Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 133
Filtrar
1.
JAMA Cardiol ; 6(4): 457-462, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33084842

RESUMEN

Importance: Patients with congenital heart disease (CHD), the most common birth defect, have increased risks for cancer. Identification of the variables that contribute to cancer risk is essential for recognizing patients with CHD who warrant longitudinal surveillance and early interventions. Objective: To compare the frequency of damaging variants in cancer risk genes among patients with CHD and control participants and identify associated clinical variables in patients with CHD who have cancer risk variants. Design, Setting, and Participants: This multicenter case-control study included participants with CHD who had previously been recruited to the Pediatric Cardiac Genomics Consortium based on presence of structural cardiac anomaly without genetic diagnosis at the time of enrollment. Permission to use published sequencing data from unaffected adult participants was obtained from 2 parent studies. Data were collected for this study from December 2010 to April 2019. Exposures: Presence of rare (allele frequency, <1 × 10-5) loss-of-function (LoF) variants in cancer risk genes. Main Outcomes and Measures: Frequency of LoF variants in cancer risk genes (defined in the Catalogue of Somatic Mutations in Cancer-Cancer Gene Consensus database), were statistically assessed by binomial tests in patients with CHD and control participants. Results: A total of 4443 individuals with CHD (mean [range] age, 13.0 [0-84] years; 2225 of 3771 with reported sex [59.0%] male) and 9808 control participants (mean [range] age, 52.1 [1-92] years; 4967 of 9808 [50.6%] male) were included. The frequency of LoF variants in regulatory cancer risk genes was significantly higher in patients with CHD than control participants (143 of 4443 [3.2%] vs 166 of 9808 [1.7%]; odds ratio [OR], 1.93 [95% CI, 1.54-2.42]; P = 1.38 × 10-12), and among CHD genes previously associated with cancer risk (58 of 4443 [1.3%] vs 18 of 9808 [0.18%]; OR, 7.2 [95% CI, 4.2-12.2]; P < 2.2 × 10-16). The LoF variants were also nominally increased in 14 constrained cancer risk genes with high expression in the developing heart. Seven of these genes (ARHGEF12, CTNNB1, LPP, MLLT4, PTEN, TCF12, and TFRC) harbored LoF variants in multiple patients with unexplained CHD. The highest rates for LoF variants in cancer risk genes occurred in patients with CHD and extracardiac anomalies (248 of 1482 individuals [16.7%]; control: 1099 of 9808 individuals [11.2%]; OR, 1.59 [95% CI, 1.37-1.85]; P = 1.3 × 10-10) and/or neurodevelopmental delay (209 of 1393 individuals [15.0%]; control: 1099 of 9808 individuals [11.2%]; OR, 1.40 [95% CI, 1.19-1.64]; P = 9.6 × 10-6). Conclusions and Relevance: Genotypes of CHD may account for increased cancer risks. In this cohort, damaging variants were prominent in the 216 genes that predominantly encode regulatory proteins. Consistent with their fundamental developmental functions, patients with CHD and damaging variants in these genes often had extracardiac manifestations. These data may also implicate cancer risk genes that are repeatedly varied in patients with unexplained CHD as CHD genes.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Cardiopatías Congénitas/complicaciones , Neoplasias/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Niño , Preescolar , Femenino , Regulación de la Expresión Génica/genética , Frecuencia de los Genes/genética , Genes Relacionados con las Neoplasias/genética , Variación Genética/genética , Cardiopatías Congénitas/genética , Humanos , Lactante , Recién Nacido , Mutación con Pérdida de Función/genética , Masculino , Persona de Mediana Edad , Neoplasias/etiología , Adulto Joven
2.
Nature ; 588(7838): 466-472, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32971526

RESUMEN

Cardiovascular disease is the leading cause of death worldwide. Advanced insights into disease mechanisms and therapeutic strategies require a deeper understanding of the molecular processes involved in the healthy heart. Knowledge of the full repertoire of cardiac cells and their gene expression profiles is a fundamental first step in this endeavour. Here, using state-of-the-art analyses of large-scale single-cell and single-nucleus transcriptomes, we characterize six anatomical adult heart regions. Our results highlight the cellular heterogeneity of cardiomyocytes, pericytes and fibroblasts, and reveal distinct atrial and ventricular subsets of cells with diverse developmental origins and specialized properties. We define the complexity of the cardiac vasculature and its changes along the arterio-venous axis. In the immune compartment, we identify cardiac-resident macrophages with inflammatory and protective transcriptional signatures. Furthermore, analyses of cell-to-cell interactions highlight different networks of macrophages, fibroblasts and cardiomyocytes between atria and ventricles that are distinct from those of skeletal muscle. Our human cardiac cell atlas improves our understanding of the human heart and provides a valuable reference for future studies.


Asunto(s)
Miocardio/citología , Análisis de la Célula Individual , Transcriptoma , Adipocitos/clasificación , Adipocitos/metabolismo , Adulto , Enzima Convertidora de Angiotensina 2/análisis , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Células Epiteliales/clasificación , Células Epiteliales/metabolismo , Epitelio , Femenino , Fibroblastos/clasificación , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Atrios Cardíacos/anatomía & histología , Atrios Cardíacos/citología , Atrios Cardíacos/inervación , Ventrículos Cardíacos/anatomía & histología , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/inervación , Homeostasis/inmunología , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Miocitos Cardíacos/clasificación , Miocitos Cardíacos/metabolismo , Neuronas/clasificación , Neuronas/metabolismo , Pericitos/clasificación , Pericitos/metabolismo , Receptores de Coronavirus/análisis , Receptores de Coronavirus/genética , Receptores de Coronavirus/metabolismo , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidad , Células del Estroma/clasificación , Células del Estroma/metabolismo
3.
Hum Genet ; 137(2): 183-193, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29417219

RESUMEN

Mosaicism due to somatic mutations can cause multiple diseases including cancer, developmental and overgrowth syndromes, neurodevelopmental disorders, autoinflammatory diseases, and atrial fibrillation. With the increased use of next generation sequencing technology, multiple tools have been developed to identify low-frequency variants, specifically from matched tumor-normal tissues in cancer studies. To investigate whether mosaic variants are implicated in congenital heart disease (CHD), we developed a pipeline using the cancer somatic variant caller MuTect to identify mosaic variants in whole-exome sequencing (WES) data from a cohort of parent/affected child trios (n = 715) and a cohort of healthy individuals (n = 416). This is a novel application of the somatic variant caller designed for cancer to WES trio data. We identified two cases with mosaic KMT2D mutations that are likely pathogenic for CHD, but conclude that, overall, mosaicism detectable in peripheral blood or saliva does not account for a significant portion of CHD etiology.


Asunto(s)
Secuenciación del Exoma , Variación Genética , Cardiopatías Congénitas/genética , Mosaicismo , Niño , Exoma/genética , Cardiopatías Congénitas/fisiopatología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Mutación , Programas Informáticos
4.
Curr Protoc Mol Biol ; 115: 23.16.1-23.16.9, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27366889

RESUMEN

RNA interference (RNAi) is a rapid approach to dissect loss-of-function phenotype for a gene of interest. However, it is challenging to perform RNAi in specific organs and tissues in vivo. Engineered viruses can provide a useful tool for delivery of small RNAs in vivo. Recombinant adeno-associated viruses (rAAVs) are the preferred method for delivering genes or gene modulators to target cells due to their high titer, low immune response, ability to transduce many types of cell, and overall safety. In this unit, we describe protocols for use of rAAVs as a cargo to deliver miRNA backbone-based shRNA controlled by a cardiac-specific promoter into the mouse heart. © 2016 by John Wiley & Sons, Inc.


Asunto(s)
Dependovirus/genética , Técnicas de Transferencia de Gen , Miocardio/metabolismo , ARN Interferente Pequeño/administración & dosificación , Animales , Ingeniería Genética , Células HEK293 , Humanos , Ratones , Interferencia de ARN , ARN Interferente Pequeño/genética , Transducción Genética
5.
Science ; 351(6273): 617-21, 2016 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-26912705

RESUMEN

Hypertrophic cardiomyopathy (HCM) is an inherited disease of heart muscle that can be caused by mutations in sarcomere proteins. Clinical diagnosis depends on an abnormal thickening of the heart, but the earliest signs of disease are hyperdynamic contraction and impaired relaxation. Whereas some in vitro studies of power generation by mutant and wild-type sarcomere proteins are consistent with mutant sarcomeres exhibiting enhanced contractile power, others are not. We identified a small molecule, MYK-461, that reduces contractility by decreasing the adenosine triphosphatase activity of the cardiac myosin heavy chain. Here we demonstrate that early, chronic administration of MYK-461 suppresses the development of ventricular hypertrophy, cardiomyocyte disarray, and myocardial fibrosis and attenuates hypertrophic and profibrotic gene expression in mice harboring heterozygous human mutations in the myosin heavy chain. These data indicate that hyperdynamic contraction is essential for HCM pathobiology and that inhibitors of sarcomere contraction may be a valuable therapeutic approach for HCM.


Asunto(s)
Adenosina Trifosfatasas/antagonistas & inhibidores , Bencilaminas/administración & dosificación , Miosinas Cardíacas/antagonistas & inhibidores , Cardiomiopatía Hipertrófica Familiar/tratamiento farmacológico , Contracción Miocárdica/efectos de los fármacos , Cadenas Pesadas de Miosina/antagonistas & inhibidores , Sarcómeros/efectos de los fármacos , Uracilo/análogos & derivados , Animales , Bencilaminas/química , Miosinas Cardíacas/genética , Cardiomiopatía Hipertrófica Familiar/patología , Cardiomiopatía Hipertrófica Familiar/fisiopatología , Células Cultivadas , Modelos Animales de Enfermedad , Fibrosis , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/patología , Heterocigoto , Humanos , Masculino , Ratones , Ratones Endogámicos , Mutación , Miocardio/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Cadenas Pesadas de Miosina/genética , Ratas , Uracilo/administración & dosificación , Uracilo/química
6.
J Clin Invest ; 125(11): 4122-34, 2015 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-26436652

RESUMEN

Cardiomyopathy is a common human disorder that is characterized by contractile dysfunction and cardiac remodeling. Genetic mutations and altered expression of genes encoding many signaling molecules and contractile proteins are associated with cardiomyopathy; however, how cardiomyocytes sense pathophysiological stresses in order to then modulate cardiac remodeling remains poorly understood. Here, we have described a regulator in the heart that harmonizes the progression of cardiac hypertrophy and dilation. We determined that expression of the myocyte-enriched protein cardiac ISL1-interacting protein (CIP, also known as MLIP) is reduced in patients with dilated cardiomyopathy. As CIP is highly conserved between human and mouse, we evaluated the effects of CIP deficiency on cardiac remodeling in mice. Deletion of the CIP-encoding gene accelerated progress from hypertrophy to heart failure in several cardiomyopathy models. Conversely, transgenic and AAV-mediated CIP overexpression prevented pathologic remodeling and preserved cardiac function. CIP deficiency combined with lamin A/C deletion resulted in severe dilated cardiomyopathy and cardiac dysfunction in the absence of stress. Transcriptome analyses of CIP-deficient hearts revealed that the p53- and FOXO1-mediated gene networks related to homeostasis are disturbed upon pressure overload stress. Moreover, FOXO1 overexpression suppressed stress-induced cardiomyocyte hypertrophy in CIP-deficient cardiomyocytes. Our studies identify CIP as a key regulator of cardiomyopathy that has potential as a therapeutic target to attenuate heart failure progression.


Asunto(s)
Cardiomegalia/fisiopatología , Proteínas Nucleares/deficiencia , Animales , Calcineurina/fisiología , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/fisiopatología , Proteínas Portadoras/genética , Proteínas Portadoras/fisiología , Proteínas Co-Represoras , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/fisiología , Redes Reguladoras de Genes , Terapia Genética , Vectores Genéticos/uso terapéutico , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/fisiopatología , Homeostasis , Humanos , Lamina Tipo A/biosíntesis , Lamina Tipo A/deficiencia , Lamina Tipo A/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Miocitos Cardíacos/patología , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Presión/efectos adversos , Ratas , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Estrés Mecánico , Transcriptoma , Remodelación Ventricular/fisiología
7.
Curr Protoc Hum Genet ; 86: 18.9.1-18.9.10, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26132004

RESUMEN

Viruses and bacteria are established as one of the main causes of human diseases from hepatitis to cancer. Recently, the presence of such pathogens has been extensively studied using human whole genome and transcriptome sequencing data. However, detecting and studying pathogens via next generation sequencing data is a challenging task in terms of time and computational resources. In this protocol we give instructions for a simple and quick method to find pathogenic DNA or RNA and detect possible integration of the pathogen genome into the host genome.


Asunto(s)
Secuencia de Bases , Biología Computacional/métodos , Genes Microbianos , Secuenciación de Nucleótidos de Alto Rendimiento , Análisis de Secuencia de ADN/métodos , Bases de Datos de Ácidos Nucleicos , Genoma Microbiano , Genómica/métodos , Humanos , Programas Informáticos
8.
Proc Natl Acad Sci U S A ; 112(29): 9046-51, 2015 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-26153423

RESUMEN

Homozygous cardiac myosin binding protein C-deficient (Mybpc(t/t)) mice develop dramatic cardiac dilation shortly after birth; heart size increases almost twofold. We have investigated the mechanism of cardiac enlargement in these hearts. Throughout embryogenesis myocytes undergo cell division while maintaining the capacity to pump blood by rapidly disassembling and reforming myofibrillar components of the sarcomere throughout cell cycle progression. Shortly after birth, myocyte cell division ceases. Cardiac MYBPC is a thick filament protein that regulates sarcomere organization and rigidity. We demonstrate that many Mybpc(t/t) myocytes undergo an additional round of cell division within 10 d postbirth compared with their wild-type counterparts, leading to increased numbers of mononuclear myocytes. Short-hairpin RNA knockdown of Mybpc3 mRNA in wild-type mice similarly extended the postnatal window of myocyte proliferation. However, adult Mybpc(t/t) myocytes are unable to fully regenerate the myocardium after injury. MYBPC has unexpected inhibitory functions during postnatal myocyte cytokinesis and cell cycle progression. We suggest that human patients with homozygous MYBPC3-null mutations develop dilated cardiomyopathy, coupled with myocyte hyperplasia (increased cell number), as observed in Mybpc(t/t) mice. Human patients, with heterozygous truncating MYBPC3 mutations, like mice with similar mutations, have hypertrophic cardiomyopathy. However, the mechanism leading to hypertrophic cardiomyopathy in heterozygous MYBPC3(+/-) individuals is myocyte hypertrophy (increased cell size), whereas the mechanism leading to cardiac dilation in homozygous Mybpc3(-/-) mice is primarily myocyte hyperplasia.


Asunto(s)
Proteínas Portadoras/metabolismo , Citocinesis , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Animales Recién Nacidos , Aurora Quinasas/metabolismo , Biomarcadores/metabolismo , Calcio/metabolismo , Recuento de Células , Diferenciación Celular , Proliferación Celular , Dependovirus/metabolismo , Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Ventrículos Cardíacos/metabolismo , Histonas/metabolismo , Humanos , Indoles/metabolismo , Ratones , Modelos Biológicos , Miocardio/citología , Miocitos Cardíacos/citología , Fosforilación , ARN Interferente Pequeño/metabolismo
9.
Proc Natl Acad Sci U S A ; 111(43): 15544-9, 2014 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-25313082

RESUMEN

Previous studies have established that a subset of head and neck tumors contains human papillomavirus (HPV) sequences and that HPV-driven head and neck cancers display distinct biological and clinical features. HPV is known to drive cancer by the actions of the E6 and E7 oncoproteins, but the molecular architecture of HPV infection and its interaction with the host genome in head and neck cancers have not been comprehensively described. We profiled a cohort of 279 head and neck cancers with next generation RNA and DNA sequencing and show that 35 (12.5%) tumors displayed evidence of high-risk HPV types 16, 33, or 35. Twenty-five cases had integration of the viral genome into one or more locations in the human genome with statistical enrichment for genic regions. Integrations had a marked impact on the human genome and were associated with alterations in DNA copy number, mRNA transcript abundance and splicing, and both inter- and intrachromosomal rearrangements. Many of these events involved genes with documented roles in cancer. Cancers with integrated vs. nonintegrated HPV displayed different patterns of DNA methylation and both human and viral gene expressions. Together, these data provide insight into the mechanisms by which HPV interacts with the human genome beyond expression of viral oncoproteins and suggest that specific integration events are an integral component of viral oncogenesis.


Asunto(s)
Genoma Humano/genética , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/virología , Interacciones Huésped-Patógeno/genética , Papillomaviridae/fisiología , Secuencia de Bases , Metilación de ADN/genética , Regulación Neoplásica de la Expresión Génica , Genes Relacionados con las Neoplasias , Humanos , Datos de Secuencia Molecular , Integración Viral/genética
10.
Circ Res ; 115(2): 227-37, 2014 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-24829265

RESUMEN

RATIONALE: Approximately 40% of hypertrophic cardiomyopathy (HCM) is caused by heterozygous missense mutations in ß-cardiac myosin heavy chain (ß-MHC). Associating disease phenotype with mutation is confounded by extensive background genetic and lifestyle/environmental differences between subjects even from the same family. OBJECTIVE: To characterize disease caused by ß-cardiac myosin heavy chain Val606Met substitution (VM) that has been identified in several HCM families with wide variation of clinical outcomes, in mice. METHODS AND RESULTS: Unlike 2 mouse lines bearing the malignant myosin mutations Arg453Cys (RC/+) or Arg719Trp (RW/+), VM/+ mice with an identical inbred genetic background lacked hallmarks of HCM such as left ventricular hypertrophy, disarray of myofibers, and interstitial fibrosis. Even homozygous VM/VM mice were indistinguishable from wild-type animals, whereas RC/RC- and RW/RW-mutant mice died within 9 days after birth. However, hypertrophic effects of the VM mutation were observed both in mice treated with cyclosporine, a known stimulator of the HCM response, and compound VM/RC heterozygous mice, which developed a severe HCM phenotype. In contrast to all heterozygous mutants, both systolic and diastolic function of VM/RC hearts was severely impaired already before the onset of cardiac remodeling. CONCLUSIONS: The VM mutation per se causes mild HCM-related phenotypes; however, in combination with other HCM activators it exacerbates the HCM phenotype. Double-mutant mice are suitable for assessing the severity of benign mutations.


Asunto(s)
Sustitución de Aminoácidos , Cardiomiopatía Hipertrófica Familiar/genética , Mutación Missense , Cadenas Pesadas de Miosina/genética , Mutación Puntual , Animales , Miosinas Cardíacas , Cardiomiopatía Hipertrófica Familiar/diagnóstico por imagen , Cardiomiopatía Hipertrófica Familiar/patología , Ciclosporina/toxicidad , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Genotipo , Humanos , Hipertrofia Ventricular Izquierda/diagnóstico por imagen , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/patología , Ratones , Modelos Moleculares , Contracción Miocárdica , Cadenas Pesadas de Miosina/fisiología , Fenotipo , Conformación Proteica , Transcripción Genética , Ultrasonografía , Miosinas Ventriculares/genética , Miosinas Ventriculares/fisiología , Remodelación Ventricular/genética , Remodelación Ventricular/fisiología
11.
Science ; 342(6154): 111-4, 2013 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-24092743

RESUMEN

Dominant mutations in sarcomere proteins such as the myosin heavy chains (MHC) are the leading genetic causes of human hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy. We found that expression of the HCM-causing cardiac MHC gene (Myh6) R403Q mutation in mice can be selectively silenced by an RNA interference (RNAi) cassette delivered by an adeno-associated virus vector. RNAi-transduced MHC(403/+) mice developed neither hypertrophy nor myocardial fibrosis, the pathologic manifestations of HCM, for at least 6 months. Because inhibition of HCM was achieved by only a 25% reduction in the levels of the mutant transcripts, we suggest that the variable clinical phenotype in HCM patients reflects allele-specific expression and that partial silencing of mutant transcripts may have therapeutic benefit.


Asunto(s)
Cardiomiopatía Hipertrófica/diagnóstico , Terapia Genética , Cadenas Pesadas de Miosina/genética , Interferencia de ARN , Alelos , Animales , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/patología , Dependovirus , Fibrosis , Silenciador del Gen , Células HEK293 , Humanos , Ratones , Mutación
12.
J Mol Cell Cardiol ; 57: 23-31, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23313350

RESUMEN

Dilated cardiomyopathy (DCM) is a disease characterized by dilation of the ventricular chambers and reduced contractile function. We examined the contractile performance of chemically-skinned ventricular strips from two heterozygous murine models of DCM-causing missense mutations of myosin, F764L/+ and S532P/+, in an α-myosin heavy chain (MyHC) background. In Ca(2+)-activated skinned myocardial strips, the maximum developed tension in F764L/+ was only ~50% that of litter-mate controls (+/+). The F764L/+ also exhibited significantly reduced rigor stiffness, loaded shortening velocity and power output. Corresponding indices for S532P/+ strips were not different from controls. Manipulation of MgATP concentration in conjunction with measures of viscoelasticity, which provides estimates of myosin detachment rate 2πc, allowed us to probe the molecular basis of changes in crossbridge kinetics that occur with the myosin mutations. By examining the response of detachment rate to varying MgATP we found the rate of MgADP release was unaffected by the myosin mutations. However, MgATP binding rate was higher in the DCM groups compared to controls (422±109mM(-1)·s(-1) in F764L/+, 483±74mM(-1)·s(-1) in S532P/+ and 303±18mM(-1)·s(-1) in +/+). In addition, the rate constant of force development, 2πb, was significantly higher in DCM groups compared to controls (at 5mM MgATP: 36.9±4.9s(-1) in F764L/+, 32.9±4.5s(-1) in S532P/+ and 18.2±1.7s(-1) in +/+). These results suggest that elevated rates of force development and MgATP binding are features of cardiac myofilament function that underlie the development of DCM.


Asunto(s)
Adenosina Trifosfato/fisiología , Cardiomiopatía Dilatada/genética , Mutación Missense , Contracción Miocárdica , Miosinas Ventriculares/genética , Animales , Calcio/fisiología , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/fisiopatología , Ventrículos Cardíacos/fisiopatología , Humanos , Técnicas In Vitro , Cinética , Ratones , Ratones Transgénicos , Miosinas Ventriculares/metabolismo
13.
Proc Natl Acad Sci U S A ; 109(35): 14087-91, 2012 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-22891333

RESUMEN

Somatic mtDNA mutations have been reported in some human tumors, but their spectrum in different malignancies and their role in cancer development remain incompletely understood. Here, we describe the breadth of somatic and inherited mutations across the mitochondrial genome by sequence analyses of paired tumor and normal tissue samples from 226 individuals with five types of cancer using whole-genome data generated by The Cancer Genome Atlas Research Network. The frequencies of deleterious tumor-specific somatic mutations found in mtDNA varied across tumor types, ranging from 13% of glioblastomas to 63% of rectal adenocarcinomas. Compared with inherited mtDNA variants, somatic mtDNA mutations were enriched for nonsynonymous vs. synonymous changes (93 vs. 15; P < 2.2E-16) and were predicted to functionally impact the encoded protein. Somatic missense mutations in tumors were distributed uniformly among the mitochondrial protein genes, but 65% of somatic truncating mutations occurred in NADH dehydrogenase 5. Analysis of staging data in colon and rectal cancers revealed that the frequency of damaging mitochondrial mutations is the same in stages I and IV tumors. In summary, these data suggest that damaging somatic mtDNA mutations occur frequently (13-63%) in these five tumor types and likely confer a selective advantage in oncogenesis.


Asunto(s)
Adenocarcinoma/genética , Neoplasias del Colon/genética , Cistadenocarcinoma Seroso/genética , Genes Mitocondriales/genética , Leucemia Mieloide Aguda/genética , Neoplasias Ováricas/genética , Neoplasias Colorrectales/genética , ADN Mitocondrial/genética , Femenino , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica/genética , Genómica , Humanos , Mutación Missense/genética
14.
PLoS One ; 6(9): e24593, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21980348

RESUMEN

BACKGROUND: Postoperative ventricular dysfunction (VnD) occurs in 9-20% of coronary artery bypass graft (CABG) surgical patients and is associated with increased postoperative morbidity and mortality. Understanding genetic causes of postoperative VnD should enhance patient risk stratification and improve treatment and prevention strategies. We aimed to determine if genetic variants associate with occurrence of in-hospital VnD after CABG surgery. METHODS: A genome-wide association study identified single nucleotide polymorphisms (SNPs) associated with postoperative VnD in male subjects of European ancestry undergoing isolated primary CABG surgery with cardiopulmonary bypass. VnD was defined as the need for ≥2 inotropes or mechanical ventricular support after CABG surgery. Validated SNPs were assessed further in two replication CABG cohorts and meta-analysis was performed. RESULTS: Over 100 SNPs were associated with VnD (P<10(-4)), with one SNP (rs17691914) encoded at 3p22.3 reaching genome-wide significance (P(additive model) = 2.14×10(-8)). Meta-analysis of validation and replication study data for 17 SNPs identified three SNPs associated with increased risk for developing postoperative VnD after adjusting for clinical risk factors. These SNPs are located at 3p22.3 (rs17691914, OR(additive model) = 2.01, P = 0.0002), 3p14.2 (rs17061085, OR(additive model) = 1.70, P = 0.0001) and 11q23.2 (rs12279572, OR(recessive model) = 2.19, P = 0.001). CONCLUSIONS: No SNPs were consistently associated with strong risk (OR(additive model)>2.1) of developing in-hospital VnD after CABG surgery. However, three genetic loci identified by meta-analysis were more modestly associated with development of postoperative VnD. Studies of larger cohorts to assess these loci as well as to define other genetic mechanisms and related biology that link genetic variants to postoperative ventricular dysfunction are warranted.


Asunto(s)
Puente de Arteria Coronaria/métodos , Estudio de Asociación del Genoma Completo , Disfunción Ventricular/genética , Alelos , Estudios de Cohortes , Femenino , Predisposición Genética a la Enfermedad , Variación Genética , Genoma Humano , Genómica , Genotipo , Humanos , Masculino , Polimorfismo de Nucleótido Simple , Reproducibilidad de los Resultados , Disfunción Ventricular/cirugía , Población Blanca
15.
N Engl J Med ; 363(6): 552-63, 2010 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-20818890

RESUMEN

BACKGROUND: Myocardial fibrosis is a hallmark of hypertrophic cardiomyopathy and a proposed substrate for arrhythmias and heart failure. In animal models, profibrotic genetic pathways are activated early, before hypertrophic remodeling. Data showing early profibrotic responses to sarcomere-gene mutations in patients with hypertrophic cardiomyopathy are lacking. METHODS: We used echocardiography, cardiac magnetic resonance imaging (MRI), and serum biomarkers of collagen metabolism, hemodynamic stress, and myocardial injury to evaluate subjects with hypertrophic cardiomyopathy and a confirmed genotype. RESULTS: The study involved 38 subjects with pathogenic sarcomere mutations and overt hypertrophic cardiomyopathy, 39 subjects with mutations but no left ventricular hypertrophy, and 30 controls who did not have mutations. Levels of serum C-terminal propeptide of type I procollagen (PICP) were significantly higher in mutation carriers without left ventricular hypertrophy and in subjects with overt hypertrophic cardiomyopathy than in controls (31% and 69% higher, respectively; P<0.001). The ratio of PICP to C-terminal telopeptide of type I collagen was increased only in subjects with overt hypertrophic cardiomyopathy, suggesting that collagen synthesis exceeds degradation. Cardiac MRI studies showed late gadolinium enhancement, indicating myocardial fibrosis, in 71% of subjects with overt hypertrophic cardiomyopathy but in none of the mutation carriers without left ventricular hypertrophy. CONCLUSIONS: Elevated levels of serum PICP indicated increased myocardial collagen synthesis in sarcomere-mutation carriers without overt disease. This profibrotic state preceded the development of left ventricular hypertrophy or fibrosis visible on MRI. (Funded by the National Institutes of Health and others.)


Asunto(s)
Cardiomiopatía Hipertrófica/complicaciones , Miocardio/patología , Fragmentos de Péptidos/sangre , Procolágeno/sangre , Adolescente , Adulto , Biomarcadores/sangre , Miosinas Cardíacas , Cardiomiopatía Hipertrófica/sangre , Cardiomiopatía Hipertrófica/genética , Proteínas Portadoras/genética , Niño , Preescolar , Colágeno/biosíntesis , Colágeno Tipo I , Ecocardiografía , Femenino , Fibrosis/diagnóstico , Fibrosis/etiología , Humanos , Hipertrofia Ventricular Izquierda/etiología , Modelos Logísticos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Mutación , Miocardio/metabolismo , Cadenas Pesadas de Miosina/genética , Péptidos , Adulto Joven
16.
J Am Coll Cardiol ; 55(4): 320-9, 2010 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-20117437

RESUMEN

OBJECTIVES: We sought to further define the role of sarcomere mutations in dilated cardiomyopathy (DCM) and associated clinical phenotypes. BACKGROUND: Mutations in several contractile proteins contribute to DCM, but definitive evidence for the roles of most sarcomere genes remains limited by the lack of robust genetic support. METHODS: Direct sequencing of 6 sarcomere genes was performed on 334 probands with DCM. A novel D230N missense mutation in the gene encoding alpha-tropomyosin (TPM1) was identified. Functional assessment was performed by the use of an in vitro reconstituted sarcomere complex to evaluate ATPase regulation and Ca(2+) affinity as correlates of contractility. RESULTS: TPM1 D230N segregated with DCM in 2 large unrelated families. This mutation altered an evolutionarily conserved residue and was absent in >1,000 control chromosomes. In vitro studies demonstrated major inhibitory effects on sarcomere function with reduced Ca(2+) sensitivity, maximum activation, and Ca(2+) affinity compared with wild-type TPM1. Clinical manifestations ranged from decompensated heart failure or sudden death in those presenting early in life to asymptomatic left ventricular dysfunction in those diagnosed during adulthood. Notably, several affected infants had remarkable improvement. CONCLUSIONS: Genetic segregation in 2 unrelated families and functional analyses conclusively establish a pathogenic role for TPM1 mutations in DCM. In vitro results demonstrate contrasting effects of DCM and hypertrophic cardiomyopathy mutations in TPM1, suggesting that specific functional consequences shape cardiac remodeling. Along with previous reports, our data support a distinctive, age-dependent phenotype with sarcomere-associated DCM where presentation early in life is associated with severe, sometimes lethal, disease. These observations have implications for the management of familial DCM.


Asunto(s)
Cardiomiopatía Dilatada/genética , Sarcómeros/genética , Adolescente , Adulto , Factores de Edad , Anciano , Calcio/metabolismo , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Persona de Mediana Edad , Mutación , Miosinas/análisis , Fenotipo , Tropomiosina/genética , Troponina/metabolismo , Remodelación Ventricular/fisiología
17.
Circ Cardiovasc Genet ; 2(5): 499-506, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20031626

RESUMEN

BACKGROUND: Atrial fibrillation (AF) is the most common adverse event following coronary artery bypass graft surgery. A recent study identified chromosome 4q25 variants associated with AF in ambulatory populations. However, their role in postoperative AF is unknown. We hypothesized that genetic variants in the 4q25 chromosomal region are independently associated with postoperative AF after coronary artery bypass graft surgery. METHODS AND RESULTS: Two prospectively collected cohorts of patients undergoing coronary artery bypass graft surgery, with or without concurrent valve surgery, at 3 US centers. From a discovery cohort of 959 patients, clinical and genomic multivariate predictors of postoperative AF were identified by genotyping 45 single-nucleotide polymorphisms (SNPs) encompassing the 4q25 locus. Three SNPs were then assessed in a separately collected validation cohort of 494 patients. After adjustment for clinical predictors of postoperative AF and multiple comparisons, rs2200733, rs13143308, and 5 other linked SNPs independently predicted postoperative AF in the discovery cohort. Additive odds ratios for the 7 associated 4q25 SNPs ranged between 1.57 and 2.17 (P=8.0x10(-4) to 3.4x10(-6)). Association with postoperative AF were measured and replicated for rs2200733 and rs13143308 in the validation cohort. CONCLUSIONS: In 2 independently collected cardiac surgery cohorts, noncoding SNPs within the chromosome 4q25 region are independently associated with postoperative AF after coronary artery bypass graft surgery after adjusting for clinical covariates and multiple comparisons.


Asunto(s)
Fibrilación Atrial/genética , Cromosomas Humanos Par 4/genética , Puente de Arteria Coronaria/efectos adversos , Variación Genética , Complicaciones Posoperatorias/etiología , Adulto , Anciano , Anciano de 80 o más Años , Fibrilación Atrial/etiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Valor Predictivo de las Pruebas , Estudios Prospectivos , Adulto Joven
18.
Circ Heart Fail ; 2(2): 121-8, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19808327

RESUMEN

BACKGROUND: Pheochromocytomas are rare tumors derived from the chromaffin cells of the adrenal medulla. Although these tumors have long been postulated to induce hypertension and cardiomyopathy through the hypersecretion of catecholamines, catecholamines alone may not fully explain the profound myocardial remodeling induced by these tumors. We sought to determine whether changes in myocardial function in pheochromocytoma-induced cardiomyopathy result solely from catecholamines secretion or from multiple pheochromocytoma-derived factors. METHODS AND RESULTS: Isolated cardiomyocytes incubated with pheochromocytoma-conditioned growth media contracted at a higher frequency than cardiomyocytes incubated with norepinephrine (NE) only. Sprague-Dawley rats and black-6 mice were implanted with agarose-encapsulated pheochromocytoma (PC12) cells, dihydroxyphenylalanine decarboxylase knock-out PC12 cells deficient in NE (PC12-KO), or NE-secreting pumps. PC12 cell implantation increased left ventricular dilation by 35+/-6% and 9.6+/-1.4% and reduced left ventricular fractional shortening by 20+/-3% and 28+/-4% in rats and mice compared with animals dosed only with NE, respectively. Elimination of NE secretion in PC12-KO cells induced neither cardiac dilation (3.9%+/-1.8% increase versus control) nor changes in (1.9%+/-0.4% reduction) fractional shortening compared to controls. CONCLUSIONS: Pheochromocytomas induce a greater degree of cardiomyopathy than equivalent doses of NE, suggesting pheochromocytoma-induced cardiomyopathy is not solely mediated by NE, rather pheochromocytoma secretory factors in combination with catecholamines act synergistically to induce greater cardiac damage than catecholamines alone.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/complicaciones , Cardiomiopatías/etiología , Feocromocitoma/complicaciones , Neoplasias de las Glándulas Suprarrenales/metabolismo , Animales , Cardiomiopatías/inducido químicamente , Cardiomiopatías/diagnóstico , Cardiomiopatías/fisiopatología , Catecolaminas/metabolismo , Modelos Animales de Enfermedad , Ecocardiografía , Femenino , Expresión Génica , Corazón/fisiopatología , Ratones , Ratones Endogámicos , Contracción Miocárdica/efectos de los fármacos , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/efectos de los fármacos , Trasplante de Neoplasias , Norepinefrina/deficiencia , Norepinefrina/metabolismo , Norepinefrina/farmacología , Células PC12/metabolismo , Feocromocitoma/metabolismo , Ratas , Ratas Sprague-Dawley , Células Tumorales Cultivadas
19.
Development ; 135(20): 3425-34, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18799547

RESUMEN

To investigate the mechanisms by which mutations in the human transcriptional co-activator EYA4 gene cause sensorineural hearing loss that can occur in association with dilated cardiomyopathy, we studied eya4 expression during zebrafish development and characterized eya4 deficiency. eya4 morphant fish embryos had reduced numbers of hair cells in the otic vesicle and lateral line neuromasts with impaired sensory responses. Analyses of candidate genes that are known to be expressed in a temporal and spatial pattern comparable to eya4 focused our analyses on atp1b2b, which encodes the beta2b subunit of the zebrafish Na+/K+-ATPase. We demonstrate atp1b2b levels are reduced in eya4 morphant fish and that morpholino oligonucleotides targeting the atp1b2b gene recapitulated the eya4 deficiency phenotypes, including heart failure, decreased sensory hair cell numbers in the otic vesicle and neuromasts, and abnormal sensory responses. Furthermore, atp1b2b overexpression rescued these phenotypes in eya4 morphant fish. We conclude that eya4 regulation of Na+/K+-ATPase is crucial for the development of mechanosensory cells and the maintenance of cardiac function in zebrafish.


Asunto(s)
Oído Interno/embriología , Regulación del Desarrollo de la Expresión Génica , ATPasa Intercambiadora de Sodio-Potasio/fisiología , Transactivadores/genética , Proteínas de Pez Cebra/genética , Pez Cebra/embriología , Animales , Oído Interno/metabolismo , Embrión no Mamífero , Proteínas del Ojo/genética , Células Ciliadas Auditivas/metabolismo , Sistema de la Línea Lateral/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/genética , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Transactivadores/metabolismo , Proteínas de Pez Cebra/metabolismo
20.
Circulation ; 116(21): 2399-408, 2007 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-17984378

RESUMEN

BACKGROUND: Cardiac myosin binding protein-C (cMyBP-C) is a thick-filament protein whose presence and phosphorylation by protein kinase A (PKA) regulates cross-bridge formation and kinetics in isolated myocardium. We tested the influence of cMyBP-C and its PKA-phosphorylation on contraction/relaxation kinetics in intact hearts and revealed its essential role in several classic properties of cardiac function. METHODS AND RESULTS: Comprehensive in situ cardiac pressure-volume analysis was performed in mice harboring a truncation mutation of cMyBP-C (cMyBP-C(t/t)) that resulted in nondetectable protein versus hearts re-expressing solely wild-type (cMyBP-C(WT:(t/t))) or mutated protein in which known PKA-phosphorylation sites were constitutively suppressed (cMyBP-C(AllP-:(t/t))). Hearts lacking cMyBP-C had faster early systolic activation, which then terminated prematurely, limiting ejection. Systole remained short at faster heart rates; thus, cMyBP-C(t/t) hearts displayed minimal rate-dependent decline in diastolic time and cardiac preload. Furthermore, prolongation of pressure relaxation by afterload was markedly blunted in cMyBP-C(t/t) hearts. All 3 properties were similarly restored to normal in cMyBP-C(WT:(t/t)) and cMyBP-C(AllP-:(t/t)) hearts, which supports independence of PKA-phosphorylation. However, the dependence of peak rate of pressure rise on preload was specifically depressed in cMyBP-C(AllP-:(t/t)) hearts, whereas cMyBP-C(t/t) and cMyBP-C(AllP-:(t/t)) hearts had similar blunted adrenergic and rate-dependent contractile reserve, which supports linkage of these behaviors to PKA-cMyBP-C modification. CONCLUSIONS: cMyBP-C is essential for major properties of cardiac function, including sustaining systole during ejection, the heart-rate dependence of the diastolic time period, and relaxation delay from increased arterial afterload. These are independent of its phosphorylation by PKA, which more specifically modulates early pressure rise rate and adrenergic/heart rate reserve.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Contracción Miocárdica/fisiología , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/farmacocinética , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/farmacocinética , Diástole/genética , Diástole/fisiología , Femenino , Frecuencia Cardíaca/genética , Frecuencia Cardíaca/fisiología , Masculino , Ratones , Mutación , Contracción Miocárdica/genética , Fosforilación , Sístole/genética , Sístole/fisiología , Función Ventricular Izquierda/genética , Función Ventricular Izquierda/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA