Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Circ Res ; 101(4): e32-42, 2007 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-17673668

RESUMEN

Apelin constitutes a novel endogenous peptide system suggested to be involved in a broad range of physiological functions, including cardiovascular function, heart development, control of fluid homeostasis, and obesity. Apelin is also a catalytic substrate for angiotensin-converting enzyme 2, the key severe acute respiratory syndrome receptor. The in vivo physiological role of Apelin is still elusive. Here we report the generation of Apelin gene-targeted mice. Apelin mutant mice are viable and fertile, appear healthy, and exhibit normal body weight, water and food intake, heart rates, and heart morphology. Intriguingly, aged Apelin knockout mice developed progressive impairment of cardiac contractility associated with systolic dysfunction in the absence of histological abnormalities. We also report that pressure overload induces upregulation of Apelin expression in the heart. Importantly, in pressure overload-induced heart failure, loss of Apelin did not significantly affect the hypertrophy response, but Apelin mutant mice developed progressive heart failure. Global gene expression arrays and hierarchical clustering of differentially expressed genes in hearts of banded Apelin(-/y) and Apelin(+/y) mice showed concerted upregulation of genes involved in extracellular matrix remodeling and muscle contraction. These genetic data show that the endogenous peptide Apelin is crucial to maintain cardiac contractility in pressure overload and aging.


Asunto(s)
Envejecimiento/fisiología , Presión Sanguínea/fisiología , Proteínas Portadoras/genética , Insuficiencia Cardíaca/fisiopatología , Contracción Miocárdica/fisiología , Adipoquinas , Envejecimiento/genética , Animales , Aorta , Apelina , Presión Sanguínea/genética , Proteínas Portadoras/metabolismo , Modelos Animales de Enfermedad , Conducta de Ingestión de Líquido , Ecocardiografía , Conducta Alimentaria , Femenino , Corazón/embriología , Corazón/fisiología , Insuficiencia Cardíaca/diagnóstico por imagen , Homeostasis/fisiología , Péptidos y Proteínas de Señalización Intercelular , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Contracción Miocárdica/genética , Obesidad/fisiopatología , ARN Mensajero/metabolismo , Índice de Severidad de la Enfermedad
2.
J Thorac Cardiovasc Surg ; 134(1): 74-81, 81.e1-2, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17599489

RESUMEN

OBJECTIVE: The effects of cold cardioplegic arrest and reperfusion on human ventricular gene expression are unknown. We tested the hypothesis that intraoperative ischemia-reperfusion under conditions of blood cardioplegic arrest would induce a unique myocardial genomic profile indicative of a cardioprotective response. METHODS: Right ventricular samples were serially acquired during surgical repair of ventricular septal defect. RESULTS: Expression profiling revealed 3 patterns of gene expression: (1) increased expression above control levels within 1 hour of cardioplegic arrest, with further amplification during early reperfusion; (2) increased expression limited to the reperfusion phase; and (3) reduced expression during reperfusion. Functional annotation and network mapping of differentially expressed genes indicated activation of multiple signaling pathways regulated by phosphatidylinositide 3'-OH kinase convergent on cellular growth and reparative programs. Also observed was increased expression of genes regulating hemoglobin synthesis, suggesting a novel cardioprotective pathway evoked during ischemia-reperfusion. CONCLUSION: Reversible myocardial ischemia-reperfusion during cardiac surgery is associated with an immediate genomic response that predicts a net cardioprotective phenotype.


Asunto(s)
Perfilación de la Expresión Génica , Defectos del Tabique Interventricular/cirugía , Isquemia Miocárdica/genética , Isquemia Miocárdica/cirugía , Reperfusión Miocárdica , Miocardio/metabolismo , Adaptación Fisiológica/genética , Análisis de Varianza , Biopsia , Puente Cardiopulmonar , Preescolar , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Expresión Génica , Globinas/genética , Defectos del Tabique Interventricular/complicaciones , Defectos del Tabique Interventricular/patología , Ventrículos Cardíacos/patología , Humanos , Lactante , Periodo Intraoperatorio , Precondicionamiento Isquémico , Análisis por Micromatrices/normas , Isquemia Miocárdica/etiología , ARN de Transferencia/aislamiento & purificación , Factor A de Crecimiento Endotelial Vascular/metabolismo
3.
Proc Natl Acad Sci U S A ; 103(7): 2304-9, 2006 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-16467148

RESUMEN

Inappropriate cardiac remodeling and repair after myocardial infarction (MI) predisposes to heart failure. Studies have reported on the potential for lineage negative, steel factor positive (c-kit+) bone marrow-derived hematopoetic stem/progenitor cells (HSPCs) to repair damaged myocardium through neovascularization and myogenesis. However, the precise contribution of the c-kit signaling pathway to the cardiac repair process has yet to be determined. In this study, we sought to directly elucidate the mechanistic contributions of c-kit+ bone marrow-derived hematopoetic stem/progenitor cells in the maintenance and repair of damaged myocardium after MI. Using c-kit-deficient mice, we demonstrate the importance of c-kit signaling in preventing ventricular dilation and hypertrophy, and the maintenance of cardiac function after MI in c-kit-deficient mice. Furthermore, we show phenotypic rescue of cardiac repair after MI of c-kit-deficient mice by bone marrow transplantation of wild-type HSPCs. The transplanted group also had reduced apoptosis and collagen deposition, along with an increase in neovascularization. To better understand the mechanisms underlying this phenotypic rescue, we investigated the gene expression pattern within the infarcted region by using microarray analysis. This analysis suggested activation of inflammatory pathways, specifically natural killer (NK) cell-mediated mobilization after MI in rescued hearts. This finding was confirmed by immunohistology and by using an NK blocker. Thus, our investigation revealed a previously uncharacterized role for c-kit signaling after infarction by mediating bone marrow-derived NK and angiogenic cell mobilization, which contributes to improved remodeling and cardiac function after MI.


Asunto(s)
Células Madre Hematopoyéticas/fisiología , Células Asesinas Naturales/fisiología , Infarto del Miocardio/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Remodelación Ventricular/genética , Animales , Células de la Médula Ósea/fisiología , Trasplante de Médula Ósea , Femenino , Perfilación de la Expresión Génica , Masculino , Ratones , Ratones Mutantes , Mutación , Neovascularización Fisiológica/genética , Proteínas Proto-Oncogénicas c-kit/genética
4.
J Thorac Cardiovasc Surg ; 130(5): 1326-32, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16256785

RESUMEN

BACKGROUND: We have recently demonstrated that remote ischemic preconditioning reduces ischemia-reperfusion injury in animal models. The mechanisms by which the remote ischemic preconditioning stimulus exerts its effect remain to be fully defined, and its effect on myocardial gene expression is unknown. We tested the hypothesis that remote ischemic preconditioning modifies myocardial gene expression immediately after the remote ischemic preconditioning stimulus (early phase) and 24 hours later (late phase). METHODS: Twenty male (C57BL/6) 10- to 12-week-old mice were randomized into 4 groups: group 1 (control, early phase; n = 5), group 2 (remote ischemic preconditioning, early phase; n = 5), group 3 (control, late phase; n = 5), and group 4 (remote ischemic preconditioning, late phase; n = 5). Groups 2 and 4 underwent remote ischemic preconditioning induced by 6 cycles of 4 minutes of occlusion and 4 minutes of reperfusion of the femoral artery. Groups 1 and 2 were killed 15 minutes after completion of sham procedure or remote ischemic preconditioning, and the hearts were removed and frozen in liquid nitrogen. Groups 3 and 4 were killed 24 hours after remote ischemic preconditioning, and the hearts were harvested in the same fashion. Gene expression was assessed by using the Affymetrix MG-430A chip (Affymetrix, Santa Clara, Calif). RESULTS: Data filtering (P < .05, analysis of variance) and hierarchic 2-way clustering identified significant differences in gene expression among the 4 groups. Genes involved in protection against oxidative stress (eg, Hadhsc, Prdx4, and Fabp4) and cytoprotection (Hsp73) were upregulated, whereas many proinflammatory genes (eg, Egr-1 and Dusp 1 and 6) were suppressed. CONCLUSION: A simple remote ischemic preconditioning stimulus modifies myocardial gene expression by upregulating cardioprotective genes and suppressing genes potentially involved in the pathogenesis of ischemia-reperfusion injury.


Asunto(s)
Regulación de la Expresión Génica , Precondicionamiento Isquémico Miocárdico/métodos , Miocardio , Animales , Masculino , Ratones , Ratones Endogámicos C57BL
5.
Physiol Genomics ; 19(1): 143-50, 2004 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-15304621

RESUMEN

Remote ischemic preconditioning (IPC) reduces tissue injury caused by ischemia-reperfusion (IR) in distant organs. We tested the hypothesis that remote IPC (rIPC) modifies inflammatory gene transcription in humans. Using a microarray method, we demonstrated that a simple model of brief forearm ischemia suppresses proinflammatory gene expression in circulating leukocytes. Genes encoding key proteins involved in cytokine synthesis, leukocyte chemotaxis, adhesion and migration, exocytosis, innate immunity signaling pathways, and apoptosis were all suppressed within 15 min (early phase IPC) and more so after 24 h (second window IPC). Changes in leukocyte CD11b expression measured by flow cytometry mirrored this pattern, with there being a significant (P = 0.01) reduction at 24 h. The results of this study show that the rIPC stimulus modifies leukocyte inflammatory gene expression. This effect may contribute to the protective effect of IPC against IR injury and may have broader implications in other inflammatory processes. This is the first study of human gene expression following rIPC stimulus. rIPC stimulus suppressed proinflammatory gene transcription in human leukocytes.


Asunto(s)
Regulación de la Expresión Génica/genética , Inflamación/genética , Precondicionamiento Isquémico , Adulto , Antígeno CD11b/genética , Femenino , Citometría de Flujo , Humanos , Inflamación/inmunología , Inflamación/patología , Leucocitos/inmunología , Leucocitos/metabolismo , Masculino , Activación Neutrófila , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/análisis , ARN Mensajero/genética , Daño por Reperfusión/genética , Daño por Reperfusión/patología , Daño por Reperfusión/prevención & control , Reproducibilidad de los Resultados
6.
Blood ; 100(6): 2175-86, 2002 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-12200383

RESUMEN

We have created a molecular resource of genes expressed in primary malignant plasma cells using a combination of cDNA library construction, 5' end single-pass sequencing, bioinformatics, and microarray analysis. In total, we identified 9732 nonredundant expressed genes. This dataset is available as the Myeloma Gene Index (www.uhnres.utoronto.ca/akstewart_lab).Predictably, the sequenced profile of myeloma cDNAs mirrored the known function of immunoglobulin-producing, high-respiratory rate, low-cycling, terminally differentiated plasma cells. Nevertheless, approximately 10% of myeloma-expressed sequences matched only entries in the database of Expressed Sequence Tags (dbEST) or the high-throughput genomic sequence (htgs) database. Numerous novel genes of potential biologic significance were identified. We therefore spotted 4300 sequenced cDNAs on glass slides creating a myeloma-enriched microarray. Several of the most highly expressed genes identified by sequencing, such as a novel putative disulfide isomerase (MGC3178), tumor rejection antigen TRA1, heat shock 70-kDa protein 5, and annexin A2, were also differentially expressed between myeloma and B lymphoma cell lines using this myeloma-enriched microarray. Furthermore, a defined subset of 34 up-regulated and 18 down-regulated genes on the array were able to differentiate myeloma from nonmyeloma cell lines. These not only include genes involved in B-cell biology such as syndecan, BCMA, PIM2, MUM1/IRF4, and XBP1, but also novel uncharacterized genes matching sequences only in the public databases. In summary, our expressed gene catalog and myeloma-enriched microarray contains numerous genes of unknown function and may complement other commercially available arrays in defining the molecular portrait of this hematopoietic malignancy. GenBank Accession numbers include BF169967-BF176369, BF185966-BF185969, and BF177280-BF177455.


Asunto(s)
ADN de Neoplasias/análisis , Biblioteca de Genes , Proteínas de la Membrana , Mieloma Múltiple/genética , Proteínas de Transferencia de Fosfolípidos , Secuencia de Aminoácidos , Anexina A2/genética , Anexina A2/metabolismo , Bases de Datos de Ácidos Nucleicos , Progresión de la Enfermedad , Perfilación de la Expresión Génica , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Glicoproteínas de Membrana , Datos de Secuencia Molecular , Mieloma Múltiple/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Células Plasmáticas , Proteína Disulfuro Isomerasas/genética , Proteína Disulfuro Isomerasas/metabolismo , Proteínas/genética , Proteínas/metabolismo , Proteoglicanos , Análisis de Secuencia de ADN , Sindecanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA