Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
Sci Adv ; 9(14): eade4110, 2023 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-37018396

RESUMEN

The liver plays a protective role in myocardial infarction (MI). However, very little is known about the mechanisms. Here, we identify mineralocorticoid receptor (MR) as a pivotal nexus that conveys communications between the liver and the heart during MI. Hepatocyte MR deficiency and MR antagonist spironolactone both improve cardiac repair after MI through regulation on hepatic fibroblast growth factor 21 (FGF21), illustrating an MR/FGF21 axis that underlies the liver-to-heart protection against MI. In addition, an upstreaming acute interleukin-6 (IL-6)/signal transducer and activator of transcription 3 (STAT3) pathway transmits the heart-to-liver signal to suppress MR expression after MI. Hepatocyte Il6 receptor deficiency and Stat3 deficiency both aggravate cardiac injury through their regulation on the MR/FGF21 axis. Therefore, we have unveiled an IL-6/STAT3/MR/FGF21 signaling axis that mediates heart-liver cross-talk during MI. Targeting the signaling axis and the cross-talk could provide new strategies to treat MI and heart failure.


Asunto(s)
Interleucina-6 , Infarto del Miocardio , Humanos , Interleucina-6/metabolismo , Factor de Transcripción STAT3/metabolismo , Infarto del Miocardio/metabolismo , Hígado/metabolismo , Receptores de Interleucina-6/metabolismo
2.
Can J Cardiol ; 39(5): 593-604, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36669686

RESUMEN

BACKGROUND: Mineralocorticoid receptor (MR) antagonists have been widely used to treat heart failure (HF). Studies have shown that MR in T cells plays important roles in hypertension and myocardial hypertrophy. However, the function of T-cell MR in myocardial infarction (MI) has not been elucidated. METHODS: In this study, we used T-cell MR knockout (TMRKO) mouse to investigate the effects of T-cell MR deficiency on MI and to explore the underlying mechanisms. Echocardiography and tissue staining were used to assess cardiac function, fibrosis, and myocardial apoptosis after MI. Flow cytometry and quantitative real-time polymerase chain reaction (qRT-PCR) were used to detect immune cell infiltration and inflammation. RESULTS: T-cell MR deficiency significantly improved cardiac function, promoted myocardial repair, and inhibited myocardial apoptosis, fibrosis, and inflammation after MI. Luminex assays revealed that TMRKO mice had significantly lower levels of interferon-gamma (IFN-γ) and interleukin-6 (IL-6) in serum and infarcted myocardium than littermate control mice. In cultured splenic T cells, MR deficiency suppressed IL-6 expression, whereas MR overexpression enhanced IL-6 expression. Chromatin immunoprecipitation (ChIP) assay demonstrated that MR bound to the MR response element on the promoter of IL-6 gene. Finally, T-cell MR deficiency significantly suppressed accumulation of macrophages in infarcted myocardium and differentiation of proinflammatory macrophages, thereby alleviating the consequences of MI. CONCLUSIONS: T-cell MR deficiency improved pathologic ventricular remodelling after MI, likely through inhibition of accumulation and differentiation of proinflammatory macrophages. At the molecular level, MR may work through IFN-γ and IL-6 in T cells to exert functions in MI.


Asunto(s)
Interleucina-6 , Infarto del Miocardio , Ratones , Animales , Remodelación Ventricular , Receptores de Mineralocorticoides/genética , Infarto del Miocardio/metabolismo , Miocardio/patología , Linfocitos T/metabolismo , Interferón gamma , Fibrosis , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL
3.
Cell Death Differ ; 30(3): 618-631, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36151473

RESUMEN

Phenotypic modulation of vascular smooth muscle cells (VSMCs) plays critical roles in the pathogenesis of aortic aneurysm (AA). The function of nuclear receptor corepressor1 (NCOR1) in regulation of VSMC phenotype and AA is unclear. Herein, using smooth muscle NCOR1 knockout mice, we demonstrated that smooth muscle NCOR1 deficiency decreased both mRNA and protein levels of contractile genes, impaired stress fibers formation and RhoA pathway activation, reduced synthesis of elastin and collagens, and induced the expression and activity of MMPs, manifesting a switch from contractile to degradative phenotype of VSMCs. NCOR1 modulated VSMC phenotype through 3 different mechanisms. First, NCOR1 deficiency increased acetylated FOXO3a to inhibit the expression of Myocd, which downregulated contractile genes. Second, deletion of NCOR1 derepressed NFAT5 to induce the expression of Rgs1, thus impeding RhoA activation. Third, NCOR1 deficiency increased the expression of Mmp12 and Mmp13 by derepressing ATF3. Finally, a mouse model combined apoE knockout mice with angiotensin II was used to study the role of smooth muscle NCOR1 in the development of AA. The results showed that smooth muscle NCOR1 deficiency increased the incidence of aortic aneurysms and exacerbated medial degeneration in angiotensin II-induced AA mouse model. Collectively, our data illustrated that NCOR1 interacts with FOXO3a, NFAT5, and ATF3 to maintain contractile phenotype of VSMCs and suppress AA development. Manipulation of smooth muscle NCOR1 may be a potential approach for AA treatment.


Asunto(s)
Aneurisma de la Aorta , Músculo Liso Vascular , Ratones , Animales , Músculo Liso Vascular/metabolismo , Angiotensina II/metabolismo , Aneurisma de la Aorta/metabolismo , Aneurisma de la Aorta/patología , Ratones Noqueados , Fenotipo , Ratones Noqueados para ApoE , Homeostasis , Células Cultivadas , Co-Represor 1 de Receptor Nuclear/metabolismo
4.
J Mol Cell Cardiol ; 167: 40-51, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35314145

RESUMEN

RATIONALE: Mineralocorticoid receptor (MR) antagonists have been clinically used to treat heart failure. However, the underlying cellular and molecular mechanisms remain incompletely understood. METHODS AND RESULTS: Using osteoblast MR knockout (MRobko) mouse in combination with myocardial infarction (MI) model, we demonstrated that MR deficiency in osteoblasts significantly improved cardiac function, promoted myocardial healing, as well as attenuated cardiac hypertrophy, fibrosis and inflammatory response after MI. Gene expression profiling using RNA sequencing revealed suppressed expression of osteocalcin (OCN) in calvaria from MRobko mice compared to littermate control (MRfl/fl) mice with or without MI. Plasma levels of undercarboxylated OCN (ucOCN) were also markedly decreased in MRobko mice compared to MRfl/fl mice. Administration of ucOCN abolished the protective effects of osteoblast MR deficiency on infarcted hearts. Mechanistically, ucOCN treatment promoted proliferation and inflammatory cytokine secretion in macrophages. Spironolactone, an MR antagonist, significantly inhibited the expression and secretion of OCN in post-MI mice. More importantly, spironolactone decreased plasma levels of ucOCN and inflammatory cytokines in heart failure patients. CONCLUSIONS: MR deficiency in osteoblasts alleviates pathological ventricular remodeling after MI, likely through its regulation on OCN. Spironolactone may work through osteoblast MR/OCN axis to exert its therapeutic effects on pathological ventricular remodeling and heart failure in mice and human patients.


Asunto(s)
Insuficiencia Cardíaca , Infarto del Miocardio , Animales , Humanos , Ratones , Antagonistas de Receptores de Mineralocorticoides/farmacología , Infarto del Miocardio/patología , Osteoblastos/metabolismo , Espironolactona , Remodelación Ventricular
5.
Genes (Basel) ; 12(3)2021 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-33809104

RESUMEN

Atrial fibrillation (AF) represents the most common type of clinical cardiac arrhythmia and substantially increases the risks of cerebral stroke, heart failure and death. Accumulating evidence has convincingly demonstrated the strong genetic basis of AF, and an increasing number of pathogenic variations in over 50 genes have been causally linked to AF. Nevertheless, AF is of pronounced genetic heterogeneity, and the genetic determinants underpinning AF in most patients remain obscure. In the current investigation, a Chinese pedigree with AF as well as ventricular arrhythmias and hypertrophic cardiomyopathy was recruited. Whole exome sequencing and bioinformatic analysis of the available family members were conducted, and a novel heterozygous variation in the KLF15 gene (encoding Krüppel-like factor 15, a transcription factor critical for cardiac electrophysiology and structural remodeling), NM_014079.4: c.685A>T; p.(Lys229*), was identified. The variation was verified by Sanger sequencing and segregated with autosomal dominant AF in the family with complete penetrance. The variation was absent from 300 unrelated healthy subjects used as controls. In functional assays using a dual-luciferase assay system, mutant KLF15 showed neither transcriptional activation of the KChIP2 promoter nor transcriptional inhibition of the CTGF promoter, alone or in the presence of TGFB1, a key player in the pathogenesis of arrhythmias and cardiomyopathies. The findings indicate KLF15 as a new causative gene responsible for AF as well as ventricular arrhythmias and hypertrophic cardiomyopathy, and they provide novel insight into the molecular mechanisms underlying cardiac arrhythmias and hypertrophic cardiomyopathy.


Asunto(s)
Arritmias Cardíacas/genética , Fibrilación Atrial/genética , Cardiomiopatías/genética , Predisposición Genética a la Enfermedad/genética , Factores de Transcripción de Tipo Kruppel/genética , Mutación/genética , Adolescente , Adulto , Anciano , Animales , Pueblo Asiatico/genética , Línea Celular , Línea Celular Tumoral , Femenino , Células HeLa , Heterocigoto , Humanos , Masculino , Ratones , Persona de Mediana Edad , Células 3T3 NIH , Linaje , Regiones Promotoras Genéticas/genética , Activación Transcripcional/genética , Adulto Joven
6.
Front Cell Infect Microbiol ; 11: 643092, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33768014

RESUMEN

Rheumatic heart disease refers to the long-term damage of heart valves and results from an autoimmune response to group A Streptococcus infection. This study aimed to analyze the microbiota composition of patients with rheumatic heart disease and explore potential function of microbiota in this disease. First, we revealed significant alterations of microbiota in feces, subgingival plaques, and saliva of the patients compared to control subjects using 16S rRNA gene sequencing. Significantly different microbial diversity was observed in all three types of samples between the patients and control subjects. In the gut, the patients possessed higher levels of genera including Bifidobacterium and Eubacterium, and lower levels of genera including Lachnospira, Bacteroides, and Faecalibacterium. Coprococcus was identified as a super-generalist in fecal samples of the patients. Significant alterations were also observed in microbiota of subgingival plaques and saliva of the patients compared to control subjects. Second, we analyzed microbiota in mitral valves of the patients and identified microbes that could potentially transmit from the gut or oral cavity to heart valves, including Streptococcus. Third, we further analyzed the data using random forest model and demonstrated that microbiota in the gut, subgingival plaque or saliva could distinguish the patients from control subjects. Finally, we identified gut/oral microbes that significantly correlated with clinical indices of rheumatic heart disease. In conclusion, patients with rheumatic heart disease manifested important alterations in microbiota that might distinguish the patients from control subjects and correlated with severity of this disease.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Cardiopatía Reumática , Heces , Humanos , Válvula Mitral , Boca , ARN Ribosómico 16S
7.
Clin Chem Lab Med ; 59(5): 955-963, 2021 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-33554560

RESUMEN

OBJECTIVES: Dilated cardiomyopathy (DCM) represents the most frequent form of cardiomyopathy, leading to heart failure, cardiac arrhythmias and death. Accumulating evidence convincingly demonstrates the crucial role of genetic defects in the pathogenesis of DCM, and over 100 culprit genes have been implicated with DCM. However, DCM is of substantial genetic heterogeneity, and the genetic determinants underpinning DCM remain largely elusive. METHODS: Whole-exome sequencing and bioinformatical analyses were implemented in a consanguineous Chinese family with DCM. A total of 380 clinically annotated control individuals and 166 more DCM index cases then underwent Sanger sequencing analysis for the identified genetic variation. The functional characteristics of the variant were delineated by utilizing a dual-luciferase assay system. RESULTS: A heterozygous variation in the MEF2A gene (encoding myocyte enhancer factor 2A, a transcription factor pivotal for embryonic cardiogenesis and postnatal cardiac adaptation), NM_001365204.1: c.718G>T; p. (Gly240*), was identified, and verified by Sanger sequencing to segregate with autosome-dominant DCM in the family with complete penetrance. The nonsense variation was neither detected in 760 control chromosomes nor found in 166 more DCM probands. Functional analyses revealed that the variant lost transactivation on the validated target genes MYH6 and FHL2, both causally linked to DCM. Furthermore, the variation nullified the synergistic activation between MEF2A and GATA4, another key transcription factor involved in DCM. CONCLUSIONS: The findings firstly indicate that MEF2A loss-of-function variation predisposes to DCM in humans, providing novel insight into the molecular mechanisms of DCM and suggesting potential implications for genetic testing and prognostic evaluation of DCM patients.


Asunto(s)
Cardiomiopatía Dilatada , Cardiomiopatía Dilatada/genética , Análisis Mutacional de ADN , Heterocigoto , Humanos , Proteínas con Homeodominio LIM , Factores de Transcripción MEF2/genética , Proteínas Musculares , Linaje , Factores de Transcripción
8.
Heart Rhythm ; 18(5): 684-693, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33429106

RESUMEN

BACKGROUND: Atrial fibrillation (AF) represents the most common clinical cardiac arrhythmia and substantially increases the risk of cerebral stroke, heart failure, and death. Although causative genes for AF have been identified, the genetic determinants for AF remain largely unclear. OBJECTIVE: This study aimed to investigate the molecular basis of AF in a Chinese kindred. METHODS: A 4-generation family with autosomal-dominant AF and other arrhythmias (atrioventricular block, sinus bradycardia, and premature ventricular contractions) was recruited. Genome-wide scan with microsatellite markers and linkage analysis as well as whole-exome sequencing analysis were performed. Electrophysiological characteristics and subcellular localization of the AF-linked mutant were analyzed using dual whole-cell patch clamps and confocal microscopy, respectively. RESULTS: A novel genetic locus for AF was mapped to chromosome 17q21.3, a 3.23-cM interval between markers D17S951 and D17S931, with a maximum 2-point logarithm of odds score of 4.2144 at marker D17S1868. Sequencing analysis revealed a heterozygous mutation in the mapping region, NM_005497.4:c.703A>T;p.(M235L), in the GJC1 gene encoding connexin45 (Cx45). The mutation cosegregated with AF in the family and was absent in 632 control individuals. The mutation decreased the coupling conductance in cell pairs (M235L/M235L, M235L/Cx45, M235L/Cx43, and M235L/Cx40), likely because of impaired subcellular localization. CONCLUSION: This study defines a novel genetic locus for AF on chromosome 17q21.3 and reveals a loss-of-function mutation in GJC1 (Cx45) contributing to AF and other cardiac arrhythmias.


Asunto(s)
Fibrilación Atrial/genética , Trastorno del Sistema de Conducción Cardíaco/genética , Conexinas/genética , ADN/genética , Electrocardiografía/métodos , Adolescente , Adulto , Anciano , Fibrilación Atrial/fisiopatología , Trastorno del Sistema de Conducción Cardíaco/fisiopatología , Niño , Conexinas/metabolismo , Análisis Mutacional de ADN , Femenino , Sistema de Conducción Cardíaco/fisiopatología , Heterocigoto , Humanos , Masculino , Persona de Mediana Edad , Mutación , Linaje , Adulto Joven
9.
Int Heart J ; 60(5): 1113-1122, 2019 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-31484864

RESUMEN

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.


Asunto(s)
Ventrículo Derecho con Doble Salida/genética , Genes Reporteros/genética , Predisposición Genética a la Enfermedad/epidemiología , Proteínas con Homeodominio LIM/genética , Mutación con Pérdida de Función/genética , Factores de Transcripción/genética , Estudios de Casos y Controles , Causalidad , Preescolar , China/epidemiología , Ventrículo Derecho con Doble Salida/diagnóstico por imagen , Femenino , Cardiopatías Congénitas/diagnóstico por imagen , Cardiopatías Congénitas/epidemiología , Cardiopatías Congénitas/genética , Heterocigoto , Hospitales Universitarios , Humanos , Incidencia , Lactante , Masculino , Mutación , Linaje , Pronóstico , Estudios Retrospectivos , Medición de Riesgo
10.
EMBO Mol Med ; 11(11): e9127, 2019 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-31532577

RESUMEN

The function of nuclear receptor corepressor 1 (NCoR1) in cardiomyocytes is unclear, and its physiological and pathological implications are unknown. Here, we found that cardiomyocyte-specific NCoR1 knockout (CMNKO) mice manifested cardiac hypertrophy at baseline and had more severe cardiac hypertrophy and dysfunction after pressure overload. Knockdown of NCoR1 exacerbated whereas overexpression mitigated phenylephrine-induced cardiomyocyte hypertrophy. Mechanistic studies revealed that myocyte enhancer factor 2a (MEF2a) and MEF2d mediated the effects of NCoR1 on cardiomyocyte hypertrophy. The receptor interaction domains (RIDs) of NCoR1 interacted with MEF2a to repress its transcriptional activity. Furthermore, NCoR1 formed a complex with MEF2a and class IIa histone deacetylases (HDACs) to suppress hypertrophy-related genes. Finally, overexpression of RIDs of NCoR1 in the heart attenuated cardiac hypertrophy and dysfunction induced by pressure overload. In conclusion, NCoR1 cooperates with MEF2 and HDACs to repress cardiac hypertrophy. Targeting NCoR1 and the MEF2/HDACs complex may be an attractive therapeutic strategy to tackle pathological cardiac hypertrophy.


Asunto(s)
Cardiomegalia/fisiopatología , Regulación de la Expresión Génica , Miocitos Cardíacos/fisiología , Co-Represor 1 de Receptor Nuclear/metabolismo , Animales , Técnicas de Silenciamiento del Gen , Técnicas de Inactivación de Genes , Redes Reguladoras de Genes , Humanos , Factores de Transcripción MEF2/metabolismo , Ratones , Ratones Noqueados , Co-Represor 1 de Receptor Nuclear/deficiencia , Unión Proteica , Mapeo de Interacción de Proteínas
11.
Int J Mol Med ; 43(4): 1839-1846, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30720060

RESUMEN

Congenital bicuspid aortic valve (BAV) represents the most common type of cardiac birth defect affecting 0.4­2% of the general population, and accounts for a markedly increased incidence of life­threatening complications, including valvulopathy and aortopathy. Accumulating evidence has demonstrated the genetic basis of BAV. However, the genetic basis for BAV in the majority of cases remains to be elucidated. In the present study, the coding regions and splicing donors/acceptors of the nuclear receptor subfamily 2 group F member 2 (NR2F2) gene, which encodes a transcription factor essential for proper cardiovascular development, were sequenced in 176 unrelated cases of congenital BAV. The available family members of the proband carrying an identified NR2F2 mutation and 280 unrelated, sex­ and ethnicity­matched healthy individuals as controls were additionally genotyped for NR2F2. The functional effect of the mutation was characterized using a dual­luciferase reporter assay system. As a result, a novel heterozygous NR2F2 mutation, NM_021005.3: c.288C>A; p.(Cys96*), was identified in a family with BAV, which was transmitted in an autosomal dominant mode with complete penetrance. The nonsense mutation was absent from the 560 control chromosomes. Functional analysis identified that the mutant NR2F2 protein had no transcriptional activity. Furthermore, the mutation disrupted the synergistic transcriptional activation between NR2F2 and transcription factor GATA­4, another transcription factor that is associated with BAV. These findings suggested NR2F2 as a novel susceptibility gene of human BAV, which reveals a novel molecular pathogenesis underpinning BAV.


Asunto(s)
Válvula Aórtica/anomalías , Factor de Transcripción COUP II/genética , Cardiopatías Congénitas/genética , Mutación con Pérdida de Función/genética , Válvula Aórtica/patología , Secuencia de Bases , Enfermedad de la Válvula Aórtica Bicúspide , Línea Celular , Femenino , Factor de Transcripción GATA4/metabolismo , Enfermedades de las Válvulas Cardíacas/patología , Humanos , Masculino , Persona de Mediana Edad , Proteínas Mutantes/metabolismo , Fenotipo , Activación Transcripcional/genética
12.
Eur J Med Genet ; 62(9): 103540, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30217752

RESUMEN

As two members of the basic helix-loop-helix family of transcription factors, HAND1 and HAND2 are both required for the embryonic cardiogenesis and postnatal ventricular structural remodeling. Recently a HAND1 mutation has been reported to cause dilated cardiomyopathy (DCM). However, the association of a HAND2 mutation with DCM is still to be ascertained. In this research, the coding regions and splicing junction sites of the HAND2 gene were sequenced in 206 unrelated patients affected with idiopathic DCM, and a new heterozygous HAND2 mutation, NM_021973.2: c.199G > T; p.(Glu67*), was discovered in an index patient with DCM. The nonsense mutation was absent in 300 unrelated, ethnically-matched healthy persons. Genetic scan of the mutation carrier's family members revealed that the genetic mutation co-segregated with DCM, which was transmitted in an autosomal dominant fashion, with complete penetrance. Functional deciphers unveiled that the mutant HAND2 protein had no transcriptional activity. In addition, the mutation abrogated the synergistic transcriptional activation between HAND2 and GATA4 or between HAND2 and NKX2.5, two other cardiac transcription factors that have been implicated in DCM. These research findings firstly suggest HAND2 as a novel gene predisposing to DCM in humans, which adds novel insight to the molecular pathogenesis of DCM, implying potential implications in the design of personized preventive and therapeutic strategies against DCM.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Cardiomiopatía Dilatada/genética , Mutación con Pérdida de Función , Adulto , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Femenino , Células HEK293 , Células HeLa , Heterocigoto , Humanos , Masculino , Persona de Mediana Edad , Penetrancia
13.
Heart Vessels ; 34(4): 658-668, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30390123

RESUMEN

Congenital heart defect (CHD) is the most common form of birth deformity and is responsible for substantial morbidity and mortality in humans. Increasing evidence has convincingly demonstrated that genetic defects play a pivotal role in the pathogenesis of CHD. However, CHD is a genetically heterogeneous disorder and the genetic basis underpinning CHD in the vast majority of cases remains elusive. This study was sought to identify the pathogenic mutation in the ISL1 gene contributing to CHD. A cohort of 210 unrelated patients with CHD and a total of 256 unrelated healthy individuals used as controls were registered. The coding exons and splicing boundaries of ISL1 were sequenced in all study subjects. The functional effect of an identified ISL1 mutation was evaluated using a dual-luciferase reporter assay system. A novel heterozygous ISL1 mutation, c.409G > T or p.E137X, was identified in an index patient with congenital patent ductus arteriosus and ventricular septal defect. Analysis of the proband's pedigree revealed that the mutation co-segregated with CHD, which was transmitted in the family in an autosomal dominant pattern with complete penetrance. The nonsense mutation was absent in 512 control chromosomes. Functional analysis unveiled that the mutant ISL1 protein failed to transactivate the promoter of MEF2C, alone or in synergy with TBX20. This study firstly implicates ISL1 loss-of-function mutation with CHD in humans, which provides novel insight into the molecular mechanism of CHD, implying potential implications for genetic counseling and individually tailored treatment of CHD patients.


Asunto(s)
ADN/genética , Cardiopatías Congénitas/genética , Proteínas con Homeodominio LIM/genética , Mutación con Pérdida de Función , Factores de Transcripción/genética , Adolescente , Adulto , Niño , Preescolar , Análisis Mutacional de ADN , Exones , Femenino , Cardiopatías Congénitas/metabolismo , Humanos , Lactante , Proteínas con Homeodominio LIM/metabolismo , Masculino , Linaje , Reacción en Cadena de la Polimerasa , Factores de Transcripción/metabolismo , Adulto Joven
14.
Int J Med Sci ; 15(13): 1564-1572, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30443179

RESUMEN

Atrial fibrillation (AF), as the most common sustained cardiac arrhythmia, is associated with substantially increased morbidity and mortality. Aggregating evidence demonstrates that genetic defects play a crucial role in the pathogenesis of AF, especially in familial AF. Nevertheless, AF is of pronounced genetic heterogeneity, and in an overwhelming majority of cases the genetic determinants underlying AF remain elusive. In the current study, 162 unrelated patients with familial AF and 238 unrelated healthy individuals served as controls were recruited. The coding exons and splicing junction sites of the SHOX2 gene, which encodes a homeobox-containing transcription factor essential for proper development and function of the cardiac conduction system, were sequenced in all study participants. The functional effect of the mutant SHOX2 protein was characterized with a dual-luciferase reporter assay system. As a result, a novel heterozygous SHOX2 mutation, c.580C>T or p.R194X, was identified in an index patient, which was absent from the 476 control chromosomes. Genetic analysis of the proband's pedigree revealed that the nonsense mutation co-segregated with AF in the family with complete penetrance. Functional assays demonstrated that the mutant SHOX2 protein had no transcriptional activity compared with its wild-type counterpart. In conclusion, this is the first report on the association of SHOX2 loss-of-function mutation with enhanced susceptibility to familial AF, which provides novel insight into the molecular mechanism underpinning AF, suggesting potential implications for genetic counseling and individualized management of AF patients.


Asunto(s)
Fibrilación Atrial/metabolismo , Proteínas de Homeodominio/metabolismo , Fibrilación Atrial/genética , Codón sin Sentido/genética , Femenino , Células HEK293 , Proteínas de Homeodominio/genética , Humanos , Masculino , Persona de Mediana Edad , Mutación , Linaje , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
15.
Heart Vessels ; 33(7): 722-732, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29445930

RESUMEN

Dilated cardiomyopathy (DCM) is a common primary myocardial disease leading to congestive heart failure, arrhythmia and sudden cardiac death. Increasing studies demonstrate substantial genetic determinants for DCM. Nevertheless, DCM is of substantial genetic heterogeneity, and the genetic basis for DCM in most patients remains unclear. The present study was sought to investigate the association of a genetic variant in the ZBTB17 gene with DCM. A cohort of 158 unrelated patients with idiopathic DCM and a total of 230 unrelated, ethnically matched healthy individuals used as controls were recruited. The coding exons and splicing boundaries of ZBTB17 were sequenced in all study participants. The functional effect of the mutant ZBTB17 was characterized by a dual-luciferase reporter assay system. A novel heterozygous ZBTB17 mutation, p.E243X, was discovered in an index patient. Genetic scan of the mutation carrier's available relatives showed that the mutation was present in all affected family members but absent in unaffected family members. Analysis of the proband's pedigree revealed that the mutation co-segregated with DCM, which was transmitted in an autosomal dominant pattern with complete penetrance. The nonsense mutation was absent in the 460 control chromosomes. Functional assays demonstrated that the truncated ZBTB17 protein had no transcriptional activity as compared with its wild-type counterpart. This study firstly associates ZBTB17 loss-of-function mutation with enhanced susceptibility to DCM in humans, which provides novel insight into the molecular mechanism underpinning DCM, implying potential implications for genetic counseling and personalized management of DCM.


Asunto(s)
Cardiomiopatía Dilatada/genética , ADN/genética , Predisposición Genética a la Enfermedad , Factores de Transcripción de Tipo Kruppel/genética , Mutación , Cardiomiopatía Dilatada/metabolismo , Análisis Mutacional de ADN , Exones , Femenino , Heterocigoto , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , Persona de Mediana Edad , Linaje , Reacción en Cadena de la Polimerasa , Dedos de Zinc
16.
Am J Cardiol ; 121(4): 469-474, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29325903

RESUMEN

Aggregating evidence suggests that genetic determinants play a pivotal role in the pathogenesis of the congenitally bicuspid aortic valve (BAV). BAV is of pronounced genetic heterogeneity, and the genetic components underlying BAV in an overwhelming majority of patients remain elusive. In the current study, the whole coding exons and adjacent introns, as well as 5' and 3' untranslated regions of the GATA4 gene, which codes for a zinc-finger transcription factor crucial for the normal development of the aortic valve, were screened by direct sequencing in 150 index patients with congenital BAV. The available family members of an identified mutation carrier and 300 unrelated, ethnically matched healthy individuals used as controls were also genotyped for GATA4. The functional effect of the mutation was characterized using a dual-luciferase reporter assay system. As a result, a novel heterozygous GATA4 mutation, p.E147X, was identified in a family with BAV transmitted in an autosomal dominant pattern. The nonsense mutation was absent in 600 control chromosomes. Functional deciphers revealed that the mutant GATA4 protein lost transcriptional activity compared with its wild-type counterpart. Furthermore, the mutation disrupted the synergistic transcriptional activation between GATA4 and NKX2.5, another transcription factor responsible for BAV. In conclusion, this study associates the GATA4 loss-of-function mutation with enhanced susceptibility to a BAV, thus providing novel insight into the molecular mechanism underpinning the BAV.


Asunto(s)
Válvula Aórtica/anomalías , Factor de Transcripción GATA4/genética , Enfermedades de las Válvulas Cardíacas/congénito , Enfermedades de las Válvulas Cardíacas/genética , Mutación con Pérdida de Función , Adolescente , Adulto , Enfermedad de la Válvula Aórtica Bicúspide , Estudios de Casos y Controles , China , Comorbilidad , Análisis Mutacional de ADN , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Persona de Mediana Edad , Linaje
17.
Eur J Med Genet ; 61(4): 197-203, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29222010

RESUMEN

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.


Asunto(s)
Factor de Transcripción COUP II/genética , Ventrículo Derecho con Doble Salida/genética , Defectos del Tabique Interventricular/genética , Mutación con Pérdida de Función , Adolescente , Adulto , Animales , Células COS , Factor de Transcripción COUP II/metabolismo , Niño , Preescolar , Chlorocebus aethiops , Ventrículo Derecho con Doble Salida/patología , Femenino , Predisposición Genética a la Enfermedad , Células HEK293 , Defectos del Tabique Interventricular/patología , Humanos , Lactante , Masculino , Persona de Mediana Edad , Penetrancia
18.
Clin Chem Lab Med ; 56(3): 502-511, 2018 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-28902616

RESUMEN

BACKGROUND: The MADS-box transcription factor myocyte enhancer factor 2C (MEF2C) is required for the cardiac development and postnatal adaptation and in mice-targeted disruption of the MEF2C gene results in dilated cardiomyopathy (DCM). However, in humans, the association of MEF2C variation with DCM remains to be investigated. METHODS: The coding regions and splicing boundaries of the MEF2C gene were sequenced in 172 unrelated patients with idiopathic DCM. The available close relatives of the index patient harboring an identified MEF2C mutation and 300 unrelated, ethnically matched healthy individuals used as controls were genotyped for MEF2C. The functional effect of the mutant MEF2C protein was characterized in contrast to its wild-type counterpart by using a dual-luciferase reporter assay system. RESULTS: A novel heterozygous MEF2C mutation, p.Y157X, was detected in an index patient with adult-onset DCM. Genetic screen of the mutation carrier's family members revealed that the mutation co-segregated with DCM, which was transmitted as an autosomal dominant trait with complete penetrance. The non-sense mutation was absent in 300 control individuals. Functional analyses unveiled that the mutant MEF2C protein had no transcriptional activity. Furthermore, the mutation abolished the synergistic transactivation between MEF2C and GATA4 as well as HAND1, two other transcription factors that have been associated with DCM. CONCLUSIONS: This study indicates MEF2C as a new gene responsible for human DCM, which provides novel insight into the mechanism underpinning DCM, suggesting potential implications for development of innovative prophylactic and therapeutic strategies for DCM, the most prevalent form of primary myocardial disease.


Asunto(s)
Cardiomiopatía Dilatada/genética , Adulto , Cardiomiopatía Dilatada/metabolismo , Femenino , Células HeLa , Humanos , Factores de Transcripción MEF2/deficiencia , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/metabolismo , Masculino , Persona de Mediana Edad , Mutación , Células Tumorales Cultivadas
19.
Int Heart J ; 58(4): 521-529, 2017 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-28690296

RESUMEN

Dilated cardiomyopathy (DCM), the most common form of primary myocardial disease, is a leading cause of congestive heart failure and the most common indication for heart transplantation. Recently, NKX2-5 mutations have been involved in the pathogenesis of familial DCM. However, the prevalence and spectrum of NKX2-5 mutations associated with sporadic DCM remain to be evaluated. In this study, the coding regions and flanking introns of the NKX2-5 gene, which encodes a cardiac transcription factor pivotal for cardiac development and structural remodeling, were sequenced in 210 unrelated patients with sporadic adult-onset DCM. A total of 300 unrelated healthy individuals used as controls were also genotyped for NKX2-5. The functional effect of the mutant NKX2-5 was investigated using a dual-luciferase reporter assay system. As a result, two novel heterozygous NKX2-5 mutations, p.R139W and p.E167X, were identified in 2 unrelated patients with sporadic adult-onset DCM, with a mutational prevalence of approximately 0.95%. The mutations were absent in 600 referential chromosomes and the altered amino acids were completely conserved evolutionarily across species. Functional assays revealed that the NKX2-5 mutants were associated with significantly reduced transcriptional activity. Furthermore, the mutations abrogated the synergistic activation between NKX2-5 and GATA4 as well as TBX20, two other cardiac key transcription factors that have been causally linked to adult-onset DCM. This study is the first to associate NKX2-5 loss-of-function mutations with enhanced susceptibility to sporadic DCM, which provides novel insight into the molecular etiology underpinning DCM, and suggests the potential implications for the genetic counseling and personalized treatment of the DCM patients.


Asunto(s)
Cardiomiopatía Dilatada/genética , ADN/genética , Proteína Homeótica Nkx-2.5/genética , Mutación , Edad de Inicio , Cardiomiopatía Dilatada/epidemiología , Cardiomiopatía Dilatada/metabolismo , China/epidemiología , Análisis Mutacional de ADN , Femenino , Estudios de Seguimiento , Genes Reporteros/genética , Genotipo , Proteína Homeótica Nkx-2.5/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Linaje , Reacción en Cadena de la Polimerasa , Prevalencia
20.
Int J Med Sci ; 14(4): 323-332, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28553164

RESUMEN

Congenital heart disease (CHD), the most common form of developmental abnormality in humans, remains a leading cause of morbidity and mortality in neonates. Genetic defects have been recognized as the predominant causes of CHD. Nevertheless, CHD is of substantial genetic heterogeneity and the genetic defects underlying CHD in most cases remain unclear. In the current study, the coding regions and splicing junction sites of the TBX20 gene, which encodes a T-box transcription factor key to cardiovascular morphogenesis, were sequenced in 175 unrelated patients with CHD, and a novel heterozygous TBX20 mutation, p.K274X, was identified in an index patient with tetralogy of Fallot (TOF). Genetic analysis of the proband's available family members showed that his father, elder brother and son had also TOF. In addition, his father and elder brother had also atrial septal defect, and his niece had persistent truncus arteriosus and ventricular septal defect. Analysis of the pedigree revealed that the mutation co-segregated with CHD transmitted in an autosomal dominant fashion, with complete penetrance. The nonsense mutation, which was absent in the 800 control chromosomes, was predicted to produce a truncated protein with only the amino terminus and partial T-box domain left. Functional analyses by using a dual-luciferase reporter assay system showed that the mutant TBX20 lost the ability to transactivate the target gene ANF. Furthermore, the mutation reduced the synergistic activation between TBX20 and NKX2.5 as well as GATA4, two other transcriptional factors previously associated with various CHD, encompassing TOF. This study firstly links TBX20 loss-of-function mutation to familial TOF or sporadic persistent truncus arteriosus, providing novel insight into the molecular pathogenesis of CHD.


Asunto(s)
Cardiopatías Congénitas/genética , Defectos del Tabique Interatrial/genética , Proteínas de Dominio T Box/genética , Tetralogía de Fallot/genética , Tronco Arterial Persistente/genética , Secuencia de Aminoácidos , Niño , Preescolar , Femenino , Factor de Transcripción GATA4/genética , Cardiopatías Congénitas/fisiopatología , Defectos del Tabique Interatrial/fisiopatología , Heterocigoto , Proteína Homeótica Nkx-2.5/genética , Humanos , Masculino , Mutación , Linaje , Tetralogía de Fallot/fisiopatología , Tronco Arterial Persistente/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA