Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Circulation ; 149(18): 1419-1434, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38357791

ABSTRACT

BACKGROUND: Clonal hematopoiesis of indeterminate potential (CHIP), a common age-associated phenomenon, associates with increased risk of both hematological malignancy and cardiovascular disease. Although CHIP is known to increase the risk of myocardial infarction and heart failure, the influence of CHIP in cardiac arrhythmias, such as atrial fibrillation (AF), is less explored. METHODS: CHIP prevalence was determined in the UK Biobank, and incident AF analysis was stratified by CHIP status and clone size using Cox proportional hazard models. Lethally irradiated mice were transplanted with hematopoietic-specific loss of Tet2, hematopoietic-specific loss of Tet2 and Nlrp3, or wild-type control and fed a Western diet, compounded with or without NLRP3 (NLR [NACHT, LRR {leucine rich repeat}] family pyrin domain containing protein 3) inhibitor, NP3-361, for 6 to 9 weeks. Mice underwent in vivo invasive electrophysiology studies and ex vivo optical mapping. Cardiomyocytes from Ldlr-/- mice with hematopoietic-specific loss of Tet2 or wild-type control and fed a Western diet were isolated to evaluate calcium signaling dynamics and analysis. Cocultures of pluripotent stem cell-derived atrial cardiomyocytes were incubated with Tet2-deficient bone marrow-derived macrophages, wild-type control, or cytokines IL-1ß (interleukin 1ß) or IL-6 (interleukin 6). RESULTS: Analysis of the UK Biobank showed individuals with CHIP, in particular TET2 CHIP, have increased incident AF. Hematopoietic-specific inactivation of Tet2 increases AF propensity in atherogenic and nonatherogenic mouse models and is associated with increased Nlrp3 expression and CaMKII (Ca2+/calmodulin-dependent protein kinase II) activation, with AF susceptibility prevented by inactivation of Nlrp3. Cardiomyocytes isolated from Ldlr-/- mice with hematopoietic inactivation of Tet2 and fed a Western diet have impaired calcium release from the sarcoplasmic reticulum into the cytosol, contributing to atrial arrhythmogenesis. Abnormal sarcoplasmic reticulum calcium release was recapitulated in cocultures of cardiomyocytes with the addition of Tet2-deficient macrophages or cytokines IL-1ß or IL-6. CONCLUSIONS: We identified a modest association between CHIP, particularly TET2 CHIP, and incident AF in the UK Biobank population. In a mouse model of AF resulting from hematopoietic-specific inactivation of Tet2, we propose altered calcium handling as an arrhythmogenic mechanism, dependent on Nlrp3 inflammasome activation. Our data are in keeping with previous studies of CHIP in cardiovascular disease, and further studies into the therapeutic potential of NLRP3 inhibition for individuals with TET2 CHIP may be warranted.


Subject(s)
Atrial Fibrillation , Clonal Hematopoiesis , DNA-Binding Proteins , Dioxygenases , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Proto-Oncogene Proteins , Animals , Dioxygenases/metabolism , Dioxygenases/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Atrial Fibrillation/metabolism , Atrial Fibrillation/etiology , Atrial Fibrillation/genetics , Atrial Fibrillation/pathology , Inflammasomes/metabolism , Humans , Mice , Clonal Hematopoiesis/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Male , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Aged , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Middle Aged , Mice, Knockout , Risk Factors
2.
Child Adolesc Psychiatry Ment Health ; 17(1): 92, 2023 Jul 19.
Article in English | MEDLINE | ID: mdl-37468975

ABSTRACT

BACKGROUND: The coronavirus disease (COVID-19) and universal mitigation strategies have fundamentally affected peoples' lives worldwide, particularly during the first two years of the pandemic. Reductions in physical activity (PA) and increased mental health (MH) problems among children and youth have been observed. This systematic review and meta-analysis investigated the relationship between physical activity (PA) and mental health (MH) among children and youth during the COVID-19 pandemic. METHODS: Four electronic databases (EMBASE, PsycINFO, PubMed, and Web of Science) were systematically searched to identify studies that (1) examined the relationship between PA and MH among children and youth (aged 2-24 years old) and (2) were published in peer-reviewed journals in English between January 2020 and December 2021. Relationships between PA and two MH aspects (i.e., negative and positive psychological responses) among children and youth at different age ranges and those with disabilities or chronic conditions (DCC) were synthesized. Meta-analyses were also performed for eligible studies to determine the pooled effect size. RESULTS: A total of 58 studies were eventually included for variable categorization, with 32 eligible for meta-analyses. Our synthesis results showed that greater PA participation was strongly related to lower negative psychological responses (i.e., anxiety, depression, stress, insomnia, fatigue, and mental health problems) and higher positive psychological responses (i.e., general well-being and vigor) in children and youth during COVID-19. The pattern and strength of relations between PA and MH outcomes varied across age ranges and health conditions, with preschoolers and those with DCC receiving less attention in the existing research. Meta-analysis results showed that the magnitude of associations of PA with negative (Fisher's z = - 0.198, p < 0.001) and positive (Fisher's z = 0.170, p < 0.001) psychological responses among children and youth was weak. These results were linked to age of participants, study quality, and reporting of PA-related information. CONCLUSIONS: PA participation and MH among children and youth deteriorated during the COVID-19 pandemic and were closely associated with each other. For the post-COVID-19 era, additional research on age- and health condition-specific relationships between PA and MH outcomes from a comprehensive perspective is warranted. (Word count: 344 words).

3.
Circ Res ; 133(4): 313-329, 2023 08 04.
Article in English | MEDLINE | ID: mdl-37449401

ABSTRACT

BACKGROUND: ZFHX3 (zinc finger homeobox 3), a gene that encodes a large transcription factor, is at the second-most significantly associated locus with atrial fibrillation (AF), but its function in the heart is unknown. This study aims to identify causative genetic variation related to AF at the ZFHX3 locus and examine the impact of Zfhx3 loss on cardiac function in mice. METHODS: CRISPR-Cas9 genome editing, chromatin immunoprecipitation, and luciferase assays in pluripotent stem cell-derived cardiomyocytes were used to identify causative genetic variation related to AF at the ZFHX3 locus. Cardiac function was assessed by echocardiography, magnetic resonance imaging, electrophysiology studies, calcium imaging, and RNA sequencing in mice with heterozygous and homozygous cardiomyocyte-restricted Zfhx3 loss (Zfhx3 Het and knockout, respectively). Human cardiac single-nucleus ATAC (assay for transposase-accessible chromatin)-sequencing data was analyzed to determine which genes in atrial cardiomyocytes are directly regulated by ZFHX3. RESULTS: We found single-nucleotide polymorphism (SNP) rs12931021 modulates an enhancer regulating ZFHX3 expression, and the AF risk allele is associated with decreased ZFHX3 transcription. We observed a gene-dose response in AF susceptibility with Zfhx3 knockout mice having higher incidence, frequency, and burden of AF than Zfhx3 Het and wild-type mice, with alterations in conduction velocity, atrial action potential duration, calcium handling and the development of atrial enlargement and thrombus, and dilated cardiomyopathy. Zfhx3 loss results in atrial-specific differential effects on genes and signaling pathways involved in cardiac pathophysiology and AF. CONCLUSIONS: Our findings implicate ZFHX3 as the causative gene at the 16q22 locus for AF, and cardiac abnormalities caused by loss of cardiac Zfhx3 are due to atrial-specific dysregulation of pathways involved in AF susceptibility. Together, these data reveal a novel and important role for Zfhx3 in the control of cardiac genes and signaling pathways essential for normal atrial function.


Subject(s)
Atrial Fibrillation , Homeodomain Proteins , Animals , Humans , Mice , Atrial Fibrillation/genetics , Calcium/metabolism , Dilatation , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Myocytes, Cardiac/metabolism , Transcription Factors/genetics
4.
Complement Ther Clin Pract ; 51: 101747, 2023 May.
Article in English | MEDLINE | ID: mdl-36996660

ABSTRACT

PURPOSE: To determine the associations of resilience with physical activity (PA) and sedentary behaviour (SB) recommendations attainment among university students by considering the sex of the participants. METHODS: This cross-sectional study recruited 352 Chinese university students (131 males, 20.8 ± 2.18 years). PA and SB were assessed using the International Physical Activity Questionnaire-Short Form. Resilience was measured using the Chinese version of the Connor-Davidson Resilience Scale with 25 items (CD-RISC-25). Attainment of PA and SB recommendations with different patterns was determined by referring to the global recommendations for adults. Mann-Whitney U tests and generalized linear models (GLMs) were used to determine sex differences in all outcomes and the contribution of resilience to the attainment of PA and SB recommendations, respectively. RESULTS: The percentage of males who attained all patterns related to vigorous PA (VPA), moderate-to-vigorous PA (MVPA), and SB recommendations was significantly higher than that of females. Males also scored significantly higher than females in the CD-RISC-25 final score (p < .01). GLMs results showed that, after adjusting for key confounders, resilience was a significant predictor of PA recommendations attainment in terms of minimum moderate PA (MPA), minimum MVPA, and adequate MVPA (all p < .05). CONCLUSION: PA (at more intense levels), SB, and resilience among university students vary by sex, with males outperforming females. Regardless of sex, resilience is an important predictor for the attainment of PA and SB recommendations. Sex-specific resilience-building interventions should be developed to foster a physically active lifestyle among this population group.


Subject(s)
Exercise , Sedentary Behavior , Adult , Humans , Male , Female , Cross-Sectional Studies , Universities , Students
5.
Nature ; 608(7921): 174-180, 2022 08.
Article in English | MEDLINE | ID: mdl-35732739

ABSTRACT

Heart failure encompasses a heterogeneous set of clinical features that converge on impaired cardiac contractile function1,2 and presents a growing public health concern. Previous work has highlighted changes in both transcription and protein expression in failing hearts3,4, but may overlook molecular changes in less prevalent cell types. Here we identify extensive molecular alterations in failing hearts at single-cell resolution by performing single-nucleus RNA sequencing of nearly 600,000 nuclei in left ventricle samples from 11 hearts with dilated cardiomyopathy and 15 hearts with hypertrophic cardiomyopathy as well as 16 non-failing hearts. The transcriptional profiles of dilated or hypertrophic cardiomyopathy hearts broadly converged at the tissue and cell-type level. Further, a subset of hearts from patients with cardiomyopathy harbour a unique population of activated fibroblasts that is almost entirely absent from non-failing samples. We performed a CRISPR-knockout screen in primary human cardiac fibroblasts to evaluate this fibrotic cell state transition; knockout of genes associated with fibroblast transition resulted in a reduction of myofibroblast cell-state transition upon TGFß1 stimulation for a subset of genes. Our results provide insights into the transcriptional diversity of the human heart in health and disease as well as new potential therapeutic targets and biomarkers for heart failure.


Subject(s)
Cardiomyopathy, Dilated , Cardiomyopathy, Hypertrophic , Cell Nucleus , Gene Expression Profiling , Heart Failure , Single-Cell Analysis , CRISPR-Cas Systems , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/pathology , Case-Control Studies , Cell Nucleus/genetics , Cells, Cultured , Gene Knockout Techniques , Heart Failure/genetics , Heart Failure/pathology , Heart Ventricles/metabolism , Heart Ventricles/pathology , Humans , Myocardium/metabolism , Myocardium/pathology , Myofibroblasts/metabolism , Myofibroblasts/pathology , RNA-Seq , Transcription, Genetic , Transforming Growth Factor beta1
6.
Circ Cardiovasc Genet ; 10(6)2017 Dec.
Article in English | MEDLINE | ID: mdl-29212899

ABSTRACT

BACKGROUND: Restrictive cardiomyopathy (RCM) is a rare cardiomyopathy characterized by impaired diastolic ventricular function resulting in a poor clinical prognosis. Rarely, heritable forms of RCM have been reported, and mutations underlying RCM have been identified in genes that govern the contractile function of the cardiomyocytes. METHODS AND RESULTS: We evaluated 8 family members across 4 generations by history, physical examination, electrocardiography, and echocardiography. Affected individuals presented with a pleitropic syndrome of progressive RCM, atrioventricular septal defects, and a high prevalence of atrial fibrillation. Exome sequencing of 5 affected members identified a single novel missense variant in a highly conserved residue of FLNC (filamin C; p.V2297M). FLNC encodes filamin C-a protein that acts as both a scaffold for the assembly and organization of the central contractile unit of striated muscle and also as a mechanosensitive signaling molecule during cell migration and shear stress. Immunohistochemical analysis of FLNC localization in cardiac tissue from an affected family member revealed a diminished localization at the z disk, whereas traditional localization at the intercalated disk was preserved. Stem cell-derived cardiomyocytes mutated to carry the effect allele had diminished contractile activity when compared with controls. CONCLUSION: We have identified a novel variant in FLNC as pathogenic variant for familial RCM-a finding that further expands on the genetic basis of this rare and morbid cardiomyopathy.


Subject(s)
Cardiomyopathy, Restrictive/genetics , Filamins/genetics , Genetic Predisposition to Disease , Mutation/genetics , Adult , Aged , Amino Acid Sequence , Base Sequence , Cardiomyopathy, Restrictive/pathology , Family , Female , Filamins/chemistry , Humans , Male , Middle Aged , Phenotype
7.
Circ Cardiovasc Genet ; 10(5)2017 Oct.
Article in English | MEDLINE | ID: mdl-28974514

ABSTRACT

BACKGROUND: Atrial fibrillation (AF) affects over 33 million individuals worldwide. Genome-wide association studies have identified at least 30 AF loci, but the mechanisms through which individual variants lead to altered disease risk have remained unclear for the majority of these loci. At the 1q24 locus, we hypothesized that the transcription factor PRRX1 could be a strong candidate gene as it is expressed in the pulmonary veins, a source of AF in many individuals. We sought to identify the molecular mechanism, whereby variation at 1q24 may lead to AF susceptibility. METHODS AND RESULTS: We sequenced a ≈158 kb region encompassing PRRX1 in 962 individuals with and without AF. We identified a broad region of association with AF at the 1q24 locus. Using in silico prediction and functional validation, we identified an enhancer that interacts with the promoter of PRRX1 in cells of cardiac lineage. Within this enhancer, we identified a single-nucleotide polymorphism, rs577676, which alters enhancer activity in a mouse atrial cell line and in embryonic zebrafish and differentially regulates PRRX1 expression in human left atria. We found that suppression of PRRX1 in human embryonic stem cell-derived cardiomyocytes and embryonic zebrafish resulted in shortening of the atrial action potential duration, a hallmark of AF. CONCLUSIONS: We have identified a functional genetic variant that alters PRRX1 expression, ultimately resulting in electrophysiological alterations in atrial myocytes that may promote AF.


Subject(s)
Action Potentials/genetics , Atrial Fibrillation , Homeodomain Proteins , Human Embryonic Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Polymorphism, Single Nucleotide , Animals , Animals, Genetically Modified , Atrial Fibrillation/genetics , Atrial Fibrillation/metabolism , Atrial Fibrillation/physiopathology , Cell Line , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Human Embryonic Stem Cells/pathology , Humans , Mice , Myocytes, Cardiac/pathology , Zebrafish
8.
Heart Rhythm ; 14(2): 284-291, 2017 02.
Article in English | MEDLINE | ID: mdl-27756709

ABSTRACT

BACKGROUND: The genetic basis of atrial fibrillation (AF) and congenital heart disease remains incompletely understood. OBJECTIVE: We sought to determine the causative mutation in a family with AF, atrial septal defects, and ventricular septal defects. METHODS: We evaluated a pedigree with 16 family members, 1 with an atrial septal defect, 1 with a ventricular septal defect, and 3 with AF; we performed whole exome sequencing in 3 affected family members. Given that early-onset AF was prominent in the family, we then screened individuals with early-onset AF, defined as an age of onset <66 years, for mutations in GATA6. Variants were functionally characterized using reporter assays in a mammalian cell line. RESULTS: Exome sequencing in 3 affected individuals identified a conserved mutation, R585L, in the transcription factor gene GATA6. In the Massachusetts General Hospital Atrial Fibrillation (MGH AF) Study, the mean age of AF onset was 47.1 ± 10.9 years; 79% of the participants were men; and there was no evidence of structural heart disease. We identified 3 GATA6 variants (P91S, A177T, and A543G). Using wild-type and mutant GATA6 constructs driving atrial natriuretic peptide promoter reporter, we found that 3 of the 4 variants had a marked upregulation of luciferase activity (R585L: 4.1-fold, P < .0001; P91S: 2.5-fold, P = .0002; A177T; 1.7-fold, P = .03). In addition, when co-overexpressed with GATA4 and MEF2C, GATA6 variants exhibited upregulation of the alpha myosin heavy chain and atrial natriuretic peptide reporter activity. CONCLUSION: Overall, we found gain-of-function mutations in GATA6 in both a family with early-onset AF and atrioventricular septal defects as well as in a family with sporadic, early-onset AF.


Subject(s)
Atrial Fibrillation , Exome Sequencing , GATA6 Transcription Factor/genetics , Heart Septal Defects, Atrial , Heart Septal Defects, Ventricular , Adult , Age of Onset , Atrial Fibrillation/diagnosis , Atrial Fibrillation/epidemiology , Atrial Fibrillation/genetics , Female , Heart Septal Defects, Atrial/diagnosis , Heart Septal Defects, Atrial/epidemiology , Heart Septal Defects, Atrial/genetics , Heart Septal Defects, Ventricular/diagnosis , Heart Septal Defects, Ventricular/epidemiology , Heart Septal Defects, Ventricular/genetics , Humans , Male , Massachusetts/epidemiology , Middle Aged , Mutation , Pedigree , Exome Sequencing/methods
9.
Am J Hum Genet ; 99(6): 1281-1291, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27866707

ABSTRACT

The most significantly associated genetic locus for atrial fibrillation (AF) is in chromosomal region 4q25, where four independent association signals have been identified. Although model-system studies suggest that altered PITX2c expression might underlie the association, the link between specific variants and the direction of effect on gene expression remains unknown for all four signals. In the present study, we analyzed the AF-associated region most proximal to PITX2 at 4q25. First, we identified candidate regulatory variants that might confer AF risk through a combination of mammalian conservation, DNase hypersensitivity, and histone modification from ENCODE and the Roadmap Epigenomics Project, as well as through in vivo analysis of enhancer activity in embryonic zebrafish. Within candidate regions, we then identified a single associated SNP, rs2595104, which displayed dramatically reduced enhancer activity with the AF risk allele. CRISPR-Cas9-mediated deletion of the rs2595104 region and editing of the rs2595104 risk allele in human stem-cell-derived cardiomyocytes resulted in diminished PITX2c expression in comparison to that of the non-risk allele. This differential activity was mediated by activating enhancer binding protein 2 alpha (TFAP2a), which bound robustly to the non-risk allele at rs2595104, but not to the risk allele, in cardiomyocytes. In sum, we found that the AF-associated SNP rs2595104 altered PITX2c expression via interaction with TFAP2a. Such a pathway could ultimately contribute to AF susceptibility at the PITX2 locus associated with AF.


Subject(s)
Atrial Fibrillation/genetics , Gene Expression Regulation , Homeodomain Proteins/genetics , Polymorphism, Single Nucleotide/genetics , Transcription Factor AP-2/metabolism , Transcription Factors/genetics , Alleles , Animals , Chromosomes, Human, Pair 4/genetics , Conserved Sequence/genetics , Deoxyribonucleases/metabolism , Enhancer Elements, Genetic/genetics , Genetic Predisposition to Disease , Histones/chemistry , Histones/metabolism , Humans , Mammals/genetics , Myocytes, Cardiac/cytology , Zebrafish/genetics , Homeobox Protein PITX2
10.
Elife ; 52016 05 10.
Article in English | MEDLINE | ID: mdl-27162171

ABSTRACT

Genetic variants identified by genome-wide association studies explain only a modest proportion of heritability, suggesting that meaningful associations lie 'hidden' below current thresholds. Here, we integrate information from association studies with epigenomic maps to demonstrate that enhancers significantly overlap known loci associated with the cardiac QT interval and QRS duration. We apply functional criteria to identify loci associated with QT interval that do not meet genome-wide significance and are missed by existing studies. We demonstrate that these 'sub-threshold' signals represent novel loci, and that epigenomic maps are effective at discriminating true biological signals from noise. We experimentally validate the molecular, gene-regulatory, cellular and organismal phenotypes of these sub-threshold loci, demonstrating that most sub-threshold loci have regulatory consequences and that genetic perturbation of nearby genes causes cardiac phenotypes in mouse. Our work provides a general approach for improving the detection of novel loci associated with complex human traits.


Subject(s)
Epigenomics , Genetic Loci , Genome-Wide Association Study , Heart Conduction System/physiology , Animals , Humans , Mice
11.
Nat Commun ; 5: 5177, 2014 Oct 08.
Article in English | MEDLINE | ID: mdl-25295779

ABSTRACT

Heart failure (HF) is associated with complicated molecular remodelling within cardiomyocytes; however, the mechanisms underlying this process remain unclear. Here we show that sorting nexin-13 (SNX13), a member of both the sorting nexin and the regulator of G protein signalling (RGS) protein families, is a potent mediator of HF. Decreased levels of SNX13 are observed in failing hearts of humans and of experimental animals. SNX13-deficient zebrafish recapitulate HF with striking cardiomyocyte apoptosis. Mechanistically, a reduction in SNX13 expression facilitates the degradative sorting of apoptosis repressor with caspase recruitment domain (ARC), which is a multifunctional inhibitor of apoptosis. Consequently, the apoptotic pathway is activated, resulting in the loss of cardiac cells and the dampening of cardiac function. The N-terminal PXA structure of SNX13 is responsible for mediating the endosomal trafficking of ARC. Thus, this study reveals that SNX13 profoundly affects cardiac performance through the SNX13-PXA-ARC-caspase signalling pathway.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Heart Failure/metabolism , Muscle Proteins/metabolism , Myocytes, Cardiac/metabolism , Sorting Nexins/metabolism , Animals , Caspase 8/metabolism , Disease Models, Animal , Humans , Mice , Zebrafish
12.
Circulation ; 130(15): 1225-35, 2014 Oct 07.
Article in English | MEDLINE | ID: mdl-25124494

ABSTRACT

BACKGROUND: Atrial fibrillation (AF) affects >30 million individuals worldwide and is associated with an increased risk of stroke, heart failure, and death. AF is highly heritable, yet the genetic basis for the arrhythmia remains incompletely understood. METHODS AND RESULTS: To identify new AF-related genes, we used a multifaceted approach, combining large-scale genotyping in 2 ethnically distinct populations, cis-eQTL (expression quantitative trait loci) mapping, and functional validation. Four novel loci were identified in individuals of European descent near the genes NEURL (rs12415501; relative risk [RR]=1.18; 95% confidence interval [CI], 1.13-1.23; P=6.5×10(-16)), GJA1 (rs13216675; RR=1.10; 95% CI, 1.06-1.14; P=2.2×10(-8)), TBX5 (rs10507248; RR=1.12; 95% CI, 1.08-1.16; P=5.7×10(-11)), and CAND2 (rs4642101; RR=1.10; 95% CI, 1.06-1.14; P=9.8×10(-9)). In Japanese, novel loci were identified near NEURL (rs6584555; RR=1.32; 95% CI, 1.26-1.39; P=2.0×10(-25)) and CUX2 (rs6490029; RR=1.12; 95% CI, 1.08-1.16; P=3.9×10(-9)). The top single-nucleotide polymorphisms or their proxies were identified as cis-eQTLs for the genes CAND2 (P=2.6×10(-19)), GJA1 (P=2.66×10(-6)), and TBX5 (P=1.36×10(-5)). Knockdown of the zebrafish orthologs of NEURL and CAND2 resulted in prolongation of the atrial action potential duration (17% and 45%, respectively). CONCLUSIONS: We have identified 5 novel loci for AF. Our results expand the diversity of genetic pathways implicated in AF and provide novel molecular targets for future biological and pharmacological investigation.


Subject(s)
Atrial Fibrillation/genetics , Connexin 43/genetics , Homeodomain Proteins/genetics , Nuclear Proteins/genetics , Repressor Proteins/genetics , T-Box Domain Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Aged , Animals , Atrial Fibrillation/ethnology , Atrial Fibrillation/physiopathology , Chromosome Mapping , Connexin 43/physiology , Europe , Female , Gene Knockdown Techniques , Genetic Loci/physiology , Genetic Predisposition to Disease/ethnology , Genotype , Homeodomain Proteins/physiology , Humans , Japan , Male , Middle Aged , Muscle Proteins , Nuclear Proteins/physiology , Quantitative Trait Loci , Repressor Proteins/physiology , T-Box Domain Proteins/physiology , Transcription Factors/genetics , Transcription Factors/physiology , Ubiquitin-Protein Ligases/physiology , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/physiology , Homeobox Protein PITX2
13.
Cell Res ; 24(8): 977-93, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25022898

ABSTRACT

Pathological cardiac hypertrophy is an inevitable forerunner of heart failure. Regardless of the etiology of cardiac hypertrophy, cardiomyocyte mitochondrial alterations are always observed in this context. The translocases of mitochondrial outer membrane (Tom) complex governs the import of mitochondrial precursor proteins to maintain mitochondrial function under pathophysiological conditions; however, its role in the development of pathological cardiac hypertrophy remains unclear. Here, we showed that Tom70 was downregulated in pathological hypertrophic hearts from humans and experimental animals. The reduction in Tom70 expression produced distinct pathological cardiomyocyte hypertrophy both in vivo and in vitro. The defective mitochondrial import of Tom70-targeted optic atrophy-1 triggered intracellular oxidative stress, which led to a pathological cellular response. Importantly, increased Tom70 levels provided cardiomyocytes with full resistance to diverse pro-hypertrophic insults. Together, these results reveal that Tom70 acts as a molecular switch that orchestrates hypertrophic stresses and mitochondrial responses to determine pathological cardiac hypertrophy.


Subject(s)
Cardiomegaly/pathology , Mitochondrial Membrane Transport Proteins/metabolism , Animals , Cardiomegaly/metabolism , Disease Models, Animal , Down-Regulation , Heart Failure/metabolism , Heart Failure/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Precursor Protein Import Complex Proteins , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Oxidative Stress , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Zebrafish/metabolism
14.
J Cell Mol Med ; 17(9): 1119-27, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23837875

ABSTRACT

Sarcolemmal Na(+) /H(+) exchanger 1 (NHE1) activity is essential for the intracellular pH (pHi ) homeostasis in cardiac myocytes. Emerging evidence indicates that sarcolemmal NHE1 dysfunction was closely related to cardiomyocyte death, but it remains unclear whether defective trafficking of NHE1 plays a role in the vital cellular signalling processes. Dynamin (DNM), a large guanosine triphosphatase (GTPase), is best known for its roles in membrane trafficking events. Herein, using co-immunoprecipitation, cell surface biotinylation and confocal microscopy techniques, we investigated the potential regulation on cardiac NHE1 activity by DNM. We identified that DNM2, a cardiac isoform of DNM, directly binds to NHE1. Overexpression of a wild-type DNM2 or a dominant-negative DNM2 mutant with defective GTPase activity in adult rat ventricular myocytes (ARVMs) facilitated or retarded the internalization of sarcolemmal NHE1, whereby reducing or increasing its activity respectively. Importantly, the increased NHE1 activity associated with DNM2 deficiency led to ARVMs apoptosis, as demonstrated by cell viability, terminal deoxynucleotidyl transferase-mediated dUTP nick-end labelling assay, Bcl-1/Bax expression and caspase-3 activity, which were effectively rescued by pharmacological inhibition of NHE1 with zoniporide. Thus, our results demonstrate that disruption of the DNM2-dependent retrograde trafficking of NHE1 contributes to cardiomyocyte apoptosis.


Subject(s)
Apoptosis , Dynamin II/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Sodium-Hydrogen Exchangers/metabolism , Animals , Cell Survival , Dynamin II/deficiency , HEK293 Cells , Heart Ventricles/cytology , Humans , Male , Protein Transport , Rats , Rats, Sprague-Dawley , Sarcolemma/metabolism
15.
Int J Cardiol ; 168(3): 2109-19, 2013 Oct 03.
Article in English | MEDLINE | ID: mdl-23410488

ABSTRACT

BACKGROUND: Heart failure (HF) is approaching an epidemic proportion and has become one of the leading causes of death. It imposes a great burden on the healthcare system and society. Remodeling of cardiomyocyte membranes has a profound role in the pathogenesis of HF. However, whether dynamin (DNM), a membrane-remodeling GTPase, is associated with HF remains unclear. METHODS AND RESULTS: Here, we identified that DNM2 is necessary for the maintenance of cardiac function. Endogenous DNM2 protein levels were gradually decreased in parallel with the progression of HF in different experimental animal models. Decreased DNM2 level was also observed in the end-stage failing human heart. DNM2-deficient zebrafish exhibited signs of notable cardiac apoptosis and eventually developed severe HF. Mechanistic study showed that DNM2 downregulation caused cardiomyocyte sarcoplasmic reticulum Ca(2+) overload and subsequent mitochondria-dependent apoptosis. These events were preceded by enhanced membrane translocation of the L-type Ca(2+) channel due to DNM2 deficiency-mediated membrane trafficking dysfunction. Furthermore, prevention of cardiomyocyte Ca(2+)-mishandling largely ameliorated the DNM2 deficiency-associated cardiomyocyte apoptosis and HF. CONCLUSIONS: DNM2 mediates HF by modulating Ca(2+)-dependent apoptotic death of cardiomyocyte. The finding may shed light on the new strategy of HF treatment.


Subject(s)
Apoptosis , Calcium Channels, L-Type/metabolism , Calcium/metabolism , DNA/genetics , Dynamin II/genetics , Heart Failure/genetics , Myocytes, Cardiac/ultrastructure , Animals , Blotting, Western , Disease Models, Animal , Dynamin II/biosynthesis , Heart Failure/metabolism , Heart Failure/pathology , Humans , In Situ Nick-End Labeling , Microscopy, Electron, Transmission , Mitochondria, Heart/metabolism , Mitochondria, Heart/ultrastructure , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/ultrastructure , Zebrafish/embryology
16.
Biochem Biophys Res Commun ; 427(1): 73-9, 2012 Oct 12.
Article in English | MEDLINE | ID: mdl-22975349

ABSTRACT

Cyclic adenosine 3',5'-monophosphate (cAMP), which is synthesized by adenylyl cyclase (AC) and degraded by phosphodiesterase (PDE), plays crucial roles in the regulation of multiple cellular functions and physiological processes. Prolyl hydroxylase domain (PHD) proteins, which belong to a family of dioxygenases that function as oxygen sensors through their hydroxylation activity, have been implicated in multiple signaling pathways. Here, we aimed to determine whether PHD played a role in regulating intracellular cAMP level in cardiomyocytes. Through the overexpression/knockdown of the PHD gene and the measurement of the cAMP content, we found that PHD2, but not PHD1 or PHD3, acts as a regulator of intracellular cAMP. In neonatal rat cardiomyocytes and H9c2 cells, the overexpression of PHD2 increased the intracellular cAMP level, whereas the PHD2 knockdown reduced it. There was no alteration in the AC expression or activity in cells that overexpressed or downregulated PHD2. The overexpression of PHD2 decreased both the protein expression and the activity of phosphodiesterase 4D (PDE4D), whereas the PHD2 knockdown increased the PDE4D expression and activity. Co-immunoprecipitation experiments revealed a direct binding between PHD2 and PDE4D and liquid chromatography-tandem mass spectrometry analyses identified the specific hydroxylation sites on PDE4D. In conclusion, PHD2 may directly interact with PDE4D to function as a novel regulator of the intracellular cAMP levels in cardiomyocytes.


Subject(s)
Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Myocytes, Cardiac/metabolism , Procollagen-Proline Dioxygenase/metabolism , Animals , Cell Line , Down-Regulation , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases , Myocytes, Cardiac/enzymology , Proteasome Endopeptidase Complex/metabolism , Rats
17.
Chin Med J (Engl) ; 125(11): 2062-4, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22884078

ABSTRACT

Coronary artery bypass grafting (CABG) is a mature procedure in treating patients with coronary artery diseases. We report a patient undergoing CABG had history of esophageal cancer and multiple underlying diseases: hypothyroidism, type 2 diabetes mellitus and hypertension. A CABG with midline sternotomy was safely performed in the presence of thyroid replacement therapy and intensive control of blood pressure and blood glucose. The patient recovered postoperatively with supportive care.


Subject(s)
Coronary Artery Bypass/methods , Diabetes Mellitus, Type 2 , Esophagectomy , Hypertension , Hypothyroidism , Aged , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...