Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 129
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Circulation ; 149(1): e1-e156, 2024 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-38033089

RESUMEN

AIM: The "2023 ACC/AHA/ACCP/HRS Guideline for the Diagnosis and Management of Atrial Fibrillation" provides recommendations to guide clinicians in the treatment of patients with atrial fibrillation. METHODS: A comprehensive literature search was conducted from May 12, 2022, to November 3, 2022, encompassing studies, reviews, and other evidence conducted on human subjects that were published in English from PubMed, EMBASE, the Cochrane Library, the Agency for Healthcare Research and Quality, and other selected databases relevant to this guideline. Additional relevant studies, published through November 2022, during the guideline writing process, were also considered by the writing committee and added to the evidence tables, where appropriate. STRUCTURE: Atrial fibrillation is the most sustained common arrhythmia, and its incidence and prevalence are increasing in the United States and globally. Recommendations from the "2014 AHA/ACC/HRS Guideline for the Management of Patients With Atrial Fibrillation" and the "2019 AHA/ACC/HRS Focused Update of the 2014 AHA/ACC/HRS Guideline for the Management of Patients With Atrial Fibrillation" have been updated with new evidence to guide clinicians. In addition, new recommendations addressing atrial fibrillation and thromboembolic risk assessment, anticoagulation, left atrial appendage occlusion, atrial fibrillation catheter or surgical ablation, and risk factor modification and atrial fibrillation prevention have been developed.


Asunto(s)
Fibrilación Atrial , Cardiología , Tromboembolia , Humanos , American Heart Association , Fibrilación Atrial/diagnóstico , Fibrilación Atrial/epidemiología , Fibrilación Atrial/terapia , Factores de Riesgo , Estados Unidos/epidemiología
2.
Int J Mol Sci ; 25(19)2024 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-39408638

RESUMEN

Genome-wide association studies have identified a locus on chromosome 10q22, where many co-inherited single nucleotide polymorphisms (SNPs) are associated with atrial fibrillation (AF). This study seeks to identify the impact of this locus on gene expression at the transcript isoform level in human left atria and to gain insight into potential causal variants. Bulk RNA sequencing was analyzed to identify myozenin 1 (MYOZ1) and synaptopodin 2-like (SYNPO2L) transcript isoforms and the association of common SNPs in this region with transcript isoform expression levels. Chromatin marks were used to suggest candidate regulatory SNPs in this region. Protein amino acid changes were examined for predicted functional consequences. Transfection of MYOZ1 and two SYNPO2L isoforms were performed to localize their encoded proteins in cardiomyocytes derived from stem cells. We identified one MYOZ1 transcript isoform and four SYNPO2L transcript isoforms, two of which encode proteins, while the other two encode long noncoding RNAs (lncRNAs). The risk allele of the strongest AF susceptibility SNP on chromosome 10q22 is associated with decreased MYOZ1 expression and increased expression of the two SNYPO2L lncRNA isoforms. There are many SNPs co-inherited with the top AF-associated SNP due to linkage disequilibrium (LD), including rs11000728, which we propose as the MYOZ1 regulatory SNP, confirmed by reporter gene transfection. In addition, this LD block includes three missense SNPs in the SYNPO2L gene, with the minor protective haplotype predicted to be detrimental to protein function. MYOZ1 and both protein isoforms of SYNPO2L were localized to the sarcomere. This is a complex locus with the potential for several SNPs in a haplotype to alter AF susceptibility by opposing effects on MYOZ1 and SYNPO2L lncRNA expression, along with effects on SYNPO2L protein function.


Asunto(s)
Fibrilación Atrial , Predisposición Genética a la Enfermedad , Polimorfismo de Nucleótido Simple , Humanos , Fibrilación Atrial/genética , Fibrilación Atrial/metabolismo , Transcriptoma , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Desequilibrio de Ligamiento , Estudio de Asociación del Genoma Completo , Isoformas de Proteínas/genética , Miocitos Cardíacos/metabolismo , ARN Largo no Codificante/genética , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Masculino
3.
Gut ; 71(1): 55-67, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-33468536

RESUMEN

OBJECTIVE: Creeping fat, the wrapping of mesenteric fat around the bowel wall, is a typical feature of Crohn's disease, and is associated with stricture formation and bowel obstruction. How creeping fat forms is unknown, and we interrogated potential mechanisms using novel intestinal tissue and cell interaction systems. DESIGN: Tissues from normal, UC, non-strictured and strictured Crohn's disease intestinal specimens were obtained. The muscularis propria matrisome was determined via proteomics. Mesenteric fat explants, primary human preadipocytes and adipocytes were used in multiple ex vivo and in vitro cell migration systems on muscularis propria muscle cell derived or native extracellular matrix. Functional experiments included integrin characterisation via flow cytometry and their inhibition with specific blocking antibodies and chemicals. RESULTS: Crohn's disease muscularis propria cells produced an extracellular matrix scaffold which is in direct spatial and functional contact with the immediately overlaid creeping fat. The scaffold contained multiple proteins, but only fibronectin production was singularly upregulated by transforming growth factor-ß1. The muscle cell-derived matrix triggered migration of preadipocytes out of mesenteric fat, fibronectin being the dominant factor responsible for their migration. Blockade of α5ß1 on the preadipocyte surface inhibited their migration out of mesenteric fat and on 3D decellularised intestinal tissue extracellular matrix. CONCLUSION: Crohn's disease creeping fat appears to result from the migration of preadipocytes out of mesenteric fat and differentiation into adipocytes in response to an increased production of fibronectin by activated muscularis propria cells. These new mechanistic insights may lead to novel approaches for prevention of creeping fat-associated stricture formation.


Asunto(s)
Adipocitos/patología , Movimiento Celular , Enfermedad de Crohn/patología , Intestinos/patología , Músculo Liso/patología , Adipogénesis/fisiología , Tejido Adiposo/patología , Diferenciación Celular , Células Cultivadas , Matriz Extracelular/patología , Fibronectinas/metabolismo , Humanos , Andamios del Tejido
4.
Circulation ; 143(8): 805-820, 2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33225722

RESUMEN

BACKGROUND: Elevated intracardiac pressure attributable to heart failure induces electrical and structural remodeling in the left atrium (LA) that begets atrial myopathy and arrhythmias. The underlying molecular pathways that drive atrial remodeling during cardiac pressure overload are poorly defined. The purpose of this study is to characterize the response of the ETV1 (ETS translocation variant 1) signaling axis in the LA during cardiac pressure overload in humans and mouse models and explore the role of ETV1 in atrial electrical and structural remodeling. METHODS: We performed gene expression profiling in 265 left atrial samples from patients who underwent cardiac surgery. Comparative gene expression profiling was performed between 2 murine models of cardiac pressure overload, transverse aortic constriction banding and angiotensin II infusion, and a genetic model of Etv1 cardiomyocyte-selective knockout (Etv1f/fMlc2aCre/+). RESULTS: Using the Cleveland Clinic biobank of human LA specimens, we found that ETV1 expression is decreased in patients with reduced ejection fraction. Consistent with its role as an important mediator of the NRG1 (Neuregulin 1) signaling pathway and activator of rapid conduction gene programming, we identified a direct correlation between ETV1 expression level and NRG1, ERBB4, SCN5A, and GJA5 levels in human LA samples. In a similar fashion to patients with heart failure, we showed that left atrial ETV1 expression is downregulated at the RNA and protein levels in murine pressure overload models. Comparative analysis of LA RNA sequencing datasets from transverse aortic constriction and angiotensin II-treated mice showed a high Pearson correlation, reflecting a highly ordered process by which the LA undergoes electrical and structural remodeling. Cardiac pressure overload produced a consistent downregulation of ErbB4, Etv1, Scn5a, and Gja5 and upregulation of profibrotic gene programming, which includes Tgfbr1/2, Igf1, and numerous collagen genes. Etv1f/fMlc2aCre/+ mice displayed atrial conduction disease and arrhythmias. Correspondingly, the LA from Etv1f/fMlc2aCre/+ mice showed downregulation of rapid conduction genes and upregulation of profibrotic gene programming, whereas analysis of a gain-of-function ETV1 RNA sequencing dataset from neonatal rat ventricular myocytes transduced with Etv1 showed reciprocal changes. CONCLUSIONS: ETV1 is downregulated in the LA during cardiac pressure overload, contributing to both electrical and structural remodeling.


Asunto(s)
Arritmias Cardíacas/patología , Proteínas de Unión al ADN/metabolismo , Atrios Cardíacos/metabolismo , Factores de Transcripción/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Angiotensina II/administración & dosificación , Angiotensina II/efectos adversos , Animales , Arritmias Cardíacas/metabolismo , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Regulación hacia Abajo , Femenino , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Neurregulina-1/genética , Neurregulina-1/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta/genética , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Remodelación Ventricular , Adulto Joven
5.
Pflugers Arch ; 473(3): 461-475, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33454842

RESUMEN

Atrial fibrillation (AF) is strongly associated with risk of stroke and heart failure. AF promotes atrial remodeling that increases risk of stroke due to left atrial thrombogenesis, and increases energy demand to support high rate electrical activity and muscle contraction. While many transcriptomic studies have assessed AF-related changes in mRNA abundance, fewer studies have assessed proteomic changes. We performed a proteomic analysis on left atrial appendage (LAA) tissues from 12 patients with a history of AF undergoing elective surgery; atrial rhythm was documented at time of surgery. Proteomic analysis was performed using liquid chromatography with mass spectrometry (LC/MS-MS). Data-dependent analysis identified 3090 unique proteins, with 408 differentially expressed between sinus rhythm and AF. Ingenuity Pathway Analysis of differentially expressed proteins identified mitochondrial dysfunction, oxidative phosphorylation, and sirtuin signaling among the most affected pathways. Increased abundance of electron transport chain (ETC) proteins in AF was accompanied by decreased expression of ETC complex assembly factors, tricarboxylic acid cycle proteins, and other key metabolic modulators. Discordant changes were also evident in the contractile unit with both up and downregulation of key components. Similar pathways were affected in a comparison of patients with a history of persistent vs. paroxysmal AF, presenting for surgery in sinus rhythm. Together, these data suggest that while the LAA attempts to meet the energetic demands of AF, an uncoordinated response may reduce ATP availability, contribute to tissue contractile and electrophysiologic heterogeneity, and promote a progression of AF from paroxysmal episodes to development of a substrate amenable to persistent arrhythmia.


Asunto(s)
Apéndice Atrial/metabolismo , Fibrilación Atrial/metabolismo , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteómica
7.
Hum Mol Genet ; 25(19): 4350-4368, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27577874

RESUMEN

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.


Asunto(s)
Enfermedades Cardiovasculares/genética , Estudio de Asociación del Genoma Completo , Ventrículos Cardíacos/fisiopatología , Canal de Sodio Activado por Voltaje NAV1.5/genética , Negro o Afroamericano/genética , Alelos , Enfermedades Cardiovasculares/mortalidad , Enfermedades Cardiovasculares/fisiopatología , Electrocardiografía , Femenino , Genotipo , Humanos , Masculino , Miocardio/patología , Polimorfismo de Nucleótido Simple/genética , Población Blanca/genética
8.
JAMA ; 320(22): 2354-2364, 2018 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-30535219

RESUMEN

Importance: Atrial fibrillation (AF) is the most common arrhythmia affecting 1% of the population. Young individuals with AF have a strong genetic association with the disease, but the mechanisms remain incompletely understood. Objective: To perform large-scale whole-genome sequencing to identify genetic variants related to AF. Design, Setting, and Participants: The National Heart, Lung, and Blood Institute's Trans-Omics for Precision Medicine Program includes longitudinal and cohort studies that underwent high-depth whole-genome sequencing between 2014 and 2017 in 18 526 individuals from the United States, Mexico, Puerto Rico, Costa Rica, Barbados, and Samoa. This case-control study included 2781 patients with early-onset AF from 9 studies and identified 4959 controls of European ancestry from the remaining participants. Results were replicated in the UK Biobank (346 546 participants) and the MyCode Study (42 782 participants). Exposures: Loss-of-function (LOF) variants in genes at AF loci and common genetic variation across the whole genome. Main Outcomes and Measures: Early-onset AF (defined as AF onset in persons <66 years of age). Due to multiple testing, the significance threshold for the rare variant analysis was P = 4.55 × 10-3. Results: Among 2781 participants with early-onset AF (the case group), 72.1% were men, and the mean (SD) age of AF onset was 48.7 (10.2) years. Participants underwent whole-genome sequencing at a mean depth of 37.8 fold and mean genome coverage of 99.1%. At least 1 LOF variant in TTN, the gene encoding the sarcomeric protein titin, was present in 2.1% of case participants compared with 1.1% in control participants (odds ratio [OR], 1.76 [95% CI, 1.04-2.97]). The proportion of individuals with early-onset AF who carried a LOF variant in TTN increased with an earlier age of AF onset (P value for trend, 4.92 × 10-4), and 6.5% of individuals with AF onset prior to age 30 carried a TTN LOF variant (OR, 5.94 [95% CI, 2.64-13.35]; P = 1.65 × 10-5). The association between TTN LOF variants and AF was replicated in an independent study of 1582 patients with early-onset AF (cases) and 41 200 control participants (OR, 2.16 [95% CI, 1.19-3.92]; P = .01). Conclusions and Relevance: In a case-control study, there was a statistically significant association between an LOF variant in the TTN gene and early-onset AF, with the variant present in a small percentage of participants with early-onset AF (the case group). Further research is necessary to understand whether this is a causal relationship.


Asunto(s)
Fibrilación Atrial/genética , Conectina/genética , Mutación con Pérdida de Función , Adulto , Edad de Inicio , Estudios de Casos y Controles , Femenino , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Heterocigoto , Humanos , Masculino , Persona de Mediana Edad , Control de Calidad
9.
Circulation ; 134(6): 486-98, 2016 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-27462069

RESUMEN

BACKGROUND: Adenosine provokes atrial fibrillation (AF) with a higher activation frequency in right atria (RA) versus left atria (LA) in patients, but the underlying molecular and functional substrates are unclear. We tested the hypothesis that adenosine-induced AF is driven by localized reentry in RA areas with highest expression of adenosine A1 receptor and its downstream GIRK (G protein-coupled inwardly rectifying potassium channels) channels (IK,Ado). METHODS: We applied biatrial optical mapping and immunoblot mapping of various atrial regions to reveal the mechanism of adenosine-induced AF in explanted failing and nonfailing human hearts (n=37). RESULTS: Optical mapping of coronary-perfused atria (n=24) revealed that adenosine perfusion (10-100 µmol/L) produced more significant shortening of action potential durations in RA (from 290±45 to 239±41 ms, 17.3±10.4%; P<0.01) than LA (from 307±24 to 286±23 ms, 6.7±6.6%; P<0.01). In 10 hearts, adenosine induced AF (317±116 s) that, when sustained (≥2 minutes), was primarily maintained by 1 to 2 localized reentrant drivers in lateral RA. Tertiapin (10-100 nmol/L), a selective GIRK channel blocker, counteracted adenosine-induced action potential duration shortening and prevented AF induction. Immunoblotting showed that the superior/middle lateral RA had significantly higher adenosine A1 receptor (2.7±1.7-fold; P<0.01) and GIRK4 (1.7±0.8-fold; P<0.05) protein expression than lateral/posterior LA. CONCLUSIONS: This study revealed a 3-fold RA-to-LA adenosine A1 receptor protein expression gradient in the human heart, leading to significantly greater RA versus LA repolarization sensitivity in response to adenosine. Sustained adenosine-induced AF is maintained by reentrant drivers localized in lateral RA regions with the highest adenosine A1 receptor/GIRK4 expression. Selective atrial GIRK channel blockade may effectively treat AF during conditions with increased endogenous adenosine.


Asunto(s)
Adenosina/toxicidad , Fibrilación Atrial/inducido químicamente , Fibrilación Atrial/metabolismo , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/biosíntesis , Atrios Cardíacos/metabolismo , Receptor de Adenosina A1/biosíntesis , Adulto , Anciano , Femenino , Regulación de la Expresión Génica , Corazón/diagnóstico por imagen , Corazón/efectos de los fármacos , Atrios Cardíacos/diagnóstico por imagen , Atrios Cardíacos/efectos de los fármacos , Sistema de Conducción Cardíaco/diagnóstico por imagen , Sistema de Conducción Cardíaco/efectos de los fármacos , Sistema de Conducción Cardíaco/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Técnicas de Cultivo de Órganos , Tomografía Computarizada por Tomografía de Emisión de Positrones
11.
Acta Cardiol Sin ; 33(2): 195-203, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28344424

RESUMEN

BACKGROUND: To investigate the left atrial (LA) size as an independent predictor of mortality following coronary artery bypass surgery (CABG). METHODS: This single center study evaluated determinants of mortality in 1070 patients who underwent isolated CABG from 2005-2014. Clinical, laboratory and demographic data were obtained from medical records. Collinearity between enlarged LA size (diameter ≥ 4 cm) and covariates was identified. The adjusted effects of enlarged LA size on 30-day mortality post CABG were tested using multiple logistic regression models. Adjusted odds ratios (OR) and 95% confidence intervals (CI) were reported. RESULTS: The mean age was 59 ± 9.8 years, and 238 patients were female. Two multivariate logistic regression models were evaluated. In Model A, mitral regurgitation (MR), ejection fraction, intensive care unit length-of-stay and variables found to be collinear with LA size as predictors of mortality were excluded. In model B, the collinear variables were included. By multivariate analysis (Model A), the statistically significant independent predictors of 30-day mortality after CABG were: enlarged LA size (OR 4.82, 95% CI 2.16-10.79), emergency CABG (OR 3.54, 95% CI 1.75-7.18), prolonged inotropic support (OR 2.79, 95% CI 1.38-5.6), diuretic use ≥ 1 month (OR 1.29, 95% CI 1.3-8.42), and use of clopidogrel within a week before surgery (OR 3.27, 95% CI 1.28-8.36. In Model B, enlarged LA and moderate MR were identified as independent predictors of 30-day mortality. CONCLUSIONS: Increased LA size is a strong independent predictor of mortality after isolated CABG.

12.
J Physiol ; 594(24): 7341-7360, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27558544

RESUMEN

KEY POINTS: Hyperammonaemia occurs in hepatic, cardiac and pulmonary diseases with increased muscle concentration of ammonia. We found that ammonia results in reduced skeletal muscle mitochondrial respiration, electron transport chain complex I dysfunction, as well as lower NAD+ /NADH ratio and ATP content. During hyperammonaemia, leak of electrons from complex III results in oxidative modification of proteins and lipids. Tricarboxylic acid cycle intermediates are decreased during hyperammonaemia, and providing a cell-permeable ester of αKG reversed the lower TCA cycle intermediate concentrations and increased ATP content. Our observations have high clinical relevance given the potential for novel approaches to reverse skeletal muscle ammonia toxicity by targeting the TCA cycle intermediates and mitochondrial ROS. ABSTRACT: Ammonia is a cytotoxic metabolite that is removed primarily by hepatic ureagenesis in humans. Hyperammonaemia occurs in advanced hepatic, cardiac and pulmonary disease, and in urea cycle enzyme deficiencies. Increased skeletal muscle ammonia uptake and metabolism are the major mechanism of non-hepatic ammonia disposal. Non-hepatic ammonia disposal occurs in the mitochondria via glutamate synthesis from α-ketoglutarate resulting in cataplerosis. We show skeletal muscle mitochondrial dysfunction during hyperammonaemia in a comprehensive array of human, rodent and cellular models. ATP synthesis, oxygen consumption, generation of reactive oxygen species with oxidative stress, and tricarboxylic acid (TCA) cycle intermediates were quantified. ATP content was lower in the skeletal muscle from cirrhotic patients, hyperammonaemic portacaval anastomosis rat, and C2C12 myotubes compared to appropriate controls. Hyperammonaemia in C2C12 myotubes resulted in impaired intact cell respiration, reduced complex I/NADH oxidase activity and electron leak occurring at complex III of the electron transport chain. Consistently, lower NAD+ /NADH ratio was observed during hyperammonaemia with reduced TCA cycle intermediates compared to controls. Generation of reactive oxygen species resulted in increased content of skeletal muscle carbonylated proteins and thiobarbituric acid reactive substances during hyperammonaemia. A cell-permeable ester of α-ketoglutarate reversed the low TCA cycle intermediates and ATP content in myotubes during hyperammonaemia. However, the mitochondrial antioxidant MitoTEMPO did not reverse the lower ATP content during hyperammonaemia. We provide for the first time evidence that skeletal muscle hyperammonaemia results in mitochondrial dysfunction and oxidative stress. Use of anaplerotic substrates to reverse ammonia-induced mitochondrial dysfunction is a novel therapeutic approach.


Asunto(s)
Hiperamonemia/metabolismo , Mitocondrias Musculares/metabolismo , Músculo Esquelético/metabolismo , Estrés Oxidativo , Adenosina Trifosfato/metabolismo , Anciano , Animales , Línea Celular , Respiración de la Célula , Creatina Quinasa/metabolismo , Femenino , Humanos , Cirrosis Hepática/metabolismo , Masculino , Persona de Mediana Edad , Cadenas Pesadas de Miosina/metabolismo , NAD/metabolismo , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo
14.
Circulation ; 130(15): 1225-35, 2014 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-25124494

RESUMEN

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.


Asunto(s)
Fibrilación Atrial/genética , Conexina 43/genética , Proteínas de Homeodominio/genética , Proteínas Nucleares/genética , Proteínas Represoras/genética , Proteínas de Dominio T Box/genética , Ubiquitina-Proteína Ligasas/genética , Anciano , Animales , Fibrilación Atrial/etnología , Fibrilación Atrial/fisiopatología , Mapeo Cromosómico , Conexina 43/fisiología , Europa (Continente) , Femenino , Técnicas de Silenciamiento del Gen , Sitios Genéticos/fisiología , Predisposición Genética a la Enfermedad/etnología , Genotipo , Proteínas de Homeodominio/fisiología , Humanos , Japón , Masculino , Persona de Mediana Edad , Proteínas Musculares , Proteínas Nucleares/fisiología , Sitios de Carácter Cuantitativo , Proteínas Represoras/fisiología , Proteínas de Dominio T Box/fisiología , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Ubiquitina-Proteína Ligasas/fisiología , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/fisiología , Proteína del Homeodomínio PITX2
15.
Magn Reson Med ; 74(5): 1370-9, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25408542

RESUMEN

PURPOSE: The aim of this study was to develop a rapid, multislice cardiac T1 mapping method in mice and to apply the method to quantify manganese (Mn(2+)) uptake in a mouse model with altered Ca(2+) channel activity. METHODS: An electrocardiography-triggered multislice saturation-recovery Look-Locker method was developed and validated both in vitro and in vivo. A two-dose study was performed to investigate the kinetics of T1 shortening, Mn(2+) relaxivity in myocardium, and the impact of Mn(2+) on cardiac function. The sensitivity of Mn(2+)-enhanced MRI in detecting subtle changes in altered Ca(2+) channel activity was evaluated in a mouse model with α-dystrobrevin knockout. RESULTS: Validation studies showed strong agreement between the current method and an established method. High Mn(2+) dose led to significantly accelerated T1 shortening. Heart rate decreased during Mn(2+) infusion, while ejection ratio increased slightly at the end of imaging protocol. No statistical difference in cardiac function was detected between the two dose groups. Mice with α-dystrobrevin knockout showed enhanced Mn(2+) uptake in vivo. In vitro patch-clamp study showed increased Ca(2+) channel activity. CONCLUSION: The saturation recovery method provides rapid T1 mapping in mouse hearts, which allowed sensitive detection of subtle changes in Mn(2+) uptake in α-dystrobrevin knockout mice.


Asunto(s)
Técnicas de Imagen Cardíaca/métodos , Proteínas Asociadas a la Distrofina/genética , Procesamiento de Imagen Asistido por Computador/métodos , Imagen por Resonancia Magnética/métodos , Manganeso/farmacocinética , Animales , Calcio/metabolismo , Proteínas Asociadas a la Distrofina/metabolismo , Corazón/anatomía & histología , Corazón/fisiología , Masculino , Ratones , Ratones Noqueados , Fantasmas de Imagen
16.
J Cardiovasc Pharmacol ; 66(6): 523-9, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26335221

RESUMEN

Atrial fibrillation (AF) is an important cause of stroke and risk factor for heart failure and death. Current pharmacologic treatments for AF have limited efficacy, and treatments that more directly target the underlying causes of AF are needed. Oxidant stress and inflammatory activation are interrelated pathways that promote atrial electrical and structural remodeling, leading to atrial ectopy, interstitial fibrosis, and increased stroke risk. This review evaluates the impact of common stressors on atrial oxidant stress and inflammatory activation and the contribution of these pathways to atrial remodeling. Recent studies suggest that integrated efforts to target the underlying risk factors, rather than the AF per se, may have a greater impact on health and outcomes than isolated efforts focused on the electrical abnormalities.


Asunto(s)
Antiinflamatorios/administración & dosificación , Antioxidantes/administración & dosificación , Fibrilación Atrial/tratamiento farmacológico , Mediadores de Inflamación/antagonistas & inhibidores , Oxidantes/antagonistas & inhibidores , Estrés Oxidativo/efectos de los fármacos , Animales , Fibrilación Atrial/metabolismo , Sistemas de Liberación de Medicamentos/tendencias , Humanos , Mediadores de Inflamación/metabolismo , Oxidantes/metabolismo , Estrés Oxidativo/fisiología
17.
Circ J ; 79(3): 495-502, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25746525

RESUMEN

Atrial fibrillation (AF) is the most common clinically relevant arrhythmia, but the methods available for treating AF and its complications (of which the most important is thrombogenesis), as well as for assessing AF risk and underlying pathophysiology, are largely limited. Emerging evidence suggests a significant role of inflammation in the pathogenesis of AF. That evidence includes elevated serum levels of inflammatory biomarkers in AF subjects, the expression of inflammatory markers in cardiac tissues of AF patients and animal models of AF, and beneficial effects of anti-inflammatory drugs in experimental AF paradigms. Inflammation is suggested to be linked to various pathological processes, such as oxidative stress, apoptosis, and fibrosis, that promote AF substrate formation. Inflammation has also been associated with endothelial dysfunction, platelet activation, and coagulation cascade activation, leading to thrombogenesis. Thus, inflammation may contribute to both the occurrence/maintenance of AF and its thromboembolic complications. Here, we review the evidence for a role of inflammation and inflammatory biomarkers in the risk management and treatment of AF. We also summarize the current knowledge of inflammation-dependent cellular and molecular mechanisms in AF pathophysiology and their potential as therapeutic targets.


Asunto(s)
Apoptosis , Fibrilación Atrial , Coagulación Sanguínea , Mediadores de Inflamación/sangre , Estrés Oxidativo , Animales , Fibrilación Atrial/sangre , Fibrilación Atrial/fisiopatología , Fibrilación Atrial/terapia , Biomarcadores , Fibrosis , Humanos , Inflamación/sangre , Inflamación/patología , Inflamación/fisiopatología
19.
JACC Clin Electrophysiol ; 10(9): 2074-2084, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39023484

RESUMEN

BACKGROUND: While sleep disorders are implicated in atrial fibrillation (AF), the interplay of physiologic alterations and symptoms remains unclear. Sleep-based phenotypes can account for this complexity and translate to actionable approaches to identify at-risk patients and therapeutic interventions. OBJECTIVES: This study hypothesized discrete phenotypes of symptoms and polysomnography (PSG)-based data differ in relation to incident AF. METHODS: Data from the STARLIT (sleep Signals, Testing, And Reports LInked to patient Traits) registry on Cleveland Clinic patients (≥18 years of age) who underwent PSG from November 27, 2004, to December 30,2015, were retrospectively examined. Phenotypes were identified using latent class analysis of symptoms and PSG-based measures of sleep-disordered breathing and sleep architecture. Phenotypes were included as the primary predictor in a multivariable-adjusted Cox proportional hazard models for incident AF. RESULTS: In our cohort (N = 43,433, age 51.8 ± 14.5 years, 51.9% male, 74.9% White), 7.3% (n = 3,166) had baseline AF. Over a 7.6- ± 3.4-year follow-up period, 8.9% (n = 3,595) developed incident AF. Five phenotypes were identified. The hypoxia subtype (n = 3,245) had 48% increased incident AF (HR: 1.48; 95% CI: 1.34-1.64), the apneas + arousals subtype (n = 4,592) had 22% increased incident AF (HR: 1.22; 95% CI: 1.10-1.35), and the short sleep + nonrapid eye movement subtype (n = 6,126) had 11% increased incident AF (HR: 1.11; 95% CI: 1.01-1.22) compared with long sleep + rapid eye movement (n = 26,809), the reference group. The hypopneas subtype (n = 2,661) did not differ from reference (HR: 0.89; 95% CI: 0.77-1.03). CONCLUSIONS: Consistent with prior evidence supporting hypoxia as an AF driver and cardiac risk of the sleepy phenotype, this constellation of symptoms and physiologic alterations illustrates vulnerability for AF development, providing potential value in enhancing our understanding of integrated sleep-specific symptoms and physiologic risk of atrial arrhythmogenesis.


Asunto(s)
Fibrilación Atrial , Fenotipo , Polisomnografía , Síndromes de la Apnea del Sueño , Humanos , Fibrilación Atrial/epidemiología , Fibrilación Atrial/fisiopatología , Masculino , Femenino , Persona de Mediana Edad , Anciano , Estudios Retrospectivos , Adulto , Síndromes de la Apnea del Sueño/epidemiología , Síndromes de la Apnea del Sueño/fisiopatología , Síndromes de la Apnea del Sueño/complicaciones , Incidencia , Sueño/fisiología , Factores de Riesgo
20.
Circ Genom Precis Med ; 17(5): e004750, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39355904

RESUMEN

BACKGROUND: Atrial fibrillation GWAS (genome-wide association studies) identified significant associations for rs1152591 and linked variants in the SYNE2 gene encoding Nesprin-2, which connects the nuclear membrane with the cytoskeleton. METHODS: Reporter gene vector transfection and CRISPR-Cas9 editing were used to identify the causal variant regulating the expression of SYNE2α1. After SYNE2 knockdown or SYNE2α1 overexpression in human stem cell-derived cardiomyocytes, nuclear phenotypes were assessed by imaging and atomic force microscopy. Gene expression was assessed by RNAseq and gene set enrichment analysis. Fura-2 AM staining assessed calcium transients. Optical mapping assessed action potential duration and conduction velocity. RESULTS: The risk allele of rs1152591 had lower promoter and enhancer activity and was significantly associated with lower expression of the short SYNE2α1 isoform in human stem cell-derived cardiomyocytes, without an effect on the expression of the full-length SYNE2 mRNA. SYNE2α1 overexpression had dominant negative effects on the nucleus with its overexpression or SYNE2 knockdown leading to increased nuclear area and decreased nuclear stiffness. Gene expression results from SYNE2α1 overexpression demonstrated both concordant and nonconcordant effects with SYNE2 knockdown. SYNE2α1 overexpression had a gain of function on electrophysiology, leading to significantly faster calcium reuptake and decreased assessed action potential duration, while SYNE2 knockdown showed both shortened assessed action potential duration and decreased conduction velocity. CONCLUSIONS: rs1152591 was identified as a causal atrial fibrillation variant, with the risk allele decreasing SYNE2α1 expression. Downstream effects of SYNE2α1 overexpression include changes in nuclear stiffness and electrophysiology, which may contribute to the mechanism for the risk allele's association with AF.


Asunto(s)
Fibrilación Atrial , Miocitos Cardíacos , Proteínas del Tejido Nervioso , Humanos , Fibrilación Atrial/genética , Fibrilación Atrial/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Núcleo Celular/metabolismo , Polimorfismo de Nucleótido Simple , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Estudio de Asociación del Genoma Completo , Alelos , Proteínas de Microfilamentos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA