Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Hum Mol Genet ; 24(11): 3181-91, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25701874

RESUMEN

Fabry disease is caused by deficient activity of lysosomal enzyme α-galactosidase A. The enzyme deficiency results in intracellular accumulation of glycosphingolipids, leading to a variety of clinical manifestations including hypertrophic cardiomyopathy and renal insufficiency. The mechanism through which glycosphingolipid accumulation causes these manifestations remains unclear. Current treatment, especially when initiated at later stage of the disease, does not produce completely satisfactory results. Elucidation of the pathogenesis of Fabry disease is therefore crucial to developing new treatments. We found increased activity of androgen receptor (AR) signaling in Fabry disease. We subsequently also found that blockade of AR signaling either through castration or AR-antagonist prevented and reversed cardiac and kidney hypertrophic phenotype in a mouse model of Fabry disease. Our findings implicate abnormal AR pathway in the pathogenesis of Fabry disease and suggest blocking AR signaling as a novel therapeutic approach.


Asunto(s)
Antagonistas de Receptores Androgénicos/farmacología , Enfermedad de Fabry/metabolismo , Hipertrofia Ventricular Izquierda/metabolismo , Enfermedades Renales/metabolismo , Receptores Androgénicos/metabolismo , Animales , Enfermedad de Fabry/tratamiento farmacológico , Femenino , Hipertrofia Ventricular Izquierda/tratamiento farmacológico , Riñón/metabolismo , Riñón/patología , Enfermedades Renales/tratamiento farmacológico , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores Androgénicos/genética , Transducción de Señal , Transcripción Genética
2.
Circ Res ; 117(11): 967-77, 2015 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-26438688

RESUMEN

RATIONALE: Inflammation in post-myocardial infarction (MI) is necessary for myocyte repair and wound healing. Unfortunately, it is also a key component of subsequent heart failure pathology. Transcription factor forkhead box O4 (FoxO4) regulates a variety of biological processes, including inflammation. However, its role in MI remains unknown. OBJECTIVE: To test the hypothesis that FoxO4 promotes early post-MI inflammation via endothelial arginase 1 (Arg1). METHODS AND RESULTS: We induced MI in wild-type and FoxO4(-/-) mice. FoxO4(-/-) mice had a significantly higher post-MI survival, better cardiac function, and reduced infarct size. FoxO4(-/-) hearts had significantly fewer neutrophils, reduced expression of cytokines, and competitive nitric oxide synthase inhibitor Arg1. We generated conditional FoxO4 knockout mice with FoxO4 deleted in cardiac mycoytes or endothelial cells. FoxO4 endothelial cell-specific knockout mice showed significant post-MI improvement of cardiac function and reduction of neutrophil accumulation and cytokine expression, whereas FoxO4 cardiac mycoyte-specific knockout mice had no significant difference in cardiac function and post-MI inflammation from those of control littermates. FoxO4 binds the Foxo-binding site in the Arg1 promoter and activates Arg1 transcription. FoxO4 knockdown in human aortic endothelial cells upregulated nitric oxide on ischemia and suppressed monocyte adhesion that can be reversed by ectopic-expression of Arg1. Furthermore, chemical inhibition of Arg1 in wild-type mice had similar cardioprotection and reduced inflammation after MI as FoxO4 inactivation and administration of nitric oxide synthase inhibitor to FoxO4 KO mice reversed the beneficial effects of FoxO4 deletion on post-MI cardiac function. CONCLUSIONS: FoxO4 activates Arg1 transcription in endothelial cells in response to MI, leading to downregulation of nitric oxide and upregulation of neutrophil infiltration to the infarct area.


Asunto(s)
Arginasa/biosíntesis , Células Endoteliales/enzimología , Factores de Transcripción Forkhead/metabolismo , Inflamación/enzimología , Infarto del Miocardio/enzimología , Miocitos Cardíacos/enzimología , Animales , Antiinflamatorios/farmacología , Arginasa/antagonistas & inhibidores , Arginasa/genética , Sitios de Unión , Adhesión Celular , Proteínas de Ciclo Celular , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , Inducción Enzimática , Factores de Transcripción Forkhead/deficiencia , Factores de Transcripción Forkhead/genética , Células HEK293 , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Inflamación/fisiopatología , Inflamación/prevención & control , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/genética , Infarto del Miocardio/inmunología , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/patología , Infiltración Neutrófila , Óxido Nítrico/metabolismo , Regiones Promotoras Genéticas , Interferencia de ARN , Transducción de Señal , Factores de Tiempo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Transfección , Células U937
3.
Proc Natl Acad Sci U S A ; 111(1): E129-38, 2014 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-24367119

RESUMEN

Mammalian skeletal muscle can remodel, repair, and regenerate itself by mobilizing satellite cells, a resident population of myogenic progenitor cells. Muscle injury and subsequent activation of myogenic progenitor cells is associated with oxidative stress. Cytoglobin is a hemoprotein expressed in response to oxidative stress in a variety of tissues, including striated muscle. In this study, we demonstrate that cytoglobin is up-regulated in activated myogenic progenitor cells, where it localizes to the nucleus and contributes to cell viability. siRNA-mediated depletion of cytoglobin from C2C12 myoblasts increased levels of reactive oxygen species and apoptotic cell death both at baseline and in response to stress stimuli. Conversely, overexpression of cytoglobin reduced reactive oxygen species levels, caspase activity, and cell death. Mice in which cytoglobin was knocked out specifically in skeletal muscle were generated to examine the role of cytoglobin in vivo. Myogenic progenitor cells isolated from these mice were severely deficient in their ability to form myotubes as compared with myogenic progenitor cells from wild-type littermates. Consistent with this finding, the capacity for muscle regeneration was severely impaired in mice deficient for skeletal-muscle cytoglobin. Collectively, these data demonstrate that cytoglobin serves an important role in muscle repair and regeneration.


Asunto(s)
Regulación de la Expresión Génica , Globinas/metabolismo , Músculos/fisiología , Regeneración/fisiología , Células Satélite del Músculo Esquelético/citología , Animales , Apoptosis , Diferenciación Celular , Línea Celular , Proliferación Celular , Supervivencia Celular , Citoglobina , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno , Células Satélite del Músculo Esquelético/patología , Células Madre/citología , Factores de Tiempo
4.
Circ Res ; 108(1): 51-9, 2011 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-21106942

RESUMEN

RATIONALE: Transgenic Notch reporter mice express enhanced green fluorescent protein in cells with C-promoter binding factor-1 response element transcriptional activity (CBF1-RE(x)4-EGFP), providing a unique and powerful tool for identifying and isolating "Notch-activated" progenitors. OBJECTIVE: We asked whether, as in other tissues of this mouse, EGFP localized and functionally tagged adult cardiac tissue progenitors, and, if so, whether this cell-based signal could serve as a quantitative and qualitative biosensor of the injury repair response of the heart. METHODS AND RESULTS: In addition to scattered endothelial and interstitial cells, Notch-activated (EGFP(+)) cells unexpectedly richly populated the adult epicardium. We used fluorescence-activated cell sorting to isolate EGFP(+) cells and excluded hematopoietic (CD45(+)) and endothelial (CD31(+)) subsets. We analyzed EGFP(+)/CD45⁻/CD31⁻ cells, a small (<2%) but distinct subpopulation, by gene expression profiling and functional analyses. We called this mixed cell pool, which had dual multipotent stromal cell and epicardial lineage signatures, Notch-activated epicardial-derived cells (NECs). Myocardial infarction and thoracic aortic banding amplified the NEC pool, increasing fibroblast differentiation. Validating the functional vitality of clonal NEC lines, serum growth factors triggered epithelial-mesenchymal transition and the immobilized Notch ligand Delta-like 1-activated downstream target genes. Moreover, cardiomyocyte coculture and engraftment in NOD-SCID (nonobese diabetic-severe combined immunodeficiency) mouse myocardium increased cardiac gene expression in NECs. CONCLUSIONS: A dynamic Notch injury response activates adult epicardium, producing a multipotent cell population that contributes to fibrosis repair.


Asunto(s)
Células Madre Multipotentes/metabolismo , Infarto del Miocardio/metabolismo , Pericardio/metabolismo , Receptores Notch/metabolismo , Animales , Proteínas de Unión al Calcio , Fibrosis , Perfilación de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Antígenos Comunes de Leucocito , Ratones , Ratones Transgénicos , Células Madre Multipotentes/patología , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Pericardio/patología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta , Receptores Notch/genética
5.
Elife ; 122023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37096669

RESUMEN

Enhancers orchestrate gene expression programs that drive multicellular development and lineage commitment. Thus, genetic variants at enhancers are thought to contribute to developmental diseases by altering cell fate commitment. However, while many variant-containing enhancers have been identified, studies to endogenously test the impact of these enhancers on lineage commitment have been lacking. We perform a single-cell CRISPRi screen to assess the endogenous roles of 25 enhancers and putative cardiac target genes implicated in genetic studies of congenital heart defects (CHDs). We identify 16 enhancers whose repression leads to deficient differentiation of human cardiomyocytes (CMs). A focused CRISPRi validation screen shows that repression of TBX5 enhancers delays the transcriptional switch from mid- to late-stage CM states. Endogenous genetic deletions of two TBX5 enhancers phenocopy epigenetic perturbations. Together, these results identify critical enhancers of cardiac development and suggest that misregulation of these enhancers could contribute to cardiac defects in human patients.


Asunto(s)
Cardiopatías Congénitas , Miocitos Cardíacos , Humanos , Miocitos Cardíacos/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos , Diferenciación Celular/genética , Cardiopatías Congénitas/genética
6.
Nat Med ; 26(11): 1788-1800, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33188278

RESUMEN

Ribonucleoprotein (RNP) granules are biomolecular condensates-liquid-liquid phase-separated droplets that organize and manage messenger RNA metabolism, cell signaling, biopolymer assembly, biochemical reactions and stress granule responses to cellular adversity. Dysregulated RNP granules drive neuromuscular degenerative disease but have not previously been linked to heart failure. By exploring the molecular basis of congenital dilated cardiomyopathy (DCM) in genome-edited pigs homozygous for an RBM20 allele encoding the pathogenic R636S variant of human RNA-binding motif protein-20 (RBM20), we discovered that RNP granules accumulated abnormally in the sarcoplasm, and we confirmed this finding in myocardium and reprogrammed cardiomyocytes from patients with DCM carrying the R636S allele. Dysregulated sarcoplasmic RBM20 RNP granules displayed liquid-like material properties, docked at precisely spaced intervals along cytoskeletal elements, promoted phase partitioning of cardiac biomolecules and fused with stress granules. Our results link dysregulated RNP granules to myocardial cellular pathobiology and heart failure in gene-edited pigs and patients with DCM caused by RBM20 mutation.


Asunto(s)
Cardiomiopatía Dilatada/genética , Miocardio/metabolismo , Proteínas de Unión al ARN/genética , Ribonucleoproteínas/genética , Alelos , Animales , Cardiomiopatía Dilatada/fisiopatología , Reprogramación Celular , Modelos Animales de Enfermedad , Femenino , Edición Génica , Humanos , Masculino , Mutación/genética , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , ARN Mensajero/genética , Retículo Sarcoplasmático/genética , Retículo Sarcoplasmático/metabolismo , Vesículas Secretoras/genética , Vesículas Secretoras/metabolismo , Porcinos
7.
Physiol Genomics ; 30(1): 44-52, 2007 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-17327495

RESUMEN

Previous studies have suggested that the heart may be capable of limited repair and regeneration in response to a focal injury, while other studies indicate that the mammalian heart has no regenerative capacity. To further explore this issue, we performed a series of superficial and transmural myocardial injuries in C57BL/6 and MRL/MpJ adult mice. At defined time intervals following the respective injury (days 3, 14, 30 and 60), we examined cardiac function using echocardiography, morphology, fluorescence-activated cell sorting for 5-bromo-2-deoxyuridine-positive cells and molecular signature using microarray analysis. We observed restoration of myocardial function in the superficial MRL cryoinjured heart and significantly less collagen deposition compared with the injured hearts of C57BL/6 mice. Following a severe transmural myocardial injury, the MRL mouse has increased survival and decreased ventricular remodeling compared with the C57BL/6 mouse but without evidence of complete regeneration. The cytoprotective program observed in the severely injured MRL heart is in part due to increased cellular proliferation, increased vasculogenesis, and decreased apoptosis that limits the extension of the injury. We conclude that MRL injured hearts have evidence of myocardial regeneration, in response to superficial injury, but the stabilized left ventricular function and improved survival observed in the MRL mouse following severe injury is not associated with complete myocardial regeneration.


Asunto(s)
Lesiones Cardíacas/fisiopatología , Miocardio/metabolismo , Regeneración/fisiología , Animales , Colágeno/metabolismo , Ecocardiografía , Citometría de Flujo , Lesiones Cardíacas/etiología , Lesiones Cardíacas/genética , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos MRL lpr , Miocardio/patología , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Cadenas Ligeras de Miosina/genética , Cadenas Ligeras de Miosina/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Regeneración/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Transcripción Genética , Remodelación Ventricular/genética , Remodelación Ventricular/fisiología , Cicatrización de Heridas/genética , Cicatrización de Heridas/fisiología , Heridas Penetrantes/complicaciones
9.
Stem Cells Dev ; 14(2): 181-7, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15910244

RESUMEN

The regenerative capacity of skeletal muscle is due to the myogenic progenitor cell population that is resident in adult skeletal muscle. To enhance our understanding of this cell population, we examined the temporal-spatial expression pattern for filamin C during murine embryogenesis, adult muscle regeneration and in selected myopathic models of human disease. Using in situ hybridization, we observed filamin C to be restricted to mesodermal lineages including the developing heart and skeletal muscle during embryogenesis. Following cardiotoxin-induced muscle injury of adult skeletal muscle, filamin C expression was dynamically regulated in activated myogenic progenitor cells and newly regenerated myotubes. This expression pattern was further supported using RT-PCR analysis of filamin C expression in differentiating C2C12 myotubes. These results support the paradigm that the regulatory mechanisms of muscle regeneration largely recapitulate the fundamental events observed during muscle development and that filamin C may function in signal transduction or cellular migration of the myogenic progenitor cell population.


Asunto(s)
Proteínas Contráctiles/biosíntesis , Proteínas Contráctiles/fisiología , Proteínas de Microfilamentos/biosíntesis , Proteínas de Microfilamentos/fisiología , Músculo Esquelético/citología , Animales , Técnicas de Cultivo de Célula , Línea Celular , Linaje de la Célula , Movimiento Celular , Proliferación Celular , Células Cultivadas , Filaminas , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Músculo Esquelético/embriología , Músculos/citología , Miocardio/metabolismo , ARN/metabolismo , Regeneración , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Células Madre/citología , Factores de Tiempo , Regulación hacia Arriba
10.
Am J Med Sci ; 330(2): 88-101, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16103789

RESUMEN

Heart failure has reached epidemic proportions in the United States. More than 5 million patients are treated for heart failure and approximately a half a million new patients are diagnosed with this disease each year in the United States. Recent pharmacological therapies have been used for the treatment of this patient population, but heart failure remains a major source of morbidity and mortality for patients. Orthotopic heart transplantation is a viable treatment option for heart failure patients; however, cardiac transplantation is limited by the donor availability. Limited donor organ availability has led to the development of alternative therapeutic strategies, including xenotransplantation, mechanical support devices, and cell transfer/tissue engineering protocols. This review highlights the current treatment modalities and emerging strategies for the treatment of advanced heart failure.


Asunto(s)
Insuficiencia Cardíaca/terapia , Trasplante de Corazón/métodos , Animales , Insuficiencia Cardíaca/cirugía , Humanos
11.
Physiol Genomics ; 14(3): 261-71, 2003 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-12799472

RESUMEN

Muscle regeneration is a complex process requiring the coordinated interaction between the myogenic progenitor cells or satellite cells, growth factors, cytokines, inflammatory components, vascular components and the extracellular matrix (ECM). Previous studies have elegantly described the physiological modulation of the regenerative process in response to muscle injury, but the molecular response that characterizes stages of the repair process remains ill-defined. The recent completion of the Human and Mouse Genome Projects and the advent of technologies such as high-density oligonucleotide array analysis facilitate an expanded analysis of complex processes such as muscle regeneration. In the present study, we define cellular and molecular events that characterize stages of muscle injury and regeneration. Utilization of transcriptional profiling strategies revealed coordinated expression of growth factors [i.e., Tgfb1, Igf1, Egf, chemokine (C-C motif) ligand 6 and 7], the fetal myogenic program (Myod1, Myf5, Myf6), and the biomatrix (procollagen genes, Mmp3, Mmp9, biglycan, periostin) during muscle regeneration. Corroboration of the transcriptional profiling analysis included quantitative real-time RT-PCR and in situ hybridization analyses of selected candidate genes. In situ hybridization studies for periostin [osteoblast-specific factor 2 (fasciclin I-like)] and biglycan revealed that these genes are restricted to mesenchymal derivatives during embryogenesis and are significantly regulated during regeneration of the injured hindlimb skeletal muscle. We conclude that muscle regeneration is a complex process that requires the coordinated modulation of the inflammatory response, myogenic progenitor cells, growth factors, and ECM for complete restoration of muscle architecture.


Asunto(s)
Matriz Extracelular/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/genética , Músculo Esquelético/fisiopatología , Análisis de Secuencia por Matrices de Oligonucleótidos , Regeneración/genética , Células 3T3 , Animales , Biglicano , Moléculas de Adhesión Celular/biosíntesis , Moléculas de Adhesión Celular/fisiología , Línea Celular , Matriz Extracelular/fisiología , Proteínas de la Matriz Extracelular , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Miembro Posterior , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Fibras Musculares Esqueléticas/química , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Músculo Esquelético/embriología , Músculo Esquelético/patología , Mioblastos/química , Mioblastos/efectos de los fármacos , Mioblastos/metabolismo , Mioblastos/patología , Proteoglicanos/biosíntesis , Proteoglicanos/fisiología , Factor de Crecimiento Transformador beta/farmacología , Factor de Crecimiento Transformador beta/fisiología , Factor de Crecimiento Transformador beta1
12.
J Histochem Cytochem ; 50(12): 1591-8, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12486081

RESUMEN

Hemoproteins are widely distributed among unicellular eukaryotes, plants, and animals. In addition to myoglobin and hemoglobin, a third hemoprotein, neuroglobin, has recently been isolated from vertebrate brain. Although the functional role of this novel member of the globin family remains unclear, neuroglobin contains a heme-binding domain and may participate in diverse processes such as oxygen transport, oxygen storage, nitric oxide detoxification, or modulation of terminal oxidase activity. In this study we utilized in situ hybridization (ISH) and RT-PCR analyses to examine the expression of neuroglobin in the normoxic and hypoxic murine brain. In the normoxic adult mouse, neuroglobin expression was observed in focal regions of the brain, including the lateral tegmental nuclei, the preoptic nucleus, amygdala, locus coeruleus, and nucleus of the solitary tract. Using ISH and RT-PCR techniques, no significant changes in neuroglobin expression in the adult murine brain was observed in response to chronic 10% oxygen. These results support the hypothesis that neuroglobin is a hemoprotein that is expressed in the brain and may have diverse functional roles.


Asunto(s)
Encéfalo/metabolismo , Globinas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Expresión Génica , Globinas/genética , Hipoxia/metabolismo , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/genética , Neuroglobina , Especificidad de Órganos , Oxígeno/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
ACS Chem Biol ; 7(6): 1067-76, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22413910

RESUMEN

Targeting native progenitors with small molecule pharmaceuticals that direct cell fate decisions is an attractive approach for regenerative medicine. Here, we show that 3,5-disubstituted isoxazoles (Isx), stem cell-modulator small molecules originally recovered in a P19 embryonal carcinoma cell-based screen, directed cardiac muscle gene expression in vivo in target tissues of adult transgenic reporter mice. Isx also stimulated adult mouse myocardial cell cycle activity. Narrowing our focus onto one target cardiac-resident progenitor population, Isx directed muscle transcriptional programs in vivo in multipotent Notch-activated epicardium-derived cells (NECs), generating Notch-activated adult cardiomyocyte-like precursors. Myocardial infarction (MI) preemptively differentiated NECs toward fibroblast lineages, overriding Isx's cardiogenic influence in this cell population. Isx dysregulated gene expression in vivo in Notch-activated repair fibroblasts, driving distinctive (pro-angiogenesis) gene programs, but failed to mitigate fibrosis or avert ventricular functional decline after MI. In NECs in vitro, Isx directed partial muscle differentiation, which included biosynthesis and assembly of sarcomeric α-actinin premyofibrils, beaded structures pathognomonic of early developing cardiomyocytes. Thus, although Isx small molecules have promising in vivo efficacy at the level of cardiac muscle gene expression in native multipotent progenitors and are first in class in this regard, a greater understanding of the dynamic interplay between fibrosis and cardiogenic small molecule signals will be required to pharmacologically enable regenerative repair of the heart.


Asunto(s)
Corazón/efectos de los fármacos , Isoxazoles/química , Isoxazoles/farmacología , Infarto del Miocardio/tratamiento farmacológico , Miocardio/metabolismo , Activación Transcripcional/efectos de los fármacos , Animales , Células Cultivadas , Expresión Génica/efectos de los fármacos , Corazón/fisiopatología , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Isoxazoles/uso terapéutico , Masculino , Ratones , Ratones Transgénicos , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/patología , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Pericardio/citología , Pericardio/metabolismo , Pericardio/patología , Pericardio/fisiopatología , Receptores Notch/metabolismo
14.
ACS Chem Neurosci ; 3(7): 557-68, 2012 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-22860225

RESUMEN

Acidosis, a critical aspect of central nervous system (CNS) pathophysiology and a metabolic corollary of the hypoxic stem cell niche, could be an expedient trigger for hippocampal neurogenesis and brain repair. We recently tracked the function of our isoxazole stem cell-modulator small molecules (Isx) through a chemical biology-target discovery strategy to GPR68, a proton (pH) sensing G protein-coupled receptor with no known function in brain. Isx and GPR68 coregulated neuronal target genes such as Bex1 (brain-enriched X-linked protein-1) in hippocampal neural progenitors (HCN cells), which further amplified GPR68 signaling by producing metabolic acid in response to Isx. To evaluate this proneurogenic small molecule/proton signaling circuit in vivo, we explored GPR68 and BEX1 expression in brain and probed brain function with Isx. We localized proton-sensing GPR68 to radial processes of hippocampal type 1 neural stem cells (NSCs) and, conversely, localized BEX1 to neurons. At the transcriptome level, Isx demonstrated unrivaled proneurogenic activity in primary hippocampal NSC cultures. In vivo, Isx pharmacologically targeted type 1 NSCs, promoting neurogenesis in young mice, depleting the progenitor pool without adversely affecting hippocampal learning and memory function. After traumatic brain injury, cerebral cortical astrocytes abundantly expressed GPR68, suggesting an additional role for proton-GPCR signaling in reactive astrogliosis. Thus, probing a novel proneurogenic synthetic small molecule's mechanism-of-action, candidate target, and pharmacological activity, we identified a new GPR68 regulatory pathway for integrating neural stem and astroglial cell functions with brain pH.


Asunto(s)
Encéfalo/fisiología , Hipocampo/fisiología , Neurogénesis/fisiología , Receptores Acoplados a Proteínas G/metabolismo , Animales , Células Cultivadas , Hipocampo/citología , Concentración de Iones de Hidrógeno , Aprendizaje por Laberinto/fisiología , Ratones , Células-Madre Neurales/metabolismo , Células-Madre Neurales/fisiología , Receptores Acoplados a Proteínas G/fisiología
15.
ACS Chem Biol ; 7(6): 1077-83, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22462679

RESUMEN

Chemical biology promises discovery of new and unexpected mechanistic pathways, protein functions and disease targets. Here, we probed the mechanism-of-action and protein targets of 3,5-disubstituted isoxazoles (Isx), cardiomyogenic small molecules that target Notch-activated epicardium-derived cells (NECs) in vivo and promote functional recovery after myocardial infarction (MI). Mechanistic studies in NECs led to an Isx-activated G(q) protein-coupled receptor (G(q)PCR) hypothesis tested in a cell-based functional target screen for GPCRs regulated by Isx. This screen identified one agonist hit, the extracellular proton/pH-sensing GPCR GPR68, confirmed through genetic gain- and loss-of-function. Overlooked until now, GPR68 expression and localization were highly regulated in early post-natal and adult post-infarct mouse heart, where GPR68-expressing cells accumulated subepicardially. Remarkably, GPR68-expressing cardiomyocytes established a proton-sensing cellular "buffer zone" surrounding the MI. Isx pharmacologically regulated gene expression (mRNAs and miRs) in this GPR68-enriched border zone, driving cardiomyogenic and pro-survival transcriptional programs in vivo. In conclusion, we tracked a (micromolar) bioactive small molecule's mechanism-of-action to a candidate target protein, GPR68, and validated this target as a previously unrecognized regulator of myocardial cellular responses to tissue acidosis, setting the stage for future (nanomolar) target-based drug lead discovery.


Asunto(s)
Isoxazoles/química , Isoxazoles/farmacología , Infarto del Miocardio/tratamiento farmacológico , Pericardio/citología , Receptores Acoplados a Proteínas G/genética , Activación Transcripcional/efectos de los fármacos , Animales , Calcio/metabolismo , Línea Celular , Células Cultivadas , Ratones , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Pericardio/efectos de los fármacos , Pericardio/metabolismo , Pericardio/patología , Receptores Notch/metabolismo
16.
Am J Physiol Cell Physiol ; 293(5): C1636-44, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17855775

RESUMEN

Xin is a muscle-specific actin binding protein of which its role and regulation within skeletal muscle is not well understood. Here we demonstrate that Xin mRNA is robustly upregulated (>16-fold) within 12 h of skeletal muscle injury and is localized to the muscle satellite cell population. RT-PCR confirmed the expression pattern of Xin during regeneration, as well as within primary muscle myoblast cultures, but not other known stem cell populations. Immunohistochemical staining of single myofibers demonstrate Xin expression colocalized with the satellite cell marker Syndecan-4 further supporting the mRNA expression of Xin in satellite cells. In situ hybridization of regenerating muscle 5-7 days postinjury illustrates Xin expression within newly regenerated myofibers. Promoter-reporter assays demonstrate that known myogenic transcription factors [myocyte enhancer factor-2 (MEF2), myogenic differentiation-1 (MyoD), and myogenic factor-5 (Myf-5)] transactivate Xin promoter constructs supporting the muscle-specific expression of Xin. To determine the role of Xin within muscle precursor cells, proliferation, migration, and differentiation analysis using Xin, short hairpin RNA (shRNA) were undertaken in C2C12 myoblasts. Reducing endogenous Xin expression resulted in a 26% increase (P < 0.05) in cell proliferation and a 20% increase (P < 0.05) in myoblast migratory capacity. Skeletal muscle myosin heavy chain protein levels were increased (P < 0.05) with Xin shRNA administration; however, this was not accompanied by changes in myoglobin protein (another marker of differentiation) nor overt morphological differences relative to differentiating control cells. Taken together, the present findings support the hypothesis that Xin is expressed within muscle satellite cells during skeletal muscle regeneration and is involved in the regulation of myoblast function.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Enfermedades Musculares/metabolismo , Proteínas Nucleares/metabolismo , Regeneración , Células Satélite del Músculo Esquelético/metabolismo , Animales , Línea Celular , Movimiento Celular , Proliferación Celular , Proteínas Cardiotóxicas de Elápidos , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Genes Reporteros , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Desarrollo de Músculos , Músculo Esquelético/embriología , Músculo Esquelético/fisiopatología , Enfermedades Musculares/inducido químicamente , Enfermedades Musculares/genética , Enfermedades Musculares/fisiopatología , Factores Reguladores Miogénicos/genética , Factores Reguladores Miogénicos/metabolismo , Proteínas Nucleares/genética , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sindecano-4/metabolismo , Factores de Tiempo , Activación Transcripcional , Regulación hacia Arriba
17.
EMBO J ; 26(7): 1902-12, 2007 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-17363903

RESUMEN

The regulation of myogenic progenitor cells during muscle regeneration is not clearly understood. We have previously shown that the Foxk1 gene, a member of the forkhead/winged helix family of transcription factors, is expressed in myogenic progenitor cells in adult skeletal muscle. In the present study, we utilize transgenic technology and demonstrate that the 4.6 kb upstream fragment of the Foxk1 gene directs beta-galactosidase expression to the myogenic progenitor cell population. We further establish that Sox15 directs Foxk1 expression to the myogenic progenitor cell population, as it binds to an evolutionarily conserved site and recruits Fhl3 to transcriptionally coactivate Foxk1 gene expression. Knockdown of endogenous Sox15 results in perturbed cell cycle kinetics and decreased Foxk1 expression. Furthermore, Sox15 mutant mice display perturbed skeletal muscle regeneration, due in part to decreased numbers of satellite cells and decreased Foxk1 expression. These studies demonstrate that Sox15, Fhl3 and Foxk1 function to coordinately regulate the myogenic progenitor cell population and skeletal muscle regeneration.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Músculo Esquelético/citología , Células Madre/citología , Factores de Transcripción/metabolismo , Transcripción Genética , Animales , Secuencia de Bases , Sitios de Unión , Ciclo Celular , Secuencia Conservada , Embrión de Mamíferos/metabolismo , Factores de Transcripción Forkhead/genética , Regulación de la Expresión Génica , Proteínas del Grupo de Alta Movilidad/química , Péptidos y Proteínas de Señalización Intracelular/química , Cinética , Proteínas con Dominio LIM , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Músculo Esquelético/fisiología , Mutagénesis , Regiones Promotoras Genéticas/genética , Unión Proteica , Estructura Terciaria de Proteína , ARN Mensajero/genética , ARN Mensajero/metabolismo , Regeneración , Factores de Transcripción SOX , Células Madre/metabolismo , Factores de Transcripción/química
18.
Am J Physiol Cell Physiol ; 290(2): C379-87, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16221735

RESUMEN

The successful use of myogenic progenitor cells for therapeutic applications requires an understanding of the intrinsic and extrinsic cues involved in their regulation. Herein we demonstrate the expression pattern and transcriptional regulation of Rad, a prototypical member of a family of novel Ras-related GTPases, during mammalian development and skeletal muscle regeneration. Rad was identified using microarray analysis, which revealed robust upregulation of its expression during skeletal muscle regeneration. Our current findings demonstrate negligible Rad expression with resting adult skeletal muscle; however, after muscle injury, Rad is expressed within the myogenic progenitor cell population. Rad expression is significantly increased and localized to the myogenic progenitor cell population during the early phases of regeneration and within the newly regenerated myofibers during the later phases of regeneration. Immunohistochemical analysis demonstrated that Rad and MyoD are coexpressed within the myogenic progenitor cell population of regenerating skeletal muscle. This expression profile of Rad during skeletal muscle regeneration is consistent with the proposed roles for Rad in the inhibition of L-type Ca(2+) channel activity and the inhibition of Rho/RhoA kinase activity. We also have demonstrated that known myogenic transcription factors (MEF2, MyoD, and Myf-5) can increase the transcriptional activity of the Rad promoter and that this ability is significantly enhanced by the presence of the Ca(2+)-dependent phosphatase calcineurin. Furthermore, this enhanced transcriptional activity appears to be dependent on the presence of a conserved NFAT binding motif within the Rad promoter. Taken together, these data define Rad as a novel factor within the myogenic progenitor cells of skeletal muscle and identify key regulators of its transcriptional activity.


Asunto(s)
Regulación de la Expresión Génica , Músculo Esquelético/fisiología , Regeneración/fisiología , Células Madre/fisiología , Proteínas ras/metabolismo , Animales , Linaje de la Célula , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Músculo Esquelético/citología , Músculo Esquelético/patología , Miocardio/citología , Miocardio/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas , Distribución Tisular , Transcripción Genética , Proteínas ras/genética
19.
Dev Biol ; 265(1): 262-75, 2004 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-14697368

RESUMEN

Stem cells are important in the maintenance and repair of adult tissues. A population of cells, termed side population (SP) cells, has stem cell characteristics as they have been shown to contribute to diverse lineages. In this study, we confirm that Abcg2 is a determinant of the SP cell phenotype. Therefore, we examined Abcg2 expression during murine embryogenesis and observed robust expression in the blood islands of the E8.5 yolk sac and in developing tissues including the heart. During the latter stages of embryogenesis, Abcg2 identifies a rare cell population in the developing organs. We further establish that the adult heart contains an Abcg2 expressing SP cell population and these progenitor cells are capable of proliferation and differentiation. We define the molecular signature of cardiac SP cells and compare it to embryonic stem cells and adult cardiomyocytes using emerging technologies. We propose that the cardiac SP cell population functions as a progenitor cell population for the development, maintenance, and repair of the heart.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/fisiología , Regulación del Desarrollo de la Expresión Génica , Corazón/fisiología , Miocardio/citología , Proteínas de Neoplasias/fisiología , Células Madre/fisiología , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/genética , Animales , Western Blotting , Células Cultivadas , Perfilación de la Expresión Génica , Inmunohistoquímica , Hibridación in Situ , Ratones , Proteínas de Neoplasias/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Saco Vitelino/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA