A novel NKX2-5 loss-of-function mutation predisposes to familial dilated cardiomyopathy and arrhythmias

  • Authors:
    • Fang Yuan
    • Xing-Biao Qiu
    • Ruo-Gu Li
    • Xin-Kai Qu
    • Juan Wang
    • Ying-Jia Xu
    • Xu Liu
    • Wei-Yi Fang
    • Yi-Qing Yang
    • De-Ning Liao
  • View Affiliations

  • Published online on: December 9, 2014     https://doi.org/10.3892/ijmm.2014.2029
  • Pages: 478-486
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Dilated cardiomyopathy (DCM) is the most prevalent type of primary myocardial disease, which is the third most common cause of heart failure and the most frequent reason for heart transplantation. Aggregating evidence demonstrates that genetic risk factors are involved in the pathogenesis of idiopathic DCM. Nevertheless, DCM is of remarkable genetic heterogeneity and the genetic defects underpinning DCM in an overwhelming majority of patients remain unknown. In the present study, the whole coding exons and splice junction sites of the NKX2-5 gene, which encodes a homeodomain transcription factor crucial for cardiac development and structural remodeling, were sequenced in 130 unrelated patients with idiopathic DCM. The available relatives of the index patient harboring an identified mutation and 200 unrelated ethnically matched healthy individuals used as controls were genotyped for the NKX2-5 gene. The functional effect of the mutant NKX2-5 was characterized in contrast to its wild-type counterpart using a dual-luciferase reporter assay system. As a result, a novel heterozygous NKX2-5 mutation, p.S146W, was identified in a family with DCM inherited as an autosomal dominant trait, which co-segregated with DCM in the family with complete penetrance. Notably, the mutation carriers also had arrhythmias, such as paroxysmal atrial fibrillation and atrioventricular block. The missense mutation was absent in 400 reference chromosomes and the altered amino acid was completely conserved evolutionarily among species. Functional analysis revealed that the NKX2-5 mutant was associated with a significantly reduced transcriptional activity. The findings expand the mutational spectrum of NKX2-5 linked to DCM and provide novel insight into the molecular mechanisms underlying DCM, contributing to the antenatal prophylaxis and allele-specific management of DCM.

Introduction

Dilated cardiomyopathy (DCM) is a primary cardiac disorder characterized by ventricular chamber enlargement and systolic dysfunction with normal ventricular wall thickness in the absence of associated conditions, such as coronary artery disease, hypertension and valve disease sufficient to cause global decreased contractility (1). It is the most frequent form of non-ischemic cardiomyopathy, affecting approximately 1 in 250 individuals (2). DCM is the most common cause of chronic congestive heart failure and sudden cardiac death in individuals between the ages of 20 and 60 years, and is the leading indication for heart transplantation in both children and adult patients worldwide (14). A variety of etiologies has been implicated in the pathogenesis of DCM, including viral myocarditis, myocardial infarction, toxic insults, alcohol abuse, nutritional deficiencies, chronic uncontrolled tachycardia or premature ventricular contractions, autoimmune abnormalities and metabolic disorders (5). However, the majority of DCM cases remain unexplained after a thorough review for secondary causes, and such DCM is defined as idiopathic DCM, among which 25–50% of DCM cases occur in at least two closely related family members, hence termed familial DCM (3). A growing body of evidence demonstrates that genetic risk factors play a pivotal role in the pathology of DCM, and mutations in over 50 DCM genes have been described as leading to DCM in patients with familial, as well as sporadic DCM (13,68). The majority of the genes linked to DCM encode sarcomeric and cytoskeletal proteins, which are involved in the generation and transmission of contractile force, and are predominantly inherited in an autosomal dominant pattern, although a few follow an autosomal recessive, X-linked or mitochondrial mode of inheritance (2). Nevertheless, these established DCM-associated genes merely account for less than a third of the studied cases and each gene has a low mutational frequency, with most occurring in <1% of patients with DCM (8). Thus, the genetic determinants responsible for DCM in an overwhelming majority of patients remain to be identified.

A number of studies have highlighted the essential roles of the cardiac transcription factors in cardiovascular development and cardiac structural remodeling, including the GATA zinc finger-containing transcription factor and the NK homeodomain transcription factor families (914), and a long list of mutations in GATA binding protein (GATA)4, GATA5, GATA6 and NK2 homeobox 5 (NKX2-5) have been associated with various congenital heart diseases and arrhythmias, including atrial septal defect, ventricular septal defect, tetralogy of Fallot, endocardial cushion defect, patent ductus arteriosus, double outlets of the right ventricle, pulmonary stenosis, hypoplastic right ventricle, atrial fibrillation and cardiac conduction block (1539). Moreover, GATA4 has also been causally implicated in the development of DCM (40,41).

Similar with GATA4, the homeobox transcription factor, NKX2-5, is abundantly expressed in the heart at various developmental stages and its high expression is also found in adult cardiomyocytes, where it mediates the expression of several crucial structural and regulatory genes, including those encoding atrial natriuretic factor (ANF), brain natriuretic peptide, connexin40, troponin I, troponin C, α- and β-myosin heavy chains (12,42). The targeted disruption of NKX2-5 in mice has been shown to lead to impaired cardiac growth and chamber formation, deranged gene regulatory networks and early embryonic lethality (4345), while mice with the heterozygous or conditional deletion of NKX2-5 at the postnatal stages have shown defects of late cardiomyocyte maturation and adult heart contraction, predisposing to progressive cardiomyopathy and congestive heart failure (4648). In human families with NKX2-5 mutations underlying congenital cardiovascular malformations, left ventricular contractile dysfunction and DCM as a late clinical manifestation have been observed in some mutation carriers (35,49). Furthermore, NKX2-5 has been shown to physically interact with GATA4 or T-box 20 (TBX20) and synergistically regulate the expression of multiple important cardiac target genes, including those encoding ANF, T- and L-type Ca2+ channels, connexin40, α-actin and leucine-rich repeat containing 10 (LRRC10) (42), and loss-of-function mutations in GATA4, LRRC10, α-actin and TBX5 have been associated with DCM (2,40,41,50,51). These observational results indicate that NKX2-5 is an alternative candidate gene for DCM.

Subjects and methods

Study subjects

A cohort of 130 genetically unrelated patients with idiopathic DCM was enrolled from the Han Chinese population. The available relatives of the index patients were also recruited. A total of 200 ethnically-matched unrelated healthy individuals were enlisted as the controls. All participants were evaluated by a detailed history and physical examination, chest radiography, electrocardiogram, echocardiography and an exercise performance test. Cardiac catheterization, angiography, endomyocardial biopsy and cardiac magnetic resonance imaging were performed only if there was a strong clinical indication. Medical records were also reviewed in the case of deceased or unavailable relatives. The diagnosis of idiopathic DCM was made according to the criteria established by the World Health Organization/International Society and Federation of Cardiology Task Force on the Classification of Cardiomyopathy: a left ventricular end-diastolic diameter >27 mm/m2 and an ejection fraction <40% or fractional shortening <25% in the absence of abnormal loading conditions, coronary artery disease, congenital heart lesions and other systemic diseases, as previously described (40,41,52). Individuals were excluded if they had insufficient echocardiographic image quality or coexistent conditions that may give rise to contractile dysfunction, such as uncontrolled systemic hypertension, coronary artery disease or valvular heart disease. Familial DCM was defined when DCM occurred in two or more first-degree family relatives. Peripheral venous blood samples were obtained from all participants. The clinical analyses were conducted with investigators blinded to the genotypes. The present study was carried out in accordance with the principles of the Declaration of Helsinki and the study protocol was approved by the Institutional Ethics committee of Shanghai Chest Hospital, Shanghai, China. Written informed consent was obtained from all participants or their guardians prior to study.

Mutational analysis

Genomic DNA was extracted from the peripheral blood lymphocytes of each participant using the Wizard Genomic DNA Purification kit (Promega, Madison, WI, USA). The whole coding region and splice junction sites of the NKX2-5 gene was sequenced in the 130 unrelated patients with idiopathic DCM. When a mutation was identified in an index patient, the available relatives of the mutation carrier and the 200 unrelated healthy controls were subsequently genotyped for NKX2-5. The referential genomic DNA sequence of NKX2-5 was derived from GenBank (accession no. NT_023133). The primer pairs used to amplify the coding exons and flanking introns of NKX2-5 by polymerase chain reaction (PCR) were designed as previously described (38). The PCR was carried out using HotStar Taq DNA Polymerase (Qiagen GmbH, Hilden, Germany) on a Veriti Thermal Cycler (Applied Biosystems, Foster, CA, USA) with standard conditions and concentrations of reagents. The amplified products were purified using the QIAquick Gel Extraction kit (Qiagen). Both strands of each PCR product were sequenced with a BigDye® Terminator version 3.1 Cycle Sequencing kit (Applied Biosystems) under an ABI PRISM 3130xl DNA Analyzer (Applied Biosystems). The DNA sequences were viewed and analyzed with the DNA Sequencing Analysis Software versoin 5.1 (Applied Biosystems). The variant was validated by re-sequencing of an independent PCR-generated amplicon from the same subject. In addition, for an identified sequence variant, the single nucleotide polymorphism (SNP; http://www.ncbi.nlm.nih.gov/SNP) and human gene mutation (HGM; http://www.hgmd.org) databases were queried to confirm its novelty.

Comparison of amino acid sequence of NKX2-5 from various species

The amino acid sequence of human NKX2-5 predicted from GenBank (Accession no. NM_004387) was aligned with that of the chimpanzee, rhesus monkey, dog, cattle, rat, mouse, zebrafish and fowl, using the online MUSCLE program, version 3.6 (http://www.ncbi.nlm.nih.gov/).

Molecular modeling

The disease-causing potential of a NKX2-5 sequence variation was predicted by MutationTaster (an online program at http://www.mutationtaster.org), which automatically provides a probability for the variation to be either a pathogenic mutation or a benign polymorphism. Of note, the P-value used in this study is the probability of the correct prediction rather than the probability of error as used in t-test statistics (i.e., a value close to 1 indicates a high accuracy of the prediction). Additionally, another online program PolyPhen-2 (http://genetics.bwh.harvard.edu/pph2) was used to evaluate the possible effects of an amino acid substitution on the structure and function of human NKX2-5.

Expression plasmids and site-directed mutagenesis

The recombinant expression plasmid, NKX2-5-pEFSA, and the ANF-luciferase (ANF-luc) reporter plasmid, which contains the 2600-bp 5′-flanking region of the ANF gene, were kindly provided by Dr Ichiro Shiojima from Chiba University School of Medicine, Chiba, Japan. The identified mutation was introduced into the wild-type NKX2-5 gene using a QuickChange II XL Site-Directed Mutagenesis kit (Stratagene, La Jolla, CA, USA) with a complementary pair of primers. The mutant was sequenced to confirm the desired mutation and to exclude any other sequence variations.

Reporter gene assays

COS-7 cells (a fibroblast-like cell line derived from monkey kidney tissue; 1×104), which were obtained from the Cell Bank of the Shanghai Institute of Life Science, Chinese Academy of Sciences, Shanghai, China, were plated onto a 24-well plate and cultured in Dulbecco’s modified Eagle’s medium supplemented with 10% fetal calf serum. The internal control reporter plasmid, pGL4.75 (hRluc/CMV; Promega), was used in transient transfection analyses to evaluate the transcriptional activity of the NKX2-5 mutant. The COS-7 cells were transfected with 0.4 μg of wild-type or mutant NKX2-5-pEFSA, 1.0 μg of ANF-luc, and 0.04 μg of pGL4.75 using PolyFect Transfection Reagent (Qiagen). For co-transfection experiments, 0.2 μg of wild-type NKX2-5-pEFSA, 0.2 μg of mutant NKX2-5-pEFSA, 1.0 μg of ANF-luc and 0.04 μg of pGL4.75 were used. Firefly luciferase and Renilla luciferase activities were measured with the Dual-Glo Luciferase assay system (Promega) 48 h after transfection. The activity of the ANF promoter was presented as the fold activation of firefly luciferase relative to Renilla luciferase. Three independent experiments were performed at minimum for wild-type and mutant NKX2-5.

Statistical analysis

Data are expressed as the means ± standard deviation (SD). Continuous variables were tested for normality of distribution, and the Student’s unpaired t-test was used to compare the numeric variables between two groups. A comparison of the categorical variables between two groups was performed using the Pearson’s χ2 test or Fisher’s exact test where appropriate. A two-tailed P-value of <0.05 was considered to indicate a statistically significantly difference.

Results

Clinical characteristics of the study participants

A total of 130 unrelated patients with idiopathic DCM were clinically evaluated in contrast to 200 control individuals. None of the patients had overt traditional risk factors for DCM. All the patients presented with the typical DCM phenotype as previously described (40,41,52). The control individuals had no evidence of structural cardiac diseases, and their echocardiogram results were normal. The baseline clinical characteristics of the study participants are summarized in Table I.

Table I

Clinical characteristics of the patients with dilated cardiomyopathy and the controls.

Table I

Clinical characteristics of the patients with dilated cardiomyopathy and the controls.

VariablesPatients (n=130)Controls (n=200)P-value
Age (years)46.4±12.247.8±11.50.2923
Male (%)60 (46.2)93 (46.5)0.9509
Family history of DCM (%)53 (40.8)0 (0)<0.0001
SBP (mmHg)116.3±14.5124.0±11.8<0.0001
DBP (mmHg)75.2±8.684.0±7.3<0.0001
HR (bpm)96.5±15.076.8±11.9<0.0001
LVEDD (mm)71.4±7.846.5±6.2<0.0001
LVESD (mm)58.5±8.335.2±6.1<0.0001
LVEF (%)38.4±9.464.2±7.8<0.0001
NYHA function class (%)
 I12 (9.2)NANA
 II40 (30.8)NANA
 III53 (40.8)NANA
 IV25 (19.2)NANA

[i] DCM, dilated cardiomyopathy; SBP, systolic blood pressure; DBP, diastolic blood pressure; HR, heart rate; LVEDD, left ventricular end-diastolic diameter; LVESD, left ventricular end-systolic diameter; LVEF, left ventricular ejection fraction; NYHA, New York Heart Association; NA, not applicable or not available.

Identification of NKX2-5 mutation

The exons and exon-intron boundaries of the NKX2-5 gene were sequenced in 130 index patients with idiopathic DCM, and a missense mutation was identified in the heterozygous state in a male DCM patient, with a mutational prevalence of approximately 0.77%. Specifically, a substitution of guanine for cytosine in the second nucleotide of codon 146 (c.437C>G), predicting the transition of serine (S) into tryptophan (W) at amino acid position 146 (p.S146W) was identified in the proband from family 1. The sequence chromatograms showing the detected heterozygous NKX2-5 mutation of c.437C>G compared with its control sequence are shown in Fig. 1. A schematic diagram of NKX2-5 protein delineating the structural domains and location of the mutation detected in the present study is presented in Fig. 2. The missense mutation was neither found in the 200 Chinese control subjects nor reported in the public databases for human sequence variations including the SNP and HGM databases, indicating that it was a novel mutation. The genetic screening of the family revealed that the mutation was present in all affected living family members, but absent in the unaffected family members examined. Analysis of the pedigree demonstrated that the mutation co-segregated with DCM transmitted as an autosomal dominant trait in the family with complete penetrance. The pedigree structure of the family is displayed in Fig. 3. The phenotypic characteristics and status of the NKX2-5 mutation of the affected living family members are listed in Table II.

Table II

The phenotypic characteristics and status of NKX2-5 mutation in the living affected pedigree members.

Table II

The phenotypic characteristics and status of NKX2-5 mutation in the living affected pedigree members.

IndividualGenderAge (years)Cardiac phenotypeLVEDD (mm)LVESD (mm)LVEF (%)LVFS (%)NKX2-5 mutation
II-1Male56DCM79633220+/−
II-3Male50DCM60423527+/−
III-2Female32DCM55404223+/−

[i] DCM, dilated cardiomyopathy; LVEDD, left ventricular end-diastolic diameter; LVESD, left ventricular end-systolic diameter; LVEF, left ventricular ejection fraction; LVFS, left ventricular fractional shortening; +/−, heterozygous mutation.

Of note, the proband’s father (I-1) also had atrial fibrillation, third-degree atrioventricular block and frequent premature ventricular contractions, and died suddenly at the age of 49; the proband’s brother (II-1) also had paroxysmal atrial fibrillation, second-degree atrioventricular block and premature ventricular contractions; and the proband (II-3) and his niece (III-2) also had premature ventricular contractions. In addition, two NKX2-5 polymorphisms, c.63A>G and c.606C>G, were observed in both DCM patients and control individuals. However, there was no significant difference in either of the two allele frequencies between the DCM patient and healthy control groups. All the sequence variants and their allele frequencies are listed in Table III.

Table III

NKX2-5 sequence variations identified in the present study.

Table III

NKX2-5 sequence variations identified in the present study.

LocationNucleotideAmino acidAllele frequency
PatientsControls
Exon 1c.63A>Gp.E21E(0.238) 62/260(0.233) 93/400
Exon 2c.437C>Gp.S146W(0.004) 1/260(0.000) 0/400
Exon 2c.606C>Gp.L202L(0.012) 3/260(0.015) 6/400
Alignment of multiple NKX2-5 protein sequences from various species

A cross-species alignment of NKX2-5 protein sequences demonstrated that the altered amino acid p.S146 was completely conserved evolutionarily in various species (Fig. 4).

Functional modeling in silico

The NKX2-5 sequence variation of c.437C>G was predicted by MutationTaster to be a pathogenic mutation with a P-value of nearly 1.0. No SNPs in the altered region were found in the MutationTaster database. This amino acid substitution was also predicted by PolyPhen-2 to be probably damaging, with a score of 1.000 (sensitivity, 0.00; specificity, 1.00).

Reduced transcriptional activity of the NKX2-5 mutant

As shown in Fig. 5, the same amount (0.4 μg) of wild-type and mutant NKX2-5 activated the ANF promoter by ~12-fold and ~3-fold, respectively. When the same amount of wild-type NKX2-5 (0.2 μg) was co-transfected with mutant NKX2-5 (0.2 μg), the induced activation of the ANF promoter was ~5-fold. These results demonstrate that the NKX2-5 mutant has a significantly reduced activation activity compared with its wild-type counterpart.

Discussion

In the present study, a heterozygous DCM-associated sequence variation, p.S146W, was identified in the NKX2-5 gene, which was not found in either 400 reference chromosomes or public databases for sequence variations including the SNP and HGM databases. This variant affected the amino acid that was completely conserved evolutionarily, and was predicted to be a causative mutation by the MutationTaster and PolyPhen-2 databases. The missense mutation co-segregated with DCM, as well as arrhythmias in the family with complete penetrance. Functional analysis unveiled that the mutant NKX2-5 was associated with significantly decreased transcriptional activity. Therefore, it is possible that NKX2-5 loss-of-function mutation predisposes these mutation carriers to DCM and arrhythmias.

It has been substantiated that NKX2-5 is an upstream regulator of multiple genes expressed during cardiac morphogenesis, including the ANF gene (42). Therefore, the functional characteristics of the NKX2-5 mutation may be deciphered by analyzing the transcriptional activity of the ANF promoter in cells expressing NKX2-5. In the present study, the functional effect of the novel NKX2-5 mutation (p.S146W) identified in the patients with familial DCM was characterized by transcriptional activity analysis and the results demonstrated that the mutant NKX2-5 was associated with a significantly diminished transcriptional activity. These findings suggest that haploinsufficiency or the dominant-negative effect caused by the NKX2-5 mutation is potentially an alternative pathological mechanism of DCM.

The association of NKX2-5 loss-of-function mutation with enhanced susceptibility to DCM has been previously investigated. Costa et al (53) performed mutational screening of NKX2-5 in 220 probands with adult-onset DCM, and identified three missense mutations in three probands in addition to three synonymous polymorphisms, with a mutational prevalence of approximately 1.36%. The three mutations included two previously reported mutations (p.R25C and p.A119S) and one novel mutation (p.I184M). The p.R25C and p.A119S mutations have been reported to result in reduced DNA binding and transactivation properties, and the former has been associated with congenital cardiovascular deformities and thyroid dysgenesis while the latter has been related to thyroid ectopy (49,5458). The novel p.I184M mutation was identified in a proband with familial DCM and genetic analysis of the family showed that the mutation was present in all affected family members. Functional analysis of p.I184M in vitro demonstrated a significant decrease in DNA binding activity despite the increased expression level of the mutant protein due to reduced degradation via the ubiquitin-proteasome system, resulting in the diminished activation of target genes. Notably, in a total of five mutation carriers of the family, one had DCM, atrial septal defect, atrial fibrillation and right bundle branch block; another had DCM, patent foramen ovale and complete heart block; a third had DCM, atrial septal defect and right bundle branch block; the other two had only tricuspid atresia and right bundle branch block, respectively (53). Similarly, in the present study, the mutation carriers also presented with DCM and arrhythmias, including atrial fibrillation and progressive cardiac conduction block, underlining the crucial role of NKX2-5 in the embryonic heart development and postnatal cardiac adaptation.

The findings that functionally defective NKX2-5 enhances the susceptibility to DCM may be partially attributed to the developmental and regenerative defects of the myocardium as well as abnormal heart remodeling (10). As a critical regulator of the cardiac gene network and heart development, NKX2-5 is highly expressed in early heart progenitor cells that commit to the cardiac lineage during embryogenesis, and continues abundant expression in the heart throughout adulthood (43,5962). Three independent NKX2-5-null mouse models showed a uniform phenotype of lethality between E9–10 associated with arrested heart tube looping morphogenesis and growth retardation, with the expression of several prominent cardiac structural and transcriptional regulatory genes downregulated (44,6367). NKX2-5 also plays an important part in the postnatal maturation and homeostasis of cardiomyocytes and the functional adaptation of adult heart. In a feline model of right-ventricular pressure overload conferred by banding of the pulmonary artery, or in adult mice with adrenergic-induced cardiac hypertrophy, the expression of NKX2-5 was upregulated, suggesting that NKX2-5 participates in the cardiac hypertrophic response during pressure overload or stress stimulation (68,69). By contrast, the expression of a dominant-negative human NKX2-5 mutant in the mouse heart under the control of α-myosin heavy chain (α-MHC) promoter induced cardiac dysfunction and degeneration, and injection of doxorubicin promoted more severe cardiac dysfunction and increased cardiomyocyte apoptosis (70,71). Furthermore, NKX2-5 has been documented to promote cardiomyocyte differentiation and modulate adult cardiac hypertrophic response through interacting with other cardiac transcription factors, such as TBX5, GATA4, serum response factor (SRF) and calmodulin binding transcription activator 2 (CAMTA2) (7276). In addition, NKX2-5 also regulates expression of gap junction protein connexin43 and sarcomere organization in postnatal cardiomyocytes (77), indicating the cardioprotective role of NKX2-5 as a survival factor in the heart.

Notably, a great number of NKX2-5 mutations have been previously associated with a wide variety of congenital cardiovascular anomalies, including atrial septal defect, ventricular septal defect, tetralogy of Fallot, double outlet right ventricle, L-transposition of the great artery and hypoplastic left heart syndrome (35,39,49,5457,7884). However, in the present study, the patients harboring the identified NKX2-5 mutation presented with DCM and arrhythmias, but without cardiovascular malformations. The remarkable discrepancy in the phenotypes of NKX2-5 genotypes may be explained by the following reasons. Firstly, considering some congenital cardiac structural aberrations may restore spontaneously, we cannot rule out the possibility that some mutation carriers had minor cardiac septal defects that closed shortly after birth on their own (26). Next, genomic imprinting results in preferential expression of the paternal or maternal allele of a certain gene, underlining the effect of proband (85). Thirdly, the nature of a mutation (loss-of-function, dominant-negative or gain-of-function effect) and its temporal and spatial effect during cardiac development (germline or somatic) are a potential explanation for this phenomenon (49,54). Fourthly, different genetic backgrounds, including possibly common SNPs altering disease susceptibility, are responsible for the marked phenotypic heterogeneity of the genotype (8688). Finally, mutations like p.S146W may be a genetic modifying factor that confers vulnerability to congenital heart diseases, rather than a direct cause, and environmental risk factors may be required for the onset of congenital cardiac abnormalities.

In conclusion, the data from the present study suggests that the NKX2-5 loss-of-function mutation contributes to the pathogenesis of DCM, suggesting potential implications in genetic testing that can help improve the care of patients and families with DCM.

Acknowledgments

The authors are extremely grateful to the participants for their participation in the study. The present study was supported in part by grants from the National Natural Science Fund of China (81270161, 81271927 and 81370301), the Natural Science Fund of Shanghai, China (13ZR1438400), the Fundamental Research Fund for Central University of Ministry of Education, China (15072), the Scientific Research Fund of Health Bureau, Shanghai, China (2010119) and the Excellent Discipline Leader Training Foundation of Pudong New Strict, Shanghai, China (PWRd2012-05).

References

1 

Garcia-Pavia P, Cobo-Marcos M, Guzzo-Merello G, Gomez-Bueno M, Bornstein B, Lara-Pezzi E, Segovia J and Alonso-Pulpon L: Genetics in dilated cardiomyopathy. Biomark Med. 7:517–533. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Hershberger RE, Hedges DJ and Morales A: Dilated cardiomyopathy: the complexity of a diverse genetic architecture. Nat Rev Cardiol. 10:531–547. 2013. View Article : Google Scholar : PubMed/NCBI

3 

McNally EM, Golbus JR and Puckelwartz MJ: Genetic mutations and mechanisms in dilated cardiomyopathy. J Clin Invest. 123:19–26. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Koutalas E, Kanoupakis E and Vardas P: Sudden cardiac death in non-ischemic dilated cardiomyopathy: a critical appraisal of existing and potential risk stratification tools. Int J Cardiol. 167:335–341. 2013. View Article : Google Scholar

5 

Yoshikawa T: Contribution of acquired factors to the pathogenesis of dilated cardiomyopathy. The cause of dilated cardiomyopathy: genetic or acquired? (Acquired-Side). Circ J. 75:1766–1773. 2011. View Article : Google Scholar

6 

Refaat MM, Lubitz SA, Makino S, Islam Z, Frangiskakis JM, Mehdi H, Gutmann R, Zhang ML, Bloom HL, MacRae CA, Dudley SC, Shalaby AA, Weiss R, McNamara DM, London B and Ellinor PT: Genetic variation in the alternative splicing regulator RBM20 is associated with dilated cardiomyopathy. Heart Rhythm. 9:390–396. 2012. View Article : Google Scholar :

7 

Wahbi K, Béhin A, Bécane HM, Leturcq F, Cossée M, Laforêt P, Stojkovic T, Carlier P, Toussaint M, Gaxotte V, Cluzel P, Eymard B and Duboc D: Dilated cardiomyopathy in patients with mutations in anoctamin 5. Int J Cardiol. 168:76–79. 2013. View Article : Google Scholar

8 

Flack E and Kannankeril PJ: The genetics of dilated cardiomyopathy. Heart Rhythm. 9:397–398. 2012. View Article : Google Scholar

9 

Pikkarainen S, Tokola H, Kerkelä R and Ruskoaho H: GATA transcription factors in the developing and adult heart. Cardiovasc Res. 63:196–207. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Oka T, Xu J and Molkentin JD: Re-employment of developmental transcription factors in adult heart disease. Semin Cell Dev Biol. 18:117–131. 2007. View Article : Google Scholar

11 

Kikuchi K, Holdway JE, Werdich AA, Anderson RM, Fang Y, Egnaczyk GF, Evans T, Macrae CA, Stainier DY and Poss KD: Primary contribution to zebrafish heart regeneration by gata4+ cardiomyocytes. Nature. 464:601–605. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Akazawa H and Komuro I: Cardiac transcription factor Csx/Nkx2–5: its role in cardiac development and diseases. Pharmacol Ther. 107:252–268. 2005. View Article : Google Scholar : PubMed/NCBI

13 

Kasahara A, Cipolat S, Chen Y, Dorn GW II and Scorrano L: Mitochondrial fusion directs cardiomyocyte differentiation via calcineurin and Notch signaling. Science. 342:734–737. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Cai H, Katoh-Kurasawa M, Muramoto T, Santhanam B, Long Y, Li L, Ueda M, Iglesias PA, Shaulsky G and Devreotes PN: Nucleocytoplasmic shuttling of a GATA transcription factor functions as a development timer. Science. 343:12495312014. View Article : Google Scholar : PubMed/NCBI

15 

Garg V, Kathiriya IS, Barnes R, Schluterman MK, King IN, Butler CA, Rothrock CR, Eapen RS, Hirayama-Yamada K, Joo K, Matsuoka R, Cohen JC and Srivastava D: GATA4 mutations cause human congenital heart defects and reveal an interaction with TBX5. Nature. 424:443–447. 2003. View Article : Google Scholar : PubMed/NCBI

16 

Rajagopal SK, Ma Q, Obler D, Shen J, Manichaikul A, Tomita-Mitchell A, Boardman K, Briggs C, Garg V, Srivastava D, Goldmuntz E, Broman KW, Benson DW, Smoot LB and Pu WT: Spectrum of heart disease associated with murine and human GATA4 mutation. J Mol Cell Cardiol. 43:677–685. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Yang YQ, Li L, Wang J, Liu XY, Chen XZ, Zhang W, Wang XZ, Jiang JQ, Liu X and Fang WY: A novel GATA4 loss-of-function mutation associated with congenital ventricular septal defect. Pediatr Cardiol. 33:539–546. 2012. View Article : Google Scholar

18 

Wang J, Sun YM and Yang YQ: Mutation spectrum of the GATA4 gene in patients with idiopathic atrial fibrillation. Mol Biol Rep. 39:8127–8135. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Yang YQ, Wang J, Liu XY, Chen XZ, Zhang W and Wang XZ: Mutation spectrum of GATA4 associated with congenital atrial septal defects. Arch Med Sci. 9:976–983. 2013. View Article : Google Scholar

20 

Yang YQ, Gharibeh L, Li RG, Xin YF, Wang J, Liu ZM, Qiu XB, Xu YJ, Xu L, Qu XK, Liu X, Fang WY, Huang RT, Xue S and Nemer G: GATA4 loss-of-function mutations underlie familial tetralogy of fallot. Hum Mutat. 34:1662–1671. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Yang YQ, Wang J, Wang XH, Wang Q, Tan HW, Zhang M, Shen FF, Jiang JQ, Fang WY and Liu X: Mutational spectrum of the GATA5 gene associated with familial atrial fibrillation. Int J Cardiol. 157:305–307. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Gu JY, Xu JH, Yu H and Yang YQ: Novel GATA5 loss-of-function mutations underlie familial atrial fibrillation. Clinics (Sao Paulo). 67:1393–1399. 2012. View Article : Google Scholar

23 

Jiang JQ, Li RG, Wang J, Liu XY, Xu YJ, Fang WY, Chen XZ, Zhang W, Wang XZ and Yang YQ: Prevalence and spectrum of GATA5 mutations associated with congenital heart disease. Int J Cardiol. 165:570–573. 2013. View Article : Google Scholar

24 

Wei D, Bao H, Zhou N, Zheng GF, Liu XY and Yang YQ: GATA5 loss-of-function mutation responsible for the congenital ventriculoseptal defect. Pediatr Cardiol. 34:504–511. 2013. View Article : Google Scholar

25 

Wei D, Bao H, Liu XY, Zhou N, Wang Q, Li RG, Xu YJ and Yang YQ: GATA5 loss-of-function mutations underlie tetralogy of fallot. Int J Med Sci. 10:34–42. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Wang XH, Huang CX, Wang Q, Li RG, Xu YJ, Liu X, Fang WY and Yang YQ: A novel GATA5 loss-of-function mutation underlies lone atrial fibrillation. Int J Mol Med. 31:43–50. 2013.

27 

Shi LM, Tao JW, Qiu XB, Wang J, Yuan F, Xu L, Liu H, Li RG, Xu YJ, Wang Q, Zheng HZ, Li X, Wang XZ, Zhang M, Qu XK and Yang YQ: GATA5 loss-of-function mutations associated with congenital bicuspid aortic valve. Int J Mol Med. 33:1219–1226. 2014.PubMed/NCBI

28 

Huang RT, Xue S, Xu YJ, Zhou M and Yang YQ: Somatic GATA5 mutations in sporadic tetralogy of Fallot. Int J Mol Med. 33:1227–1235. 2014.PubMed/NCBI

29 

Zheng GF, Wei D, Zhao H, Zhou N, Yang YQ and Liu XY: A novel GATA6 mutation associated with congenital ventricular septal defect. Int J Mol Med. 29:1065–1071. 2012.PubMed/NCBI

30 

Wang J, Luo XJ, Xin YF, Liu Y, Liu ZM, Wang Q, Li RG, Fang WY, Wang XZ and Yang YQ: Novel GATA6 mutations associated with congenital ventricular septal defect or tetralogy of fallot. DNA Cell Biol. 31:1610–1617. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Yang YQ, Wang XH, Tan HW, Jiang WF, Fang WY and Liu X: Prevalence and spectrum of GATA6 mutations associated with familial atrial fibrillation. Int J Cardiol. 155:494–496. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Yang YQ, Li L, Wang J, Zhang XL, Li RG, Xu YJ, Tan HW, Wang XH, Jiang JQ, Fang WY and Liu X: GATA6 loss-of-function mutation in atrial fibrillation. Eur J Med Genet. 55:520–526. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Li J, Liu WD, Yang ZL and Yang YQ: Novel GATA6 loss-of-function mutation responsible for familial atrial fibrillation. Int J Mol Med. 30:783–790. 2012.PubMed/NCBI

34 

Huang RT, Xue S, Xu YJ and Yang YQ: Somatic mutations in the GATA6 gene underlie sporadic tetralogy of Fallot. Int J Mol Med. 31:51–58. 2013.

35 

Schott JJ, Benson DW, Basson CT, Pease W, Silberbach GM, Moak JP, Maron BJ, Seidman CE and Seidman JG: Congenital heart disease caused by mutations in the transcription factor NKX2-5. Science. 281:108–111. 1998. View Article : Google Scholar : PubMed/NCBI

36 

Guntheroth W, Chun L, Patton KK, Matsushita MM, Page RL and Raskind WH: Wenckebach periodicity at rest that normalizes with tachycardia in a family with a NKX2.5 mutation. Am J Cardiol. 110:1646–1650. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Xie WH, Chang C, Xu YJ, Li RG, Qu XK, Fang WY, Liu X and Yang YQ: Prevalence and spectrum of Nkx2.5 mutations associated with idiopathic atrial fibrillation. Clinics (Sao Paulo). 68:777–784. 2013. View Article : Google Scholar

38 

Huang RT, Xue S, Xu YJ, Zhou M and Yang YQ: A novel NKX2.5 loss-of-function mutation responsible for familial atrial fibrillation. Int J Mol Med. 31:1119–1126. 2013.PubMed/NCBI

39 

McCulley DJ and Black BL: Transcription factor pathways and congenital heart disease. Curr Top Dev Biol. 100:253–277. 2012. View Article : Google Scholar : PubMed/NCBI

40 

Li RG, Li L, Qiu XB, Yuan F, Xu L, Li X, Xu YJ, Jiang WF, Jiang JQ, Liu X, Fang WY, Zhang M, Peng LY, Qu XK and Yang YQ: GATA4 loss-of-function mutation underlies familial dilated cardiomyopathy. Biochem Biophys Res Commun. 439:591–596. 2013. View Article : Google Scholar : PubMed/NCBI

41 

Zhao L, Xu JH, Xu WJ, Yu H, Wang Q, Zheng HZ, Jiang WF, Jiang JF and Yang YQ: A novel GATA4 loss-of-function mutation responsible for familial dilated cardiomyopathy. Int J Mol Med. 33:654–660. 2014.

42 

Brody MJ, Cho E, Mysliwiec MR, Kim TG, Carlson CD, Lee KH and Lee Y: Lrrc10 is a novel cardiac-specific target gene of Nkx2-5 and GATA4. J Mol Cell Cardiol. 62:237–246. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Lints TJ, Parsons LM, Hartley L, Lyons I and Harvey RP: Nkx-2.5: a novel murine homeobox gene expressed in early heart progenitor cells and their myogenic descendants. Development. 119:419–431. 1993.PubMed/NCBI

44 

Lyons I, Parsons LM, Hartley L, Li R, Andrews JE, Robb L and Harvey RP: Myogenic and morphogenetic defects in the heart tubes of murine embryos lacking the homeo box gene Nkx2-5. Genes Dev. 9:1654–1666. 1995. View Article : Google Scholar : PubMed/NCBI

45 

Prall OW, Menon MK, Solloway MJ, Watanabe Y, Zaffran S, Bajolle F, Biben C, McBride JJ, Robertson BR, Chaulet H, Stennard FA, Wise N, Schaft D, Wolstein O, Furtado MB, Shiratori H, Chien KR, Hamada H, Black BL, Saga Y, Robertson EJ, Buckingham ME and Harvey RP: An Nkx2-5/Bmp2/Smad1 negative feedback loop controls heart progenitor specification and proliferation. Cell. 128:947–959. 2007. View Article : Google Scholar : PubMed/NCBI

46 

Pashmforoush M, Lu JT, Chen H, Amand TS, Kondo R, Pradervand S, Evans SM, Clark B, Feramisco JR, Giles W, Ho SY, Benson DW, Silberbach M, Shou W and Chien KR: Nkx2-5 pathways and congenital heart disease; loss of ventricular myocyte lineage specification leads to progressive cardiomy-opathy and complete heart block. Cell. 117:373–386. 2004. View Article : Google Scholar : PubMed/NCBI

47 

Briggs LE, Takeda M, Cuadra AE, Wakimoto H, Marks MH, Walker AJ, Seki T, Oh SP, Lu JT, Sumners C, Raizada MK, Horikoshi N, Weinberg EO, Yasui K, Ikeda Y, Chien KR and Kasahara H: Perinatal loss of Nkx2-5 results in rapid conduction and contraction defects. Circ Res. 103:580–590. 2008. View Article : Google Scholar : PubMed/NCBI

48 

Takeda M, Briggs LE, Wakimoto H, Marks MH, Warren SA, Lu JT, Weinberg EO, Robertson KD, Chien KR and Kasahara H: Slow progressive conduction and contraction defects in loss of Nkx2-5 mice after cardiomyocyte terminal differentiation. Lab Invest. 89:983–993. 2009. View Article : Google Scholar : PubMed/NCBI

49 

Benson DW, Silberbach GM, Kavanaugh-McHugh A, Cottrill C, Zhang Y, Riggs S, Smalls O, Johnson MC, Watson MS, Seidman JG, Seidman CE, Plowden J and Kugler JD: Mutations in the cardiac transcription factor NKX2.5 affect diverse cardiac developmental pathways. J Clin Invest. 104:1567–1573. 1999. View Article : Google Scholar : PubMed/NCBI

50 

Kirk EP, Sunde M, Costa MW, Rankin SA, Wolstein O, Castro ML, Butler TL, Hyun C, Guo G, Otway R, Mackay JP, Waddell LB, Cole AD, Hayward C, Keogh A, Macdonald P, Griffiths L, Fatkin D, Sholler GF, Zorn AM, Feneley MP, Winlaw DS and Harvey RP: Mutations in cardiac T-box factor gene TBX20 are associated with diverse cardiac pathologies, including defects of septation and valvulogenesis and cardiomyopathy. Am J Hum Genet. 81:280–291. 2007. View Article : Google Scholar : PubMed/NCBI

51 

Brody MJ, Hacker TA, Patel JR, Feng L, Sadoshima J, Tevosian SG, Balijepalli RC, Moss RL and Lee Y: Ablation of the cardiac-specific gene leucine-rich repeat containing 10 (Lrrc10) results in dilated cardiomyopathy. PLoS One. 7:e516212012. View Article : Google Scholar : PubMed/NCBI

52 

Elliott P, O’Mahony C, Syrris P, Evans A, Rivera Sorensen C, Sheppard MN, Carr-White G, Pantazis A and McKenna WJ: Prevalence of desmosomal protein gene mutations in patients with dilated cardiomyopathy. Circ Cardiovasc Genet. 3:314–322. 2010. View Article : Google Scholar : PubMed/NCBI

53 

Costa MW, Guo G, Wolstein O, Vale M, Castro ML, Wang L, Otway R, Riek P, Cochrane N, Furtado M, Semsarian C, Weintraub RG, Yeoh T, Hayward C, Keogh A, Macdonald P, Feneley M, Graham RM, Seidman JG, Seidman CE, Rosenthal N, Fatkin D and Harvey RP: Functional characterization of a novel mutation in NKX2-5 associated with congenital heart disease and adult-onset cardiomyopathy. Circ Cardiovasc Genet. 6:238–247. 2013. View Article : Google Scholar : PubMed/NCBI

54 

Kasahara H, Lee B, Schott JJ, Benson DW, Seidman JG, Seidman CE and Izumo S: Loss of function and inhibitory effects of human CSX/NKX2.5 homeoprotein mutations associated with congenital heart disease. J Clin Invest. 106:299–308. 2000. View Article : Google Scholar : PubMed/NCBI

55 

Goldmuntz E, Geiger E and Benson DW: NKX2.5 mutations in patients with tetralogy of fallot. Circulation. 104:2565–2568. 2001. View Article : Google Scholar : PubMed/NCBI

56 

Stallmeyer B, Fenge H, Nowak-Göttl U and Schulze-Bahr E: Mutational spectrum in the cardiac transcription factor gene NKX2.5 (CSX) associated with congenital heart disease. Clin Genet. 78:533–540. 2010. View Article : Google Scholar : PubMed/NCBI

57 

Beffagna G, Cecchetto A, Dal Bianco L, Lorenzon A, Angelini A, Padalino M, Vida V, Bhattacharya S, Stellin G, Rampazzo A and Daliento L: R25C mutation in the NKX2.5 gene in Italian patients affected with non-syndromic and syndromic congenital heart disease. J Cardiovasc Med (Hagerstown). 14:582–586. 2013. View Article : Google Scholar

58 

Dentice M, Cordeddu V, Rosica A, Ferrara AM, Santarpia L, Salvatore D, Chiovato L, Perri A, Moschini L, Fazzini C, Olivieri A, Costa P, Stoppioni V, Baserga M, De Felice M, Sorcini M, Fenzi G, Di Lauro R, Tartaglia M and Macchia PE: Missense mutation in the transcription factor NKX2-5: a novel molecular event in the pathogenesis of thyroid dysgenesis. J Clin Endocrinol Metab. 91:1428–1433. 2006. View Article : Google Scholar : PubMed/NCBI

59 

Komuro I and Izumo S: Csx: a murine homeobox-containing gene specifically expressed in the developing heart. Proc Natl Acad Sci USA. 90:814581491993. View Article : Google Scholar

60 

Kasahara H, Bartunkova S, Schinke M, Tanaka M and Izumo S: Cardiac and extracardiac expression of Csx/Nkx2.5 homeodomain protein. Circ Res. 82:936–946. 1998. View Article : Google Scholar : PubMed/NCBI

61 

Stanley EG, Biben C, Elefanty A, Barnett L, Koentgen F, Robb L and Harvey RP: Efficient Cre-mediated deletion in cardiac progenitor cells conferred by a 3’UTR-ires-Cre allele of the homeobox gene Nkx2-5. Int J Dev Biol. 46:431–439. 2002.

62 

Schlesinger J, Schueler M, Grunert M, Fischer JJ, Zhang Q, Krueger T, Lange M, Tönjes M, Dunkel I and Sperling SR: The cardiac transcription network modulated by Gata4, Mef2a, Nkx2.5, Srf, histone modifications, and microRNAs. PLoS Genet. 7:e10013132011. View Article : Google Scholar : PubMed/NCBI

63 

Shin CH, Liu ZP, Passier R, Zhang CL, Wang DZ, Harris TM, Yamagishi H, Richardson JA, Childs G and Olson EN: Modulation of cardiac growth and development by HOP, an unusual homeodomain protein. Cell. 110:725–735. 2002. View Article : Google Scholar : PubMed/NCBI

64 

Tanaka M, Chen Z, Bartunkova S, Yamasaki N and Izumo S: The cardiac homeobox gene Csx/Nkx2.5 lies genetically upstream of multiple genes essential for heart development. Development. 126:1269–1280. 1999.PubMed/NCBI

65 

Biben C, Weber R, Kesteven S, Stanley E, McDonald L, Elliott DA, Barnett L, Köentgen F, Robb L, Feneley M and Harvey RP: Cardiac septal and valvular dysmorphogenesis in mice heterozygous for mutations in the homeobox gene Nkx2-5. Circ Res. 87:888–895. 2000. View Article : Google Scholar : PubMed/NCBI

66 

Biben C and Harvey RP: Homeodomain factor Nkx2-5 controls left/right asymmetric expression of bHLH gene eHand during murine heart development. Genes Dev. 11:1357–1369. 1997. View Article : Google Scholar : PubMed/NCBI

67 

Zou Y, Evans S, Chen J, Kuo HC, Harvey RP and Chien KR: CARP, a cardiac ankyrin repeat protein, is downstream in the Nkx2–5 homeobox gene pathway. Development. 124:793–804. 1997.PubMed/NCBI

68 

Thompson JT, Rackley MS and O’Brien TX: Upregulation of the cardiac homeobox gene Nkx2-5 (CSX) in feline right ventricular pressure overload. Am J Physiol. 274:H1569–H1573. 1998.PubMed/NCBI

69 

Saadane N, Alpert L and Chalifour LE: Expression of immediate early genes, GATA-4, and Nkx-2.5 in adrenergic-induced cardiac hypertrophy and during regression in adult mice. Br J Pharmacol. 127:1165–1176. 1999. View Article : Google Scholar : PubMed/NCBI

70 

Toko H, Zhu W, Takimoto E, Shiojima I, Hiroi Y, Zou Y, Oka T, Akazawa H, Mizukami M, Sakamoto M, Terasaki F, Kitaura Y, Takano H, Nagai T, Nagai R and Komuro I: Csx/Nkx2-5 is required for homeostasis and survival of cardiac myocytes in the adult heart. J Biol Chem. 277:24735–24743. 2002. View Article : Google Scholar : PubMed/NCBI

71 

Kasahara H, Wakimoto H, Liu M, Maguire CT, Converso KL, Shioi T, Huang WY, Manning WJ, Paul D, Lawitts J, Berul CI and Izumo S: Progressive atrioventricular conduction defects and heart failure in mice expressing a mutant Csx/Nkx2.5 homeoprotein. J Clin Invest. 108:189–201. 2001. View Article : Google Scholar : PubMed/NCBI

72 

Hiroi Y, Kudoh S, Monzen K, Ikeda Y, Yazaki Y, Nagai R and Komuro I: Tbx5 associates with Nkx2-5 and synergistically promotes cardiomyocyte differentiation. Nat Genet. 28:276–280. 2001. View Article : Google Scholar : PubMed/NCBI

73 

Sepulveda JL, Belaguli N, Nigam V, Chen CY, Nemer M and Schwartz RJ: GATA-4 and Nkx-2.5 coactivate Nkx-2 DNA binding targets: role for regulating early cardiac gene expression. Mol Cell Biol. 18:3405–3415. 1998.PubMed/NCBI

74 

Durocher D, Charron F, Warren R, Schwartz RJ and Nemer M: The cardiac transcription factors Nkx2-5 and GATA-4 are mutual cofactors. EMBO J. 16:5687–5696. 1997. View Article : Google Scholar : PubMed/NCBI

75 

Chen CY and Schwartz RJ: Recruitment of the tinman homolog Nkx-2.5 by serum response factor activates cardiac alpha-actin gene transcription. Mol Cell Biol. 16:6372–6384. 1996.PubMed/NCBI

76 

Song K, Backs J, McAnally J, Qi X, Gerard RD, Richardson JA, Hill JA, Bassel-Duby R and Olson EN: The transcriptional coactivator CAMTA2 stimulates cardiac growth by opposing class II histone deacetylases. Cell. 125:453–466. 2006. View Article : Google Scholar : PubMed/NCBI

77 

Kasahara H, Ueyama T, Wakimoto H, Liu MK, Maguire CT, Converso KL, Kang PM, Manning WJ, Lawitts J, Paul DL, Berul CI and Izumo S: Nkx2. 5 homeoprotein regulates expression of gap junction protein connexin 43 and sarcomere organization in postnatal cardiomyocytes. J Mol Cell Cardiol. 35:243–256. 2003. View Article : Google Scholar : PubMed/NCBI

78 

Gutierrez-Roelens I, Sluysmans T, Gewillig M, Devriendt K and Vikkula M: Progressive AV-block and anomalous venous return among cardiac anomalies associated with two novel missense mutations in the CSX/NKX2-5 gene. Hum Mutat. 20:75–76. 2002. View Article : Google Scholar : PubMed/NCBI

79 

Elliott DA, Kirk EP, Yeoh T, Chandar S, McKenzie F, Taylor P, Grossfeld P, Fatkin D, Jones O, Hayes P, Feneley M and Harvey RP: Cardiac homeobox gene NKX2-5 mutations and congenital heart disease: associations with atrial septal defect and hypoplastic left heart syndrome. J Am Coll Cardiol. 41:2072–2076. 2003. View Article : Google Scholar : PubMed/NCBI

80 

McElhinney DB, Geiger E, Blinder J, Benson DW and Goldmuntz E: NKX2.5 mutations in patients with congenital heart disease. J Am Coll Cardiol. 42:1650–1655. 2003. View Article : Google Scholar : PubMed/NCBI

81 

Reamon-Buettner SM and Borlak J: NKX2-5: an update on this hypermutable homeodomain protein and its role in human congenital heart disease (CHD). Hum Mutat. 31:1185–1194. 2010. View Article : Google Scholar : PubMed/NCBI

82 

Wang J, Xin YF, Liu XY, Liu ZM, Wang XZ and Yang YQ: A novel NKX2-5 mutation in familial ventricular septal defect. Int J Mol Med. 27:369–375. 2011.

83 

Qin X, Xing Q, Ma L, Meng H, Liu Y, Pang S and Yan B: Genetic analysis of an enhancer of the NKX2-5 gene in ventricular septal defects. Gene. 508:106–109. 2012. View Article : Google Scholar : PubMed/NCBI

84 

Huang W, Meng H, Qiao Y, Pang S, Chen D and Yan B: Two novel and functional DNA sequence variants within an upstream enhancer of the human NKX2-5 gene in ventricular septal defects. Gene. 524:152–155. 2013. View Article : Google Scholar : PubMed/NCBI

85 

Yan H, Yuan W, Velculescu VE, Vogelstein B and Kinzler KW: Allelic variation in human gene expression. Science. 297:11432002. View Article : Google Scholar : PubMed/NCBI

86 

Weisfeld-Adams JD, Edelmann L, Gadi IK and Mehta L: Phenotypic heterogeneity in a family with a small atypical microduplication of chromosome 22q11.2 involving TBX1. Eur J Med Genet. 55:732–736. 2012. View Article : Google Scholar : PubMed/NCBI

87 

Smith H, Galmes R, Gogolina E, Straatman-Iwanowska A, Reay K, Banushi B, Bruce CK, Cullinane AR, Romero R, Chang R, Ackermann O, Baumann C, Cangul H, Cakmak Celik F, Aygun C, Coward R, Dionisi-Vici C, Sibbles B, Inward C, Kim CA, Klumperman J, Knisely AS, Watson SP and Gissen P: Associations among genotype, clinical phenotype, and intracellular localization of trafficking proteins in ARC syndrome. Hum Mutat. 33:1656–1664. 2012. View Article : Google Scholar : PubMed/NCBI

88 

Soemedi R, Wilson IJ, Bentham J, Darlay R, Töpf A, Zelenika D, Cosgrove C, Setchfield K, Thornborough C, Granados-Riveron J, Blue GM, Breckpot J, Hellens S, Zwolinkski S, Glen E, Mamasoula C, Rahman TJ, Hall D, Rauch A, Devriendt K, Gewillig M, O’ Sullivan J, Winlaw DS, Bu’Lock F, Brook JD, Bhattacharya S, Lathrop M, Santibanez-Koref M, Cordell HJ, Goodship JA and Keavney BD: Contribution of global rare copy-number variants to the risk of sporadic congenital heart disease. Am J Hum Genet. 91:489–501. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2015
Volume 35 Issue 2

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yuan F, Qiu X, Li R, Qu X, Wang J, Xu Y, Liu X, Fang W, Yang Y, Liao D, Liao D, et al: A novel NKX2-5 loss-of-function mutation predisposes to familial dilated cardiomyopathy and arrhythmias. Int J Mol Med 35: 478-486, 2015
APA
Yuan, F., Qiu, X., Li, R., Qu, X., Wang, J., Xu, Y. ... Liao, D. (2015). A novel NKX2-5 loss-of-function mutation predisposes to familial dilated cardiomyopathy and arrhythmias. International Journal of Molecular Medicine, 35, 478-486. https://doi.org/10.3892/ijmm.2014.2029
MLA
Yuan, F., Qiu, X., Li, R., Qu, X., Wang, J., Xu, Y., Liu, X., Fang, W., Yang, Y., Liao, D."A novel NKX2-5 loss-of-function mutation predisposes to familial dilated cardiomyopathy and arrhythmias". International Journal of Molecular Medicine 35.2 (2015): 478-486.
Chicago
Yuan, F., Qiu, X., Li, R., Qu, X., Wang, J., Xu, Y., Liu, X., Fang, W., Yang, Y., Liao, D."A novel NKX2-5 loss-of-function mutation predisposes to familial dilated cardiomyopathy and arrhythmias". International Journal of Molecular Medicine 35, no. 2 (2015): 478-486. https://doi.org/10.3892/ijmm.2014.2029