Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
Circulation ; 145(8): 606-619, 2022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35113653

RESUMO

BACKGROUND: The pathogenic missense variant p.G125R in TBX5 (T-box transcription factor 5) causes Holt-Oram syndrome (also known as hand-heart syndrome) and early onset of atrial fibrillation. Revealing how an altered key developmental transcription factor modulates cardiac physiology in vivo will provide unique insights into the mechanisms underlying atrial fibrillation in these patients. METHODS: We analyzed ECGs of an extended family pedigree of Holt-Oram syndrome patients. Next, we introduced the TBX5-p.G125R variant in the mouse genome (Tbx5G125R) and performed electrophysiologic analyses (ECG, optical mapping, patch clamp, intracellular calcium measurements), transcriptomics (single-nuclei and tissue RNA sequencing), and epigenetic profiling (assay for transposase-accessible chromatin using sequencing, H3K27ac [histone H3 lysine 27 acetylation] CUT&RUN [cleavage under targets and release under nuclease sequencing]). RESULTS: We discovered high incidence of atrial extra systoles and atrioventricular conduction disturbances in Holt-Oram syndrome patients. Tbx5G125R/+ mice were morphologically unaffected and displayed variable RR intervals, atrial extra systoles, and susceptibility to atrial fibrillation, reminiscent of TBX5-p.G125R patients. Atrial conduction velocity was not affected but systolic and diastolic intracellular calcium concentrations were decreased and action potentials were prolonged in isolated cardiomyocytes of Tbx5G125R/+ mice compared with controls. Transcriptional profiling of atria revealed the most profound transcriptional changes in cardiomyocytes versus other cell types, and identified over a thousand coding and noncoding transcripts that were differentially expressed. Epigenetic profiling uncovered thousands of TBX5-p.G125R-sensitive, putative regulatory elements (including enhancers) that gained accessibility in atrial cardiomyocytes. The majority of sites with increased accessibility were occupied by Tbx5. The small group of sites with reduced accessibility was enriched for DNA-binding motifs of members of the SP (specificity protein) and KLF (Krüppel-like factor) families of transcription factors. These data show that Tbx5-p.G125R induces changes in regulatory element activity, alters transcriptional regulation, and changes cardiomyocyte behavior, possibly caused by altered DNA binding and cooperativity properties. CONCLUSIONS: Our data reveal that a disease-causing missense variant in TBX5 induces profound changes in the atrial transcriptional regulatory network and epigenetic state in vivo, leading to arrhythmia reminiscent of those seen in human TBX5-p.G125R variant carriers.


Assuntos
Anormalidades Múltiplas , Regulação da Expressão Gênica , Cardiopatias Congênitas , Comunicação Interatrial , Heterozigoto , Deformidades Congênitas das Extremidades Inferiores , Mutação de Sentido Incorreto , Linhagem , Proteínas com Domínio T , Deformidades Congênitas das Extremidades Superiores , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/metabolismo , Substituição de Aminoácidos , Animais , Fibrilação Atrial/genética , Fibrilação Atrial/metabolismo , Feminino , Átrios do Coração/metabolismo , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/metabolismo , Comunicação Interatrial/genética , Comunicação Interatrial/metabolismo , Humanos , Deformidades Congênitas das Extremidades Inferiores/genética , Deformidades Congênitas das Extremidades Inferiores/metabolismo , Masculino , Camundongos , Camundongos Mutantes , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Deformidades Congênitas das Extremidades Superiores/genética , Deformidades Congênitas das Extremidades Superiores/metabolismo
2.
Cardiovasc Res ; 118(3): 859-871, 2022 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-33956078

RESUMO

AIMS: Congenital heart disease (CHD) frequently occurs in newborns due to abnormal formation of the heart or major blood vessels. Mutations in the GATA4 gene, which encodes GATA binding protein 4, are responsible for atrial septal defect (ASD), a common CHD. This study aims to gain insights into the molecular mechanisms of CHD using human-induced pluripotent stem cells (iPSCs) from a family cohort with ASD. METHODS AND RESULTS: Patient-specific iPSCs possess the same genetic information as the donor and can differentiate into various cell types from all three germ layers in vitro, thus presenting a promising approach for disease modelling and molecular mechanism research. Here, we generated a patient-specific iPSC line (iPSC-G4T280M) from a family cohort carrying a hereditary ASD mutation in GATA4 gene (T280M), as well as a human embryonic stem cell line (ESC-G4T280M) carrying the isogenic T280M mutation using the CRISPR/Cas9 genome editing method. The GATA4-mutant iPSCs and ESCs were then differentiated into cardiomyocytes (CMs) to model GATA4 mutation-associated ASD. We observed an obvious defect in cell proliferation in cardiomyocytes derived from both GATA4T280M-mutant iPSCs (iPSC-G4T280M-CMs) and ESCs (ESC-G4T280M-CMs), while the impaired proliferation ability of iPSC-G4T280M-CMs could be restored by gene correction. Integrated analysis of RNA-Seq and ChIP-Seq data indicated that FGF16 is a direct target of wild-type GATA4. However, the T280M mutation obstructed GATA4 occupancy at the FGF16 promoter region, leading to impaired activation of FGF16 transcription. Overexpression of FGF16 in GATA4-mutant cardiomyocytes rescued the cell proliferation defect. The direct relationship between GATA4T280M and ASD was demonstrated in a human iPSC model for the first time. CONCLUSIONS: In summary, our study revealed the molecular mechanism of the GATA4T280M mutation in ASD. Understanding the roles of the GATA4-FGF16 axis in iPSC-CMs will shed light on heart development and provide novel insights for the treatment of ASD and other CHD disorders.


Assuntos
Fatores de Crescimento de Fibroblastos , Comunicação Interatrial , Células-Tronco Pluripotentes Induzidas , Linhagem Celular , Células-Tronco Embrionárias , Fatores de Crescimento de Fibroblastos/genética , Fator de Transcrição GATA4/genética , Comunicação Interatrial/genética , Comunicação Interatrial/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Recém-Nascido , Mutação , Miócitos Cardíacos/metabolismo
3.
Sci Rep ; 11(1): 20082, 2021 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-34635725

RESUMO

Invariant Natural Killer T (iNKT) cells respond to the ligation of lipid antigen-CD1d complexes via their T-cell receptor and are implicated in various immunometabolic diseases. We considered that immunometabolic factors might affect iNKT cell function. To this end, we investigated iNKT cell phenotype and function in a cohort of adolescents with chronic disease and immunometabolic abnormalities. We analyzed peripheral blood iNKT cells of adolescents with cystic fibrosis (CF, n = 24), corrected coarctation of the aorta (CoA, n = 25), juvenile idiopathic arthritis (JIA, n = 20), obesity (OB, n = 20), and corrected atrial septal defect (ASD, n = 25) as controls. To study transcriptional differences, we performed RNA sequencing on a subset of obese patients and controls. Finally, we performed standardized co-culture experiments using patient plasma, to investigate the effect of plasma factors on iNKT cell function. We found comparable iNKT cell numbers across patient groups, except for reduced iNKT cell numbers in JIA patients. Upon ex-vivo activation, we observed enhanced IFN-γ/IL-4 cytokine ratios in iNKT cells of obese adolescents versus controls. The Th1-skewed iNKT cell cytokine profile of obese adolescents was not explained by a distinct transcriptional profile of the iNKT cells. Co-culture experiments with patient plasma revealed that across all patient groups, obesity-associated plasma factors including LDL-cholesterol, leptin, and fatty-acid binding protein 4 (FABP4) coincided with higher IFN-γ production, whereas high HDL-cholesterol and insulin sensitivity (QUICKI) coincided with higher IL-4 production. LDL and HDL supplementation in co-culture studies confirmed the effects of lipoproteins on iNKT cell cytokine production. These results suggest that circulating immunometabolic factors such as lipoproteins may be involved in Th1 skewing of the iNKT cell cytokine response in immunometabolic disease.


Assuntos
Artrite Juvenil/imunologia , Fibrose Cística/imunologia , Comunicação Interatrial/imunologia , Células T Matadoras Naturais/imunologia , Obesidade/fisiopatologia , Células Th1/imunologia , Adolescente , Artrite Juvenil/metabolismo , Artrite Juvenil/patologia , Estudos de Casos e Controles , Doença Crônica , Estudos Transversais , Fibrose Cística/metabolismo , Fibrose Cística/patologia , Citocinas/metabolismo , Feminino , Comunicação Interatrial/metabolismo , Comunicação Interatrial/patologia , Humanos , Interferon gama/metabolismo , Masculino
4.
Eur J Med Genet ; 64(11): 104314, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34481090

RESUMO

OBJECTIVE: Atrial septal defect, secundum (ASD Ⅱ, OMIM: 603642) is the second common congenital heart defect (CHD) in China. However, the genetic etiology of familial ASD II remains elusive. METHODS AND RESULTS: Using whole-exome sequencing (WES) and Sanger sequencing, we identified a novel myosin heavy chain 6 (MYH6) gene insertion variation, NM_002471.3: c.5465_5470dup (Arg1822_Glu1823dup), in a large Chinese Han family with ASD II. The variant Arg1822_Glu1823dup co-segregated with the disease in this family with autosomal dominant inheritance. The insertion variant located in the coiled-coil domain of the MYH6 protein, which is highly conserved across homologous myosin proteins and species. In transfected myoblast C2C12 cell lines, the MYH6 Arg1822_Glu1823dup variant significantly impaired myofibril formation and increased apoptosis but did not significantly reduce cell viability. Furthermore, molecular simulations revealed that the Arg1822_Glu1823dup variant impaired the myosin α-helix, increasing the stability of the coiled-coil myosin dimer, suggesting that this variant has an effect on the coiled-coil domain self-aggregation. These findings indicate that Arg1822_Glu1823dup variant plays a crucial role in the pathogenesis of ASD II. CONCLUSION: Our findings expand the spectrum of MYH6 variations associated with familial ASD II and may provide a molecular basis in genetic counseling and prenatal diagnosis for this Chinses family.


Assuntos
Miosinas Cardíacas/genética , Comunicação Interatrial/genética , Mutagênese Insercional , Cadeias Pesadas de Miosina/genética , Adulto , Animais , Apoptose , Miosinas Cardíacas/química , Miosinas Cardíacas/metabolismo , Linhagem Celular , Sobrevivência Celular , Criança , Feminino , Comunicação Interatrial/metabolismo , Comunicação Interatrial/patologia , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Mioblastos/metabolismo , Cadeias Pesadas de Miosina/química , Cadeias Pesadas de Miosina/metabolismo , Linhagem , Conformação Proteica em alfa-Hélice , Estabilidade Proteica
5.
BMC Cardiovasc Disord ; 21(1): 321, 2021 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-34193080

RESUMO

BACKGROUND: Congenital heart disease (CHD) is the leading cause of mortality from birth defects. In adult CHD patients with successful surgical repair, cardiac complications including heart failure develop at late stage, likely due to genetic causes. To date, many mutations in cardiac developmental genes have been associated with CHD. Recently, regulatory variants in genes have been linked to many human diseases. Although mutations and splicing variants in GATA4 gene have been reported in CHD patients, few regulatory variants of GATA4 gene are identified in CHD patients. METHODS: GATA4 gene regulatory region was investigated in the patients with atrial septal defects (ASD) (n = 332) and ethnic-matched controls (n = 336). RESULTS: Five heterozygous regulatory variants including four SNPs [g.31360 T>C (rs372004083), g.31436G>A, g.31437C>A (rs769262495), g.31487C>G (rs1053351749) and g.31856C>T (rs1385460518)] were only identified in ASD patients. Functional analysis indicated that the regulatory variants significantly affected the transcriptional activity of GATA4 gene promoter. Furthermore, two of the five regulatory variants have evidently effected on transcription factor binding sites. CONCLUSIONS: Our data suggested that GATA4 gene regulatory variants may confer ASD susceptibility by decreasing GATA4 levels.


Assuntos
Fator de Transcrição GATA4/genética , Comunicação Interatrial/genética , Polimorfismo de Nucleotídeo Único , Adolescente , Adulto , Animais , Sítios de Ligação , Estudos de Casos e Controles , Linhagem Celular , Criança , Pré-Escolar , Regulação para Baixo , Feminino , Fator de Transcrição GATA4/metabolismo , Predisposição Genética para Doença , Comunicação Interatrial/diagnóstico por imagem , Comunicação Interatrial/metabolismo , Heterozigoto , Humanos , Lactente , Masculino , Miócitos Cardíacos/metabolismo , Fenótipo , Regiões Promotoras Genéticas , Ratos , Transcrição Gênica , Adulto Jovem
6.
Cardiovasc Res ; 116(8): 1458-1472, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31688894

RESUMO

AIMS: Increased Ankyrin Repeat Domain 1 (ANKRD1) levels linked to gain of function mutations have been associated to total anomalous pulmonary venous return and adult cardiomyopathy occurrence in humans. The link between increased ANKRD1 level and cardiac structural and functional disease is not understood. To get insight into this problem, we have generated a gain of function ANKRD1 mouse model by overexpressing ANKRD1 in the myocardium. METHODS AND RESULTS: Ankrd1 is expressed non-homogeneously in the embryonic myocardium, with a dynamic nucleo-sarcomeric localization in developing cardiomyocytes. ANKRD1 transgenic mice present sinus venosus defect, which originates during development by impaired remodelling of early embryonic heart. Adult transgenic hearts develop diastolic dysfunction with preserved ejection fraction, which progressively evolves into heart failure, as shown histologically and haemodynamically. Transgenic cardiomyocyte structure, sarcomeric assembly, and stability are progressively impaired from embryonic to adult life. Postnatal transgenic myofibrils also present characteristic functional alterations: impaired compliance at neonatal stage and impaired lusitropism in adult hearts. Altogether, our combined analyses suggest that impaired embryonic remodelling and adult heart dysfunction in ANKRD1 transgenic mice present a common ground of initial cardiomyocyte defects, which are exacerbated postnatally. Molecular analysis showed transient activation of GATA4-Nkx2.5 transcription in early transgenic embryos and subsequent dynamic transcriptional modulation within titin gene. CONCLUSIONS: ANKRD1 is a fine mediator of cardiomyocyte response to haemodynamic load in the developing and adult heart. Increased ANKRD1 levels are sufficient to initiate an altered cellular phenotype, which is progressively exacerbated into a pathological organ response by the high ventricular workload during postnatal life. Our study defines for the first time a unifying picture for ANKRD1 role in heart development and disease and provides the first mechanistic link between ANKRD1 overexpression and cardiac disease onset.


Assuntos
Comunicação Interatrial/metabolismo , Proteínas Musculares/metabolismo , Miocárdio/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Disfunção Ventricular Esquerda/metabolismo , Função Ventricular Esquerda , Animais , Diástole , Feminino , Fator de Transcrição GATA4/genética , Fator de Transcrição GATA4/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Comunicação Interatrial/genética , Comunicação Interatrial/patologia , Comunicação Interatrial/fisiopatologia , Proteína Homeobox Nkx-2.5/genética , Proteína Homeobox Nkx-2.5/metabolismo , Masculino , Camundongos Transgênicos , Proteínas Musculares/genética , Miocárdio/patologia , Proteínas Nucleares/genética , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Proteínas Repressoras/genética , Regulação para Cima , Disfunção Ventricular Esquerda/genética , Disfunção Ventricular Esquerda/patologia , Disfunção Ventricular Esquerda/fisiopatologia
7.
Circ Genom Precis Med ; 12(8): e002491, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31430208

RESUMO

BACKGROUND: Familial atrial septal defect (ASD) has previously been attributed primarily to mutations in cardiac transcription factors. Here, we report a large, multi-generational family (78 members) with ASD combined with a late-onset dilated cardiomyopathy and further characterize the consequences of mutant α-actin. METHODS: We combined a genome-wide linkage analysis with cell biology, microscopy, and molecular biology tools to characterize a novel ACTC1 (cardiac α-actin) mutation identified in association with ASD and late-onset dilated cardiomyopathy in a large, multi-generational family. RESULTS: Using a genome-wide linkage analysis, the ASD disease locus was mapped to chromosome 15q14 harboring the ACTC1 gene. In 15 affected family members, a heterozygous, nonsynonymous, and fully penetrant mutation (p. Gly247Asp) was identified in exon 5 of ACTC1 that was absent in all healthy family members (n=63). In silico tools predicted deleterious consequences of this variant that was found absent in control databases. Ultrastructural analysis of myocardial tissue of one of the mutation carriers showed sarcomeric disarray, myofibrillar degeneration, and increased apoptosis, while cardiac proteomics revealed a significant increase in extracellular matrix proteins. Consistently, structural defects and increased apoptosis were also observed in neonatal rat ventricular cardiomyocytes overexpressing the mutant, but not native human ACTC1. Molecular dynamics studies and additional mechanistic analyses in cardiomyocytes confirmed actin polymerization/turnover defects, thereby affecting contractility. CONCLUSIONS: A combined phenotype of ASD and late-onset heart failure was caused by a heterozygous, nonsynonymous ACTC1 mutation. Mechanistically, we found a shared molecular mechanism of defective actin signaling and polymerization in both cardiac development and contractile function. Detection of ACTC1 mutations in patients with ASD may thus have further clinical implications with regard to monitoring for (late-onset) dilated cardiomyopathy.


Assuntos
Actinas/genética , Cardiomiopatia Dilatada/genética , Comunicação Interatrial/genética , Actinas/química , Actinas/metabolismo , Idade de Início , Animais , Apoptose , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/patologia , Cardiomiopatia Dilatada/fisiopatologia , Feminino , Comunicação Interatrial/metabolismo , Comunicação Interatrial/fisiopatologia , Humanos , Masculino , Pessoa de Meia-Idade , Simulação de Dinâmica Molecular , Mutação de Sentido Incorreto , Miócitos Cardíacos/metabolismo , Linhagem , Ratos
8.
Med Sci Monit ; 25: 2756-2763, 2019 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-30982828

RESUMO

BACKGROUND The NKX2 gene family is made up of core transcription factors that are involved in the morphogenesis of the vertebrate heart. NKx2-5 plays a pivotal role in mouse cardiogenesis, and mutations in NKx2-5 result in an abnormal structure and function of the heart, including atrial septal defect and cardiac electrophysiological abnormalities. MATERIAL AND METHODS To investigate the genetic variation of NKX2-5 in Chinese patients with sporadic atrial septal defect, we sequenced the full length of the NKX2-5 gene in the participants of the study. Four hundred thirty-nine patients and 567 healthy unrelated individuals were recruited. Genomic DNA was extracted from the peripheral blood leukocytes of the participants. DNA samples from the participants were amplified by multiplex PCR and sequenced on an Illumina HiSeq platform. Variations were detected by comparison with a standard reference genome and annotation with a variant effect predictor. RESULTS Thirty variations were detected in Chinese patients with sporadic atrial septal defect, and 6 single nucleotide polymorphisms (SNPs) had a frequency greater than 1%. Among the 30 variations, the SNPs rs2277923 and rs3729753 were extremely prominent, with a high frequency and odds ratio in patients. CONCLUSIONS Single nucleotide variations are the prominent genetic variations of NKX2-5 in Chinese patients with sporadic atrial septal defect. The SNPs rs2277923 and rs3729753 are prominent single nucleotide variations (SNVs) in Chinese patients with sporadic atrial septal defect.


Assuntos
Comunicação Interatrial/genética , Proteína Homeobox Nkx-2.5/genética , Povo Asiático/genética , Sequência de Bases , China/epidemiologia , Análise Mutacional de DNA , Feminino , Genes Homeobox , Comunicação Interatrial/sangue , Comunicação Interatrial/epidemiologia , Comunicação Interatrial/metabolismo , Proteína Homeobox Nkx-2.5/sangue , Proteína Homeobox Nkx-2.5/metabolismo , Humanos , Masculino , Mutação , Polimorfismo de Nucleotídeo Único , Análise de Sequência de DNA/métodos , Fatores de Transcrição/genética
9.
Elife ; 72018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-30067223

RESUMO

In historical attempts to treat morning sickness, use of the drug thalidomide led to the birth of thousands of children with severe birth defects. Despite their teratogenicity, thalidomide and related IMiD drugs are now a mainstay of cancer treatment; however, the molecular basis underlying the pleiotropic biology and characteristic birth defects remains unknown. Here we show that IMiDs disrupt a broad transcriptional network through induced degradation of several C2H2 zinc finger transcription factors, including SALL4, a member of the spalt-like family of developmental transcription factors. Strikingly, heterozygous loss of function mutations in SALL4 result in a human developmental condition that phenocopies thalidomide-induced birth defects such as absence of thumbs, phocomelia, defects in ear and eye development, and congenital heart disease. We find that thalidomide induces degradation of SALL4 exclusively in humans, primates, and rabbits, but not in rodents or fish, providing a mechanistic link for the species-specific pathogenesis of thalidomide syndrome.


Assuntos
Síndrome da Retração Ocular/metabolismo , Proteólise/efeitos dos fármacos , Talidomida/farmacologia , Fatores de Transcrição/metabolismo , Anormalidades Múltiplas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Sequência de Aminoácidos , Dedos de Zinco CYS2-HIS2 , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Células HEK293 , Cardiopatias Congênitas/metabolismo , Comunicação Interatrial/metabolismo , Humanos , Deformidades Congênitas das Extremidades Inferiores/metabolismo , Peptídeo Hidrolases/metabolismo , Fenótipo , Ligação Proteica/efeitos dos fármacos , Reprodutibilidade dos Testes , Especificidade da Espécie , Especificidade por Substrato , Teratogênicos/toxicidade , Talidomida/química , Fatores de Transcrição/química , Ubiquitina-Proteína Ligases/metabolismo , Deformidades Congênitas das Extremidades Superiores/metabolismo
10.
Hum Mol Genet ; 27(21): 3747-3760, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30016433

RESUMO

The arterial and venous poles of the mammalian heart are hotspots of congenital heart defects (CHD) such as those observed in 22q11.2 deletion (or DiGeorge) and Holt-Oram syndromes. These regions of the heart are derived from late differentiating cardiac progenitor cells of the Second Heart Field (SHF) located in pharyngeal mesoderm contiguous with the elongating heart tube. The T-box transcription factor Tbx1, encoded by the major 22q11.2 deletion syndrome gene, regulates SHF addition to both cardiac poles from a common progenitor population. Despite the significance of this cellular addition the mechanisms regulating the deployment of common progenitor cells to alternate cardiac poles remain poorly understood. Here we demonstrate that Tbx5, mutated in Holt-Oram syndrome and essential for venous pole development, is activated in Tbx1 expressing cells in the posterior region of the SHF at early stages of heart tube elongation. A subset of the SHF transcriptional program, including Tbx1 expression, is subsequently downregulated in Tbx5 expressing cells, generating a transcriptional boundary between Tbx1-positive arterial pole and Tbx5-positive venous pole progenitor cell populations. We show that normal downregulation of the definitive arterial pole progenitor cell program in the posterior SHF is dependent on both Tbx1 and Tbx5. Furthermore, retinoic acid (RA) signaling is required for Tbx5 activation in Tbx1-positive cells and blocking RA signaling at the time of Tbx5 activation results in atrioventricular septal defects at fetal stages. Our results reveal sequential steps of cardiac progenitor cell patterning and provide mechanistic insights into the origin of common forms of CHD.


Assuntos
Anormalidades Múltiplas/metabolismo , Vasos Coronários/metabolismo , Síndrome de DiGeorge/metabolismo , Cardiopatias Congênitas/metabolismo , Comunicação Interatrial/metabolismo , Deformidades Congênitas das Extremidades Inferiores/metabolismo , Transdução de Sinais , Células-Tronco/metabolismo , Proteínas com Domínio T/metabolismo , Tretinoína/metabolismo , Deformidades Congênitas das Extremidades Superiores/metabolismo , Anormalidades Múltiplas/genética , Animais , Síndrome de DiGeorge/genética , Regulação da Expressão Gênica no Desenvolvimento , Cardiopatias Congênitas/genética , Defeitos dos Septos Cardíacos/genética , Defeitos dos Septos Cardíacos/metabolismo , Comunicação Interatrial/genética , Deformidades Congênitas das Extremidades Inferiores/genética , Camundongos , Camundongos Transgênicos , Deformidades Congênitas das Extremidades Superiores/genética
11.
Mol Med Rep ; 15(4): 2247-2254, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28259982

RESUMO

Congenital atrial septal defect (ASD) and progressive atriventricular block (AVB) are the two most common phenotypes linked to NK2 homeobox 5 (NKX2.5) mutations in animals and humans. However, the prevalence and spectrum of NKX2.5 mutation in patients with ASD and AVB remain to be elucidated. In the present study, the coding exons and flanking introns of the NKX2.5 gene, which encodes a homeobox­containing transcription factor essential for development of the heart, were sequenced in a cohort of 62 unrelated patients with ASD and AVB, and subsequently in a mutation carrier's available family members. As controls, 300 unrelated, ethnically­matched healthy individuals were recruited, who were also genotyped for NKX2.5. The functional consequence of the mutant NKX2.5 was evaluated in contrast to its wild­type counterpart using a dual­luciferase reporter assay system. As a result, a novel heterozygous NKX2.5 mutation, p.Q181X, was identified in an index patient with ASD and AVB, with a prevalence of ~1.61%. Genetic analysis of the proband's pedigree revealed that the mutation co­segregated with ASD and AVB with complete penetrance. The nonsense mutation, which eliminated partial homeobox and the carboxyl terminus, was absent in the 600 control chromosomes. Functional evaluation showed that the NKX2.5 mutant had no transcriptional activity. Furthermore, the mutation disrupted the synergistic activation between NKX2.5 and GATA binding protein 4, another cardiac core transcription factor associated with ASD. The results of the present study expand the spectrum of NKX2.5 mutations linked to ASD and AVB, and indicated that NKX2.5 loss­of­function mutations are an uncommon cause of ASD and AVB in humans.


Assuntos
Bloqueio Atrioventricular/genética , Comunicação Interatrial/genética , Proteína Homeobox Nkx-2.5/genética , Mutação , Adolescente , Adulto , Sequência de Aminoácidos , Animais , Bloqueio Atrioventricular/metabolismo , Células COS , Chlorocebus aethiops , Feminino , Fator de Transcrição GATA4/metabolismo , Comunicação Interatrial/metabolismo , Proteína Homeobox Nkx-2.5/química , Proteína Homeobox Nkx-2.5/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Alinhamento de Sequência , Adulto Jovem
12.
Hum Mol Genet ; 26(5): 942-954, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28164238

RESUMO

TBX5, a member of the T-box family of transcription factors, is a dosage sensitive regulator of heart development. Mutations in TBX5 are responsible for Holt-Oram Syndrome, an autosomal dominant disease with variable and partially penetrant cardiac defects suggestive of the existence of genetic and environmental modifiers. KLF13, a member of the Krüppel-like family of zinc finger proteins is co-expressed with TBX5 in several cardiac cells including atrial cardiomyocytes and cells of the interatrial septum. We report that KLF13 interacts physically and functionally with TBX5 to synergistically activate transcription of cardiac genes. We show that TBX5 contacts KLF13 via its T-domain and find that several disease-causing mutations therein have decreased KLF13 interaction. Whereas Klf13 heterozygote mice have no detectable cardiac defects, loss of a Klf13 allele in Tbx5 heterozygote mice significantly increases the penetrance of TBX5-dependent cardiac abnormalities including atrial, atrial-ventricular and ventricular septal defects. The results reveal for the first time combinatorial interaction between a T-box protein and a KLF family member and its importance for heart and possibly other organ development. The data also suggest that, in human, KLF13 may be a genetic modifier of the Holt-Oram Syndrome gene TBX5.


Assuntos
Anormalidades Múltiplas/genética , Proteínas de Ciclo Celular/genética , Átrios do Coração/metabolismo , Cardiopatias Congênitas/genética , Comunicação Interatrial/genética , Fatores de Transcrição Kruppel-Like/genética , Deformidades Congênitas das Extremidades Inferiores/genética , Proteínas Repressoras/genética , Proteínas com Domínio T/genética , Deformidades Congênitas das Extremidades Superiores/genética , Anormalidades Múltiplas/metabolismo , Anormalidades Múltiplas/patologia , Animais , Proteínas de Ciclo Celular/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica , Átrios do Coração/patologia , Cardiopatias Congênitas/metabolismo , Cardiopatias Congênitas/patologia , Comunicação Interatrial/metabolismo , Comunicação Interatrial/patologia , Ventrículos do Coração/metabolismo , Ventrículos do Coração/patologia , Heterozigoto , Humanos , Fatores de Transcrição Kruppel-Like/metabolismo , Deformidades Congênitas das Extremidades Inferiores/metabolismo , Deformidades Congênitas das Extremidades Inferiores/patologia , Camundongos , Mutação , Ligação Proteica , Domínios Proteicos/genética , Mapas de Interação de Proteínas/genética , Proteínas Repressoras/metabolismo , Proteínas com Domínio T/metabolismo , Ativação Transcricional/genética , Deformidades Congênitas das Extremidades Superiores/metabolismo , Deformidades Congênitas das Extremidades Superiores/patologia
13.
Sci Rep ; 6: 36749, 2016 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-27827456

RESUMO

A previous report showed that the consumption of glutathione through oxidative stress activates the glutathione synthetic pathway, which is accompanied by production of ophthalmic acid from 2-aminobutyric acid (2-AB). We conducted a comprehensive quantification of serum metabolites using gas chromatography-mass spectrometry in patients with atrial septal defect to find clues for understanding myocardial metabolic regulation, and demonstrated that circulating 2-AB levels reflect hemodynamic changes. However, the metabolism and pathophysiological role of 2-AB remains unclear. We revealed that 2-AB is generated by an amino group transfer reaction to 2-oxobutyric acid, a byproduct of cysteine biosynthesis from cystathionine. Because cysteine is a rate-limiting substrate for glutathione synthesis, we hypothesized that 2-AB reflects glutathione compensation against oxidative stress. A murine cardiomyopathy model induced by doxorubicin supported our hypothesis, i.e., increased reactive oxygen species are accompanied by 2-AB accumulation and compensatory maintenance of myocardial glutathione levels. Intriguingly, we also found that 2-AB increases intracellular glutathione levels by activating AMPK and exerts protective effects against oxidative stress. Finally, we demonstrated that oral administration of 2-AB efficiently raises both circulating and myocardial glutathione levels and protects against doxorubicin-induced cardiomyopathy in mice. This is the first study to demonstrate that 2-AB modulates glutathione homeostasis in the myocardium.


Assuntos
Aminobutiratos/metabolismo , Cardiomegalia/metabolismo , Glutationa/metabolismo , Comunicação Interatrial/metabolismo , Homeostase , Miocárdio/metabolismo , Animais , Cardiomegalia/patologia , Modelos Animais de Doenças , Feminino , Comunicação Interatrial/patologia , Humanos , Masculino , Camundongos , Miocárdio/patologia
14.
Mol Med Rep ; 14(4): 3307-14, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27510170

RESUMO

Dilated cardiomyopathy (DCM) is the most prevalent form of primary cardiomyopathy in humans and is a leading cause of heart failure and sudden cardiac death. Genetic abnormalities have been demonstrated to be a major contributor to the development of DCM. However, DCM is a genetically heterogeneous disease, and the genetic basis underlying DCM in a significant proportion of patients remains unclear. In the current study, the coding exons and splicing junction sites of the T­Box 20 (TBX20) gene, which encodes a T­box transcription factor essential for cardiac morphogenesis and structural remodeling, were sequenced in 115 unrelated patients with idiopathic DCM, and a novel heterozygous mutation, p.E143X, was identified in one patient. Genetic analysis of the mutation carrier's pedigree indicated that the nonsense mutation was present in all the living family members with DCM, and also in a female patient with a congenital atrial septal defect. The mutation, which was predicted to generate a truncated protein with only the N­terminus and a fraction of the T­box domain remaining, was absent in 800 control chromosomes. Functional assays using a dual­luciferase reporter assay system revealed that the truncated TBX20 protein had no transcriptional activity in contrast to its wild­type counterpart. Furthermore, the mutation abolished the synergistic activation between TBX20 and NK2 homeobox 5, or between TBX20 and GATA binding protein 4. The observations of the current study expand the mutation spectrum of TBX20 associated with DCM and congenital heart disease (CHD), which provide novel insight into the molecular mechanisms underlying DCM and CHD, suggesting the potential implications for the effective and personalized treatment of these diseases.


Assuntos
Cardiomiopatia Dilatada/genética , Comunicação Interatrial/genética , Mutação de Sentido Incorreto , Proteínas com Domínio T/genética , Adulto , Cardiomiopatia Dilatada/metabolismo , Estudos de Casos e Controles , Feminino , Fator de Transcrição GATA4/metabolismo , Predisposição Genética para Doença , Comunicação Interatrial/metabolismo , Heterozigoto , Proteína Homeobox Nkx-2.5/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Mutação Puntual , Proteínas com Domínio T/metabolismo
15.
Mol Med Rep ; 13(5): 4349-56, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27035640

RESUMO

Previous genome-wide association studies have demonstrated that single nucleotide polymorphisms in T­box (TBX)5 are associated with increased susceptibility to atrial fibrillation (AF), and a recent study has causally linked a TBX5 mutation to atypical Holt-Oram syndrome and paroxysmal AF. However, the prevalence and spectrum of TBX5 mutations in patients with AF remain to be elucidated. In the present study, a cohort of 190 unrelated patients with idiopathic AF were prospectively recruited, with 400 unrelated healthy individuals recruited as controls. The coding exons and flanking introns of the TBX5 gene were sequenced in the participants. The functional characteristics of the mutant TBX5 were delineated in contrast with its wild­type counterpart using a dual­luciferase reporter assay system. As a result, a novel heterozygous TBX5 mutation, p.P132S, was identified in an index patient with AF, with a mutational prevalence of ~0.53%. Genetic analysis of the proband's family showed that the mutation co­segregated with AF, and was transmitted in an autosomal dominant pattern. The missense mutation was absent in the 800 control chromosomes, and the altered amino acid was completely evolutionarily conserved across species. Functional analyses revealed that the mutant TBX5 had significantly reduced transcriptional activity. Furthermore, the mutation markedly decreased the synergistic activation between TBX5 and NK2 homeobox 5, another transcription factor which has been causatively linked to AF. The present study was the first, to the best of our knowledge, to report on the association between a TBX5 loss­of­function mutation and increased susceptibility to AF. These results provide novel insight into the molecular mechanism underpinning AF, and have potential implications in the development of novel prophylactic and therapeutic strategies for AF, the most common form of sustained cardiac arrhythmia.


Assuntos
Anormalidades Múltiplas/genética , Fibrilação Atrial/genética , Predisposição Genética para Doença , Cardiopatias Congênitas/genética , Comunicação Interatrial/genética , Deformidades Congênitas das Extremidades Inferiores/genética , Mutação de Sentido Incorreto , Proteínas com Domínio T/genética , Deformidades Congênitas das Extremidades Superiores/genética , Anormalidades Múltiplas/metabolismo , Adulto , Fibrilação Atrial/metabolismo , Linhagem Celular , Feminino , Estudo de Associação Genômica Ampla , Cardiopatias Congênitas/metabolismo , Comunicação Interatrial/metabolismo , Proteína Homeobox Nkx-2.5/genética , Proteína Homeobox Nkx-2.5/metabolismo , Humanos , Deformidades Congênitas das Extremidades Inferiores/metabolismo , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Proteínas com Domínio T/metabolismo , Deformidades Congênitas das Extremidades Superiores/metabolismo
16.
Hum Mol Genet ; 25(6): 1140-51, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26744331

RESUMO

Atrial septal defects (ASDs) are a common human congenital heart disease (CHD) that can be induced by genetic abnormalities. Our previous studies have demonstrated a genetic interaction between Tbx5 and Osr1 in the second heart field (SHF) for atrial septation. We hypothesized that Osr1 and Tbx5 share a common signaling networking and downstream targets for atrial septation. To identify this molecular networks, we acquired the RNA-Seq transcriptome data from the posterior SHF of wild-type, Tbx5(+/) (-), Osr1(+/-), Osr1(-/-) and Tbx5(+/-)/Osr1(+/-) mutant embryos. Gene set analysis was used to identify the Kyoto Encyclopedia of Genes and Genomes pathways that were affected by the doses of Tbx5 and Osr1. A gene network module involving Tbx5 and Osr1 was identified using a non-parametric distance metric, distance correlation. A subset of 10 core genes and gene-gene interactions in the network module were validated by gene expression alterations in posterior second heart field (pSHF) of Tbx5 and Osr1 transgenic mouse embryos, a time-course gene expression change during P19CL6 cell differentiation. Pcsk6 was one of the network module genes that were linked to Tbx5. We validated the direct regulation of Tbx5 on Pcsk6 using immunohistochemical staining of pSHF, ChIP-quantitative polymerase chain reaction and luciferase reporter assay. Importantly, we identified Pcsk6 as a novel gene associated with ASD via a human genotyping study of an ASD family. In summary, our study implicated a gene network involving Tbx5, Osr1 and Pcsk6 interaction in SHF for atrial septation, providing a molecular framework for understanding the role of Tbx5 in CHD ontogeny.


Assuntos
Redes Reguladoras de Genes , Comunicação Interatrial/genética , Proteínas Serina-Treonina Quinases/genética , Serina Endopeptidases/genética , Proteínas com Domínio T/genética , Animais , Células HEK293 , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/metabolismo , Comunicação Interatrial/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Linhagem , Pró-Proteína Convertases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Serina Endopeptidases/metabolismo , Transdução de Sinais , Proteínas com Domínio T/metabolismo
17.
Drug Metab Dispos ; 44(1): 75-82, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26470914

RESUMO

The neuromuscular blocking agent cisatracurium is frequently used adjunctively in anesthesia to facilitate endotracheal intubation and to provide muscle relaxation during surgery. We aimed to determine the pharmacokinetics (PK)/pharmacodynamics (PD) of cisatracurium in patients with congenital heart defects (CHDs), such as ventricular septal defects and atrial septal defects, and to assess the effects of CHDs on the PK/PD profiles of cisatracurium. A modified two-compartment model with drug clearance from both compartments was best fitted to the PK data to determine the PK parameters. The model suggested that septal defects significantly lowered the rate of cisatracurium distribution from the central to peripheral compartment. The intercompartment rate constants k12 and k21 were significantly reduced (35%-60%, P < 0.05) in patients with ventricular septal defects and in patients with atrial septal defects compared with control patients. Consistently, septal defects caused a marked increase (160%-175%, P < 0.001) in the distribution half-life. Furthermore, significantly delayed pharmacodynamic responses to cisatracurium were observed in patients with septal defects. The onset time (i.e., the time to maximal neuromuscular block) was prolonged from 2.2 minutes to 5.0 minutes. PK/PD modeling suggested that reduced concentrations of cisatracurium in the effect compartment due to poorer distribution were the main cause of lagged pharmacodynamic responses. In conclusion, cisatracurium PK/PD were significantly altered in patients with septal defects. Our study should be of use in clinical practice for the administration of cisatracurium to patients with CHDs.


Assuntos
Atracúrio/análogos & derivados , Comunicação Interatrial/metabolismo , Comunicação Interventricular/metabolismo , Bloqueadores Neuromusculares/farmacocinética , Junção Neuromuscular/efeitos dos fármacos , Adulto , Atracúrio/administração & dosagem , Atracúrio/sangue , Atracúrio/farmacocinética , Feminino , Comunicação Interatrial/sangue , Comunicação Interventricular/sangue , Humanos , Injeções Intravenosas , Masculino , Taxa de Depuração Metabólica , Pessoa de Meia-Idade , Modelos Biológicos , Bloqueadores Neuromusculares/administração & dosagem , Bloqueadores Neuromusculares/sangue , Monitoração Neuromuscular , Distribuição Tecidual , Adulto Jovem
18.
Biomed Res Int ; 2015: 539805, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26504810

RESUMO

Mitochondrial heat shock proteins, such as HSP60, are chaperones responsible for the folding, transport, and quality control of mitochondrial matrix proteins and are essential for maintaining life. Both prosurvival and proapoptotic roles have been proposed for HSP60, and HSP60 is reportedly involved in the initiation of autoimmune, metabolic, and cardiovascular diseases. The role of HSP60 in pathogenesis of these diseases remains unclear, partly because of the lack of mouse models expressing HSP60. In this study we generated HSP60 conditional transgenic mice suitable for investigating in vivo outcomes by expressing HSP60 at the targeted organ in disease models. Ubiquitous HSP60 induction in the embryonic stage caused neonatal death in mice at postnatal day 1. A high incidence of atrial septal defects was observed in HSP60-expressing mice, with increased apoptosis and myocyte degeneration that possibly contributed to massive hemorrhage and sponge-like cardiac muscles. Our results showed that neonatal heart failure through HSP60 induction likely involves developmental defects and excessive apoptosis. The conditional HSP60 mouse model is useful for studying crucial biological questions concerning HSP60.


Assuntos
Chaperonina 60/genética , Insuficiência Cardíaca/etiologia , Proteínas Mitocondriais/genética , Animais , Animais Recém-Nascidos , Apoptose/genética , Chaperonina 60/metabolismo , Modelos Animais de Doenças , Feminino , Insuficiência Cardíaca/congênito , Insuficiência Cardíaca/genética , Comunicação Interatrial/genética , Comunicação Interatrial/metabolismo , Comunicação Interatrial/patologia , Imuno-Histoquímica , Imageamento por Ressonância Magnética , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Proteínas Mitocondriais/metabolismo , Miocárdio/metabolismo , Miocárdio/patologia , Gravidez , Regulação para Cima
19.
Life Sci ; 137: 105-15, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26165749

RESUMO

AIMS: Regenerative therapies based on resident human cardiac progenitor cells (hCPCs) are a promising alternative to medical treatments for patients with myocardial infarction. However, hCPCs are rare in human heart and finding efficient source and proper surface marker for isolation of these cells would make them a good candidate for therapy. MAIN METHODS: We have isolated 5.34∗10(6)±2.04∗10(5)/g viable cells from 35 heart tissue samples of 23 patients with congenital heart disease obtained during their heart surgery along with 6 samples from 3 normal subjects during cardiac biopsy. KEY FINDINGS: According to FACS analysis, younger ages, atrial specimen and disease with increased pulmonary vascular resistance were associated with higher percentage of c-kit(+) (CD117) hCPCs. Analysis for other stemness markers revealed increased CD133(+) cells in the hearts of patients with congenital heart disease. By using both immune-labeling and PCR, we demonstrated that these cells express key cardiac lineage and endothelial transcription factors and structural proteins during in vitro differentiation and do express stemness transcription factors in undifferentiated state. Another novel datum of potentially relevant interest is their ability in promoting greater myocardial regeneration and better survival in rat model of myocardial infarction following transplantation. SIGNIFICANCE: Our results could provide evidence for conditions associated with enriched hCPCs in patients with congenital heart disease. Moreover, we showed presence of a significant number of CD133 expressing cardiogenic stem-like cardiac precursors in the heart of patients with congenital heart disease, which could be isolated and stored for future regenerative therapies in these patients.


Assuntos
Comunicação Interatrial/patologia , Comunicação Interventricular/patologia , Mioblastos Cardíacos/citologia , Miócitos Cardíacos/citologia , Antígeno AC133 , Adolescente , Animais , Antígenos CD/metabolismo , Procedimentos Cirúrgicos Cardíacos , Diferenciação Celular , Células Cultivadas , Criança , Feminino , Expressão Gênica , Glicoproteínas/metabolismo , Comunicação Interatrial/metabolismo , Comunicação Interventricular/metabolismo , Humanos , Separação Imunomagnética , Antígeno Ki-67/metabolismo , Masculino , Mioblastos Cardíacos/metabolismo , Infarto do Miocárdio/terapia , Miócitos Cardíacos/metabolismo , Peptídeos/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Ratos , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
20.
Int J Mol Med ; 35(6): 1545-53, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25873328

RESUMO

Our previous study indicated that 8 patients from a family with a history of congenital heart disease had simple atrial septal defect (ASD) and carried the same mutation at codon 310 in the GATA4 gene. In the present study, to identify the functional defects caused by this mutation in an in vivo model, the transgene DNA constructs were microinjected into mice to generate a transgenic mouse model. The mice were genotyped using PCR and DNA sequencing. Protein expression was measured by western blot analysis. qPCR was used to determine the copy number of the transgenes. The heart tissue was fixed and sectioned by conventional procedures. The Vevo 2000 system was used to perform echocardiography on the mice. The expression of GATA4 target genes was measured using the real-time PCR system. The incidence of ASD in the heterozygous transgenic mice was found to be greater than that in the wild-type control mice (P<0.05). In addition, the expression of α-myosin heavy chain (α-MHC) in the heart tissues from the homozygous mice was lower than that in the heart tissues from their wild-type littermates (P<0.05). In conclusion, these results suggest that the introduction of GATA4 M310V negatively affects the normal expression of α-MHC. In accordance with previous findings on GATA4 mutation screening and in vitro experiments, this study confirms that GATA4 M310V mutation may lead to the development of the congenital heart defect, ASD.


Assuntos
Fator de Transcrição GATA4/metabolismo , Comunicação Interatrial/metabolismo , Comunicação Interatrial/fisiopatologia , Mutação de Sentido Incorreto , Substituição de Aminoácidos , Animais , Modelos Animais de Doenças , Eletrocardiografia , Fator de Transcrição GATA4/genética , Comunicação Interatrial/genética , Comunicação Interatrial/patologia , Humanos , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA