Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
BMC Med Genomics ; 13(1): 175, 2020 11 20.
Article in English | MEDLINE | ID: mdl-33218365

ABSTRACT

BACKGROUND: Triphalangeal thumb-polysyndactyly syndrome (TPT-PS) is a rare well-defined autosomal dominant disorder characterized by long thumbs with three phalanges combined with pre- and postaxial polydactyly/syndactyly of limbs. By now, the syndrome has been reported in several large families from different ethnic backgrounds, with a high degree of inter- and intrafamilial variability. The genome locus responsible for TPT-PS has been mapped to the 7q36.3 region harboring a long-range sonic hedgehog (SHH) regulatory sequence (ZRS). Both single-nucleotide variants and complete duplications of ZRS were shown to cause TPT-PS and similar limb phenotypes. TPT-PS usually forms as isolated limb pathology not associated with additional malformations, in particular, with cardiovascular abnormalities. CASE PRESENTATION: Here we report on a rare Russian neonatal case of TPT-PS combined with severe congenital heart disease, namely double outlet right ventricle, and microphthalmia with optic disc coloboma. Pedigree analysis revealed TPT-PS of various expressivity in 10 family members throughout five generations, while the cardiac defect and the eye pathology were detected only in the proband. To extend the knowledge on genotype-phenotype spectrum of TPT-PS, the careful clinical and genomic analysis of the family was performed. High-resolution array-based comparative genomic hybridization (array-CGH) revealed a ~ 300 kb microduplication of 7q36.3 locus (arr[GRCh37] 7q36.3(156385810_156684811) × 3) that co-segregated with TPT-PS in the proband and her mother. The duplication encompassed three genes including LMBR1, the intron 5 of which is known to harbor ZRS. Based on whole-exome sequencing data, no additional pathogenic mutations or variants of uncertain clinical significance were found in morbid cardiac genes or genes associated with a microphthalmia/anophthalmia/coloboma spectrum of ocular malformations. CONCLUSIONS: The results support the previous data, indicating that complete ZRS duplication underlies TPT-PS, and suggest a broader phenotypic impact of the 7q36.3 microduplication. Potential involvement of the 7q36.3 microduplication in the patient's cardiac and eye malformations is discussed. However, the contribution of some additional genetic/epigenetic factors to the complex patient`s phenotype cannot be excluded entirely. Further comprehensive functional studies are needed to prove the possible involvement of the 7q36.3 locus in congenital heart disease and eye pathology.


Subject(s)
Abnormalities, Multiple/genetics , Chromosomes, Human, Pair 7/genetics , Coloboma/genetics , Congenital Abnormalities/genetics , Double Outlet Right Ventricle/genetics , Gene Duplication , Mandibulofacial Dysostosis/genetics , Microphthalmos/genetics , Optic Disk/abnormalities , Adult , Chromosomes, Human, Pair 7/ultrastructure , Comparative Genomic Hybridization , Female , Humans , Infant , Male , Membrane Proteins/genetics , Pedigree , Phenotype , Polymorphism, Single Nucleotide , Syndrome , Umbilical Arteries/abnormalities
2.
Int Heart J ; 60(5): 1113-1122, 2019 Sep 27.
Article in English | MEDLINE | ID: mdl-31484864

ABSTRACT

Occurring in about 1% of all live births, congenital heart defects (CHDs) represent the most frequent type of developmental abnormality and account for remarkably increased infant morbidity and mortality. Aggregating studies demonstrate that genetic components have a key role in the occurrence of CHDs. Nevertheless, due to pronounced genetic heterogeneity, the genetic causes of CHDs remain unclear in most patients. In this research, 114 unrelated patients affected with CHDs and 218 unrelated individuals without CHDs served as controls were recruited. The coding regions and splicing donors/acceptors of the ISL1 gene, which codes for a transcription factor required for proper cardiovascular development, were screened for mutations by sequencing in all study participants. The functional characteristics of an identified ISL1 mutation were delineated with a dual-luciferase reporter assay system. As a result, a new heterozygous ISL1 mutation, NM_002202.2: c.225C>G; p. (Tyr75*), was discovered in an index patient with double outlet right ventricle and ventricular septal defect. Analysis of the proband's family unveiled that the mutation co-segregated with the CHD phenotype. The nonsense mutation was absent in the 436 control chromosomes. Biological analysis showed that the mutant ISL1 protein had no transcriptional activity. Furthermore, the mutation nullified the synergistic activation between ISL1 and TBX20, another CHD-associated transcription factor. This research for the first time links an ISL1 loss-of-function mutation to double outlet right ventricle in humans, which adds insight to the molecular pathogenesis underpinning CHDs, suggesting potential implications for timely personalized management of CHD patients.


Subject(s)
Double Outlet Right Ventricle/genetics , Genes, Reporter/genetics , Genetic Predisposition to Disease/epidemiology , LIM-Homeodomain Proteins/genetics , Loss of Function Mutation/genetics , Transcription Factors/genetics , Case-Control Studies , Causality , Child, Preschool , China/epidemiology , Double Outlet Right Ventricle/diagnostic imaging , Female , Heart Defects, Congenital/diagnostic imaging , Heart Defects, Congenital/epidemiology , Heart Defects, Congenital/genetics , Heterozygote , Hospitals, University , Humans , Incidence , Infant , Male , Mutation , Pedigree , Prognosis , Retrospective Studies , Risk Assessment
3.
Pediatr Cardiol ; 40(7): 1339-1344, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31338559

ABSTRACT

Hand1 is a basic Helix-loop-Helix transcription factor that exhibits post-translationally regulated dimer partner choice that allows for a diverse set of Hand1 transcriptional complexes. Indeed, when Hand1 phosphoregulation is altered, conditionally activated hypophorylation (Hand1PO4-) and phosphorylation mimic (Hand1PO4+) Hand1 alleles disrupt both craniofacial and limb morphogenesis with 100% penetrance. Interestingly, activation of conditional Hand1 Phosphomutant alleles within post-migratory neural crest cells produce heart defects that include ventricular septal defects, double-outlet right ventricle, persistent truncus arteriosus with partial penetrance. Single versus double-lobed thymus is a distinguishing feature between Wnt1-Cre;Hand1PO4-/+ and Wnt1-Cre;Hand1PO4+/+ mice. These data show that although Hand1 dimer regulation plays critical and consistent roles in disrupting craniofacial and limb morphogenesis, Hand1 dimer regulation during cardiac outflow track formation is less critical for normal morphogenesis. This review will present the OFT phenotypes observed in Hand1 Phosphomutant mice, and discuss possible mechanisms of how penetrance differences within the same tissues within the same embryos could be variable.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Double Outlet Right Ventricle/genetics , Neural Crest/abnormalities , Alleles , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Humans , Mice , Morphogenesis , Phenotype , Transcription, Genetic
4.
BMC Genomics ; 19(1): 428, 2018 Jun 04.
Article in English | MEDLINE | ID: mdl-29866040

ABSTRACT

BACKGROUND: Congenital heart disease (CHD) is the leading non-infectious cause of death in infants. Monozygotic (MZ) twins share nearly all of their genetic variants before and after birth. Nevertheless, MZ twins are sometimes discordant for common complex diseases. The goal of this study is to identify genomic and epigenomic differences between a pair of twins discordant for a form of congenital heart disease, double outlet right ventricle (DORV). RESULTS: A monoamniotic monozygotic (MZ) twin pair discordant for DORV were subjected to genome-wide sequencing and methylation analysis. We identified few genomic differences but 1566 differentially methylated regions (DMRs) between the MZ twins. Twenty percent (312/1566) of the DMRs are located within 2 kb upstream of transcription start sites (TSS), containing 121 binding sites of transcription factors. Particularly, ZIC3 and NR2F2 are found to have hypermethylated promoters in both the diseased twin and additional patients suffering from DORV. CONCLUSIONS: The results showed a high correlation between hypermethylated promoters at ZIC3 and NR2F2 and down-regulated gene expression levels of these two genes in patients with DORV compared to normal controls, providing new insight into the potential mechanism of this rare form of CHD.


Subject(s)
Double Outlet Right Ventricle/genetics , Epigenomics , Twins, Monozygotic/genetics , COUP Transcription Factor II/genetics , Child, Preschool , DNA Methylation , Epigenesis, Genetic , Female , Gene Ontology , Homeodomain Proteins/genetics , Humans , Infant , Male , Promoter Regions, Genetic/genetics , Transcription Factors/genetics
5.
Mol Biol Rep ; 45(5): 1507-1513, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29923154

ABSTRACT

Ventricular septal defect (VSD) including outlet VSD of double outlet right ventricle (DORV) and perimembranous VSD are among the most common congenital heart diseases found at birth. HOXB1 encodes a homeodomain transcription factor essential for normal cardiac outflow tract development. The aim of the present study was to investigate the possible genetic effect of sequence variations in HOXB1 on VSD. The coding regions and splice junctions of the HOXB1 gene were sequenced in 57 unrelated VSD patients. As a result, a homozygous c.74_82dup (p.Pro28delinsHisSerAlaPro) variant was identified in one individual with DORV. We also identified five previously reported polymorphisms (rs35114525, rs12946855, rs14534040, rs12939811, and rs7207109) in 18 patients (12 DORV and 6 perimembranous VSD). Our study did not show any pathogenic alterations in the coding region of HOXB1 among patients with VSD. To our knowledge this is the first study investigating the role of HOXB1 in nonsyndromic VSD, which provide more insight on the etiology of this disease.


Subject(s)
Double Outlet Right Ventricle/genetics , Heart Septal Defects, Ventricular/genetics , Homeodomain Proteins/genetics , Child , Child, Preschool , Cohort Studies , Female , Heart Defects, Congenital/genetics , Heart Defects, Congenital/physiopathology , Heart Septal Defects, Ventricular/physiopathology , Homeodomain Proteins/physiology , Humans , Male , Transcription Factors
6.
Pediatr Cardiol ; 39(4): 794-804, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29468350

ABSTRACT

Congenital heart defect (CHD) represents the most prevalent birth defect, and accounts for substantial morbidity and mortality in humans. Aggregating evidence demonstrates the genetic basis for CHD. However, CHD is a heterogeneous disease, and the genetic determinants underlying CHD in most patients remain unknown. In the present study, a cohort of 186 unrelated cases with CHD and 300 unrelated control individuals were recruited. The coding exons and flanking introns of the MEF2C gene, which encodes a transcription factor crucial for proper cardiovascular development, were sequenced in all study participants. The functional effect of an identified MEF2C mutation was characterized using a dual-luciferase reporter assay system. As a result, a novel heterozygous MEF2C mutation, p.R15C, was detected in an index patient with congenital double outlet right ventricle (DORV) as well as ventricular septal defect. Analysis of the proband's pedigree showed that the mutation co-segregated with CHD with complete penetrance. The missense mutation, which changed the evolutionarily conserved amino acid, was absent in 300 control individuals. Functional deciphers revealed that the mutant MEF2C protein had a significantly decreased transcriptional activity. Furthermore, the mutation significantly reduced the synergistic activation between MEF2C and GATA4, another transcription factor linked to CHD. This study firstly associates MEF2C loss-of-function mutation with DORV in humans, which provides novel insight into the molecular pathogenesis of CHD, suggesting potential implications for genetic counseling and personalized treatment of CHD patients.


Subject(s)
Double Outlet Right Ventricle/genetics , Heart Septal Defects, Ventricular/genetics , Adolescent , Asian People , Child , Child, Preschool , Cohort Studies , Female , Genes, Reporter , Heterozygote , Humans , Infant , Infant, Newborn , MEF2 Transcription Factors/genetics , Male , Mutagenesis, Site-Directed/methods , Mutation, Missense , Pedigree , Polymerase Chain Reaction
7.
Eur J Med Genet ; 61(4): 197-203, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29222010

ABSTRACT

Congenital heart defect (CHD) is the most common type of birth defect in humans and a leading cause of infant morbidity and mortality. Previous studies have demonstrated that genetic defects play a pivotal role in the pathogenesis of CHD. However, the genetic basis of CHD remains poorly understood due to substantial genetic heterogeneity. In this study, the coding exons and splicing boundaries of the NR2F2 gene, which encodes a pleiotropic transcription factor required for normal cardiovascular development, were sequenced in 168 unrelated patients with CHD, and a novel mutation (c.247G > T, equivalent to p.G83X) was detected in a patient with double outlet right ventricle as well as ventricular septal defect. Genetic scanning of the mutation carrier's relatives available showed that the mutation was present in all affected family members but absent in unaffected family members. Analysis of the index patient's pedigree displayed that the mutation co-segregated with CHD, which was transmitted as an autosomal dominant trait with complete penetrance. The nonsense mutation was absent in 230 unrelated, ethnically-matched healthy individuals used as controls. Functional deciphers by using a dual-luciferase reporter assay system revealed that the mutant NR2F2 protein had no transcriptional activity as compared with its wild-type counterpart. Furthermore, the mutation abrogated the synergistic transcriptional activation between NR2F2 and GATA4, another core cardiac transcription factor associated with CHD. This study firstly associates NR2F2 loss-of-function mutation with an increased susceptibility to double outlet right ventricle in humans, which provides further significant insight into the molecular mechanisms underpinning CHD, suggesting potential implications for genetic counseling of CHD families and personalized treatment of CHD patients.


Subject(s)
COUP Transcription Factor II/genetics , Double Outlet Right Ventricle/genetics , Heart Septal Defects, Ventricular/genetics , Loss of Function Mutation , Adolescent , Adult , Animals , COS Cells , COUP Transcription Factor II/metabolism , Child , Child, Preschool , Chlorocebus aethiops , Double Outlet Right Ventricle/pathology , Female , Genetic Predisposition to Disease , HEK293 Cells , Heart Septal Defects, Ventricular/pathology , Humans , Infant , Male , Middle Aged , Penetrance
8.
Mol Med Rep ; 16(3): 2747-2754, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28677747

ABSTRACT

Congenital heart disease (CHD) is the most common form of birth defect in humans, and remains a leading non­infectious cause of infant mortality worldwide. An increasing number of studies have demonstrated that genetic defects serve a pivotal role in the pathogenesis of CHD, and mutations in >60 genes have been causally associated with CHD. CHD is a heterogeneous disease and the genetic basis of CHD in the majority of patients remains poorly understood. In the present study, the coding exons and flanking introns of the mesoderm posterior 1 (MESP1) gene, which encodes a basic helix­loop­helix transcription factor required for normal cardiovascular development, were sequenced in 178 unrelated patients with CHD. The available relatives of the index patient carrying an identified mutation and 200 unrelated, ethnically­matched healthy individuals, who were used as controls, were genotyped for MESP1. The functional characteristics of the MESP1 mutation were determined using a dual­luciferase reporter assay system. As a result, a novel de novo heterozygous MESP1 mutation, p.Q118X, was identified in an index patient with double outlet right ventricle (DORV) and a ventricular septal defect. The nonsense mutation was absent in the 400 reference chromosomes and the altered amino acid was completely conserved evolutionarily across species. Functional assays indicated that the mutant MESP1 protein had no transcriptional activity when compared with its wild­type counterpart. The present study firstly provided experimental evidence supporting the concept that a MESP1 loss­of­function mutation may contribute to the development of DORV in humans, which presents a significant insight into the molecular pathogenesis of CHD. The results highlight the potential implications for the genetic counseling and personalized treatment of patients with CHD.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Double Outlet Right Ventricle/genetics , Mutation, Missense , Adolescent , Adult , Amino Acid Sequence , Animals , Basic Helix-Loop-Helix Transcription Factors/chemistry , Child , Child, Preschool , Double Outlet Right Ventricle/pathology , Female , HEK293 Cells , Humans , Infant , Infant, Newborn , Male , Sequence Alignment , Transcriptional Activation , Young Adult
9.
Am J Med Genet A ; 173(9): 2446-2450, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28636109

ABSTRACT

Xq25q26 duplication syndrome has been reported in individuals with clinical features such as short stature, intellectual disability, syndromic facial appearance, small hands and feet, and genital abnormalities. The symptoms are related to critical chromosome regions including Xq26.1-26.3. In this particular syndrome, no patient with congenital heart disease was previously reported. Here, we report a 6-year-old boy with typical symptoms of Xq25q26 duplication syndrome and double outlet right ventricle (DORV) with pulmonary atresia (PA). He had the common duplicated region of Xq25q26 duplication syndrome extending to the distal region including the MOSPD1 locus. MOSPD1 regulates transforming growth factor beta (TGFß) 2,3 and may be responsible for cardiac development including DORV. In the patient's lymphocytes, mRNA expression of TGFß2 was lower than control, and might cause DORV as it does in TGFß2-deficient mice. Therefore, MOSPD1 is a possible candidate gene for DORV, probably in combination with GPC3. Further studies of the combined functions of MOSPD1 and GPC3 are needed, and identification of additional patients with MOSPD1 and GPC3 duplication should be pursued.


Subject(s)
Double Outlet Right Ventricle/genetics , Glypicans/genetics , Membrane Proteins/genetics , Sex Chromosome Disorders/genetics , Trisomy/genetics , Child , Chromosome Duplication/genetics , Chromosomes, Human, X/genetics , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/physiopathology , Double Outlet Right Ventricle/physiopathology , Dwarfism/genetics , Dwarfism/physiopathology , Ear/abnormalities , Ear/physiopathology , Humans , Intellectual Disability/genetics , Intellectual Disability/physiopathology , Intracellular Signaling Peptides and Proteins , Male , Neck/abnormalities , Neck/physiopathology , Sex Chromosome Aberrations , Sex Chromosome Disorders/physiopathology , Thorax/abnormalities , Thorax/physiopathology , Transforming Growth Factor beta/genetics , Trisomy/physiopathology
10.
Taiwan J Obstet Gynecol ; 56(1): 102-105, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28254208

ABSTRACT

OBJECTIVE: We present molecular cytogenetic characterization of Jacobsen syndrome (11q23.3-q25 deletion) in a fetus associated with double outlet right ventricle (DORV), hypoplastic left heart syndrome (HLHS), and ductus venosus (DV) agenesis on prenatal ultrasound. CASE REPORT: A 26-year-old woman underwent prenatal ultrasound examination at 22 weeks of gestation, which revealed intrauterine growth restriction, short femurs, DORV, HLHS, DV agenesis, single umbilical artery, and curly fourth toe of the left foot. The parents elected to terminate the pregnancy, and a 500-g female fetus was delivered at 23 weeks of gestation with facial dysmorphism, bilateral camptodactyly, and hammertoes. The parental karyotypes were normal. Cytogenetic analysis of the cord blood and umbilical cord revealed a karyotype of 46,XX,del(11)(q23). Array comparative genomic hybridization analysis of the DNA extracted from the umbilical cord revealed a 14.38-Mb deletion of 11q23.3-q25 encompassing BSX, ETS1, FLI1, and ARHGAP32. Metaphase fluorescence in situ hybridization analysis using the probes RP11-209L12 (11q25) and RP11-25M7 (11q11) showed a distal 11q deletion in the aberrant chromosome 11 in 17/17 cells examined. CONCLUSION: Prenatal diagnosis of DORV, HLHS, DV agenesis associated with intrauterine growth restriction and short limbs should include a differential diagnosis of Jacobsen syndrome.


Subject(s)
Chromosome Deletion , Double Outlet Right Ventricle/genetics , Hypoplastic Left Heart Syndrome/genetics , Jacobsen Distal 11q Deletion Syndrome/genetics , Abnormalities, Multiple/genetics , Adult , Chromosomes, Human, Pair 11 , Comparative Genomic Hybridization , Double Outlet Right Ventricle/diagnostic imaging , Female , Fetal Growth Retardation/genetics , Gestational Age , Humans , Hypoplastic Left Heart Syndrome/diagnostic imaging , In Situ Hybridization, Fluorescence , Karyotyping , Pregnancy , Ultrasonography, Prenatal , Umbilical Veins/abnormalities , Vena Cava, Inferior/abnormalities
11.
Int J Mol Med ; 39(3): 711-718, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28112363

ABSTRACT

Congenital heart defects (CHDs), a wide variety of developmental abnormalities in the structures of the heart and the great thoracic blood vessels, are the most common form of birth defect in humans worldwide. CHDs are accountable for substantial morbidity and are still the leading cause of birth defect­related deaths. Recent studies have demonstrated the pivotal roles of genetic defects in the pathogenesis of CHDs, and a great number of genetic mutations have been associated with CHDs. Nevertheless, CHDs are a genetically heterogeneous disorder and the genetic basis underlying CHDs in an overwhelming majority of cases remains unclear. In the present study, the coding exons and flanking introns of the heart and neural crest derivatives expressed transcript 1 (HAND1) gene, which encodes a basic helix­loop­helix transcription factor crucial for cardiovascular development, were sequenced in 158 unrelated patients with CHDs, and a de novo heterozygous mutation, p.K132X, was identified in a patient with double outlet right ventricle (DORV), as well as ventricular septal defect. The nonsense mutation, which was predicted to produce a truncated HAND1 protein lacking 84 carboxyl­terminal amino acids, was absent in 600 control chromosomes. Functional analyses revealed that the HAND1 K132X mutant had no transcriptional activity. Furthermore, the mutation disrupted the synergistic activation between HAND1 and GATA binding protein 4 (GATA4), another cardiac core transcription factor causally linked to CHDs. To the best of our knowledge, this is the first report on the association of HAND1 loss­of­function mutation with an enhanced susceptibility to DORV in humans. These findings expand the phenotypic spectrum linked to HAND1 mutations, suggesting potential implications for the development of novelo prophylactic and therapeutic strategies for DORV.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Double Outlet Right Ventricle/genetics , Mutation , Amino Acid Sequence , Amino Acid Substitution , Animals , Basic Helix-Loop-Helix Transcription Factors/chemistry , Case-Control Studies , Cell Line , Child , Child, Preschool , DNA Mutational Analysis , Double Outlet Right Ventricle/diagnosis , Double Outlet Right Ventricle/surgery , Female , Gene Silencing , Humans , Infant , Infant, Newborn , Male , Mice , Phenotype
12.
Am J Med Genet A ; 167A(5): 1134-41, 2015 May.
Article in English | MEDLINE | ID: mdl-25810372

ABSTRACT

We describe a 6-year-old male, diagnosed at birth with double outlet right ventricle (DORV), anterior aorta, multiple ventricular septal defects, pulmonary stenosis, microcephaly and mildly dysmorphic craniofacial findings. Chromosomal analysis showed a normal male karyotype but on subsequent array comparative genomic hybridization (array CGH) analysis a de novo 2.5 Mb loss in chromosome 13q at 13q33.3q34, together with an inherited gain at 4p12, were detected. The propositus underwent placement of a Blalock Taussig shunt and subsequently a Glenn and Fontan operation was performed. In this report we propose that COL4A1 and COL4A2 may be candidate genes for congenital heart disease (CHD) in individuals with a deletion in 13q within the 6Mb critical region for cardiac development proposed by Huang et al., [2012].


Subject(s)
Chromosome Deletion , Craniofacial Abnormalities/genetics , Double Outlet Right Ventricle/genetics , Heart Defects, Congenital/genetics , Microcephaly/genetics , Child , Chromosomes, Human, Pair 13/genetics , Collagen Type IV/genetics , Comparative Genomic Hybridization , Craniofacial Abnormalities/physiopathology , Developmental Disabilities/genetics , Developmental Disabilities/physiopathology , Double Outlet Right Ventricle/physiopathology , Genetic Association Studies , Heart Defects, Congenital/physiopathology , Humans , Male , Microcephaly/physiopathology , Serine Endopeptidases/genetics
13.
Int J Mol Med ; 35(4): 1058-66, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25625280

ABSTRACT

Congenital heart disease (CHD), the most prevalent birth defect in humans worldwide, is still a leading non­infectious 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.


Subject(s)
Double Outlet Right Ventricle/genetics , Genetic Association Studies , Mutation , T-Box Domain Proteins/genetics , Amino Acid Sequence , Amino Acid Substitution , Child , Child, Preschool , DNA Mutational Analysis , Double Outlet Right Ventricle/diagnosis , Female , Genetic Variation , Humans , Male , Molecular Sequence Data , Pedigree , Phenotype , T-Box Domain Proteins/chemistry , Transcription, Genetic
14.
Eur J Med Genet ; 57(10): 579-86, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25195019

ABSTRACT

Congenital heart disease (CHD) is the most common form of birth defect and is the most prevalent non-infectious cause of infant death. A growing body of evidence documents that genetic defects are involved in the pathogenesis of CHD. However, CHD is a genetically heterogeneous disease and the genetic basis underpinning CHD in an overwhelming majority of patients remain unclear. In this study, the coding exons and flanking introns of the Nkx2.6 gene, which codes for a homeodomain-containing transcription factor important for normal cardiovascular development, were sequenced in 320 unrelated patients with CHD, and two novel heterozygous Nkx2.6 mutations, p.V176M and p.K177X, were identified in two unrelated patients with CHD, respectively, including a patient with tetralogy of Fallot and a patient with double outlet of right ventricle and ventricular septal defect. The mutations were absent in 400 control chromosomes and the altered amino acids were completely conserved evolutionarily across species. Due to unknown transcriptional targets of Nkx2.6, the functional consequences of the identified mutations at transcriptional activity were evaluated by using Nkx2.5 as a surrogate. Alignment between human Nkx2.6 and Nkx2.5 proteins showed that V176M-mutant Nkx2.6 was equivalent to V182M-mutant Nkx2.5 and K177X-mutant Nkx2.6 was equal to K183X-mutant Nkx2.5, and introduction of V182M or K183X into Nkx2.5 significantly diminished its transcriptional activating function when compared with its wild-type counterpart. To our knowledge, this is the first report on the association of Nkx2.6 loss-of-function mutation with increased susceptibility to tetralogy of Fallot or double outlet of right ventricle and ventricular septal defect, providing novel insight into the molecular mechanism of CHD.


Subject(s)
Heart Defects, Congenital/genetics , Homeodomain Proteins/genetics , Mutation , Asian People/genetics , Double Outlet Right Ventricle/genetics , Female , Heart Septal Defects, Ventricular/genetics , Homeobox Protein Nkx-2.5 , Humans , Male , Tetralogy of Fallot/genetics , Transcription Factors/genetics
15.
Morphologie ; 98(321): 86-9, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24656633

ABSTRACT

The Wolf-Hirschhorn syndrome (WHS) encompasses deletions at the distal part of the short arm of one chromosome 4 (4p16 region). Clinical signs frequently include a typical facial appearance, mental retardation, intrauterine and postnatal growth retardation, hypotonia with decreased muscle bulk and seizures besides congenital heart malformations, midline defects, urinary tract malformations and brain, hearing and ophthalmologic malformations. Pathogenesis of WHS is multigenic and many factors are involved in prediction of prognosis such as extent of deletion, the occurrence of severe chromosome anomalies, the severe of seizures, the existence of serious internal, mainly cardiac, abnormalities and the degree of mental retardation. The phenotype of adult with WHS is in general similar to that of childhood being facial dysmorphism, growth retardation and mental retardation the rule in both adults and children. Avoid long-term complications and provide rehabilitation programs and genetic counseling may be essential in these patients.


Subject(s)
Wolf-Hirschhorn Syndrome/pathology , Adolescent , Chromosome Deletion , Chromosomes, Human, Pair 4/genetics , Chromosomes, Human, Pair 4/ultrastructure , Chromosomes, Human, Pair 8/genetics , Chromosomes, Human, Pair 8/ultrastructure , Double Outlet Right Ventricle/genetics , Epilepsy, Generalized/genetics , Facies , Female , Hallux Valgus/genetics , Humans , In Situ Hybridization, Fluorescence , Intellectual Disability/genetics , Kyphosis/genetics , Male , Phenotype , Translocation, Genetic , Wolf-Hirschhorn Syndrome/genetics
16.
Mol Biol Rep ; 41(4): 2671-7, 2014.
Article in English | MEDLINE | ID: mdl-24469719

ABSTRACT

Tetralogy of Fallot (TOF) and double outlet right ventricle (DORV) are two common subtypes of conotruncal defects. Recent reports have implicated mutations in the zinc finger protein, FOG family member 2 (ZFPM2/FOG2) as a cause of TOF/DORV, but no current literature focuses on the relationship between ZFPM2/FOG2 gene and non-syndromic TOF and DORV in Chinese Han population. The purpose of this study was to estimate the occurrence and the prevalence of ZFPM2/FOG2 genetic variants in Chinese Han population with non-syndromic TOF and DORV and to investigate genotype-phenotype correlations in individuals with ZFPM2/FOG2 mutations. The whole exons of ZFPM2/FOG2 were sequenced in 98 non-syndromic TOF/DORV patients and 200 control subjects. All the six variants (G2482A, G1552A, A2107C, C452T, C3239T, C1208G) changed the amino acid (p.Val828Met, p.Ala518Thr, p.Met703Leu, p.Thr151Ile, p.Ser1080Phe, p.Ala403Gly), in which four variants (G2482A, C452T, G1552A, C3239T) were not reported before and absent in control subjects. Further analysis revealed that only occurrences of variants G2482A and A2107C had statistical significance compared to the control group (P < 0.05). In conclusion, our results provide strong evidence regarding the susceptibility of the ZFPM2 gene to the development of non-syndromic TOF/DORV. It suggests that ZFPM2/FOG2 genetic variants may be a novel potential bio-markers and treatment targets for the non-syndromic TOF and DORV.


Subject(s)
DNA-Binding Proteins/genetics , Double Outlet Right Ventricle/genetics , Genetic Variation , Tetralogy of Fallot/genetics , Transcription Factors/genetics , Alleles , Amino Acid Sequence , Amino Acid Substitution , Case-Control Studies , Child , Child, Preschool , DNA Mutational Analysis , Exons , Female , Gene Order , Genetic Linkage , Genotype , Humans , Infant , Male , Molecular Sequence Data , Polymorphism, Single Nucleotide
17.
Circ Res ; 114(6): 957-65, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24478334

ABSTRACT

RATIONALE: Our previous study has shown that yes-associated protein (YAP) plays a crucial role in the phenotypic modulation of vascular smooth muscle cells (SMCs) in response to arterial injury. However, the role of YAP in vascular SMC development is unknown. OBJECTIVE: The goal of this study was to investigate the functional role of YAP in cardiovascular development in mice and determine the mechanisms underlying YAP's actions. METHODS AND RESULTS: YAP was deleted in cardiomyocytes and vascular SMCs by crossing YAP flox mice with SM22α-Cre transgenic mice. Cardiac/SMC-specific deletion of YAP directed by SM22α-Cre resulted in perinatal lethality in mice because of profound cardiac defects including hypoplastic myocardium, membranous ventricular septal defect, and double outlet right ventricle. The cardiac/SMC-specific YAP knockout mice also displayed severe vascular abnormalities including hypoplastic arterial wall, short/absent brachiocephalic artery, and retroesophageal right subclavian artery. Deletion of YAP in mouse vascular SMCs induced expression of a subset of cell cycle arrest genes including G-protein-coupled receptor 132 (Gpr132). Silencing Gpr132 promoted SMC proliferation, whereas overexpression of Gpr132 attenuated SMC growth by arresting cell cycle in G0/G1 phase, suggesting that ablation of YAP-induced impairment of SMC proliferation was mediated, at least in part, by induction of Gpr132 expression. Mechanistically, YAP recruited the epigenetic repressor histone deacetylase-4 to suppress Gpr132 gene expression via a muscle CAT element in the Gpr132 gene. CONCLUSIONS: YAP plays a critical role in cardiac/SMC proliferation during cardiovascular development by epigenetically regulating expression of a set of cell cycle suppressors.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Cardiovascular Abnormalities/genetics , Fetal Heart/physiology , Gene Expression Regulation, Developmental/physiology , Myocytes, Cardiac/cytology , Myocytes, Smooth Muscle/cytology , Phosphoproteins/physiology , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Aneurysm/genetics , Animals , Brachiocephalic Trunk/abnormalities , Cardiovascular Abnormalities/embryology , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Cell Division , Cells, Cultured , Double Outlet Right Ventricle/embryology , Double Outlet Right Ventricle/genetics , Gene Expression Regulation, Developmental/genetics , Genes, Lethal , Genes, cdc , Heart Septal Defects, Ventricular/embryology , Heart Septal Defects, Ventricular/genetics , Histone Deacetylases/metabolism , Histone Deacetylases/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Muscle, Smooth, Vascular/pathology , Myocytes, Cardiac/pathology , Myocytes, Smooth Muscle/pathology , Phosphoproteins/deficiency , Phosphoproteins/genetics , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering/pharmacology , Rats , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/biosynthesis , Receptors, G-Protein-Coupled/genetics , Subclavian Artery/abnormalities , YAP-Signaling Proteins
18.
Matrix Biol ; 35: 253-65, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24200809

ABSTRACT

Impaired heparan sulfate (HS) synthesis in vertebrate development causes complex malformations due to the functional disruption of multiple HS-binding growth factors and morphogens. Here, we report developmental heart defects in mice bearing a targeted disruption of the HS-generating enzyme GlcNAc N-deacetylase/GlcN N-sulfotransferase 1 (NDST1), including ventricular septal defects (VSD), persistent truncus arteriosus (PTA), double outlet right ventricle (DORV), and retroesophageal right subclavian artery (RERSC). These defects closely resemble cardiac anomalies observed in mice made deficient in the cardiogenic regulator fibroblast growth factor 8 (FGF8). Consistent with this, we show that HS-dependent FGF8/FGF-receptor2C assembly and FGF8-dependent ERK-phosphorylation are strongly reduced in NDST1(-/-) embryonic cells and tissues. Moreover, WNT1-Cre/LoxP-mediated conditional targeting of NDST function in neural crest cells (NCCs) revealed that their impaired HS-dependent development contributes strongly to the observed cardiac defects. These findings raise the possibility that defects in HS biosynthesis may contribute to congenital heart defects in humans that represent the most common type of birth defect.


Subject(s)
Heart Defects, Congenital/genetics , Heart/embryology , Heparitin Sulfate/metabolism , Neural Crest/metabolism , Organogenesis/physiology , Animals , DNA Primers/genetics , Double Outlet Right Ventricle/genetics , Fibroblast Growth Factor 8/genetics , Fibroblast Growth Factor 8/metabolism , Heart Defects, Congenital/pathology , Heart Septal Defects, Ventricular/genetics , Heparitin Sulfate/biosynthesis , Immunohistochemistry , Mice , Mice, Knockout , Neural Crest/embryology , Reverse Transcriptase Polymerase Chain Reaction , Subclavian Artery/abnormalities , Sulfotransferases/genetics , Sulfotransferases/metabolism , Truncus Arteriosus, Persistent/genetics
19.
Pediatrics ; 132(1): 161-5, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23733790

ABSTRACT

A prenatal diagnosis of ductal-dependent, complex congenital heart disease was made in a fetus with trisomy 18. The parents requested that the genetic diagnosis be excluded from all medical and surgical decision-making and that all life-prolonging therapies be made available to their infant. There was conflict among the medical team about what threshold of neonatal benefit could outweigh maternal and neonatal treatment burdens. A prenatal ethics consultation was requested.


Subject(s)
Chromosomes, Human, Pair 18/genetics , Double Outlet Right Ventricle/diagnosis , Double Outlet Right Ventricle/genetics , Double Outlet Right Ventricle/therapy , Ethics, Medical , Heart Ventricles/abnormalities , Prenatal Diagnosis/ethics , Trisomy , Abortion, Eugenic , Animals , Cesarean Section/ethics , Cooperative Behavior , Cost of Illness , Echocardiography , Ethics Consultation , Female , Fetal Death , Fetal Growth Retardation/diagnosis , Humans , Infant, Newborn , Interdisciplinary Communication , Life Support Care/ethics , Parental Consent/ethics , Pregnancy , Religion and Medicine , Risk Assessment , Treatment Refusal , Ultrasonography, Prenatal
20.
Am J Med Genet A ; 161A(4): 792-802, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23427188

ABSTRACT

Disease causing mutations for heterotaxy syndrome were first identified in the X-linked laterality gene, ZIC3. Mutations typically result in males with situs ambiguus and complex congenital heart disease; however affected females and one male with isolated d-transposition of the great arteries (d-TGA) have been reported. We hypothesized that a subset of patients with heart defects common to heterotaxy but without laterality defects would have ZIC3 mutations. We also sought to estimate the prevalence of ZIC3 mutations in sporadic heterotaxy. Patients with TGA (n = 169), double outlet right ventricle (DORV; n = 89), common atrioventricular canal (CAVC; n = 41), and heterotaxy (n = 54) underwent sequencing of ZIC3 exons. We tested 90 patients with tetralogy of Fallot (TOF) to correlate genotype with phenotype. Three potentially disease-related missense mutations were detected: c.49G > T (Gly17Cys) in a female with isolated DORV, c.98C > T (Ala33Val) in a male with isolated d-TGA, and c.841C > T (His281Tyr) in a female with sporadic heterotaxy. We also identified a novel insertion (CPFP333ins) in a family with heterotaxy. All were absent in 200 control patients and the 1000 Genomes Project (n = 629). No significant mutations were found in patients with TOF. Functional studies demonstrated reduced transcriptional activity of the ZIC3 His281Tyr mutant protein. ZIC3 mutations were rarely identified in isolated DORV and d-TGA suggesting that a subset of DORV and d-TGA may fall within the spectrum of laterality defects. ZIC3 mutations were found in 3.7% of patients with sporadic heterotaxy; therefore testing should be considered in patients with heterotaxy.


Subject(s)
Double Outlet Right Ventricle/diagnosis , Homeodomain Proteins/genetics , Mutation , Phenotype , Transcription Factors/genetics , Transposition of Great Vessels/diagnosis , Transposition of Great Vessels/genetics , Amino Acid Sequence , Amino Acid Substitution , Animals , Base Sequence , Cell Line , Double Outlet Right Ventricle/genetics , Female , Gene Expression , Genes, Reporter , Genotype , Homeodomain Proteins/metabolism , Humans , Male , Mice , Molecular Sequence Data , Open Reading Frames , Pedigree , Sequence Alignment , Transcription Factors/metabolism , Trinucleotide Repeat Expansion
SELECTION OF CITATIONS
SEARCH DETAIL
...