TBX20 loss-of-function mutation contributes to double outlet right ventricle

  • Authors:
    • Yun Pan
    • Rui Geng
    • Ning Zhou
    • Gui-Fen Zheng
    • Hong Zhao
    • Juan Wang
    • Cui-Mei Zhao
    • Xing-Biao Qiu
    • Yi-Qing Yang
    • Xing-Yuan Liu
  • View Affiliations

  • Published online on: January 22, 2015     https://doi.org/10.3892/ijmm.2015.2077
  • Pages: 1058-1066
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Congenital heart disease (CHD), the most prevalent birth defect in humans worldwide, is still a leading non‑infectious cause of infant morbidity and mortality. Increasing evidence demonstrates that genetic risk factors play a key role in the pathogenesis of CHD, and more than 50 genes have been linked to various types of CHD. Nevertheless, CHD is a heterogeneous disorder and the genetic components underpinning CHD in an overwhelming majority of cases remain unknown. In the present study, the entire coding exons and flanking introns of the TBX20 gene, which codes for a T-box transcription factor essential for the proper development of the heart, were sequenced in a cohort of 146 unrelated patients with CHD. The available relatives of the index patient harboring an identified mutation and 200 unrelated ethnically matched healthy individuals used as the controls were also genotyped for TBX20. The functional characteristics of the TBX20 mutation were assayed by using a dual-luciferase reporter assay system. As a result, a novel heterozygous TBX20 mutation, p.R143W, was identified in an index patient with double outlet right ventricle (DORV). Genetic analyses of the pedigree of the proband revealed that in the family, the mutation co-segregated with DORV transmitted in an autosomal dominant pattern with complete penetrance. The missense mutation was absent in 400 control chromosomes and the altered amino acid was completely conserved evolutionarily across species. Functional analysis revealed that mutant TBX20 had a significantly diminished transcriptional activity compared with its wild-type counterpart. To the best of our knowledge, this study is the first to report the association of TBX20 loss-of-function mutation with increased susceptibility to DORV in humans, which provides novel insight into the molecular mechanisms responsible for CHD, suggesting potential implications for the antenatal prophylaxis of CHD.

Introduction

Congenital heart disease (CHD), a structural defect that arises from the abnormal formation of the heart or intrathoracic major blood vessel, is the most common form of birth defect in humans, affecting approximately 1% of all live births and accounting for as high as 10% of early miscarriages (13). Presently, CHD is still the leading non-infectious cause of infant morbidity and mortality worldwide, with approximately 27% of neonates who die of birth defects having cardiovascular developmental abnormalities (1). Congenital cardiovascular deformities are usually categorized into 25 distinct clinical types, of which 21 designate specific anatomic or hemodynamic lesions, including atrial septal defect, ventricular septal defect, tetraology of Fallot, patent ductus arteriosus, double outlet right ventricle (DORV), endocardial cushion defect, aortic stenosis, coarctation of the aorta, transposition of the great arteries, abnormal pulmonary venous connection, univentricular heart and hypoplastic left heart syndrome (1). Various types of CHD may occur separately or in combination, giving rise to a degraded quality of life, decreased exercise tolerance, delayed fetal brain development or brain injury, thromboembolic events, brain abscess or infective endocarditis, hypothyroidism, pulmonary arterial hypertension or Eisenmenger’s syndrome, cardiac dysfunction or heart failure, arrhythmias and even cardiac death (427). Although great advances in pediatric cardiovascular care have allowed most newborns with CHD to survive into adulthood, resulting in an increasing number of adults living with CHD, the surviving patients have a significantly increased incidence of late complications and sudden cardiac death (1228). Furthermore, due to the continued improvement of health outcomes for children with CHD, substantial mortality rates in CHD have shifted away from the young and towards adults, with a steady increase in age at death (29). Hence, CHD has imposed a vast economic burden on the families of patients and health care systems, and the socioeconomic burden is expected to be heavier in the future with the increasing number of adults with CHD (30). Despite the high prevalence and important clinical significance, the molecular mechanisms underlying CHD in a significant proportion of patients remain poorly understood.

The etiology of CHD is complex and is associated with both environmental and genetic causes (3134). Non-genetic risk factors for CHD mainly involve paternal characteristics and conditions, the use of maternal therapeutic and non-therapeutic drugs, and parental environmental exposures (3133), while an increasing number of studies on human genetics have demonstrated that genetic defects play an important role in the pathogenesis of CHD, and to date, a long list of mutations in >60 genes have been linked to isolated CHD or syndromic CHD, where CHD is part of the phenotypes (3455). Nevertheless, CHD is a heterogeneous disease, and the genetic origin underpinning CHD in an overwhelming majority of patients remains to be unveiled.

T-box transcription factors, which are characterized by the presence of a highly conserved sequence-specific DNA-binding domain termed the ‘T-box’, have been demonstrated to be essential for proper organ development (56). To date, at least 7 members of the T-box gene family, including TBX20 as an ancient member of the T-box superfamily, have been shown to be highly expressed in embryonic heart tissues in humans and vertebrates (56). In developing mouse embryos, TBX20 has been shown to be expressed in cardiac progenitor cells, as well as in the developing myocardium and endothelial cells associated with endocardial cushions, the precursor structures for the cardiac valves and the atrioventricular septum (57). In mice, the complete ablation of TBX20 has been shown to result in the death of embryos at mid-gestation with grossly abnormal heart morphogenesis, while heterozygous TBX20-knockout mice have shown mild atrial septal anomalies, including an increased prevalence of patent foramen ovale and primum atrial septal aneurysm, as well as a genetic predisposition to atrial septal defect (5860). Additionally, in entirely embryonic stem cell-derived mouse embryos, the knockdown of TBX20 using RNA interference has been shown to cause various defects in heart formation, including hypoplasia of the outflow tract, persistent truncus arteriosus and hypoplastic right ventricle (61). In humans, several mutations in TBX20 have been associated with atrial septal defect, ventricular septal defect and tetralogy of Fallot (6267). These results warrant the screening of TBX20 for a causative mutation in another subset of patients with CHD.

Materials and methods

Study participants

A cohort of 146 unrelated subjects affected by non-syndromic CHD was enrolled from the Chinese population in this study. The available relatives of the index patient harboring an identified TBX20 mutation were also included. All patients underwent a comprehensive clinical evaluation, including an evaluation of individual and medical histories, a detailed physical examination, 12-lead electrocardiogram, and two-dimensional transthoracic echocardiography with color flow Doppler. A transesophageal echocardiography or a cardiac catheterization examination was performed only when there was a strong clinical indication. The diagnosis of CHD was made by imaging and/or direct view during cardiac surgery. The patients with known chromosomal abnormalities or syndromic CHD, such as Turner syndrome, Down syndrome, Di George syndrome, Marfan syndrome and Holt-Oram syndrome were excluded from this study. The controls comprised 200 unrelated non-CHD individuals from the same geographic area, who were matched to the CHD patients in age, gender and ethnicity.

The present study was performed in accordance with the principles outlined in the Declaration of Helsinki of 1975 and as revised in 2008. The study protocol was reviewed and approved by the Ethics Committee of Tongji Hospital, Tongji University, Shanghai, China (ethical approval number for cases and controls: LL(H)-09-07; date of approval: July 27, 2009). Prior to the commencement of the study, all participants or their guardians provided written informed consent to the use of their blood specimens for genetic analysis.

Mutational scanning of TBX20

Peripheral venous blood samples were obtained from all the study participants. Genomic DNA was isolated from the white blood cells of each participant using the Wizard Genomic DNA Purification kit (Promega, Madison, WI, USA). According to the referential genomic DNA sequence of the TBX20 gene (GenBank accession no. NG_015805.1), the primers used to amplify the coding exons and flanking introns of TBX20 by polymerase chain reaction (PCR) were designed as shown in Table I. PCR was performed using HotStar TaqDNA Polymerase (Qiagen, Hilden, Germany) on a Verti Thermal Cycler (Applied Biosystems, Foster City, CA, USA) under standard conditions. Both strands of each PCR product were sequenced using a BigDye® Terminator version 3.1 Cycle Sequencing kit under an ABI PRISM 3130 XL DNA Analyzer (both from Applied Biosystems). For an identified sequence variation, the public databases for human sequence variations, including single nucleotide polymorphism (SNP; http://www.ncbi.nlm.nih.gov/SNP) and human gene mutation (HGM; http://www.hgmd.org) databases, were used to confirm its novelty.

Table I

The primers used to amplify the coding exons and flanking introns of TBX20.

Table I

The primers used to amplify the coding exons and flanking introns of TBX20.

ExonForward primerReverse primerSize (bp)
1 5′-tgtgcagctctggagtcgtt-3′ 5′-atctcccacccgcgatgtatg-3′420
2 5′-gggtcatccctacagcctgc-3′ 5′-agcaccctcaactacccagg-3′467
3 5′-agaggggtttgtggaccgga-3′ 5′-tgtccaggcttggaatgctct-3′435
4 5′-gcgtttgatcgaagcagacca-3′ 5′-cctcagggaacctgcacagt-3′482
5 5′-tggttctttcttgcctcactgt-3′ 5′-aggcagattggggtaggtgt-3′497
6 5′-caacctccagcctgtcctca-3′ 5′-tgtacaaggaatggggtgcag-3′446
7 5′-tcatggaatttcatattctt-3′ 5′-actggggccacttcctctat-3′919
8 5′-gccagtctgtgggagtgtac-3′ 5′-tggatctggattctcccatt-3′632
Multiple alignment of TBX20 amino acid sequences among species

Using the online Muscle program (version 3.6; http://www.ebi.ac.uk/Tools/msa/muscle/), the TBX20 human amino acid sequence was aligned with that of the chimpanzee, monkey, dog, cattle, mouse, fowl, zebrafish and frog.

Prediction of the causative potential of a TBX20 sequence variation

The disease-causing potential of a TBX20 sequence variation was predicted using MutationTaster (an online program at http://www.mutationtaster.org), which yielded a probability for a variation to be either a causative mutation or a benign polymorphism. Additionally, another online program PolyPhen-2 (http://genetics.bwh.harvard.edu/pph2/) was used to predict the possible pathogenic effect of an amino acid substitution occurring in the TBX20 protein.

Plasmid constructs and site-directed mutagenesis

Human heart cDNA was prepared as previously described (68). The full-length wild-type cDNA of the human TBX20 gene was amplified by PCR using PfuUltra High-Fidelity DNA Polymerase (Stratagene, La Jolla, CA, USA) and a pair of specific primers (5′-agtgctagcatggagttcacggcgtcccc-3′ and 5′-tgagcggccgctcatacaaatggcgtcatca-3′). The PCR fragment with a length of 1,364 base pairs (bp) was doubly digested by endonuclease NheI and NotI (Takara, Liaoning, China). The digested product was fractionated by 1.5% agarose gel electrophoresis, purified using the QIAquick Gel Extraction kit (Qiagen), and then subcloned into pcDNA3.1 (Promega) to construct the recombinant eukaryotic expression vector, pcDNA3.1-TBX20. The atrial natriuretic factor (ANF)-luciferase reporter gene, which contains the 2,600 bp 5′-flanking region of the ANF gene, namely ANF-luc, was kindly provided by Dr Ichiro Shiojima, from the Department of Cardiovascular Science and Medicine, Chiba University Graduate School of Medicine, Chiba, Japan. The identified mutation was introduced into wild-type TBX20 using the QuickChange II XL Site-Directed Mutagenesis kit (Stratagene) with a complementary pair of primers. The mutant was sequenced to confirm the desired mutation and to exclude any other sequence variations.

Reporter gene assay of mutant TBX20

Chinese hamster ovary (CHO) cells (from our cell bank) were cultured in Dulbecco’s modified Eagle’s medium supplemented with 10% fetal calf serum and maintained in 12-well plates under standard conditions for 48 h following transfection. One day prior to transfection, the CHO cells were plated at a density of 2×105 cells/well. PolyFect Transfection Reagent (Qiagen) was used for the transfection of the CHO cells according to the manufacturer’s instructions. The ANF-luc reporter plasmid and an internal control reporter plasmid pGL4.75 (hRluc/CMV; Promega) were used in the transient transfection assay to evaluate the transcriptional activity of the TBX20 mutant. Twenty-four hours after plating, the CHO cells were transfected with 0.8 μg of the wild-type or mutant pcDNA3.1-TBX20 expression vector, 1.0 μg of the ANF-luc reporter construct and 0.04 μg of the pGL4.75 control reporter vector. For co-transfection experiments, 0.4 μg of wild-type pcDNA3.1-TBX20 together with 0.4 μg of mutant pcDNA3.1-TBX20 or 0.4 μg of the empty pcDNA3.1 vector were used in the presence of 1.0 μg of ANF-luc and 0.04 μg of pGL4.75. The transfected cells were washed once the following day with medium before being refreshed with new medium. Forty-eight hours after transfection, the cells were lysed, and the Firefly luciferase and Renilla luciferase activities were measured using the Dual-Glo luciferase assay system (Promega). The activity of the ANF promoter was presented as the fold activation of Firefly luciferase relative to Renilla luciferase. Three independent experiments were conducted at minimum for wild-type and mutant TBX20.

Statistical analysis

Quantitative data are expressed as the means ± standard deviation (SD). The Student’s unpaired t-test was used to compare numeric variables between 2 groups. Comparisons of the categorical variables between 2 groups were made using Pearson’s χ2 test or Fisher’s exact test where appropriate. A two-tailed value of P<0.05 was considered to indicate a statistically significant difference.

Results

Clinical characteristics of the study population

A cohort of 146 unrelated patients with CHD (75 males and 71 females, with 32 having a family history of CHD) was clinically evaluated in contrast to a total of 200 unrelated non-CHD control individuals (102 males and 98 females, with no family history of CHD). All the patients were confirmed to suffer from CHD, while the control individuals had no evidence of structural cardiac abnormalities. None of them had established environmental risk factors for CHD, such as maternal illness and drug use in the first trimester of pregnancy, parental smoking and long-term exposure to toxicants and ionizing radiation. There was no difference in either gender or ethnicity between the patient and the control groups. The baseline clinical characteristics of the 146 patients with CHD are summarized in Table II.

Table II

Clinical characteristics of the 146 unrelated patients with CHD.

Table II

Clinical characteristics of the 146 unrelated patients with CHD.

ParameterStatistics
Male gender (%)75 (51)
Age (years)5±2
Positive family history (%)32 (22)
Distribution of different types of CHD
 Isolated CHD (%)66 (45)
 VSD (%)22 (15)
 ASD (%)20 (14)
 PDA (%)16 (11)
 ECD (%)5 (3)
 DORV (%)3 (2)
 Complex CHD (%)80 (55)
 TOF (%)19 (13)
 ASD + VSD (%)11 (8)
 DORV + VSD (%)10 (7)
 ASD + PDA (%)9 (6)
 VSD + PDA (%)9 (6)
 ECD + TGA (%)8 (5)
 TGA + VSD (%)4 (3)
 Other complication of CHD (%)10 (7)
Incidence of arrhythmia
 Atrial fibrillation (%)3 (2)
 Atrioventricular block (%)5 (3)
Treatment
 Surgical repair (%)93 (64)
 Catheter-based closure (%)35 (24)
 Follow-up (%)18 (12)

[i] CHD, congenital heart disease; VSD, ventricular septal defect; ASD, atrial septal defect; PDA, patent ductus arteriosus; ECD, endocardial cushion defect; DORV, double outlet of right ventricle; TOF, tetralogy of Fallot; TGA, transposition of great arteries.

Identification of a novel TBX20 mutation

By direct PCR sequencing of the TBX20 gene in the 146 unrelated patients with CHD, a novel mutation was identified in 1 patient, with a mutational prevalence of approximately 0.68%. Specifically, a substitution of thymine for guanine in the first nucleotide of codon 134 (c.400G>T), predicting the transition of arginine into tryptophane at amino acid position 134 (p.R134W), was detected in the index patient from family 1. The sequence chromatograms showing the discovered heterozygous TBX20 mutation compared with its control sequence are presented in Fig. 1. A schematic diagram depicting the location of the identified mutation in T-box structural domain of the TBX20 protein is presented in Fig. 2. The missense mutation was neither found in the 400 control chromosomes nor reported in the SNP and HGM databases (consulted again on November 25, 2014). Genetic screening of the family of the mutation carrier revealed that in the family, the mutation was present in all affected family members available, but absent in the unaffected family members examined. Analysis of the pedigree displayed that in the family, the mutation co-segregated with CHD transmitted in an autosomal dominant mode with complete penetrance (Fig. 3). In addition, the grandfather (I-1) and father (II-5) of the proband also had congenital ventricular septal defect. The phenotypic characteristics and status of the TBX20 mutation of the affected family members are listed in Table III.

Table III

Phenotypic characteristics and status of the TBX20 mutation in the affected family members.

Table III

Phenotypic characteristics and status of the TBX20 mutation in the affected family members.

IndividualGenderAge (years)Cardiac phenotypeTBX20 mutation
Family 1R134W
I-1M49aDORV, VSDNA
II-5M26DORV, VSD+/−
III-3F0DORV+/−

a Age at death. M, male; F, female; DORV, double outlet right ventricle; VSD, ventricular septal defect; NA, not available or not applicable; +/−, heterozygoty.

Alignment of multiple TBX20 amino acid sequences across species

As illustrated in Fig. 4, a cross-species alignment of the TBX20 amino acid sequences revealed that the altered amino acid arginine at position 134 was completely conserved evolutionarily.

Disease-causing potential of the TBX20 variation

The TBX20 sequence variation of c.400G>T was predicted to be causative by MutationTaster, with a P-value of 1.000. No SNPs in the altered region were found in the MutationTaster database. Moreover, the amino acid substitution, p.R134W, which occurred in TBX20 was also predicted to be possibly damaging by PolyPhen-2, with a score of 1.000 (sensitivity, 0.00; specificity, 1.00).

Diminished transcriptional activity of the R134W mutant of TBX20

The wild-type TBX20 (WT) and the R134W-mutant TBX20 (R134W) activated the ANF promoter by approximately 8- and 2-fold, respectively (Fig. 5). When wild-type TBX20 was co-expressed with the same amount of R134W-mutant TBX20, the induced activation of the ANF promoter was approximately 3-fold. These results indicate that the mutant TBX20 has a significantly reduced transcriptional activity compared with its wild-type counterpart.

Discussion

In the present study, a novel heterozygous mutation in TBX20, p.R134W, was identified in a family with DORV. In the family, the mutant allele was present in all the affected living family members, but absent in the unaffected relatives examined and 400 referral chromosomes from an ethnically-matched control population. A cross-species alignment of the TBX20 amino acid sequences revealed that the affected amino acid was completely conserved evolutionarily. The identified TBX20 sequence variation was predicted to be a pathogenic mutation, and functional analysis revealed that the mutant TBX20 was associated with a significantly decreased transcriptional activity. Therefore, it is very likely that functionally impaired TBX20 contributes to DORV in this family.

The human TBX20 gene maps on chromosome 7p14.3, which contains 8 exons and spans approximately 52 kb of the genomic DNA sequence, encoding a T-box transcription factor with 447 amino acids (GenBank accession no. NG_015805.1; NP_001071121.1). The T-box domain is a novel type of DNA-binding domain, which is responsible for DNA sequence recognition and binding to the consensus motif of GGTGT within the promoters of target genes, and is also required for interaction with co-factors (57). The identified mutation (p.R134W) is located in the T-box DNA binding domain (109-288 AA) of TBX20 (Fig. 2), and it is thus anticipated to exert an influence on the transcriptional activity of TBX20, possibly by interfering with the specific binding to target DNA sites of TBX20.

It has previously been verified that TBX20 regulates multiple downstream genes expressed during cardiac morphogenesis, including ANF, fibroblast growth factor 10 (FGF10), connexin 40 (Cx40) and connexin 45 (Cx45), of which ANF is the most extensively characterized T-box target gene in the heart (69). Hence, the functional characteristics of the TBX20 mutation may be explored by assaying the transcriptional activity of the ANF promoter. In this study, the functional effect of the novel p.R134W mutation of TBX20 identified in patients with familial DORV were deciphered by transcriptional activity assay and the results revealed a diminished transcriptional activation of the ANF promoter. These data indicate that genetically compromised TBX20 is potentially an alternative molecular pathogenesis of DORV.

It has been substantiated that TBX20 physically interacts with multiple cardiac transcription factors, including NK2 homeobox 5 (NKX2-5), GATA binding protein (GATA)4 and GATA5, collaborating to synergistically activate cardiac gene expression during embryogenesis (57), and multiple mutations in NKX2-5 (44,7073), GATA4 (37,40,41,48,7479) and GATA5 (42,43,46,49,50) have been causally linked to various types of CHD, which suggests that TBX20 loss-of-function mutation predisposes to CHD, possibly by reducing the synergistic activation of cardiac target genes with these partners.

It is interesting to note that ventricular septal defect was also observed in 2 mutation carriers with DORV. In fact, 9 mutations in the coding region of TBX20 (Fig. 2) have been previously associated with various congenital cardiovascular deformities. In a previous study, Kirk et al (62) screened the entire coding region of TBX20 by direct DNA sequencing in 353 probands with CHD, and found 2 heterozygous mutations (p.I152M and p.Q195X) within exons encoding the T-box DNA-binding domain in 2 probands with familial CHD, with a mutational prevalence of approximately 0.57%. Specifically, in family 1 carrying the missense change, p.I152M, which segregated with CHD over 3 generations, the proband had atrial septal defect; her grandmother had a small ventricular septal defect, and her mother had a large patent foramen ovale with a permanent left-to-right blood shunt. In family 2, the nonsense change, p.Q195X, which truncates TBX20 within the T-box DNA-binding domain, was present in 2 living affected individuals, of whom the proband had a small atrial septal defect, as well as mild coarctation of the aorta, and his mother had marked mitral valve prolapse with mild regurgitation. Functional assays revealed that I152M-mutant TBX20 had reduced function while Q195X-mutant TBX20 was severely compromised and most likely effectively null (62). In another study, using PCR sequencing, Liu et al (63) screened the exons 2 to 6 of TBX20 (which code for the T-box DNA-binding domain) in 203 unrelated patients with CHD, and discovered 3 non-synonymous mutations of p.A63T, p.I121F and p.T262M in 3 patients atrial septal defect, total anomalous pulmonary venous connection and tetralogy of Fallot, respectively, with a mutational prevalence of approximately 1.48%. However, a functional analysis of these new sequence variants remains to be carried out. In their study, Qian et al (64) scanned TBX20 in 192 unrelated children with CHD, and identified 2 mutations in 4 children, with a mutational prevalence of approximately 2.08%, including p.H186D found in 2 unrelated children (1 child had an atrioventicular canal with secundum and primum atrial septal defects, as well as a cleft mitral valve with moderate mitral regurgitation; another child had pentalogy of Fallot) and p.L197P identified in a female with pentalogy of Fallot and in another female with isolated atrial septal defect. However, the functional effects of the 2 mutations have not been characterized. Previously, Posch et al (65) sequenced the coding region of TBX20 in 170 unrelated patients with secundum atrial septal defect, and found a novel mutation, p.I121M in an index patient, with a mutational prevalence of approximately 0.59%. Genetic analysis of the pedigree of the proband revealed that the mutation co-segregated with CHD in the third generation kindred. A functional analysis demonstrated that TBX20-I121M resulted in a significantly enhanced transcriptional activity, which was further increased in the presence of co-transcription factors GATA4/5 and NKX2-5. By whole-exome sequencing in combination with a CHD-related gene filter, Liu et al (67) analyzed a family of 3 generations with atrial septal defect, and identified a novel TBX20 mutation, p.D176N. This mutation was predicted to be deleterious by bioinformatics programs (SIFT, PolyPhen-2 and MutationTaster), but experimental research of the mutation was not performed. These CHD-associated TBX20 mutations mentioned above are summarized in Table IV. Taken together, these findings highlight the exquisite sensitivity of the developing cardiovascular system to the level of TBX20, suggesting the pivotal role of TBX20 in the cardiovascular development.

Table IV

Summary of the TBX20 mutations previously identified in patients with congenital heart disease.

Table IV

Summary of the TBX20 mutations previously identified in patients with congenital heart disease.

Authors/(Refs.)Nucleotide changeAmino acid changeCardiac defect
Kirk et al (62) c.456C>G
c.583C>T
p.I152M
p.Q195X
ASD, VSD, PFO
ASD, CoA, MVP, MR
Liu et al (63) c.187G>A
c.361A>T
c.785C>T
p.A63T
p.I121F
p.T262M
ASD
TAPVC, ASD
TOF, PFO, PDA
Qian et al (64) c.597C>G
c.601T>C
p.H186D
p.L197P
ASD, TOF, CMV, MR
ASD, TOF
Posch et al (65)c.374C>Gp.I121MASD, TOF, CVD
Liu et al (67)c.526G>Ap.D176NASD

[i] ASD, atrial septal defect; VSD, ventricular septal defect; PFO, patent foramen ovale; CoA, coarctation of the aorta; MVP, mitral valve prolapse; MR, mitral regurgitation; TAPVC, total anomalous pulmonary venous connection; TOF, tetralogy of Fallot; PDA, patent ductus arteriosus; CMV, cleft mitral valve; CVD, cardiac valve defect.

In conclusion, to the very best of our knowledge, this is the first study on the association of TBX20 loss-of-function mutation with increased susceptibility to DORV in humans, which provides novel insight into the molecular mechanisms underlying DORV and suggests the potential implications for genetic counseling and medical care of the families affected by DORV.

Acknowledgments

The authors are greatly thankful to the participants for their participation in this investigation. This study was supported by the grants from the National Natural Science Fund of China (81270161, 81470372 and 81271927), the Natural Science Fund of Shanghai, China (15ZR1438100), the Key Program for Basic Research of Shanghai, China (14JC1405500), and the Key Project for Basic Research of Shanghai Chest Hospital, China (2014YZDH10102).

References

1 

Go AS, Mozaffarian D, Roger VL, Benjamin EJ, Berry JD, Blaha MJ, Dai S, Ford ES, Fox CS, Franco S, Fullerton HJ, Gillespie C, Hailpern SM, Heit JA, Howard VJ, Huffman MD, Judd SE, Kissela BM, Kittner SJ, Lackland DT, Lichtman JH, Lisabeth LD, Mackey RH, Magid DJ, Marcus GM, Marelli A, Matchar DB, McGuire DK, Mohler ER III, Moy CS, Mussolino ME, Neumar RW, Nichol G, Pandey DK, Paynter NP, Reeves MJ, Sorlie PD, Stein J, Towfighi A, Turan TN, Virani SS, Wong ND, Woo D and Turner MB; American Heart Association Statistics Committee and Stroke Statistics Subcommittee: Heart disease and stroke statistics - 2014 update: a report from the American Heart Association. Circulation. 129:e28–e292. 2014. View Article : Google Scholar

2 

van der Linde D, Konings EE, Slager MA, Witsenburg M, Helbing WA, Takkenberg JJ and Roos-Hesselink JW: Birth prevalence of congenital heart disease worldwide: a systematic review and meta-analysis. J Am Coll Cardiol. 58:2241–2247. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Zheng JY, Tian HT, Zhu ZM, Li B, Han L, Jiang SL, Chen Y, Li DT, He JC, Zhao Z, Cao Y, Qiu YG and Li TC: Prevalence of symptomatic congenital heart disease in Tibetan school children. Am J Cardiol. 112:1468–1470. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Müller J, Berner A, Ewert P and Hager A: Reduced health-related quality of life in older patients with congenital heart disease: a cross sectional study in 2360 patients. Int J Cardiol. 175:358–362. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Garcia Guerra G, Joffe AR, Robertson CM, Atallah J, Alton G, Sauve RS, Dinu IA, Ross DB and Rebeyka IM; Western Canadian Complex Pediatric Therapies Follow-up Group: Health-related quality of life experienced by children with chromosomal abnormalities and congenital heart defects. Pediatr Cardiol. 35:536–541. 2014. View Article : Google Scholar

6 

Cha KS, Cho KI, Seo JS, Choi JH, Park YH, Yang DH, Hong GR and Kim DS: Effects of inhaled iloprost on exercise capacity, quality of life, and cardiac function in patients with pulmonary arterial hypertension secondary to congenital heart disease (the Eisenmenger syndrome) (from the EIGER Study). Am J Cardiol. 112:1834–1839. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Müller J, Engelhardt A, Fratz S, Eicken A, Ewert P and Hager A: Improved exercise performance and quality of life after percutaneous pulmonary valve implantation. Int J Cardiol. 173:388–392. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Khalil A, Suff N, Thilaganathan B, Hurrell A, Cooper D and Carvalho JS: Brain abnormalities and neurodevelopmental delay in congenital heart disease: systematic review and meta-analysis. Ultrasound Obstet Gynecol. 43:14–24. 2014. View Article : Google Scholar

9 

Mulkey SB, Swearingen CJ, Melguizo MS, Schmitz ML, Ou X, Ramakrishnaiah RH, Glasier CM, Bradley Schaefer G and Bhutta AT: Multi-tiered analysis of brain injury in neonates with congenital heart disease. Pediatr Cardiol. 34:1772–1784. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Giglia TM, Massicotte MP, Tweddell JS, Barst RJ, Bauman M, Erickson CC, Feltes TF, Foster E, Hinoki K, Ichord RN, Kreutzer J, McCrindle BW, Newburger JW, Tabbutt S, Todd JL and Webb CL; American Heart Association Congenital Heart Defects Committee of the Council on Cardiovascular Disease in the Young; Council on Cardiovascular and Stroke Nursing; Council on Epidemiology and Prevention; Stroke Council: Prevention and treatment of thrombosis in pediatric and congenital heart disease: a scientific statement from the American Heart Association. Circulation. 128:2622–2703. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Rushani D, Kaufman JS, Ionescu-Ittu R, et al: Infective endocarditis in children with congenital heart disease: cumulative incidence and predictors. Circulation. 128:1412–1419. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Martinez-Quintana E, Rodriguez-Gonzalez F and Nieto-Lago V: Subclinical hypothyroidism in grown-up congenital heart disease patients. Pediatr Cardiol. 34:912–917. 2013. View Article : Google Scholar

13 

van Riel AC, Schuuring MJ, van Hessen ID, Zwinderman AH, Cozijnsen L, Reichert CL, Hoorntje JC, Wagenaar LJ, Post MC, van Dijk AP, Hoendermis ES, Mulder BJ and Bouma BJ: Contemporary prevalence of pulmonary arterial hypertension in adult congenital heart disease following the updated clinical classification. Int J Cardiol. 174:299–305. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Barst RJ, Ivy DD, Foreman AJ, McGoon MD and Rosenzweig EB: Four- and seven-year outcomes of patients with congenital heart disease-associated pulmonary arterial hypertension (from the REVEAL Registry). Am J Cardiol. 113:147–155. 2014. View Article : Google Scholar

15 

Dimopoulos K, Wort SJ and Gatzoulis MA: Pulmonary hypertension related to congenital heart disease: a call for action. Eur Heart J. 35:691–700. 2014. View Article : Google Scholar

16 

Friedman KG, McElhinney DB, Rhodes J, Powell AJ, Colan SD, Lock JE and Brown DW: Left ventricular diastolic function in children and young adults with congenital aortic valve disease. Am J Cardiol. 111:243–249. 2013. View Article : Google Scholar :

17 

Schuck R, Abd El Rahman MY, Rentzsch A, Hui W, Weng Y, Alexi-Meskishvili V, Lange PE, Berger F and Abdul-Khaliq H: Altered right ventricular function in the long-term follow-up evaluation of patients after delayed aortic reimplantation of the anomalous left coronary artery from the pulmonary artery. Pediatr Cardiol. 35:530–535. 2014. View Article : Google Scholar

18 

Zomer AC, Vaartjes I, van der Velde ET, de Jong HM, Konings TC, Wagenaar LJ, Heesen WF, Eerens F, Baur LH, Grobbee DE and Mulder BJ: Heart failure admissions in adults with congenital heart disease; risk factors and prognosis. Int J Cardiol. 168:2487–2493. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Stefanescu A, Macklin EA, Lin E, Dudzinski DM, Johnson J, Kennedy KF, Jacoby D, DeFaria Yeh D, Lewis GD, Yeh RW, Liberthson R, Lui G and Bhatt AB: Usefulness of the Seattle Heart Failure Model to identify adults with congenital heart disease at high risk of poor outcome. Am J Cardiol. 113:865–870. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Fahed AC, Roberts AE, Mital S and Lakdawala NK: Heart failure in congenital heart disease: a confluence of acquired and congenital. Heart Fail Clin. 10:219–227. 2014. View Article : Google Scholar

21 

Ueda A, Adachi I, McCarthy KP, Li W, Ho SY and Uemura H: Substrates of atrial arrhythmias: histological insights from patients with congenital heart disease. Int J Cardiol. 168:2481–2486. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Mondésert B, Abadir S and Khairy P: Arrhythmias in adult congenital heart disease: the year in review. Curr Opin Cardiol. 28:354–359. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Priromprintr B, Rhodes J, Silka MJ and Batra AS: Prevalence of arrhythmias during exercise stress testing in patients with congenital heart disease and severe right ventricular conduit dysfunction. Am J Cardiol. 114:468–472. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Khairy P, Van Hare GF, Balaji S, Berul CI, Cecchin F, Cohen MI, Daniels CJ, Deal BJ, Dearani JA, Groot Nd, Dubin AM, Harris L, Janousek J, Kanter RJ, Karpawich PP, Perry JC, Seslar SP, Shah MJ, Silka MJ, Triedman JK, Walsh EP and Warnes CA: PACES/HRS Expert Consensus Statement on the Recognition and Management of Arrhythmias in Adult Congenital Heart Disease: developed in partnership between the Pediatric and Congenital Electrophysiology Society (PACES) and the Heart Rhythm Society (HRS). Endorsed by the governing bodies of PACES, HRS, the American College of Cardiology (ACC), the American Heart Association (AHA), the European Heart Rhythm Association (EHRA), the Canadian Heart Rhythm Society (CHRS), and the International Society for Adult Congenital Heart Disease (ISACHD). Heart Rhythm. 11:e102–e165. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Moller JH and Anderson RC: A 43- to 54-year follow-up of 1,000 patients with congenital heart disease. Am J Cardiol. 111:1496–1500. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Koyak Z, Harris L, de Groot JR, Silversides CK, Oechslin EN, Bouma BJ, Budts W, Zwinderman AH, Van Gelder IC and Mulder BJ: Sudden cardiac death in adult congenital heart disease. Circulation. 126:1944–1954. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Walsh EP: Sudden death in adult congenital heart disease: risk stratification in 2014. Heart Rhythm. 11:1735–1742. 2014. View Article : Google Scholar : PubMed/NCBI

28 

van der Bom T, Zomer AC, Zwinderman AH, Meijboom FJ, Bouma BJ and Mulder BJ: The changing epidemiology of congenital heart disease. Nat Rev Cardiol. 8:50–60. 2011. View Article : Google Scholar

29 

Khairy P, Ionescu-Ittu R, Mackie AS, Abrahamowicz M, Pilote L and Marelli AJ: Changing mortality in congenital heart disease. J Am Coll Cardiol. 56:1149–1157. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Verheugt CL, Uiterwaal CS, van der Velde ET, Meijboom FJ, Pieper PG, Sieswerda GT, Plokker HW, Grobbee DE and Mulder BJ: The emerging burden of hospital admissions of adults with congenital heart disease. Heart. 96:872–878. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Patel SS and Burns TL: Nongenetic risk factors and congenital heart defects. Pediatr Cardiol. 34:1535–1555. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Gorini F, Chiappa E, Gargani L and Picano E: Potential effects of environmental chemical contamination in congenital heart disease. Pediatr Cardiol. 35:559–568. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Wang C, Zhou K, Xie L, Li Y, Zhan Y, Qiao L, Qin C, Liu R and Hua Y: Maternal medication use, fetal 3435 C>T polymorphism of the ABCB1 gene, and risk of isolated septal defects in a Han Chinese population. Pediatr Cardiol. 35:1132–1141. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Andersen TA, Troelsen KL and Larsen LA: Of mice and men: molecular genetics of congenital heart disease. Cell Mol Life Sci. 71:1327–1352. 2014. View Article : Google Scholar :

35 

Zaidi S, Choi M, Wakimoto H, Ma L, Jiang J, Overton JD, Romano-Adesman A, Bjornson RD, Breitbart RE, Brown KK, Carriero NJ, Cheung YH, Deanfield J, DePalma S, Fakhro KA, Glessner J, Hakonarson H, Italia MJ, Kaltman JR, Kaski J, Kim R, Kline JK, Lee T, Leipzig J, Lopez A, Mane SM, Mitchell LE, Newburger JW, Parfenov M, Pe’er I, Porter G, Roberts AE, Sachidanandam R, Sanders SJ, Seiden HS, State MW, Subramanian S, Tikhonova IR, Wang W, Warburton D, White PS, Williams IA, Zhao H, Seidman JG, Brueckner M, Chung WK, Gelb BD, Goldmuntz E, Seidman CE and Lifton RP: De novo mutations in histone-modifying genes in congenital heart disease. Nature. 498:220–223. 2013. View Article : Google Scholar : PubMed/NCBI

36 

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.

37 

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

38 

Wang J, Xin YF, Xu WJ, Liu ZM, Qiu XB, Qu XK, Xu L, Li X and Yang YQ: Prevalence and spectrum of PITX2c mutations associated with congenital heart disease. DNA Cell Biol. 32:708–716. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Lahm H, Deutsch MA, Dreßen M, Doppler S, Werner A, Hörer J, Cleuziou J, Schreiber C, Böhm J, Laugwitz KL, Lange R and Krane M: Mutational analysis of the human MESP1 gene in patients with congenital heart disease reveals a highly variable sequence in exon 1. Eur J Med Genet. 56:591–598. 2013. View Article : Google Scholar : PubMed/NCBI

40 

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

41 

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

42 

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

43 

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

44 

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

45 

Yuan F, Zhao L, Wang J, Zhang W, Li X, Qiu XB, Li RG, Xu YJ, Xu L, Qu XK, Fang WY and Yang YQ: PITX2c loss-of-function mutations responsible for congenital atrial septal defects. Int J Med Sci. 10:1422–1429. 2013. View Article : Google Scholar : PubMed/NCBI

46 

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

47 

Al Turki S, Manickaraj AK, Mercer CL, Gerety SS, Hitz MP, Lindsay S, D’Alessandro LC, Swaminathan GJ, Bentham J, Arndt AK, Low J, Breckpot J, Gewillig M, Thienpont B, Abdul-Khaliq H, Harnack C, Hoff K, Kramer HH, Schubert S, Siebert R, Toka O, Cosgrove C, Watkins H, Lucassen AM, O’Kelly IM, Salmon AP, Bu’lock FA, Granados-Riveron J, Setchfield K, Thornborough C, Brook JD, Mulder B, Klaassen S, Bhattacharya S, Devriendt K, Fitzpatrick DF; UK10K Consortium; Wilson DI, Mital S and Hurles ME: Rare variants in NR2F2 cause congenital heart defects in humans. Am J Hum Genet. 94:5745–5785. 2014.

48 

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.

49 

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

50 

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

51 

Wang X, Ji W, Wang J, Zhao P, Guo Y, Xu R, Chen S and Sun K: Identification of two novel GATA6 mutations in patients with nonsyndromic conotruncal heart defects. Mol Med Rep. 10:743–748. 2014.PubMed/NCBI

52 

Zhao L, Ni SH, Liu XY, Wei D, Yuan F, Xu L, Li X, Li RG, Qu XK, Xu YJ, Fang WY, Yang YQ and Qiu XB: Prevalence and spectrum of Nkx2.6 mutations in patients with congenital heart disease. Eur J Med Genet. 57:579–586. 2014. View Article : Google Scholar : PubMed/NCBI

53 

Cowan J, Tariq M and Ware SM: Genetic and functional analyses of ZIC3 variants in congenital heart disease. Hum Mutat. 35:66–75. 2014. View Article : Google Scholar :

54 

Wang J, Zhang DF, Sun YM and Yang YQ: A novel PITX2c loss-of-function mutation associated with familial atrial fibrillation. Eur J Med Genet. 57:25–31. 2014. View Article : Google Scholar

55 

Wei D, Gong XH, Qiu G, Wang J and Yang YQ: Novel PITX2c loss-of-function mutations associated with complex congenital heart disease. Int J Mol Med. 33:1201–1208. 2014.PubMed/NCBI

56 

Stennard FA and Harvey RP: T-box transcription factors and their roles in regulatory hierarchies in the developing heart. Development. 132:4897–4910. 2005. View Article : Google Scholar : PubMed/NCBI

57 

Stennard FA, Costa MW, Elliott DA, Rankin S, Haast SJP, Lai D, McDonald LPA, Niederreither K, Dolle P, Bruneau BG, Zorn AM and Harvey RP: Cardiac T-box factor Tbx20 directly interacts with Nkx2-5, GATA4, and GATA5 in regulation of gene expression in the developing heart. Dev Biol. 262:206–224. 2003. View Article : Google Scholar : PubMed/NCBI

58 

Stennard FA, Costa MW, Lai D, Biben C, Furtado M, Solloway MJ, McCulley DJ, Leimena C, Preis JI, Dunwoodie SL, Elliott DE, Prall OW, Black BL, Fatkin D and Harvey RP: Murine T-box transcription factor Tbx20 acts as a repressor during heart development, and is essential for adult heart integrity, function and adaptation. Development. 132:2451–2462. 2005. View Article : Google Scholar : PubMed/NCBI

59 

Cai CL, Zhou W, Yang L, Bu L, Qyang Y, Zhang X, Li X, Rosenfeld MG, Chen J and Evans S: T-box genes coordinate regional rates of proliferation and regional specification during cardiogenesis. Development. 132:2475–2487. 2005. View Article : Google Scholar : PubMed/NCBI

60 

Singh MK, Christoffels VM, Dias JM, Trowe MO, Petry M, Schuster-Gossler K, Burger A, Ericson J and Kispert A: Tbx20 is essential for cardiac chamber differentiation and repression of Tbx2. Development. 132:2697–2707. 2005. View Article : Google Scholar : PubMed/NCBI

61 

Takeuchi JJ, Mileikovskaia M, Koshiba-Takeuchi K, Heidt AB, Mori AD, Arruda EP, Gertsensein M, Georges R, Davidson L, Mo R, Hui CC, Henkelman RM, Nemer M, Black BL, Nagy A and Bruneau BG: Tbx20 dose-dependently regulates transcription factor networks required for mouse heart and motoneuron development. Development. 132:2463–2474. 2005. View Article : Google Scholar : PubMed/NCBI

62 

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

63 

Liu C, Shen A, Li X, Jiao W, Zhang X and Li Z: T-box transcription factor TBX20 mutations in Chinese patients with congenital heart disease. Eur J Med Genet. 51:580–587. 2008. View Article : Google Scholar : PubMed/NCBI

64 

Qian L, Mohapatra B, Akasaka T, Liu J, Ocorr K, Towbin JA and Bodmer R: Transcription factor neuromancer/TBX20 is required for cardiac function in Drosophila with implications for human heart disease. Proc Natl Acad Sci USA. 105:19833–19838. 2008. View Article : Google Scholar : PubMed/NCBI

65 

Posch MG, Gramlich M, Sunde M, Schmitt KR, Lee SH, Richter S, Kersten A, Perrot A, Panek AN, Al Khatib IH, Nemer G, Mégarbané A, Dietz R, Stiller B, Berger F, Harvey RP and Ozcelik C: A gain-of-function TBX20 mutation causes congenital atrial septal defects, patent foramen ovale and cardiac valve defects. J Med Genet. 47:230–235. 2010. View Article : Google Scholar :

66 

Qiao Y, Wanyan H, Xing Q, Xie W, Pang S, Shan J and Yan B: Genetic analysis of the TBX20 gene promoter region in patients with ventricular septal defects. Gene. 500:28–31. 2012. View Article : Google Scholar : PubMed/NCBI

67 

Liu JJ, Fan LL, Chen JL, Tan ZP and Yang YF: A novel variant in TBX20 (p.D176N) identified by whole-exome sequencing in combination with a congenital heart disease related gene filter is associated with familial atrial septal defect. J Zhejiang Univ Sci B. 15:830–837. 2014. View Article : Google Scholar : PubMed/NCBI

68 

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.

69 

Plageman TF and Yutzey KE: T-box genes and heart development: putting the ‘T’ in heart. Dev Dyn. 232:11–20. 2005. View Article : Google Scholar

70 

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

71 

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

72 

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

73 

Perera JL, Johnson NM, Judge DP and Crosson JE: Novel and highly lethal NKX2.5 missense mutation in a family with sudden death and ventricular arrhythmia. Pediatr Cardiol. 35:1206–1212. 2014. View Article : Google Scholar : PubMed/NCBI

74 

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

75 

Nemer G, Fadlalah F, Usta J, Nemer M, Dbaibo G, Obeid M and Bitar F: A novel mutation in the GATA4 gene in patients with Tetralogy of Fallot. Hum Mutat. 27:293–294. 2006. View Article : Google Scholar : PubMed/NCBI

76 

Tomita-Mitchell A, Maslen CL, Morris CD, Garg V and Goldmuntz E: GATA4 sequence variants in patients with congenital heart disease. J Med Genet. 44:779–783. 2007. View Article : Google Scholar : PubMed/NCBI

77 

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

78 

Wang E, Sun S, Qiao B, Duan W, Huang G, An Y, Xu S, Zheng Y, Su Z, Gu X, Jin L and Wang H: Identification of functional mutations in GATA4 in patients with congenital heart disease. PLoS One. 8:e621382013. View Article : Google Scholar : PubMed/NCBI

79 

Xiang R, Fan LL, Huang H, Cao BB, Li XP, Peng DQ and Xia K: A novel mutation of GATA4 (K319E) is responsible for familial atrial septal defect and pulmonary valve stenosis. Gene. 534:320–323. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2015
Volume 35 Issue 4

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
Pan Y, Geng R, Zhou N, Zheng G, Zhao H, Wang J, Zhao C, Qiu X, Yang Y, Liu X, Liu X, et al: TBX20 loss-of-function mutation contributes to double outlet right ventricle. Int J Mol Med 35: 1058-1066, 2015
APA
Pan, Y., Geng, R., Zhou, N., Zheng, G., Zhao, H., Wang, J. ... Liu, X. (2015). TBX20 loss-of-function mutation contributes to double outlet right ventricle. International Journal of Molecular Medicine, 35, 1058-1066. https://doi.org/10.3892/ijmm.2015.2077
MLA
Pan, Y., Geng, R., Zhou, N., Zheng, G., Zhao, H., Wang, J., Zhao, C., Qiu, X., Yang, Y., Liu, X."TBX20 loss-of-function mutation contributes to double outlet right ventricle". International Journal of Molecular Medicine 35.4 (2015): 1058-1066.
Chicago
Pan, Y., Geng, R., Zhou, N., Zheng, G., Zhao, H., Wang, J., Zhao, C., Qiu, X., Yang, Y., Liu, X."TBX20 loss-of-function mutation contributes to double outlet right ventricle". International Journal of Molecular Medicine 35, no. 4 (2015): 1058-1066. https://doi.org/10.3892/ijmm.2015.2077