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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Circ Res ; 133(4): 313-329, 2023 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-37449401

RESUMEN

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.


Asunto(s)
Fibrilación Atrial , Proteínas de Homeodominio , Animales , Humanos , Ratones , Fibrilación Atrial/genética , Calcio/metabolismo , Dilatación , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Miocitos Cardíacos/metabolismo , Factores de Transcripción/genética
2.
PLoS Biol ; 19(4): e3001144, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33872299

RESUMEN

Delineating human cardiac pathologies and their basic molecular mechanisms relies on research conducted in model organisms. Yet translating findings from preclinical models to humans present a significant challenge, in part due to differences in cardiac protein expression between humans and model organisms. Proteins immediately determine cellular function, yet their large-scale investigation in hearts has lagged behind those of genes and transcripts. Here, we set out to bridge this knowledge gap: By analyzing protein profiles in humans and commonly used model organisms across cardiac chambers, we determine their commonalities and regional differences. We analyzed cardiac tissue from each chamber of human, pig, horse, rat, mouse, and zebrafish in biological replicates. Using mass spectrometry-based proteomics workflows, we measured and evaluated the abundance of approximately 7,000 proteins in each species. The resulting knowledgebase of cardiac protein signatures is accessible through an online database: atlas.cardiacproteomics.com. Our combined analysis allows for quantitative evaluation of protein abundances across cardiac chambers, as well as comparisons of cardiac protein profiles across model organisms. Up to a quarter of proteins with differential abundances between atria and ventricles showed opposite chamber-specific enrichment between species; these included numerous proteins implicated in cardiac disease. The generated proteomics resource facilitates translational prospects of cardiac studies from model organisms to humans by comparisons of disease-linked protein networks across species.


Asunto(s)
Miocardio/metabolismo , Proteoma/metabolismo , Animales , Corazón/fisiología , Ventrículos Cardíacos/química , Ventrículos Cardíacos/metabolismo , Caballos , Humanos , Ratones , Modelos Animales , Miocardio/química , Especificidad de Órganos , Procesamiento Proteico-Postraduccional , Proteoma/análisis , Proteómica/métodos , Ratas , Especificidad de la Especie , Porcinos , Pez Cebra
3.
Hum Mutat ; 41(12): 2195-2204, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33131149

RESUMEN

The identification of a pathogenic SCN5A variant confers an increased risk of conduction defects and ventricular arrhythmias (VA) in Brugada syndrome (BrS). However, specific aspects of sodium channel function that influence clinical phenotype have not been defined. A systematic literature search identified SCN5A variants associated with BrS. Sodium current (INa ) functional parameters (peak current, decay, steady-state activation and inactivation, and recovery from inactivation) and clinical features (conduction abnormalities [CA], spontaneous VA or family history of sudden cardiac death [SCD], and spontaneous BrS electrocardiogram [ECG]) were extracted. A total of 561 SCN5A variants associated with BrS were identified, for which data on channel function and clinical phenotype were available in 142. In the primary analysis, no relationship was found between any aspect of channel function and CA, VA/SCD, or spontaneous BrS ECG pattern. Sensitivity analyses including only variants graded pathogenic or likely pathogenic suggested that reduction in peak current and positive shift in steady-state activation were weakly associated with CA and VA/SCD, although sensitivity and specificity remained low. The relationship between in vitro assessment of channel function and BrS clinical phenotype is weak. The assessment of channel function does not enhance risk stratification. Caution is needed when extrapolating functional testing to the likelihood of variant pathogenicity.


Asunto(s)
Síndrome de Brugada/genética , Síndrome de Brugada/patología , Mutación/genética , Canal de Sodio Activado por Voltaje NAV1.5/genética , Arritmias Cardíacas/genética , Síndrome de Brugada/diagnóstico por imagen , Electrocardiografía , Sistema de Conducción Cardíaco/patología , Humanos , Fenotipo
4.
Circ Res ; 118(1): 56-72, 2016 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-26503464

RESUMEN

RATIONALE: More than 25 million individuals have heart failure worldwide, with ≈4000 patients currently awaiting heart transplantation in the United States. Donor organ shortage and allograft rejection remain major limitations with only ≈2500 hearts transplanted each year. As a theoretical alternative to allotransplantation, patient-derived bioartificial myocardium could provide functional support and ultimately impact the treatment of heart failure. OBJECTIVE: The objective of this study is to translate previous work to human scale and clinically relevant cells for the bioengineering of functional myocardial tissue based on the combination of human cardiac matrix and human induced pluripotent stem cell-derived cardiomyocytes. METHODS AND RESULTS: To provide a clinically relevant tissue scaffold, we translated perfusion-decellularization to human scale and obtained biocompatible human acellular cardiac scaffolds with preserved extracellular matrix composition, architecture, and perfusable coronary vasculature. We then repopulated this native human cardiac matrix with cardiomyocytes derived from nontransgenic human induced pluripotent stem cells and generated tissues of increasing 3-dimensional complexity. We maintained such cardiac tissue constructs in culture for 120 days to demonstrate definitive sarcomeric structure, cell and matrix deformation, contractile force, and electrical conduction. To show that functional myocardial tissue of human scale can be built on this platform, we then partially recellularized human whole-heart scaffolds with human induced pluripotent stem cell-derived cardiomyocytes. Under biomimetic culture, the seeded constructs developed force-generating human myocardial tissue and showed electrical conductivity, left ventricular pressure development, and metabolic function. CONCLUSIONS: Native cardiac extracellular matrix scaffolds maintain matrix components and structure to support the seeding and engraftment of human induced pluripotent stem cell-derived cardiomyocytes and enable the bioengineering of functional human myocardial-like tissue of multiple complexities.


Asunto(s)
Bioingeniería/métodos , Matriz Extracelular/fisiología , Miocardio/citología , Células Madre Pluripotentes/fisiología , Adulto , Anciano , Diferenciación Celular/fisiología , Femenino , Humanos , Masculino , Persona de Mediana Edad
5.
Cardiovasc Res ; 120(8): 927-942, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38661182

RESUMEN

AIMS: In patients with heart failure (HF), concomitant sinus node dysfunction (SND) is an important predictor of mortality, yet its molecular underpinnings are poorly understood. Using proteomics, this study aimed to dissect the protein and phosphorylation remodelling within the sinus node in an animal model of HF with concurrent SND. METHODS AND RESULTS: We acquired deep sinus node proteomes and phosphoproteomes in mice with heart failure and SND and report extensive remodelling. Intersecting the measured (phospho)proteome changes with human genomics pharmacovigilance data, highlighted downregulated proteins involved in electrical activity such as the pacemaker ion channel, Hcn4. We confirmed the importance of ion channel downregulation for sinus node physiology using computer modelling. Guided by the proteomics data, we hypothesized that an inflammatory response may drive the electrophysiological remodeling underlying SND in heart failure. In support of this, experimentally induced inflammation downregulated Hcn4 and slowed pacemaking in the isolated sinus node. From the proteomics data we identified proinflammatory cytokine-like protein galectin-3 as a potential target to mitigate the effect. Indeed, in vivo suppression of galectin-3 in the animal model of heart failure prevented SND. CONCLUSION: Collectively, we outline the protein and phosphorylation remodeling of SND in heart failure, we highlight a role for inflammation in electrophysiological remodelling of the sinus node, and we present galectin-3 signalling as a target to ameliorate SND in heart failure.


Asunto(s)
Modelos Animales de Enfermedad , Insuficiencia Cardíaca , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Ratones Endogámicos C57BL , Proteómica , Síndrome del Seno Enfermo , Nodo Sinoatrial , Animales , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Nodo Sinoatrial/metabolismo , Nodo Sinoatrial/fisiopatología , Fosforilación , Síndrome del Seno Enfermo/metabolismo , Síndrome del Seno Enfermo/fisiopatología , Síndrome del Seno Enfermo/genética , Masculino , Mediadores de Inflamación/metabolismo , Inflamación/metabolismo , Inflamación/fisiopatología , Inflamación/patología , Frecuencia Cardíaca , Canales de Potasio/metabolismo , Canales de Potasio/genética , Simulación por Computador , Modelos Cardiovasculares , Humanos , Transducción de Señal , Potenciales de Acción
6.
Nat Cardiovasc Res ; 2(7): 673-692, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38666184

RESUMEN

Protein-protein interactions are essential for normal cellular processes and signaling events. Defining these interaction networks is therefore crucial for understanding complex cellular functions and interpretation of disease-associated gene variants. We need to build a comprehensive picture of the interactions, their affinities and interdependencies in the specific organ to decipher hitherto poorly understood signaling mechanisms through ion channels. Here we report the experimental identification of the ensemble of protein interactors for 13 types of ion channels in murine cardiac tissue. Of these, we validated the functional importance of ten interactors on cardiac electrophysiology through genetic knockouts in zebrafish, gene silencing in mice, super-resolution microscopy and patch clamp experiments. Furthermore, we establish a computational framework to reconstruct human cardiomyocyte ion channel networks from deep proteome mapping of human heart tissue and human heart single-cell gene expression data. Finally, we integrate the ion channel interactome with human population genetics data to identify proteins that influence the electrocardiogram (ECG). We demonstrate that the combined channel network is enriched for proteins influencing the ECG, with 44% of the network proteins significantly associated with an ECG phenotype. Altogether, we define interactomes of 13 major cardiac ion channels, contextualize their relevance to human electrophysiology and validate functional roles of ten interactors, including two regulators of the sodium current (epsin-2 and gelsolin). Overall, our data provide a roadmap for our understanding of the molecular machinery that regulates cardiac electrophysiology.

7.
Circulation ; 123(1): 23-30, 2011 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-21098441

RESUMEN

BACKGROUND: Genetic long QT (LQT) syndrome is a life-threatening disorder caused by mutations that result in prolongation of cardiac repolarization. Recent work has demonstrated that a zebrafish model of LQT syndrome faithfully recapitulates several features of human disease, including prolongation of ventricular action potential duration, spontaneous early afterdepolarizations, and 2:1 atrioventricular block in early stages of development. Because of their transparency, small size, and absorption of small molecules from their environment, zebrafish are amenable to high-throughput chemical screens. We describe a small-molecule screen using the zebrafish KCNH2 mutant breakdance to identify compounds that can rescue the LQT type 2 phenotype. METHODS AND RESULTS: Zebrafish breakdance embryos were exposed to test compounds at 48 hours of development and scored for rescue of 2:1 atrioventricular block at 72 hours in a 96-well format. Only compounds that suppressed the LQT phenotype in 3 of 3 fish were considered hits. Screen compounds were obtained from commercially available small-molecule libraries (Prestwick and Chembridge). Initial hits were confirmed with dose-response testing and time-course studies. Optical mapping with the voltage-sensitive dye di-4 ANEPPS was performed to measure compound effects on cardiac action potential durations. Screening of 1200 small molecules resulted in the identification of flurandrenolide and 2-methoxy-N-(4-methylphenyl) benzamide (2-MMB) as compounds that reproducibly suppressed the LQT phenotype. Optical mapping confirmed that treatment with each compound caused shortening of ventricular action potential durations. Structure activity studies and steroid receptor knockdown suggest that flurandrenolide functions via the glucocorticoid signaling pathway. CONCLUSIONS: Using a zebrafish model of LQT type 2 syndrome in a high-throughput chemical screen, we have identified 2 compounds, flurandrenolide and the novel compound 2-MMB, as small molecules that rescue the zebrafish LQT type 2 syndrome by shortening the ventricular action potential duration. We provide evidence that flurandrenolide functions via the glucocorticoid receptor-mediated pathway. These 2 molecules and future discoveries from this screen should yield novel tools for the study of cardiac electrophysiology and may lead to novel therapeutics for human LQT patients.


Asunto(s)
Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/prevención & control , Proteínas de Pez Cebra/genética , Potenciales de Acción/fisiología , Animales , Animales Modificados Genéticamente , Células COS , Chlorocebus aethiops , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/genética , Flurandrenolona/uso terapéutico , Técnicas de Silenciamiento del Gen/métodos , Células HEK293 , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Síndrome de QT Prolongado/fisiopatología , Mutación/genética , Pez Cebra
8.
Fetal Pediatr Pathol ; 30(2): 106-10, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21391750

RESUMEN

Intussusception is the leading cause of intestinal obstruction in children and is almost invariably idiopathic. Occasionally, there is a lead point for the intussusception. Intussusception caused by heterotopic pancreas (HPT) as the lead point is exceedingly rare. We report a case of intussusception caused by HPT in a child. Clinical and pathologic features and the successful medical and surgical management of the case are discussed.


Asunto(s)
Coristoma/complicaciones , Coristoma/patología , Enfermedades del Íleon/etiología , Intususcepción/etiología , Páncreas/patología , Niño , Coristoma/cirugía , Humanos , Enfermedades del Íleon/patología , Enfermedades del Íleon/cirugía , Lactante , Intususcepción/patología , Intususcepción/cirugía , Masculino
9.
JACC CardioOncol ; 3(1): 88-97, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34396309

RESUMEN

BACKGROUND: Ibrutinib is a protein kinase inhibitor that has been widely successful in treating multiple common variations of B-cell cancers. However, an unfortunate side effect of ibrutinib is that it predisposes patients to development of atrial fibrillation. OBJECTIVES: The purpose of this study was to assess other commonly prescribed protein kinase inhibitors for similar pro-arrhythmic liability. METHODS: This study comprehensively evaluated data from the U.S. Food and Drug Administration adverse events reporting system and determined the reporting of cardiac arrhythmia attributed to kinase inhibitor therapy using a multivariable logistic regression model. We evaluated 3,663,300 case reports containing 23,067 cases of atrial fibrillation and 66,262 cases of cardiac arrhythmia. In total, 32 protein kinase inhibitors were evaluated, almost all of which are oncotherapeutics. RESULTS: Seven protein kinase inhibitors were associated with a significant increase in the odds of atrial fibrillation (ibrutinib, ponatinib, nilotinib, ribociclib, trametinib, osimertinib, and idelalisib). Assessment of broader pro-arrhythmic toxicity suggested a ventricular-specific liability for nilotinib and a bradyarrhythmia risk with alectinib and crizotinib. CONCLUSIONS: Compounds that result in the inhibition of a number of protein kinases are associated with an increased risk of cardiac rhythm disturbances. The mechanisms driving the arrhythmogenic effects remain to be discovered, but this study presents an important step in identifying and prioritizing the study of these protein kinase signaling pathways.

10.
Proc Natl Acad Sci U S A ; 104(46): 17977-82, 2007 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-17951430

RESUMEN

The consumption of garlic is inversely correlated with the progression of cardiovascular disease, although the responsible mechanisms remain unclear. Here we show that human RBCs convert garlic-derived organic polysulfides into hydrogen sulfide (H(2)S), an endogenous cardioprotective vascular cell signaling molecule. This H(2)S production, measured in real time by a novel polarographic H(2)S sensor, is supported by glucose-maintained cytosolic glutathione levels and is to a large extent reliant on reduced thiols in or on the RBC membrane. H(2)S production from organic polysulfides is facilitated by allyl substituents and by increasing numbers of tethering sulfur atoms. Allyl-substituted polysulfides undergo nucleophilic substitution at the alpha carbon of the allyl substituent, thereby forming a hydropolysulfide (RS(n)H), a key intermediate during the formation of H(2)S. Organic polysulfides (R-S(n)-R'; n > 2) also undergo nucleophilic substitution at a sulfur atom, yielding RS(n)H and H(2)S. Intact aorta rings, under physiologically relevant oxygen levels, also metabolize garlic-derived organic polysulfides to liberate H(2)S. The vasoactivity of garlic compounds is synchronous with H(2)S production, and their potency to mediate relaxation increases with H(2)S yield, strongly supporting our hypothesis that H(2)S mediates the vasoactivity of garlic. Our results also suggest that the capacity to produce H(2)S can be used to standardize garlic dietary supplements.


Asunto(s)
Eritrocitos/efectos de los fármacos , Ajo/química , Sulfuro de Hidrógeno/farmacología , Acetilcisteína/farmacología , Cromatografía Líquida de Alta Presión , Electroquímica , Eritrocitos/metabolismo , Glutatión/sangre , Disulfuro de Glutatión/sangre , Humanos , Sulfuro de Hidrógeno/sangre
11.
Circ Genom Precis Med ; 11(5): e001663, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29752399

RESUMEN

BACKGROUND: Genetic variants at the SCN5A/SCN10A locus are strongly associated with electrocardiographic PR and QRS intervals. While SCN5A is the canonical cardiac sodium channel gene, the role of SCN10A in cardiac conduction is less well characterized. METHODS: We sequenced the SCN10A locus in 3699 European-ancestry individuals to identify variants associated with cardiac conduction, and replicated our findings in 21,000 individuals of European ancestry. We examined association with expression in human atrial tissue. We explored the biophysical effect of variation on channel function using cellular electrophysiology. RESULTS: We identified 2 intronic single nucleotide polymorphisms in high linkage disequilibrium (r 2=0.86) with each other to be the strongest signals for PR (rs10428132, ß=-4.74, P=1.52×10-14) and QRS intervals (rs6599251, QRS ß=-0.73; P=1.2×10-4), respectively. Although these variants were not associated with SCN5A or SCN10A expression in human atrial tissue (n=490), they were in high linkage disequilibrium (r 2≥0.72) with a common SCN10A missense variant, rs6795970 (V1073A). In total, we identified 7 missense variants, 4 of which (I962V, P1045T, V1073A, and L1092P) were associated with cardiac conduction. These 4 missense variants cluster in the cytoplasmic linker of the second and third domains of the SCN10A protein and together form 6 common haplotypes. Using cellular electrophysiology, we found that haplotypes associated with shorter PR intervals had a significantly larger percentage of late current compared with wild-type (I962V+V1073A+L1092P, 20.2±3.3%, P=0.03, and I962V+V1073A, 22.4±0.8%, P=0.0004 versus wild-type 11.7±1.6%), and the haplotype associated with the longest PR interval had a significantly smaller late current percentage (P1045T, 6.4±1.2%, P=0.03). CONCLUSIONS: Our findings suggest an association between genetic variation in SCN10A, the late sodium current, and alterations in cardiac conduction.


Asunto(s)
Estudios de Asociación Genética , Sistema de Conducción Cardíaco/metabolismo , Activación del Canal Iónico/genética , Canal de Sodio Activado por Voltaje NAV1.8/genética , Polimorfismo de Nucleótido Simple/genética , Fenómenos Biofísicos , Electrocardiografía , Haplotipos/genética , Humanos , Mutación Missense/genética , Sitios de Carácter Cuantitativo/genética
12.
Sci Rep ; 7(1): 11839, 2017 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-28928472

RESUMEN

Optogenetics is a powerful research approach that allows localized optical modulation of selected cells within an animal via the expression of genetically encoded photo-excitable ion channels. Commonly used optogenetic techniques rely on the expression of microbial opsin variants, which have many excellent features but suffer from various degrees of blue spectral overlap and limited channel conductance. Here, we expand the optogenetics toolbox in the form of a tunable, high-conductance vertebrate cation channel, zTrpa1b, coupled with photo-activated channel ligands, such as optovin and 4g6. Our results demonstrate that zTrpa1b/ligand pairing offers high light sensitivity, millisecond-scale response latency in vivo, as well as adjustable channel off latency. Exogenous in vivo expression of zTrpa1b in sensory neurons allowed subcellular photo-activation, enabling light-dependent motor control. zTrpa1b/ligand was also suitable for cardiomyocyte pacing, as shown in experiments performed on zebrafish hearts in vivo as well as in human stem cell-derived cardiomyocytes in vitro. Therefore, zTrpa1b/optovin represents a novel tool for flexible, high-conductance optogenetics.


Asunto(s)
Optogenética/instrumentación , Optogenética/métodos , Canal Catiónico TRPA1 , Proteínas de Pez Cebra , Pez Cebra , Animales , Células HEK293 , Sistema de Conducción Cardíaco/metabolismo , Humanos , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Canal Catiónico TRPA1/química , Canal Catiónico TRPA1/genética , Canal Catiónico TRPA1/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/química , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
13.
Circ Cardiovasc Genet ; 10(6)2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29212899

RESUMEN

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.


Asunto(s)
Cardiomiopatía Restrictiva/genética , Filaminas/genética , Predisposición Genética a la Enfermedad , Mutación/genética , Adulto , Anciano , Secuencia de Aminoácidos , Secuencia de Bases , Cardiomiopatía Restrictiva/patología , Familia , Femenino , Filaminas/química , Humanos , Masculino , Persona de Mediana Edad , Fenotipo
14.
Circ Cardiovasc Genet ; 10(5)2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28974514

RESUMEN

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.


Asunto(s)
Potenciales de Acción/genética , Fibrilación Atrial , Proteínas de Homeodominio , Células Madre Embrionarias Humanas/metabolismo , Miocitos Cardíacos/metabolismo , Polimorfismo de Nucleótido Simple , Animales , Animales Modificados Genéticamente , Fibrilación Atrial/genética , Fibrilación Atrial/metabolismo , Fibrilación Atrial/fisiopatología , Línea Celular , Proteínas de Homeodominio/biosíntesis , Proteínas de Homeodominio/genética , Células Madre Embrionarias Humanas/patología , Humanos , Ratones , Miocitos Cardíacos/patología , Pez Cebra
15.
Stem Cell Reports ; 2(2): 163-70, 2014 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-24527390

RESUMEN

In addition to their promise in regenerative medicine, pluripotent stem cells have proved to be faithful models of many human diseases. In particular, patient-specific stem cell-derived cardiomyocytes recapitulate key features of several life-threatening cardiac arrhythmia syndromes. For both modeling and regenerative approaches, phenotyping of stem cell-derived tissues is critical. Cellular phenotyping has largely relied upon expression of lineage markers rather than physiologic attributes. This is especially true for cardiomyocytes, in part because electrophysiological recordings are labor intensive. Likewise, most optical voltage indicators suffer from phototoxicity, which damages cells and degrades signal quality. Here we present the use of a genetically encoded fluorescent voltage indicator, ArcLight, which we demonstrate can faithfully report transmembrane potentials in human stem cell-derived cardiomyocytes. We demonstrate the application of this fluorescent sensor in high-throughput, serial phenotyping of differentiating cardiomyocyte populations and in screening for drug-induced cardiotoxicity.


Asunto(s)
Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Fenotipo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Potenciales de Acción/efectos de los fármacos , Biomarcadores , Diferenciación Celular , Línea Celular , Medios de Cultivo Condicionados/farmacología , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Expresión Génica , Genes Reporteros , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Reproducibilidad de los Resultados
16.
Cardiovasc Res ; 101(2): 326-34, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24296650

RESUMEN

BACKGROUND: A recent genome-wide association study identified a susceptibility locus for atrial fibrillation at the KCNN3 gene. Since the KCNN3 gene encodes for a small conductance calcium-activated potassium channel, we hypothesized that overexpression of the SK3 channel increases susceptibility to cardiac arrhythmias. METHODS AND RESULTS: We characterized the cardiac electrophysiological phenotype of a mouse line with overexpression of the SK3 channel. We generated homozygote (SK3(T/T)) and heterozygote (SK3(+/T)) mice with overexpression of the channel and compared them with wild-type (WT) controls. We observed a high incidence of sudden death among SK3(T/T) mice (7 of 19 SK3(T/T) mice). Ambulatory monitoring demonstrated that sudden death was due to heart block and bradyarrhythmias. SK3(T/T) mice displayed normal body weight, temperature, and cardiac function on echocardiography; however, histological analysis demonstrated that these mice have abnormal atrioventricular node morphology. Optical mapping demonstrated that SK3(T/T) mice have slower ventricular conduction compared with WT controls (SK3(T/T) vs. WT; 0.45 ± 0.04 vs. 0.60 ± 0.09 mm/ms, P = 0.001). Programmed stimulation in 1-month-old SK3(T/T) mice demonstrated inducible atrial arrhythmias (50% of SK3(T/T) vs. 0% of WT mice) and also a shorter atrioventricular nodal refractory period (SK3(T/T) vs. WT; 43 ± 6 vs. 52 ± 9 ms, P = 0.02). Three-month-old SK3(T/T) mice on the other hand displayed a trend towards a more prolonged atrioventricular nodal refractory period (SK3(T/T) vs. WT; 61 ± 1 vs. 52 ± 6 ms, P = 0.06). CONCLUSION: Overexpression of the SK3 channel causes an increased risk of sudden death associated with bradyarrhythmias and heart block, possibly due to atrioventricular nodal dysfunction.


Asunto(s)
Nodo Atrioventricular/metabolismo , Bradicardia/metabolismo , Muerte Súbita Cardíaca/etiología , Bloqueo Cardíaco/metabolismo , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Potenciales de Acción , Animales , Nodo Atrioventricular/anomalías , Nodo Atrioventricular/fisiopatología , Bradicardia/genética , Bradicardia/fisiopatología , Estimulación Cardíaca Artificial , Conexina 43/metabolismo , Electrocardiografía Ambulatoria , Predisposición Genética a la Enfermedad , Bloqueo Cardíaco/genética , Bloqueo Cardíaco/fisiopatología , Heterocigoto , Homocigoto , Ratones , Ratones Transgénicos , Fenotipo , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/genética , Factores de Tiempo , Regulación hacia Arriba , Imagen de Colorante Sensible al Voltaje
17.
Circ Arrhythm Electrophysiol ; 2(5): 571-9, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19843926

RESUMEN

BACKGROUND: Conventional right ventricular (RV) apex pacing can lead to adverse clinical outcome associated with asynchronous activation and reduced left ventricular (LV) pump function. We investigated to what extent alternate RV (septum) and LV (septum, apex) pacing sites improve LV electric activation, mechanics, hemodynamic performance, and efficiency over 4 months of pacing. METHODS AND RESULTS: After AV nodal ablation, mongrel dogs were randomized to receive 16 weeks of VDD pacing at the RV apex, RV septum, LV apex, or LV septum (transventricular septal approach). Electric activation maps (combined epicardial contact and endocardial noncontact) showed that RV apical and RV septal pacing induced significantly greater electric desynchronization than LV apical and LV septal pacing. RV apex and RV septal pacing also significantly increased mechanical dyssynchrony, discoordination (MRI tagging) and blood flow redistribution (microspheres) and reduced LV contractility, relaxation, and myocardial efficiency (stroke work/myocardial oxygen consumption). In contrast, LV apical and LV septal pacing did not significantly alter these parameters as compared with the values during intrinsic conduction. At 16 weeks, acute intrasubject comparison showed that single-site LV apical and LV septal pacing generally resulted in similar or better contractility, relaxation, and efficiency as compared with acute biventricular pacing. CONCLUSIONS: Acute and chronic LV apical and LV septal pacing maintain regional cardiac mechanics, contractility, relaxation, and efficiency near native levels, whereas RV apical or RV septal pacing diminish these variables. Acute LV apical and LV septal pacing tend to maintain or improve contractility and efficiency compared with biventricular pacing.


Asunto(s)
Estimulación Cardíaca Artificial/métodos , Sistema de Conducción Cardíaco/fisiología , Función Ventricular Izquierda/fisiología , Tabique Interventricular/fisiología , Análisis de Varianza , Animales , Volumen Cardíaco/fisiología , Perros , Contracción Miocárdica/fisiología , Consumo de Oxígeno/fisiología , Distribución Aleatoria , Volumen Sistólico/fisiología , Tabique Interventricular/cirugía
18.
Am J Physiol Heart Circ Physiol ; 295(3): H1270-H1278, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18660447

RESUMEN

Acute ventricular loading by volume inflation reversibly slows epicardial electrical conduction, but the underlying mechanism remains unclear. This study investigated the potential contributions of stretch-activated currents, alterations in resting membrane potential, or changes in intercellular resistance and membrane capacitance. Conduction velocity was assessed using optical mapping of isolated rabbit hearts at end-diastolic pressures of 0 and 30 mmHg. The addition of 50 microM Gd3+ (a stretch-activated channel blocker) to the perfusate had no effect on slowing. The effect of volume loading on conduction velocity was independent of changes in resting membrane potential created by altering the perfusate potassium concentration between 1.5 and 8 mM. Bidomain model analysis of optically recorded membrane potential responses to a unipolar stimulus suggested that the cross-fiber space constant and membrane capacitance both increased with loading (21%, P = 0.006, and 56%, P = 0.004, respectively), and these changes, when implemented in a resistively coupled one-dimensional network model, were consistent with the observed slowing (14%, P = 0.005). In conclusion, conduction slowing during ventricular volume loading is not attributable to stretch-activated currents or altered resting membrane potential, but a reduction of intercellular resistance with a concurrent increase of effective membrane capacitance results in a net slowing of conduction.


Asunto(s)
Volumen Cardíaco/fisiología , Sistema de Conducción Cardíaco/fisiología , Corazón/fisiología , Contracción Miocárdica/fisiología , Algoritmos , Animales , Espacio Extracelular/metabolismo , Gadolinio/farmacología , Ventrículos Cardíacos , Técnicas In Vitro , Potenciales de la Membrana/fisiología , Modelos Cardiovasculares , Potasio/metabolismo , Conejos , Capacitancia Vascular/fisiología
20.
J Cardiovasc Electrophysiol ; 14(7): 739-49, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12930255

RESUMEN

INTRODUCTION: Mechanical stimulation can induce electrophysiologic changes in cardiac myocytes, but how mechanoelectric feedback in the intact heart affects action potential propagation remains unclear. METHODS AND RESULTS: Changes in action potential propagation and repolarization with increased left ventricular end-diastolic pressure from 0 to 30 mmHg were investigated using optical mapping in isolated perfused rabbit hearts. With respect to 0 mmHg, epicardial strain at 30 mmHg in the anterior left ventricle averaged 0.040 +/- 0.004 in the muscle fiber direction and 0.032 +/- 0.006 in the cross-fiber direction. An increase in ventricular loading increased average epicardial activation time by 25%+/- 3% (P < 0.0001) and correspondingly decreased average apparent surface conduction velocity by 16%+/- 7% (P = 0.007). Ventricular loading did not significantly alter action potential duration at 20% repolarization (APD20) but did at 80% repolarization (APD80), from 179 +/- 7 msec to 207 +/- 5 msec (P < 0.0001). The dispersion of APD20 was decreased with loading from 19 +/- 2 msec to 13 +/- 2 msec (P = 0.024), whereas the dispersion of APD80 was not significantly changed. These electrophysiologic changes with ventricular loading were not affected by the nonspecific stretch-activated channel blocker streptomycin (200 microM) and were not attributable to changes in myocardial perfusion or the presence of an electromechanical decoupling agent (butanedione monoxime) during optical mapping. CONCLUSION: Acute loading of the left ventricle of the isolated rabbit heart decreased apparent epicardial conduction velocity and increased action potential duration by a load-dependent mechanism that may not involve stretch-activated channels.


Asunto(s)
Potenciales de Acción/fisiología , Mapeo del Potencial de Superficie Corporal/métodos , Sistema de Conducción Cardíaco/fisiología , Contracción Miocárdica/fisiología , Conducción Nerviosa/fisiología , Pericardio/fisiología , Función Ventricular Izquierda/fisiología , Potenciales de Acción/efectos de los fármacos , Animales , Elasticidad , Retroalimentación , Sistema de Conducción Cardíaco/efectos de los fármacos , Técnicas In Vitro , Mecanotransducción Celular/efectos de los fármacos , Mecanotransducción Celular/fisiología , Contracción Miocárdica/efectos de los fármacos , Conducción Nerviosa/efectos de los fármacos , Pericardio/efectos de los fármacos , Conejos , Estreptomicina/farmacología , Estrés Mecánico , Volumen Sistólico , Función Ventricular Izquierda/efectos de los fármacos , Presión Ventricular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA