therapy as
Reconstruction Trial has provided information on outcomes a treatment for SV defects such as HLHS. In 2015, intracoro-
nary injection of umbilical cord blood-derived mononuclear
in this patient population.6,116–123 In the initial comparison of
cells or intracoronary administration of autologous cardio-
Blalock–Taussig versus RV–pulmonary artery shunt types for
sphere-derived cells was successfully used in HLHS patients.
SV, transplant-free survival at 12 months was 74% and 64%,
In the case of cardiosphere-derived cells infusion, RV ejec-
respectively, with no significant differences seen at 3 years.6,119
tion fraction remained persistently improved during 36-month
Mortality with stage I palliation (Norwood) ranges from 7%
follow-up.130,131 Additional clinical trials are in progress using
to 19% and between stage I and stage II palliation, 4% to
sources of progenitor cell types that have had reliable and safe
15%. Interestingly, genetic abnormalities were identified as
results in earlier studies, including allogenic mesenchymal
independent risk factors, suggesting that some degree of the
stem cells, bone marrow–derived cells, c-kit+ cells, or um-
variation observed in HF occurrence in severity is attributable
bilical cord cells.132 Although there is optimism that stem cell
to genetic factors.122 SV patients with Fontan circulation, in
therapy represents a novel direction that could provide signifi-
which the venous return bypasses the SV and is directed to the
cant gains to management in the pediatric population, there is
pulmonary artery, face many medical issues, including growth
much still to be learned about the cell-type–specific require-
problems, hemodynamic compromise, including cyanosis,
ments, the timing of administration, the long-term impact on
pathway obstructions and valve dysfunction, arrhythmias, or
cardiac status, and the mechanisms of action.132–135 Induced
pleural effusions, and ascites.124 Fontan circulation results in
pluripotent stem cells are another mechanism for studying and
progressive multiorgan system dysfunction, and Fontan fail-
modeling patient-specific disease processes.
ure requires cardiac transplantation. Studies to determine sur-
vival of different types of SV malformations, such as HLHS Medical Therapy and Pharmacogenomics
versus tricuspid atresia, are ongoing. In this complex context, Medical therapy for HF in patients with CHD is typically ex-
it is difficult to isolate the cause and impact of ventricular dys- trapolated from adults with ischemic HF. There are limited
function and HF, but the potential use of this knowledge is data to support use in children. For example, a recent trial of
substantial. For example, the ability to stratify patients into valsartan, an angiotensin II receptor blocker, in patients with
Hinton and Ware   Heart Failure in Congenital Heart Disease   989

a systemic RV failed to show any beneficial effect on the pri- The results demonstrated an association with renin–angioten-
mary end point of RV ejection fraction.136 Similarly, a ran- sin–aldosterone genotype and failure of reverse remodeling
domized trial of enalapril in infants with SV did not alter HF after surgery. The limitations of this important study were that
severity or improve growth or ventricular function.117 Taken it consisted of a relatively small population and was biased
together, these observations suggest that pediatric HF may toward patients surviving past stage I palliation. In 2014, the
have different causes and additional contributing factors and same renin–angiotensin–aldosterone single-nucleotide poly-
therefore will require new and different therapies. morphism was shown to be independently associated with
Pharmacogenomics is defined as the association between increased tachyarrhythmia after CHD surgery (odds ratio, 1.6;
genetic factors and drug response. Interindividual variation in 95% confidence interval, 1.1–2.3; P=0.02).138 Thus, there is
drug response can lead to therapeutic failure (eg, ultrarapid preliminary evidence for the potential use of pharmacogenet-
metabolizers) and adverse drug responses (poor metaboliz- ics in the CHD population, but larger studies with increased
ers). Whereas numerous pharmacogenomic clinical trials have numbers are required. Ultimately, individual risk stratification
occurred in the adult HF population, very few studies have and personalized care are dependent on our ability to (1) de-
been performed in the CHD population.137 Specific challenges fine primary causes of CHD, (2) identify secondary genetic
to the study of pharmacogenomics in the CHD population factors that lead to susceptibility to or protection from myo-
include variability in underlying cause (genetic variabil- cardial dysfunction in the context of CHD and environmen-
ity), heterogeneity in the types of CHDs and their treatment tal/mechanical stressors, and (3) select appropriate medical
(phenotypic variability), small sample sizes, frequent dosing therapy based on predicted pharmacogenomic response. It is
Downloaded from http://circres.ahajournals.org/ by guest on June 24, 2018

changes with age and changes in drug responsiveness with the combinatorial interaction of these primary, secondary, and
age. Furthermore, there is significant variability in the treat- tertiary genetic influences which ultimately determines the
ment and management of these patients at different centers, clinical phenotype and outcome in a multifactorial fashion.
leading to challenges for multisite studies.
To date, the only study to address HF or ventricular remod- Summary
eling in CHD was performed by Mital et al,7 with a combina- Pediatric HF is a clinical entity that results from an inabil-
torial approach to investigate the impact of 5 single-nucleotide ity of the heart to meet working demands. Although shar-
polymorphisms in genes within the renin–angiotensin–aldo- ing many metabolic and molecular features with adults with
sterone pathways as part of the PHN SV Reconstruction Trial. HF, as shown in Figure 3, children have underlying causes,

Table 4.  Research Approaches to Investigate Congenital Heart Disease and Heart Failure
Approach Benefits/Questions Answered Limitations Example
Basic In vivo Modeling anatomy and physiology; Cost and time; may not predict Large animal—surgical models and
functional testing; genetic manipulation human biology bioprosthetics139
(some model organisms) Small animal—mouse models of disease44
In vitro Faster and less expensive functional Nonphysiological; no 3D heart Cell signaling,47 expression studies, siRNA,
testing than in vivo structure tissue engineering
Translational Genetic/ Disease causation; individual variation Costly; sample size requirements GWAS, CMA, WES,66 WGS,
genomic Pharmacogenomics
Epigenetic Gene regulation; disease-specific Heterogeneous cell types; miRNA,140,141 lncRNA,79 piRNA, chromatin
alterations throughput and efficiency remodeling
Omics Large data sets; comprehensive; Large data sets; descriptive Transcriptome (microarray, RNAseq),
hypothesis generating regulome (ChIPseq), microbiome/
metabolome, proteome72,142
Companion Personalized medicine; risk stratification Association and validation difficult WES/Biomarker pairs143
diagnostics
Stem cell Personalized/patient specific Labor intensive; reprogramming iPS cell, cardiac progenitor cell134,144
artifacts mesenchymal derived
Systems Unbiased comprehensive analyses Dependent on accurate annotation Bioinformatics70
biology and curation in databases
Clinical Epidemiology/ Disease incidence and determinants; Not mechanistic; may not Registry61,145
population evidence-based practice and analysis determine causality
health
Clinical trial Direct comparisons in patients Costly; multiple regulatory layers; Drug trial,117,136,146 surgical outcome122
study size requirements difficult in
pediatrics
3D indicates 3-dimensional; ChIPseq, chromatin immunoprecipitation sequencing; CMA, chromosome microarray analysis; GWAS, genome-wide association study;
iPS, cell-induced pluripotent stem cell; lncRNA, long noncoding RNA; miRNA, microRNA; piRNA, Piwi-interacting RNA, RNASeq RNA sequencing; siRNA, small interfering
RNA; WES, whole exome sequencing; and WGS, whole genome sequencing.
990  Circulation Research  March 17, 2017

presentations, and disease courses that may differ from adults. incorporated into the ACC/AHA 2005 Guidelines for the Diagnosis and
Management of Heart Failure in Adults A Report of the American College
Thus, a fundamental question is whether HF in the pediat-
of Cardiology Foundation/American Heart Association Task Force on
ric population is the same disease process as it is in adults. Practice Guidelines Developed in Collaboration With the International
What is the evidence for standard HF therapy in pediatric Society for Heart and Lung Transplantation. J Am Coll Cardiol.
CHD patients? Do standard HF prognostic tools work in these 2009;53:e1–e90. doi: 10.1016/j.jacc.2008.11.013.
3. Roger VL. Epidemiology of heart failure. Circ Res. 2013;113:646–659.
patients? How do differences in the myocardium in infancy doi: 10.1161/CIRCRESAHA.113.300268.
and early childhood affect management and therapeutics? 4. Ross RD, Daniels SR, Schwartz DC, Hannon DW, Shukla R, Kaplan S.
Additional research is needed to organize existing data and Plasma norepinephrine levels in infants and children with congestive heart
leverage established cohorts to answer these questions. For failure. Am J Cardiol. 1987;59:911–914.
5. Connolly D, Rutkowski M, Auslender M, Artman M. The New York
example, analyzing outcomes within subgroups of large co- University Pediatric Heart Failure Index: a new method of quantifying
horts with CHD is necessary to understand the common clini- chronic heart failure severity in children. J Pediatr. 2001;138:644–648.
cal characteristics and molecular features of patients whose doi: 10.1067/mpd.2001.114020.
6. Ohye RG, Sleeper LA, Mahony L, et al; Pediatric Heart Network
ventricular dysfunction does not resolve or progresses to HF. Investigators. Comparison of shunt types in the Norwood procedure
There is preliminary evidence that the myocardium has dis- for single-ventricle lesions. N Engl J Med. 2010;362:1980–1992. doi:
tinct molecular properties based on age and location (RV ver- 10.1056/NEJMoa0912461.
sus LV). Furthermore, responsiveness of the myocardium to 7. Mital S, Chung WK, Colan SD, Sleeper LA, Manlhiot C, Arrington CB,
Cnota JF, Graham EM, Mitchell ME, Goldmuntz E, Li JS, Levine JC,
medication varies based on age. Additional investigation of Lee TM, Margossian R, Hsu DT; Pediatric Heart Network Investigators.
these differences is necessary to risk stratify and customize Renin-angiotensin-aldosterone genotype influences ventricular remodel-
Downloaded from http://circres.ahajournals.org/ by guest on June 24, 2018

therapy. Stem cell therapy holds great promise as a novel ap- ing in infants with single ventricle. Circulation. 2011;123:2353–2362. doi:
10.1161/CIRCULATIONAHA.110.004341.
proach to HF in CHD, but basic, translational, and clinical
8. Giglia TM, Jenkins KJ, Matitiau A, Mandell VS, Sanders SP, Mayer JE
research is necessary to understand how to apply it. Future Jr, Lock JE. Influence of right heart size on outcome in pulmonary atresia
research investigating the shared developmental mechanisms with intact ventricular septum. Circulation. 1993;88:2248–2256.
leading to both CHD and cardiomyopathy will likely be infor- 9. Webster G, Zhang J, Rosenthal D. Comparison of the epidemiology and
co-morbidities of heart failure in the pediatric and adult populations: a
mative for understanding genetic and epigenetic pathways that retrospective, cross-sectional study. BMC Cardiovasc Disord. 2006;6:23.
increase susceptibility to HF. doi: 10.1186/1471-2261-6-23.
Numerous research approaches have been used to in- 10. Rossano JW, Shaddy RE. Heart failure in children: etiology and treatment.
vestigate HF (Table 4), and the research community is well J Pediatr. 2014;165:228–233. doi: 10.1016/j.jpeds.2014.04.055.
11. Rossano JW, Kim JJ, Decker JA, Price JF, Zafar F, Graves DE, Morales
positioned to leverage these resources. Basic research in HF DL, Heinle JS, Bozkurt B, Towbin JA, Denfield SW, Dreyer WJ, Jefferies
is substantial, and although not a focus of this review, there JL. Prevalence, morbidity, and mortality of heart failure-related hospital-
is a clear need for additional animal models of HF in CHD. izations in children in the United States: a population-based study. J Card
Fail. 2012;18:459–470. doi: 10.1016/j.cardfail.2012.03.001.
Genetic and genomic approaches are progressing rapidly, but 12. Lipshultz SE, Sleeper LA, Towbin JA, Lowe AM, Orav EJ, Cox GF, Lurie
we need standardized biorepositories that combine harmo- PR, McCoy KL, McDonald MA, Messere JE, Colan SD. The incidence
nized data collection to realize improved treatment strategies. of pediatric cardiomyopathy in two regions of the United States. N Engl J
Secondary studies from large multicenter trials are needed to Med. 2003;348:1647–1655. doi: 10.1056/NEJMoa021715.
13. Hoffman JI, Kaplan S. The incidence of congenital heart disease. J Am
further characterize disease in those cohorts. Increasing the Coll Cardiol. 2002;39:1890–1900.
emphasis on integration of the different components of HF 14. Massin MM, Astadicko I, Dessy H. Epidemiology of heart failure in a
may accelerate the identification of new therapeutic windows tertiary pediatric center. Clin Cardiol. 2008;31:388–391. doi: 10.1002/
clc.20262.
(Figure 3). Ultimately, multifaceted and combined research
15. Sommers C, Nagel BH, Neudorf U, Schmaltz AA. [Congestive heart fail-
approaches will advance medical care by allowing risk strati- ure in childhood. An epidemiologic study]. Herz. 2005;30:652–662. doi:
fication by age, lesion, and underlying cause and will lead to 10.1007/s00059-005-2596-6.
improved protocols for medical intervention and therapy. 16. Norozi K, Wessel A, Alpers V, Arnhold JO, Geyer S, Zoege M, Buchhorn
R. Incidence and risk distribution of heart failure in adolescents and
adults with congenital heart disease after cardiac surgery. Am J Cardiol.
Sources of Funding 2006;97:1238–1243. doi: 10.1016/j.amjcard.2005.10.065.
This work is supported in part by the National Institutes of Health, 17. Gurvitz M, Burns KM, Brindis R, et al. Emerging research directions in adult
an American Heart Association Established Investigator Award, congenital heart disease: a report from an NHLBI/ACHA working group. J
the March of Dimes, and the Indiana University Health—Indiana Am Coll Cardiol. 2016;67:1956–1964. doi: 10.1016/j.jacc.2016.01.062.
University School of Medicine Strategic Research Initiative and 18. Lui GK, Fernandes S, McElhinney DB. Management of cardiovascular
Physician Scientist Initiative (S.M. Ware). risk factors in adults with congenital heart disease. J Am Heart Assoc.
2014;3:e001076. doi: 10.1161/JAHA.114.001076.
19. Parrott A, Ware SM. The role of the geneticist and genetic counselor in
Disclosures an ACHD Clinic. Prog Pediatr Cardiol. 2012;34:15–20. doi: 10.1016/j.
None. ppedcard.2012.05.004.
20. Webb G, Mulder BJ, Aboulhosn J, et al. The care of adults with con-
genital heart disease across the globe: current assessment and future per-
References spective: a position statement from the International Society for Adult
1. Kirk R, Dipchand AI, Rosenthal DN, et al. The International Society Congenital Heart Disease (ISACHD). Int J Cardiol. 2015;195:326–333.
for Heart and Lung Transplantation Guidelines for the management of doi: 10.1016/j.ijcard.2015.04.230.
pediatric heart failure: Executive summary. [Corrected]. J Heart Lung 21. Wessels MW, Willems PJ. Mutations in sarcomeric protein genes not only
Transplant. 2014;33:888–909. doi: 10.1016/j.healun.2014.06.002. lead to cardiomyopathy but also to congenital cardiovascular malforma-
2. Hunt SA, Abraham WT, Chin MH, Feldman AM, Francis GS, Ganiats TG, tions. Clin Genet. 2008;74:16–19. doi: 10.1111/j.1399-0004.2008.00985.x.
Jessup M, Konstam MA, Mancini DM, Michl K, Oates JA, Rahko PS, 22. Postma AV, van Engelen K, van de Meerakker J, et al. Mutations in the
Silver MA, Stevenson LW, Yancy CW; American College of Cardiology sarcomere gene MYH7 in Ebstein anomaly. Circ Cardiovasc Genet.
Foundation; American Heart Association. 2009 Focused update 2011;4:43–50. doi: 10.1161/CIRCGENETICS.110.957985.
Hinton and Ware   Heart Failure in Congenital Heart Disease   991

23. Arbustini E, Favalli V, Narula N, Serio A, Grasso M. Left ventricular 45. Hahn A, Lauriol J, Thul J, Behnke-Hall K, Logeswaran T, Schänzer A,
noncompaction: a distinct genetic cardiomyopathy? J Am Coll Cardiol. Böğürcü N, Garvalov BK, Zenker M, Gelb BD, von Gerlach S, Kandolf
2016;68:949–966. doi: 10.1016/j.jacc.2016.05.096. R, Kontaridis MI, Schranz D. Rapidly progressive hypertrophic cardio-
24. Lorca R, Martín M, Reguero JJ, Díaz-Molina B, Morís C, Lambert JL, myopathy in an infant with Noonan syndrome with multiple lentigi-
Astudillo A. Left ventricle non-compaction: the still misdiagnosed cardiomy- nes: palliative treatment with a rapamycin analog. Am J Med Genet A.
opathy. Int J Cardiol. 2016;223:420–421. doi: 10.1016/j.ijcard.2016.08.200. 2015;167A:744–751. doi: 10.1002/ajmg.a.36982.
25. Stähli BE, Gebhard C, Biaggi P, Klaassen S, Valsangiacomo Buechel E, 46. Dietz HC, Cutting GR, Pyeritz RE, Maslen CL, Sakai LY, Corson GM,
Attenhofer Jost CH, Jenni R, Tanner FC, Greutmann M. Left ventricu- Puffenberger EG, Hamosh A, Nanthakumar EJ, Curristin SM. Marfan
lar non-compaction: prevalence in congenital heart disease. Int J Cardiol. syndrome caused by a recurrent de novo missense mutation in the fibrillin
2013;167:2477–2481. doi: 10.1016/j.ijcard.2012.05.095. gene. Nature. 1991;352:337–339. doi: 10.1038/352337a0.
26. Luxán G, Casanova JC, Martínez-Poveda B, et al. Mutations in the
47. Cook JR, Carta L, Bénard L, Chemaly ER, Chiu E, Rao SK, Hampton
NOTCH pathway regulator MIB1 cause left ventricular noncompaction TG, Yurchenco P, Costa KD, Hajjar RJ, Ramirez F; GenTAC Registry
cardiomyopathy. Nat Med. 2013;19:193–201. doi: 10.1038/nm.3046. Consortium. Abnormal muscle mechanosignaling triggers cardiomyopa-
27. Zhang W, Chen H, Qu X, Chang CP, Shou W. Molecular mechanism thy in mice with Marfan syndrome. J Clin Invest. 2014;124:1329–1339.
of ventricular trabeculation/compaction and the pathogenesis of the left doi: 10.1172/JCI71059.
ventricular noncompaction cardiomyopathy (LVNC). Am J Med Genet C 48. Tae HJ, Petrashevskaya N, Marshall S, Krawczyk M, Talan M. Cardiac
Semin Med Genet. 2013;163C:144–156. doi: 10.1002/ajmg.c.31369. remodeling in the mouse model of Marfan syndrome develops into two
28. Chen H, Zhang W, Sun X, et al. Fkbp1a controls ventricular myocardium distinctive phenotypes. Am J Physiol Heart Circ Physiol. 2016;310:H290–
trabeculation and compaction by regulating endocardial Notch1 activity. H299. doi: 10.1152/ajpheart.00354.2015.
Development. 2013;140:1946–1957. doi: 10.1242/dev.089920. 49. Tworetzky W, McElhinney DB, Brook MM, Reddy VM, Hanley FL,
29. Ching YH, Ghosh TK, Cross SJ, et al. Mutation in myosin heavy chain Silverman NH. Echocardiographic diagnosis alone for the complete repair
6 causes atrial septal defect. Nat Genet. 2005;37:423–428. doi: 10.1038/ of major congenital heart defects. J Am Coll Cardiol. 1999;33:228–233.
ng1526. 50. Friedman WF, George BL. Treatment of congestive heart failure by alter-
Downloaded from http://circres.ahajournals.org/ by guest on June 24, 2018

30. Matsson H, Eason J, Bookwalter CS, et al. Alpha-cardiac actin mutations ing loading conditions of the heart. J Pediatr. 1985;106:697–706.
produce atrial septal defects. Hum Mol Genet. 2008;17:256–265. doi: 51. Anderson PA. Maturation and cardiac contractility. Cardiol Clin.

10.1093/hmg/ddm302. 1989;7:209–225.
31. Monserrat L, Hermida-Prieto M, Fernandez X, Rodríguez I, Dumont 52. Wong J, Lamata P, Rathod RH, Bertaud S, Dedieu N, Bellsham-Revell
C, Cazón L, Cuesta MG, Gonzalez-Juanatey C, Peteiro J, Alvarez N, H, Pushparajah K, Razavi R, Hussain T, Schaeffter T, Powell AJ, Geva
Penas-Lado M, Castro-Beiras A. Mutation in the alpha-cardiac actin gene T, Greil GF. Right ventricular morphology and function following stage I
associated with apical hypertrophic cardiomyopathy, left ventricular non- palliation with a modified Blalock-Taussig shunt versus a right ventricle-
compaction, and septal defects. Eur Heart J. 2007;28:1953–1961. doi: to-pulmonary artery conduit. Eur J Cardiothorac Surg. 2017;51:50–57.
10.1093/eurheartj/ehm239.
doi: 10.1093/ejcts/ezw227.
32. Preuss C, Andelfinger G. Genetics of heart failure in congenital heart dis-
53. Abbott ME. The Atlas of Congenital Cardiac Disease. New York: American
ease. Can J Cardiol. 2013;29:803–810. doi: 10.1016/j.cjca.2013.03.011.
Heart Association; 1936.
33. Azhar M, Ware SM. Genetic and developmental basis of cardiovas-

54. Fyler DC, Rudolph AM, Wittenborg MH, Nadas AS. Ventricular septal
cular malformations. Clin Perinatol. 2016;43:39–53. doi: 10.1016/j.
defect in infants and children; a correlation of clinical, physiologic, and
clp.2015.11.002.
autopsy data. Circulation. 1958;18:833–851.
34. Lalani SR, Belmont JW. Genetic basis of congenital cardiovascular

55. Botto LD, Lin AE, Riehle-Colarusso T, Malik S, Correa A; National Birth
malformations. Eur J Med Genet. 2014;57:402–413. doi: 10.1016/j.
Defects Prevention Study. Seeking causes: classifying and evaluating con-
ejmg.2014.04.010.
genital heart defects in etiologic studies. Birth Defects Res A Clin Mol
35. Cowan JR, Ware SM. Genetics and genetic testing in congenital heart dis-
Teratol. 2007;79:714–727. doi: 10.1002/bdra.20403.
ease. Clin Perinatol. 2015;42:373–393, ix. doi: 10.1016/j.clp.2015.02.009.
56. Franklin RC, Jacobs JP, Krogmann ON, Béland MJ, Aiello VD, Colan SD,
36. Lander J, Ware SM. Copy number variation in congenital heart defects.
Elliott MJ, William Gaynor J, Kurosawa H, Maruszewski B, Stellin G,
Curr Genet Med Rep. 2014;2:168–178.
Tchervenkov CI, Walters Iii HL, Weinberg P, Anderson RH. Nomenclature
37. Vissers LE, van Ravenswaaij CM, Admiraal R, Hurst JA, de Vries

BB, Janssen IM, van der Vliet WA, Huys EH, de Jong PJ, Hamel BC, for congenital and paediatric cardiac disease: historical perspectives and
Schoenmakers EF, Brunner HG, Veltman JA, van Kessel AG. Mutations The International Pediatric and Congenital Cardiac Code. Cardiol Young.
in a new member of the chromodomain gene family cause CHARGE syn- 2008;18(suppl 2):70–80. doi: 10.1017/S1047951108002795.
drome. Nat Genet. 2004;36:955–957. doi: 10.1038/ng1407. 57. Brookes AJ, Robinson PN. Human genotype-phenotype databases: aims,
38. Metcalfe K, Rucka AK, Smoot L, et al. Elastin: mutational spectrum in challenges and opportunities. Nat Rev Genet. 2015;16:702–715. doi:
supravalvular aortic stenosis. Eur J Hum Genet. 2000;8:955–963. doi: 10.1038/nrg3932.
10.1038/sj.ejhg.5200564. 58. Freimer N, Sabatti C. The human phenome project. Nat Genet.

39. Tassabehji M, Metcalfe K, Donnai D, Hurst J, Reardon W, Burch M, Read 2003;34:15–21. doi: 10.1038/ng0503-15.
AP. Elastin: genomic structure and point mutations in patients with supra- 59. Lanktree MB, Hassell RG, Lahiry P, Hegele RA. Phenomics: expand-
valvular aortic stenosis. Hum Mol Genet. 1997;6:1029–1036. ing the role of clinical evaluation in genomic studies. J Investig Med.
40. Jerome LA, Papaioannou VE. DiGeorge syndrome phenotype in mice 2010;58:700–706. doi: 10.231/JIM.0b013e3181d844f7.
mutant for the T-box gene, Tbx1. Nat Genet. 2001;27:286–291. doi: 60. Robinson PN. Deep phenotyping for precision medicine. Hum Mutat.
10.1038/85845. 2012;33:777–780. doi: 10.1002/humu.22080.
41. Lindsay EA, Vitelli F, Su H, Morishima M, Huynh T, Pramparo T, Jurecic 61. Hinton RB, McBride KL, Bleyl SB, Bowles NE, Border WL, Garg V,
V, Ogunrinu G, Sutherland HF, Scambler PJ, Bradley A, Baldini A. Tbx1 Smolarek TA, Lalani SR, Ware SM. Rationale for the cytogenomics of
haploinsufficiency in the DiGeorge syndrome region causes aortic arch cardiovascular malformations consortium: a phenotype intensive registry
defects in mice. Nature. 2001;410:97–101. doi: 10.1038/35065105. based approach. J Cardiovasc Dev Dis. 2015;2:76–92.
42. Tartaglia M, Mehler EL, Goldberg R, Zampino G, Brunner HG, Kremer 62. Fahed AC, Roberts AE, Mital S, Lakdawala NK. Heart failure in congeni-
H, van der Burgt I, Crosby AH, Ion A, Jeffery S, Kalidas K, Patton MA, tal heart disease: a confluence of acquired and congenital. Heart Fail Clin.
Kucherlapati RS, Gelb BD. Mutations in PTPN11, encoding the pro- 2014;10:219–227. doi: 10.1016/j.hfc.2013.09.017.
tein tyrosine phosphatase SHP-2, cause Noonan syndrome. Nat Genet. 63. Blue GM, Kirk EP, Giannoulatou E, Dunwoodie SL, Ho JW, Hilton DC,
2001;29:465–468. doi: 10.1038/ng772. White SM, Sholler GF, Harvey RP, Winlaw DS. Targeted next-gener-
43. Prendiville TW, Gauvreau K, Tworog-Dube E, Patkin L, Kucherlapati RS, ation sequencing identifies pathogenic variants in familial congenital
Roberts AE, Lacro RV. Cardiovascular disease in Noonan syndrome. Arch heart disease. J Am Coll Cardiol. 2014;64:2498–2506. doi: 10.1016/j.
Dis Child. 2014;99:629–634. doi: 10.1136/archdischild-2013-305047. jacc.2014.09.048.
44. Marin TM, Keith K, Davies B, Conner DA, Guha P, Kalaitzidis D, Wu 64. Jia Y, Louw JJ, Breckpot J, Callewaert B, Barrea C, Sznajer Y, Gewillig
X, Lauriol J, Wang B, Bauer M, Bronson R, Franchini KG, Neel BG, M, Souche E, Dehaspe L, Vermeesch JR, Lambrechts D, Devriendt K,
Kontaridis MI. Rapamycin reverses hypertrophic cardiomyopathy in a Corveleyn A. The diagnostic value of next generation sequencing in
mouse model of LEOPARD syndrome-associated PTPN11 mutation. J familial nonsyndromic congenital heart defects. Am J Med Genet A.
Clin Invest. 2011;121:1026–1043. doi: 10.1172/JCI44972. 2015;167A:1822–1829. doi: 10.1002/ajmg.a.37108.
992  Circulation Research  March 17, 2017

65. Arrington CB, Bleyl SB, Matsunami N, Bonnell GD, Otterud BE, Nielsen 89. Hansson A, Hance N, Dufour E, Rantanen A, Hultenby K, Clayton
DC, Stevens J, Levy S, Leppert MF, Bowles NE. Exome analysis of a DA, Wibom R, Larsson NG. A switch in metabolism precedes in-
family with pleiotropic congenital heart disease. Circ Cardiovasc Genet. creased mitochondrial biogenesis in respiratory chain-deficient mouse
2012;5:175–182. doi: 10.1161/CIRCGENETICS.111.961797. hearts. Proc Natl Acad Sci USA. 2004;101:3136–3141. doi: 10.1073/
66. Sifrim A, Hitz MP, Wilsdon A, et al; INTERVAL Study; UK10K
pnas.0308710100.
Consortium; Deciphering Developmental Disorders Study. Distinct genet- 90. Lai L, Leone TC, Zechner C, Schaeffer PJ, Kelly SM, Flanagan DP,
ic architectures for syndromic and nonsyndromic congenital heart defects Medeiros DM, Kovacs A, Kelly DP. Transcriptional coactivators PGC-
identified by exome sequencing. Nat Genet. 2016;48:1060–1065. doi: 1alpha and PGC-lbeta control overlapping programs required for peri-
10.1038/ng.3627. natal maturation of the heart. Genes Dev. 2008;22:1948–1961. doi:
67. Homsy J, Zaidi S, Shen Y, et al. De novo mutations in congenital heart 10.1101/gad.1661708.
disease with neurodevelopmental and other congenital anomalies. Science. 91. Kimura W, Xiao F, Canseco DC, et al. Hypoxia fate mapping identifies
2015;350:1262–1266. doi: 10.1126/science.aac9396. cycling cardiomyocytes in the adult heart. Nature. 2015;523:226–230.
68. Zaidi S, Choi M, Wakimoto H, et al. De novo mutations in histone-mod- doi: 10.1038/nature14582.
ifying genes in congenital heart disease. Nature. 2013;498:220–223. doi: 92. Puente BN, Kimura W, Muralidhar SA, et al. The oxygen-rich postnatal
10.1038/nature12141. environment induces cardiomyocyte cell-cycle arrest through DNA dam-
69. Lage K, Møllgård K, Greenway S, et al. Dissecting spatio-temporal pro- age response. Cell. 2014;157:565–579. doi: 10.1016/j.cell.2014.03.032.
tein networks driving human heart development and related disorders. Mol 93. Anderson PA, Malouf NN, Oakeley AE, Pagani ED, Allen PD. Troponin
Syst Biol. 2010;6:381. doi: 10.1038/msb.2010.36. t isoform expression in humans. A comparison among normal and fail-
70. Lage K, Greenway SC, Rosenfeld JA, et al. Genetic and environmental ing adult heart, fetal heart, and adult and fetal skeletal muscle. Circ Res.
risk factors in congenital heart disease functionally converge in pro- 1991;69:1226–1233.
tein networks driving heart development. Proc Natl Acad Sci USA. 94. Boheler KR, Carrier L, de la Bastie D, Allen PD, Komajda M, Mercadier
2012;109:14035–14040. doi: 10.1073/pnas.1210730109. JJ, Schwartz K. Skeletal actin mRNA increases in the human heart during
71. Rana MS, Christoffels VM, Moorman AF. A molecular and genetic outline ontogenic development and is the major isoform of control and failing
Downloaded from http://circres.ahajournals.org/ by guest on June 24, 2018

of cardiac morphogenesis. Acta Physiol. 2013;207:588–615. doi: 10.1111/ adult hearts. J Clin Invest. 1991;88:323–330. doi: 10.1172/JCI115295.
apha.12061. 95. Izumo S, Nadal-Ginard B, Mahdavi V. Protooncogene induction and re-
72. Kathiriya IS, Nora EP, Bruneau BG. Investigating the transcriptional con- programming of cardiac gene expression produced by pressure overload.
trol of cardiovascular development. Circ Res. 2015;116:700–714. doi: Proc Natl Acad Sci USA. 1988;85:339–343.
10.1161/CIRCRESAHA.116.302832. 96. Miyata S, Minobe W, Bristow MR, Leinwand LA. Myosin heavy chain
73. Dewey FE, Wheeler MT, Ashley EA. Systems biology of heart failure, isoform expression in the failing and nonfailing human heart. Circ Res.
challenges and hopes. Curr Opin Cardiol. 2011;26:314–321. doi: 10.1097/ 2000;86:386–390.
HCO.0b013e328346597d. 97. Nakao K, Minobe W, Roden R, Bristow MR, Leinwand LA. Myosin
74. Kumarswamy R, Thum T. Non-coding RNAs in cardiac remodeling and heavy chain gene expression in human heart failure. J Clin Invest.
heart failure. Circ Res. 2013;113:676–689. doi: 10.1161/CIRCRESAHA. 1997;100:2362–2370. doi: 10.1172/JCI119776.
113.300226. 98. Thum T, Galuppo P, Wolf C, Fiedler J, Kneitz S, van Laake LW,
75. Bauters C, Kumarswamy R, Holzmann A, Bretthauer J, Anker SD, Pinet Doevendans PA, Mummery CL, Borlak J, Haverich A, Gross C,
F, Thum T. Circulating miR-133a and miR-423-5p fail as biomarkers for Engelhardt S, Ertl G, Bauersachs J. MicroRNAs in the human heart:
left ventricular remodeling after myocardial infarction. Int J Cardiol. a clue to fetal gene reprogramming in heart failure. Circulation.
2013;168:1837–1840. doi: 10.1016/j.ijcard.2012.12.074. 2007;116:258–267. doi: 10.1161/CIRCULATIONAHA.107.687947.
76. van Rooij E, Kauppinen S. Development of microRNA therapeutics
99. Siedner S, Krüger M, Schroeter M, Metzler D, Roell W, Fleischmann
is coming of age. EMBO Mol Med. 2014;6:851–864. doi: 10.15252/ BK, Hescheler J, Pfitzer G, Stehle R. Developmental changes in con-
emmm.201100899. tractility and sarcomeric proteins from the early embryonic to the adult
77. Stauffer BL, Russell G, Nunley K, Miyamoto SD, Sucharov CC. miR- stage in the mouse heart. J Physiol. 2003;548:493–505. doi: 10.1113/
NA expression in pediatric failing human heart. J Mol Cell Cardiol. jphysiol.2002.036509.
2013;57:43–46. doi: 10.1016/j.yjmcc.2013.01.005. 100. Yin Z, Ren J, Guo W. Sarcomeric protein isoform transitions in cardiac
78. Kumarswamy R, Bauters C, Volkmann I, Maury F, Fetisch J, Holzmann muscle: a journey to heart failure. Biochim Biophys Acta. 2015;1852:47–
A, Lemesle G, de Groote P, Pinet F, Thum T. Circulating long noncoding 52. doi: 10.1016/j.bbadis.2014.11.003.
RNA, LIPCAR, predicts survival in patients with heart failure. Circ Res. 101. Hsu DT, Pearson GD. Heart failure in children: part I: history, etiol-
2014;114:1569–1575. doi: 10.1161/CIRCRESAHA.114.303915. ogy, and pathophysiology. Circ Heart Fail. 2009;2:63–70. doi: 10.1161/
79. Viereck J, Kumarswamy R, Foinquinos A, et al. Long noncoding RNA CIRCHEARTFAILURE.108.820217.
Chast promotes cardiac remodeling. Sci Transl Med. 2016;8:326ra22. doi: 102. Spinale FG, Zile MR. Integrating the myocardial matrix into heart fail-
10.1126/scitranslmed.aaf1475. ure recognition and management. Circ Res. 2013;113:725–738. doi:
80. Braunwald E. Research advances in heart failure: a compendium. Circ 10.1161/CIRCRESAHA.113.300309.
Res. 2013;113:633–645. doi: 10.1161/CIRCRESAHA.113.302254. 103. Porrello ER, Johnson BA, Aurora AB, Simpson E, Nam YJ, Matkovich
81. Ingwall JS, Weiss RG. Is the failing heart energy starved? On using chemi- SJ, Dorn GW II, van Rooij E, Olson EN. MiR-15 family regulates post-
cal energy to support cardiac function. Circ Res. 2004;95:135–145. doi: natal mitotic arrest of cardiomyocytes. Circ Res. 2011;109:670–679. doi:
10.1161/01.RES.0000137170.41939.d9. 10.1161/CIRCRESAHA.111.248880.
82. Neubauer S. The failing heart–an engine out of fuel. N Engl J Med. 104. Soonpaa MH, Koh GY, Pajak L, Jing S, Wang H, Franklin MT, Kim
2007;356:1140–1151. doi: 10.1056/NEJMra063052. KK, Field LJ. Cyclin D1 overexpression promotes cardiomyocyte
83. Stanley WC, Recchia FA, Lopaschuk GD. Myocardial substrate metabo- DNA synthesis and multinucleation in transgenic mice. J Clin Invest.
lism in the normal and failing heart. Physiol Rev. 2005;85:1093–1129. doi: 1997;99:2644–2654. doi: 10.1172/JCI119453.
10.1152/physrev.00006.2004. 105. Lymperopoulos A, Rengo G, Koch WJ. Adrenergic nervous system in
84. Taegtmeyer H. Switching metabolic genes to build a better heart.
heart failure: pathophysiology and therapy. Circ Res. 2013;113:739–753.
Circulation. 2002;106:2043–2045. doi: 10.1161/CIRCRESAHA.113.300308.
85. Crouser ED. Mitochondrial dysfunction in septic shock and multiple or- 106. Miyamoto SD, Stauffer BL, Nakano S, Sobus R, Nunley K, Nelson P,
gan dysfunction syndrome. Mitochondrion. 2004;4:729–741. Sucharov CC. Beta-adrenergic adaptation in paediatric idiopathic dilated
86. Lehman JJ, Kelly DP. Transcriptional activation of energy metabolic
cardiomyopathy. Eur Heart J. 2014;35:33–41. doi: 10.1093/eurheartj/
switches in the developing and hypertrophied heart. Clin Exp Pharmacol ehs229.
Physiol. 2002;29:339–345. 107. Nakano SJ, Miyamoto SD, Movsesian M, Nelson P, Stauffer BL,

87. Fisher DJ, Heymann MA, Rudolph AM. Myocardial consumption of oxy- Sucharov CC. Age-related differences in phosphodiesterase activ-
gen and carbohydrates in newborn sheep. Pediatr Res. 1981;15:843–846. ity and effects of chronic phosphodiesterase inhibition in idiopathic
88. Pejznochova M, Tesarova M, Hansikova H, Magner M, Honzik T,
dilated cardiomyopathy. Circ Heart Fail. 2015;8:57–63. doi: 10.1161/
Vinsova K, Hajkova Z, Havlickova V, Zeman J. Mitochondrial DNA con- CIRCHEARTFAILURE.114.001218.
tent and expression of genes involved in mtDNA transcription, regula- 108. Nakano SJ, Sucharov J, van Dusen R, Cecil M, Nunley K, Wickers S,
tion and maintenance during human fetal development. Mitochondrion. Karimpur-Fard A, Stauffer BL, Miyamoto SD, Sucharov CC. Cardiac
2010;10:321–329. doi: 10.1016/j.mito.2010.01.006. adenylyl cyclase and phosphodiesterase expression profiles vary by age,
Hinton and Ware   Heart Failure in Congenital Heart Disease   993

disease, and chronic phosphodiesterase inhibitor treatment. J Card Fail. 125. Rutledge JM, Nihill MR, Fraser CD, Smith OE, McMahon CJ, Bezold
2017;23:72–80. doi: 10.1016/j.cardfail.2016.07.429. LI. Outcome of 121 patients with congenitally corrected transposition
109. Nakano SJ, Nelson P, Sucharov CC, Miyamoto SD. Myocardial re- of the great arteries. Pediatr Cardiol. 2002;23:137–145. doi: 10.1007/
sponse to milrinone in single right ventricle heart disease. J Pediatr. s00246-001-0037-8.
2016;174:199.e5–203.e5. doi: 10.1016/j.jpeds.2016.04.009. 126. Graham TP Jr, Bernard YD, Mellen BG, et al. Long-term outcome in
110. Brown DW, Mangeot C, Anderson JB, Peterson LE, King EC, Lihn congenitally corrected transposition of the great arteries: a multi-institu-
SL, Neish SR, Fleishman C, Phelps C, Hanke S, Beekman RH; Lannon tional study. J Am Coll Cardiol. 2000;36:255–261.
CM, National Pediatric Cardiology Quality Improvement Collaborative. 127. Reddy S, Osorio JC, Duque AM, Kaufman BD, Phillips AB, Chen JM,
Digoxin use is associated with reduced interstage mortality in patients Quaegebeur J, Mosca RS, Mital S. Failure of right ventricular adaptation
with no history of arrhythmia after stage i palliation for single ven- in children with tetralogy of Fallot. Circulation. 2006;114:I37–I42. doi:
tricle heart disease. J Am Heart Assoc. 2016;5:e002376. doi: 10.1161/ 10.1161/CIRCULATIONAHA.105.001248.
JAHA.115.002376. 128. Jeewa A, Manickaraj AK, Mertens L, Manlhiot C, Kinnear C, Mondal
111. Anderson JB, Beekman RH III, Kugler JD, Rosenthal GL, Jenkins
T, Smythe J, Rosenberg H, Lougheed J, McCrindle BW, van Arsdell G,
KJ, Klitzner TS, Martin GR, Neish SR, Brown DW, Mangeot C, Redington AN, Mital S. Genetic determinants of right-ventricular remod-
King E, Peterson LE, Provost L, Lannon C; National Pediatric eling after tetralogy of Fallot repair. Pediatr Res. 2012;72:407–413. doi:
Cardiology Quality Improvement Collaborative. Improvement in in- 10.1038/pr.2012.95.
terstage survival in a national pediatric cardiology learning network. 129. Kaufman BD, Desai M, Reddy S, Osorio JC, Chen JM, Mosca RS,
Circ Cardiovasc Qual Outcomes. 2015;8:428–436. doi: 10.1161/ Ferrante AW, Mital S. Genomic profiling of left and right ventricular hy-
CIRCOUTCOMES.115.001956. pertrophy in congenital heart disease. J Card Fail. 2008;14:760–767. doi:
112. Anderson JB, Beekman RH III, Kugler JD, Rosenthal GL, Jenkins
10.1016/j.cardfail.2008.06.002.
KJ, Klitzner TS, Martin GR, Neish SR, Darbie L, King E, Lannon C; 130. Burkhart HM, Qureshi MY, Peral SC, O’Leary PW, Olson TM, Cetta
National Pediatric Cardiology Quality Improvement Collaborative. Use F, Nelson TJ; Wanek Program Clinical Pipeline Group. Regenerative
of a learning network to improve variation in interstage weight gain af- therapy for hypoplastic left heart syndrome: first report of intraoperative
Downloaded from http://circres.ahajournals.org/ by guest on June 24, 2018

ter the Norwood operation. Congenit Heart Dis. 2014;9:512–520. doi: intramyocardial injection of autologous umbilical-cord blood-derived
10.1111/chd.12232. cells. J Thorac Cardiovasc Surg. 2015;149:e35–e37. doi: 10.1016/j.
113. Ohman EM, Douglas PS, Dean LB, Ginsburg GS. Philanthropy for sci- jtcvs.2014.10.093.
ence: is it a viable option? Circ Res. 2016;119:1057–1059. doi: 10.1161/ 131. Tarui S, Ishigami S, Ousaka D, Kasahara S, Ohtsuki S, Sano S, Oh H.
CIRCRESAHA.116.309657. Transcoronary infusion of cardiac progenitor cells in hypoplastic left
114. Hannan RL, Ybarra MA, Ojito JW, Alonso FA, Rossi AF, Burke RP. heart syndrome: three-year follow-up of the Transcoronary Infusion of
Complex neonatal single ventricle palliation using antegrade cerebral Cardiac Progenitor Cells in Patients With Single-Ventricle Physiology
perfusion. Ann Thorac Surg. 2006;82:1278–1284; discussion 1284–1275. (TICAP) trial. J Thorac Cardiovasc Surg. 2015;150:1198–1207, 1208.
115. Hehir DA, Dominguez TE, Ballweg JA, Ravishankar C, Marino BS, Bird e1. doi: 10.1016/j.jtcvs.2015.06.076.
GL, Nicolson SC, Spray TL, Gaynor JW, Tabbutt S. Risk factors for in- 132. Wehman B, Siddiqui OT, Mishra R, Sharma S, Kaushal S. Stem cell
terstage death after stage 1 reconstruction of hypoplastic left heart syn- therapy for CHD: towards translation. Cardiol Young. 2015;25(suppl
drome and variants. J Thorac Cardiovasc Surg. 2008;136:94–99, 99.e1. 2):58–66. doi: 10.1017/S1047951115000840.
doi: 10.1016/j.jtcvs.2007.12.012. 133. Mishra R, Vijayan K, Colletti EJ, Harrington DA, Matthiesen TS,

116. Burch PT, Gerstenberger E, Ravishankar C, et al; Pediatric Heart
Simpson D, Goh SK, Walker BL, Almeida-Porada G, Wang D, Backer
Network Investigators. Longitudinal assessment of growth in hypoplastic CL, Dudley SC Jr, Wold LE, Kaushal S. Characterization and functional-
left heart syndrome: results from the single ventricle reconstruction trial. ity of cardiac progenitor cells in congenital heart patients. Circulation.
J Am Heart Assoc. 2014;3:e000079. doi: 10.1161/JAHA.114.000079. 2011;123:364–373. doi: 10.1161/CIRCULATIONAHA.110.971622.
117. Hsu DT, Zak V, Mahony L, et al; Pediatric Heart Network Investigators. 134. Agarwal U, Smith AW, French KM, et al. Age-dependent effect of pe-
Enalapril in infants with single ventricle: results of a multicenter diatric cardiac progenitor cells after juvenile heart failure. Stem Cells
randomized trial. Circulation. 2010;122:333–340. doi: 10.1161/ Transl Med. 2016;5:883–892. doi: 10.5966/sctm.2015-0241.
CIRCULATIONAHA.109.927988. 135. Sanganalmath SK, Bolli R. Cell therapy for heart failure: a compre-
118. Newburger JW, Sleeper LA, Bellinger DC, et al; Pediatric Heart Network hensive overview of experimental and clinical studies, current chal-
Investigators. Early developmental outcome in children with hypoplas- lenges, and future directions. Circ Res. 2013;113:810–834. doi: 10.1161/
tic left heart syndrome and related anomalies: the single ventricle re- CIRCRESAHA.113.300219.
construction trial. Circulation. 2012;125:2081–2091. doi: 10.1161/ 136. van der Bom T, Winter MM, Bouma BJ, Groenink M, Vliegen HW, Pieper
CIRCULATIONAHA.111.064113. PG, van Dijk AP, Sieswerda GT, Roos-Hesselink JW, Zwinderman AH,
119. Newburger JW, Sleeper LA, Frommelt PC, et al; Pediatric Heart Network Mulder BJ. Effect of valsartan on systemic right ventricular function:
Investigators. Transplantation-free survival and interventions at 3 years a double-blind, randomized, placebo-controlled pilot trial. Circulation.
in the single ventricle reconstruction trial. Circulation. 2014;129:2013– 2013;127:322–330. doi: 10.1161/CIRCULATIONAHA.112.135392.
2020. doi: 10.1161/CIRCULATIONAHA.113.006191. 137. Dorn GW II. Adrenergic signaling polymorphisms and their impact on
120. Ravishankar C, Zak V, Williams IA, et al; Pediatric Heart Network cardiovascular disease. Physiol Rev. 2010;90:1013–1062. doi: 10.1152/
Investigators. Association of impaired linear growth and worse neurode- physrev.00001.2010.
velopmental outcome in infants with single ventricle physiology: a report 138. Smith AH, Flack EC, Borgman KY, Owen JP, Fish FA, Bichell DP,
from the pediatric heart network infant single ventricle trial. J Pediatr. Kannankeril PJ. A common angiotensin-converting enzyme polymor-
2013;162:250.e2–256.e2. doi: 10.1016/j.jpeds.2012.07.048. phism and preoperative angiotensin-converting enzyme inhibition modi-
121. Schwartz SM, Lu M, Ohye RG, et al; Pediatric Heart Network
fy risk of tachyarrhythmias after congenital heart surgery. Heart Rhythm.
Investigators. Risk factors for prolonged length of stay after the stage 2 2014;11:637–643. doi: 10.1016/j.hrthm.2014.01.005.
procedure in the single-ventricle reconstruction trial. J Thorac Cardiovasc 139. Zafar F, Hinton RB, Moore RA, Baker RS, Bryant R III, Narmoneva
Surg. 2014;147:1791–1798, 1798.e1. doi: 10.1016/j.jtcvs.2013.07.063. DA, Taylor MD, Morales DL. Physiological growth, remodeling po-
122. Tabbutt S, Ghanayem N, Ravishankar C, et al; Pediatric Heart Network tential, and preserved function of a novel bioprosthetic tricuspid valve:
Investigators. Risk factors for hospital morbidity and mortality after the tubular bioprosthesis made of small intestinal submucosa-derived extra-
Norwood procedure: a report from the Pediatric Heart Network Single cellular matrix. J Am Coll Cardiol. 2015;66:877–888. doi: 10.1016/j.
Ventricle Reconstruction trial. J Thorac Cardiovasc Surg. 2012;144:882– jacc.2015.06.1091.
895. doi: 10.1016/j.jtcvs.2012.05.019. 140. Sucharov CC, Sucharov J, Karimpour-Fard A, Nunley K, Stauffer BL,
123. Williams RV, Ravishankar C, Zak V, Evans F, Atz AM, Border WL, Miyamoto SD. Micro-RNA expression in hypoplastic left heart syn-
Levine J, Li JS, Mahony L, Mital S, Pearson GD, Prakash A, Hsu DT; drome. J Card Fail. 2015;21:83–88. doi: 10.1016/j.cardfail.2014.09.013.
Pediatric Heart Network Investigators. Birth weight and prematurity 141. Tijsen AJ, Pinto YM, Creemers EE. Non-cardiomyocyte microRNAs
in infants with single ventricle physiology: pediatric heart network in- in heart failure. Cardiovasc Res. 2012;93:573–582. doi: 10.1093/cvr/
fant single ventricle trial screened population. Congenit Heart Dis. cvr344.
2010;5:96–103. doi: 10.1111/j.1747-0803.2009.00369.x. 142. Miyamoto SD, Stauffer BL, Polk J, Medway A, Friedrich M, Haubold K,
124. Deal BJ, Jacobs ML. Management of the failing Fontan circulation. Peterson V, Nunley K, Nelson P, Sobus R, Stenmark KR, Sucharov CC.
Heart. 2012;98:1098–1104. doi: 10.1136/heartjnl-2011-301133. Gene expression and β-adrenergic signaling are altered in hypoplastic
994  Circulation Research  March 17, 2017

left heart syndrome. J Heart Lung Transplant. 2014;33:785–793. doi: 145. Raissadati A, Nieminen H, Haukka J, Sairanen H, Jokinen E. Late causes
10.1016/j.healun.2014.02.030. of death after pediatric cardiac surgery: a 60-year population-based study.
143. Hinton RB. Advances in the treatment of aortic valve disease: is it time J Am Coll Cardiol. 2016;68:487–498. doi: 10.1016/j.jacc.2016.05.038.
for companion diagnostics? Curr Opin Pediatr. 2014;26:546–552. doi: 146. Shaddy RE, Boucek MM, Hsu DT, Boucek RJ, Canter CE, Mahony L,
10.1097/MOP.0000000000000137. Ross RD, Pahl E, Blume ED, Dodd DA, Rosenthal DN, Burr J, LaSalle
144. Ghazizadeh Z, Vahdat S, Fattahi F, Fonoudi H, Omrani G, Gholampour B, Holubkov R, Lukas MA, Tani LY; Pediatric Carvedilol Study Group.
M, Aghdami N. Isolation and characterization of cardiogenic, stem-like Carvedilol for children and adolescents with heart failure: a random-
cardiac precursors from heart samples of patients with congenital heart ized controlled trial. JAMA. 2007;298:1171–1179. doi: 10.1001/
disease. Life Sci. 2015;137:105–115. doi: 10.1016/j.lfs.2015.07.006. jama.298.10.1171.
Downloaded from http://circres.ahajournals.org/ by guest on June 24, 2018
Heart Failure in Pediatric Patients With Congenital Heart Disease
Robert B. Hinton and Stephanie M. Ware
Downloaded from http://circres.ahajournals.org/ by guest on June 24, 2018

Circ Res. 2017;120:978-994


doi: 10.1161/CIRCRESAHA.116.308996
Circulation Research is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231
Copyright © 2017 American Heart Association, Inc. All rights reserved.
Print ISSN: 0009-7330. Online ISSN: 1524-4571

The online version of this article, along with updated information and services, is located on the
World Wide Web at:
http://circres.ahajournals.org/content/120/6/978

Permissions: Requests for permissions to reproduce figures, tables, or portions of articles originally published
in Circulation Research can be obtained via RightsLink, a service of the Copyright Clearance Center, not the
Editorial Office. Once the online version of the published article for which permission is being requested is
located, click Request Permissions in the middle column of the Web page under Services. Further information
about this process is available in the Permissions and Rights Question and Answer document.

Reprints: Information about reprints can be found online at:


http://www.lww.com/reprints

Subscriptions: Information about subscribing to Circulation Research is online at:


http://circres.ahajournals.org//subscriptions/

También podría gustarte