Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
Am J Physiol Heart Circ Physiol ; 323(6): H1091-H1107, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36269647

RESUMO

Many anticancer therapies cause serious cardiovascular complications that degrade quality of life and cause early mortality in treated patients. Specifically, doxorubicin is known as an effective anticancer agent that causes cardiomyopathy in treated patients. There has been growing interest in defining the role of endothelial cells in cardiac damage by doxorubicin. We have shown in the present study that endothelial nuclei accumulate more intravenously administered doxorubicin than other cardiac cell types. Doxorubicin enhanced cardiac production of the transforming growth factor-ß (TGF-ß) ligands and nuclear translocation of phospho-Smad3 in both cultured and in vivo cardiac endothelial cells. To examine the role of the TGF-ß/mothers against decapentaplegic homolog 3 (Smad3) pathway in cardiac damage by doxorubicin, we used both Smad3 shRNA stable endothelial cell lines and Smad3-knockout mice. We demonstrated using endothelial transcriptome analysis that upregulation of the TGF-ß and inflammatory cytokine/cytokine receptor pathways, as well as suppression of cell cycle and angiogenesis by doxorubicin, were alleviated in Smad3-deficient endothelial cells. The results of transcriptomic analysis were validated using qPCR, immunoblotting, and ex vivo aortic ring sprouting assays. Similarly, increased cardiac expression of cytokines and chemokines observed in treated wild-type mice was diminished in treated Smad3-knockout animals. We also detected increased end-diastolic diameter and depressed systolic function in doxorubicin-treated wild-type but not Smad3-knockout mice. This work provides evidence for the critical role of the canonical TGF-ß/Smad3 pathway in cardiac damage by doxorubicin.NEW & NOTEWORTHY Microvascular endothelial cells in the heart accumulate more intravenously administered doxorubicin than nonendothelial cardiac cell types. The treatment enhanced the TGF-ß/Smad3 pathway and elicited endothelial cell senescence and inflammatory responses followed by adverse cardiac remodeling and dysfunction in wild-type but not Smad3-deficient animals. Our study suggests that the TGF-ß/Smad3 pathway contributes to the development of doxorubicin cardiomyopathy and the potential value of novel approaches to ameliorate cardiotoxicity by targeting the Smad3 transcription factor.


Assuntos
Cardiomiopatias , Células Endoteliais , Camundongos , Animais , Células Endoteliais/metabolismo , Qualidade de Vida , Proteína Smad3/genética , Proteína Smad3/metabolismo , Doxorrubicina/toxicidade , Fator de Crescimento Transformador beta/metabolismo , Camundongos Knockout
2.
Physiol Genomics ; 52(8): 358-368, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32716698

RESUMO

MicroRNAs (miRNAs) are powerful regulators of protein expression. Many play important roles in cardiac development and disease. While several miRNAs and targets have been well characterized, the abundance of miRNAs and the numerous potential targets for each suggest that the vast majority of these interactions have yet to be described. The goal of this study was to characterize miRNA expression in the mouse heart after coronary artery ligation (LIG) and identify novel mRNA targets altered during the initial response to ischemic stress. We performed small RNA sequencing (RNA-Seq) of ischemic heart tissue 1 day and 3 days after ligation and identified 182 differentially expressed miRNAs. We then selected relevant mRNA targets from all potential targets by correlating miRNA and mRNA expression from a corresponding RNA-Seq data set. From this analysis we chose to focus, as proof of principle, on two miRNAs from the miR-125 family, miR-125a and miR-351, and two of their potential mRNA targets, Xin actin-binding repeat-containing protein 1 (XIRP1) and factor inhibiting hypoxia-inducible factor (FIH). We found miR-125a to be less abundant and XIRP1 more abundant after ligation. In contrast, the related murine miRNA miR-351 was substantially upregulated in response to ischemic injury, and FIH expression correspondingly decreased. Luciferase reporter assays confirmed direct interactions between these miRNAs and targets. In summary, we utilized a correlative analysis strategy combining miRNA and mRNA expression data to identify functional miRNA-mRNA relationships in the heart after ligation. These findings provide insight into the response to ischemic injury and suggest future therapeutic targets.


Assuntos
Proteínas do Citoesqueleto/genética , Proteínas de Ligação a DNA/genética , MicroRNAs/genética , Oxigenases de Função Mista/genética , Infarto do Miocárdio/genética , Regulação para Cima/genética , Animais , Proteínas do Citoesqueleto/metabolismo , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/metabolismo , Oxigenases de Função Mista/metabolismo , Infarto do Miocárdio/metabolismo , Ligação Proteica , RNA Mensageiro/genética , RNA-Seq , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/genética
3.
Am J Physiol Heart Circ Physiol ; 316(3): H554-H565, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30575439

RESUMO

The principal regulator of cellular response to low oxygen is hypoxia-inducible factor (HIF)-1, which is stabilized in several forms of heart failure. Our laboratory developed a mouse strain in which a stable form of HIF-1 can be inducibly expressed in cardiomyocytes. Strikingly, these mice show a rapid decrease in cardiac contractility and a rapid loss of SERCA2 protein, which is also seen in heart failure. Interestingly, while the SERCA2 transcript decreased, it did not fully account for the observed decrease in protein. We therefore investigated whether HIF-1-regulated microRNA could impair SERCA translation. Multiple screening analyses identified the microRNA miR-29c to be substantially upregulated upon HIF-1 induction and to have complementarity to SERCA, and therefore be a potential regulator of SERCA2 expression in hypoxia. Subsequent evaluation confirmed that miR-29c reduced SERCA2 expression and Ca2+ reuptake. Additionally, administration of an antagonist sequence (antimir) improved cardiac contractility and SERCA2 expression in HIF transgenic mice. To extend the significance of these findings, we examined miR-29c expression in physiological hypoxia. Surprisingly, miR-29c decreased in these settings. We also treated mice with antimir before infarction to see if further suppression of miR-29c could improve cardiac function. While no improvement in contractility or SERCA2 was observed, reduction of heart size after infarction indicated that the antimir could modulate cardiac physiology. These results demonstrate that while a HIF-1-regulated microRNA, miR-29c, can reduce SERCA2 expression and contractility, additional factors in the ischemic milieu may limit these effects. Efforts to develop miRNA-based therapies will need to explore and account for these additional countervailing effects. NEW & NOTEWORTHY Our study demonstrated hypoxia-inducible factor-1-dependent upregulation of miR-29c, which, in turn, inhibited SERCA2 expression and reduced cardiac contractility in a transgenic overexpression system. Interestingly, these results were not recapitulated in a murine myocardial infarction model. These results underscore the complexity of the pathological environment and highlight the need for therapeutic target validation in physiologically relevant models.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , MicroRNAs/metabolismo , Contração Miocárdica , Miócitos Cardíacos/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , Animais , Cálcio/metabolismo , Hipóxia Celular , Células HEK293 , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Miócitos Cardíacos/fisiologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo
4.
Physiol Genomics ; 50(7): 479-494, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29652636

RESUMO

Alternative splicing of RNA is an underexplored area of transcriptional response. We expect that early changes in alternatively spliced genes may be important for responses to cardiac injury. Hypoxia inducible factor 1 (HIF1) is a key transcription factor that rapidly responds to loss of oxygen through alteration of metabolism and angiogenesis. The goal of this study was to investigate the transcriptional response after myocardial infarction (MI) and to identify novel, hypoxia-driven changes, including alternative splicing. After ligation of the left anterior descending artery in mice, we observed an abrupt loss of cardiac contractility and upregulation of hypoxic signaling. We then performed RNA sequencing on ischemic heart tissue 1 and 3 days after infarct to assess early transcriptional changes and identified 89 transcripts with altered splicing. Of particular interest was the switch in Pkm isoform expression (pyruvate kinase, muscle). The usually predominant Pkm1 isoform was less abundant in ischemic hearts, while Pkm2 and associated splicing factors (hnRNPA1, hnRNPA2B1, Ptbp1) rapidly increased. Despite increased Pkm2 expression, total pyruvate kinase activity remained reduced in ischemic myocardial tissue. We also demonstrated HIF1 binding to PKM by chromatin immunoprecipitation, indicating a direct role for HIF1 in mediating this isoform switch. Our study provides a new, detailed characterization of the early transcriptome after MI. From this analysis, we identified an HIF1-mediated alternative splicing event in the PKM gene. Pkm1 and Pkm2 play distinct roles in glycolytic metabolism and the upregulation of Pkm2 is likely to have important consequences for ATP synthesis in infarcted cardiac muscle.


Assuntos
Perfilação da Expressão Gênica , Fator 1 Induzível por Hipóxia/genética , Infarto do Miocárdio/genética , Piruvato Quinase/genética , Processamento Alternativo , Animais , Glicólise/genética , Humanos , Hipóxia , Fator 1 Induzível por Hipóxia/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/fisiopatologia , Piruvato Quinase/metabolismo
5.
Hum Mol Genet ; 25(19): 4350-4368, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27577874

RESUMO

The electrocardiographic QRS duration, a measure of ventricular depolarization and conduction, is associated with cardiovascular mortality. While single nucleotide polymorphisms (SNPs) associated with QRS duration have been identified at 22 loci in populations of European descent, the genetic architecture of QRS duration in non-European populations is largely unknown. We therefore performed a genome-wide association study (GWAS) meta-analysis of QRS duration in 13,031 African Americans from ten cohorts and a transethnic GWAS meta-analysis with additional results from populations of European descent. In the African American GWAS, a single genome-wide significant SNP association was identified (rs3922844, P = 4 × 10-14) in intron 16 of SCN5A, a voltage-gated cardiac sodium channel gene. The QRS-prolonging rs3922844 C allele was also associated with decreased SCN5A RNA expression in human atrial tissue (P = 1.1 × 10-4). High density genotyping revealed that the SCN5A association region in African Americans was confined to intron 16. Transethnic GWAS meta-analysis identified novel SNP associations on chromosome 18 in MYL12A (rs1662342, P = 4.9 × 10-8) and chromosome 1 near CD1E and SPTA1 (rs7547997, P = 7.9 × 10-9). The 22 QRS loci previously identified in populations of European descent were enriched for significant SNP associations with QRS duration in African Americans (P = 9.9 × 10-7), and index SNP associations in or near SCN5A, SCN10A, CDKN1A, NFIA, HAND1, TBX5 and SETBP1 replicated in African Americans. In summary, rs3922844 was associated with QRS duration and SCN5A expression, two novel QRS loci were identified using transethnic meta-analysis, and a significant proportion of QRS-SNP associations discovered in populations of European descent were transferable to African Americans when adequate power was achieved.


Assuntos
Doenças Cardiovasculares/genética , Estudo de Associação Genômica Ampla , Ventrículos do Coração/fisiopatologia , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Negro ou Afro-Americano/genética , Alelos , Doenças Cardiovasculares/mortalidade , Doenças Cardiovasculares/fisiopatologia , Eletrocardiografia , Feminino , Genótipo , Humanos , Masculino , Miocárdio/patologia , Polimorfismo de Nucleotídeo Único/genética , População Branca/genética
6.
Proc Natl Acad Sci U S A ; 111(23): 8673-8, 2014 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-24912186

RESUMO

The bone marrow environment is among the most hypoxic in the body, but how hypoxia affects bone formation is not known. Because low oxygen tension stabilizes hypoxia-inducible factor alpha (HIFα) proteins, we have investigated the effect of expressing a stabilized form of HIF1α in osteoblast precursors. Brief stabilization of HIF1α in SP7-positive cells in postnatal mice dramatically stimulated cancellous bone formation via marked expansion of the osteoblast population. Remarkably, concomitant deletion of vascular endothelial growth factor A (VEGFA) in the mouse did not diminish bone accrual caused by HIF1α stabilization. Thus, HIF1α-driven bone formation is independent of VEGFA up-regulation and increased angiogenesis. On the other hand, HIF1α stabilization stimulated glycolysis in bone through up-regulation of key glycolytic enzymes including pyruvate dehydrogenase kinase 1 (PDK1). Pharmacological inhibition of PDK1 completely reversed HIF1α-driven bone formation in vivo. Thus, HIF1α stimulates osteoblast formation through direct activation of glycolysis, and alterations in cellular metabolism may be a broadly applicable mechanism for regulating cell differentiation.


Assuntos
Glicólise/fisiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Osteogênese/fisiologia , Regulação para Cima , Animais , Western Blotting , Medula Óssea/metabolismo , Células da Medula Óssea/metabolismo , Osso e Ossos/citologia , Osso e Ossos/metabolismo , Hipóxia Celular , Feminino , Glicólise/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Microscopia Confocal , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteogênese/genética , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Piruvato Desidrogenase Quinase de Transferência de Acetil , Fator de Transcrição Sp7 , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
J Mol Cell Cardiol ; 90: 129-38, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26686989

RESUMO

Elevated ALK4/5 ligands including TGF-ß and activins have been linked to cardiovascular remodeling and heart failure. Doxorubicin (Dox) is commonly used as a model of cardiomyopathy, a condition that often precedes cardiovascular remodeling and heart failure. In 7-8-week-old C57Bl/6 male mice treated with Dox we found decreased capillary density, increased levels of ALK4/5 ligand and Smad2/3 transcripts, and increased expression of Smad2/3 transcriptional targets. Human cardiac microvascular endothelial cells (HCMVEC) treated with Dox also showed increased levels of ALK4/5 ligands, Smad2/3 transcriptional targets, a decrease in proliferation and suppression of vascular network formation in a HCMVEC and human cardiac fibroblasts co-culture assay. Our hypothesis is that the deleterious effects of Dox on endothelial cells are mediated in part by the activation of the TGF-ß pathway. We used the inhibitor of ALK4/5 kinases SB431542 (SB) in concert with Dox to ascertain the role of TGF-ß pathway activation in doxorubicin induced endothelial cell defects. SB prevented the suppression of HCMVEC proliferation in the presence of TGF-ß2 and activin A, and alleviated the inhibition of HCMVEC proliferation by Dox. SB also prevented the suppression of vascular network formation in co-cultures of HCMVEC and human cardiac fibroblasts treated with Dox. Our results show that the inhibition of the TGF-ß pathway alleviates the detrimental effects of Dox on endothelial cells in vitro.


Assuntos
Doxorrubicina/farmacologia , Células Endoteliais/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Fator de Crescimento Transformador beta2/farmacologia , Receptores de Ativinas Tipo I/genética , Receptores de Ativinas Tipo I/metabolismo , Ativinas/genética , Ativinas/metabolismo , Ativinas/farmacologia , Animais , Benzamidas/farmacologia , Linhagem Celular , Técnicas de Cocultura , Dioxóis/farmacologia , Doxorrubicina/antagonistas & inibidores , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miocárdio/citologia , Miocárdio/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína Smad3/genética , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta2/antagonistas & inibidores , Fator de Crescimento Transformador beta2/genética , Fator de Crescimento Transformador beta2/metabolismo
8.
J Mol Cell Cardiol ; 82: 194-200, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25681585

RESUMO

These studies have examined the effect of hypoxia inducible factor 1α (HIF-1α) on nucleotide metabolism in the ischemic heart using a genetic mouse model with heart-specific and regulated expression of a stable form of HIF-1α. We find that AMP deaminase (AMPD), the entry point of the purine nucleotide cycle (PNC), is induced by HIF-1α at the level of mRNA, protein, and activity. AMP that accumulates during ischemia can be metabolized to adenosine by 5'-nucleotidase or to IMP by AMPD. Consistent with the finding of AMPD induction, adenosine accumulation during ischemia was much attenuated in HIF-1α-expressing hearts. Further investigation of nucleotide salvage enzymes found that hypoxanthine phosphoribosyl transferase (HPRT) is also upregulated in HIF-1α-expressing hearts. Treatment of hearts with an inhibitor of the PNC, hadacidin, hastens the fall of the adenylate energy charge during ischemia and the accumulation of AMP. The results provide new insight into the role of the PNC in the heart, especially as it relates to ischemia, and indicate that HIF-1α regulates nucleotide metabolism as a compensatory response to hypoxia.


Assuntos
Coração/fisiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Miocárdio/metabolismo , AMP Desaminase/genética , AMP Desaminase/metabolismo , Animais , Expressão Gênica , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Isquemia/genética , Isquemia/metabolismo , Camundongos , Camundongos Transgênicos , Modelos Animais , Nucleotídeos/metabolismo , Fatores de Tempo
9.
Cardiovasc Diabetol ; 14: 73, 2015 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-26047815

RESUMO

BACKGROUND: Diabetes promotes maladaptive changes in the endothelium that lead to its dysfunction and contribute to the vascular pathology of diabetes. We have previously reported the up-regulation of galectin-3, a ß-galactoside-binding lectin, in the endothelium and sera of diabetic mice, implicating this molecule in diabetic vasculopathy and suggesting its potential as a biomarker of the disease. Therefore, we sought to assess the role of galectin-3 in the vascular pathology of diabetes. METHODS: Galectin-3 knockout mice (KO) and wild-type mice (WT) were fed either a high-fat diet (HFD) (60 % fat calories) to produce insulin resistant diabetes, or standard chow (12 % fat calories), and their metabolic and endothelial responses were measured. After 8 weeks, the aortic and skeletal muscle endothelia were isolated by fluorescence sorting of CD105(+)/CD45(-) cells and comprehensive transcriptional analyses were performed. Transcripts differentially dysregulated by HFD in KO endothelium compared to WT were confirmed by semi-quantitative RT-PCR, and protein expression was determined by immunofluorescence of aortic and muscle tissue. Ingenuity® Pathway Analysis was used to identify pathways up-regulated by HFD in the KO, such as the coagulation cascade, and measurements of blood clotting activity were performed to confirm these results. RESULTS: KO mice demonstrate greater hyperglycemia and impaired glucose tolerance but lower insulin levels on HFD compared to WT. KO mice demonstrate a more robust transcriptional response to HFD in the vascular endothelium compared to WT. Transcripts dysregulated in the KO endothelium after HFD are involved in glucose uptake and insulin signaling, vasoregulation, coagulation, and atherogenesis. One of the most down-regulated transcripts in the endothelium of the KO after HFD was the glucose transporter, Glut4/Slc2a4. GLUT4 immunofluorescence confirmed lower protein abundance in the endothelium and muscle of the HFD-fed KO. Prothrombin time was decreased in the diabetic KO indicating increased coagulation activity. CONCLUSIONS: Galectin-3 deficiency leads to exacerbated metabolic derangement and endothelial dysfunction. The impaired tissue uptake of glucose in KO mice can be attributed to the reduced expression of GLUT4. Enhanced coagulation activity in the diabetic KO suggests a protective role for galectin-3 against thrombosis. These studies demonstrate that galectin-3 deficiency contributes both to the pathogenesis of diabetes and the associated vasculopathy.


Assuntos
Diabetes Mellitus Tipo 2/genética , Angiopatias Diabéticas/genética , Dieta Hiperlipídica , Endotélio Vascular/metabolismo , Galectina 3/genética , RNA Mensageiro/metabolismo , Animais , Aorta/metabolismo , Glicemia/metabolismo , Diabetes Mellitus Tipo 2/complicações , Angiopatias Diabéticas/etiologia , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Transportador de Glucose Tipo 4/genética , Transportador de Glucose Tipo 4/metabolismo , Resistência à Insulina , Masculino , Camundongos Knockout , Músculo Esquelético/metabolismo , Fosforilação , Tempo de Protrombina , Proteínas Proto-Oncogênicas c-akt/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcriptoma
10.
PLoS Genet ; 8(8): e1002870, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22912591

RESUMO

The QT interval (QT) is heritable and its prolongation is a risk factor for ventricular tachyarrhythmias and sudden death. Most genetic studies of QT have examined European ancestral populations; however, the increased genetic diversity in African Americans provides opportunities to narrow association signals and identify population-specific variants. We therefore evaluated 6,670 SNPs spanning eleven previously identified QT loci in 8,644 African American participants from two Population Architecture using Genomics and Epidemiology (PAGE) studies: the Atherosclerosis Risk in Communities study and Women's Health Initiative Clinical Trial. Of the fifteen known independent QT variants at the eleven previously identified loci, six were significantly associated with QT in African American populations (P≤1.20×10(-4)): ATP1B1, PLN1, KCNQ1, NDRG4, and two NOS1AP independent signals. We also identified three population-specific signals significantly associated with QT in African Americans (P≤1.37×10(-5)): one at NOS1AP and two at ATP1B1. Linkage disequilibrium (LD) patterns in African Americans assisted in narrowing the region likely to contain the functional variants for several loci. For example, African American LD patterns showed that 0 SNPs were in LD with NOS1AP signal rs12143842, compared with European LD patterns that indicated 87 SNPs, which spanned 114.2 Kb, were in LD with rs12143842. Finally, bioinformatic-based characterization of the nine African American signals pointed to functional candidates located exclusively within non-coding regions, including predicted binding sites for transcription factors such as TBX5, which has been implicated in cardiac structure and conductance. In this detailed evaluation of QT loci, we identified several African Americans SNPs that better define the association with QT and successfully narrowed intervals surrounding established loci. These results demonstrate that the same loci influence variation in QT across multiple populations, that novel signals exist in African Americans, and that the SNPs identified as strong candidates for functional evaluation implicate gene regulatory dysfunction in QT prolongation.


Assuntos
Negro ou Afro-Americano , Locos de Características Quantitativas , Característica Quantitativa Herdável , Taquicardia/etnologia , Taquicardia/genética , População Branca , Idoso , Biologia Computacional , Eletrocardiografia , Feminino , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Humanos , Desequilíbrio de Ligação , Masculino , Metagenômica , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Fatores de Risco , Estados Unidos/epidemiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA