Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-33443158

RESUMEN

The sinus node (SAN) is the primary pacemaker of the human heart, and abnormalities in its structure or function cause sick sinus syndrome, the most common reason for electronic pacemaker implantation. Here we report that transcription factor GATA6, whose mutations in humans are linked to arrhythmia, is highly expressed in the SAN and its haploinsufficiency in mice results in hypoplastic SANs and rhythm abnormalities. Cell-specific deletion reveals a requirement for GATA6 in various SAN lineages. Mechanistically, GATA6 directly activates key regulators of the SAN genetic program in conduction and nonconduction cells, such as TBX3 and EDN1, respectively. The data identify GATA6 as an important regulator of the SAN and provide a molecular basis for understanding the conduction abnormalities associated with GATA6 mutations in humans. They also suggest that GATA6 may be a potential modifier of the cardiac pacemaker.


Asunto(s)
Factor de Transcripción GATA6/metabolismo , Frecuencia Cardíaca/fisiología , Nodo Sinoatrial/embriología , Animales , Arritmias Cardíacas/fisiopatología , Diferenciación Celular/genética , Factor de Transcripción GATA6/genética , Regulación del Desarrollo de la Expresión Génica/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Organogénesis , Nodo Sinoatrial/fisiología , Proteínas de Dominio T Box/genética
2.
Circulation ; 138(10): 1025-1038, 2018 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-29567669

RESUMEN

BACKGROUND: Bicuspid aortic valve (BAV), the most common congenital heart defect affecting 1% to 2% of the population, is a major risk factor for premature aortic valve disease and accounts for the majority of valve replacement. The genetic basis and mechanisms of BAV etiology and pathogenesis remain largely undefined. METHODS: Cardiac structure and function was assessed in mice lacking a Gata6 allele. Human GATA6 gene variants were analyzed in 452 BAV cases from the BAV consortium and 1849 controls from the Framingham GWAS (Genome Wide Association Study). GATA6 expression was determined in mice and human tissues using quantitative real-time polymerase chain reaction and immunohistochemistry. Mechanistic studies were carried out in cultured cells. RESULTS: Gata6 heterozygous mice have highly penetrant right-left (RL)-type BAV, the most frequent type in humans. GATA6 transcript levels are lower in human BAV compared with normal tricuspid valves. Mechanistically, Gata6 haploinsufficiency disrupts valve remodeling and extracellular matrix composition through dysregulation of important signaling molecules, including matrix metalloproteinase 9. Cell-specific inactivation of Gata6 reveals an essential role for GATA6 in secondary heart field myocytes because loss of 1 Gata6 allele from Isl- 1-positive cells-but not from endothelial or neural crest cells-recapitulates the phenotype of Gata6 heterozygous mice. CONCLUSIONS: The data identify a new cellular and molecular mechanism underlying BAV. The availability of an animal model for the most frequent human BAV opens the way for the elucidation of BAV pathogenesis and the development of much needed therapies.


Asunto(s)
Válvula Aórtica/anomalías , Válvula Aórtica/metabolismo , Factor de Transcripción GATA6/genética , Haploinsuficiencia , Enfermedades de las Válvulas Cardíacas/genética , Animales , Válvula Aórtica/patología , Válvula Aórtica/fisiopatología , Enfermedad de la Válvula Aórtica Bicúspide , Estudios de Casos y Controles , Células Cultivadas , Modelos Animales de Enfermedad , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Femenino , Factor de Transcripción GATA6/deficiencia , Predisposición Genética a la Enfermedad , Enfermedades de las Válvulas Cardíacas/metabolismo , Enfermedades de las Válvulas Cardíacas/patología , Enfermedades de las Válvulas Cardíacas/fisiopatología , Heterocigoto , Humanos , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína Wnt1/genética , Proteína Wnt1/metabolismo
3.
Hum Mol Genet ; 26(5): 942-954, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28164238

RESUMEN

TBX5, a member of the T-box family of transcription factors, is a dosage sensitive regulator of heart development. Mutations in TBX5 are responsible for Holt-Oram Syndrome, an autosomal dominant disease with variable and partially penetrant cardiac defects suggestive of the existence of genetic and environmental modifiers. KLF13, a member of the Krüppel-like family of zinc finger proteins is co-expressed with TBX5 in several cardiac cells including atrial cardiomyocytes and cells of the interatrial septum. We report that KLF13 interacts physically and functionally with TBX5 to synergistically activate transcription of cardiac genes. We show that TBX5 contacts KLF13 via its T-domain and find that several disease-causing mutations therein have decreased KLF13 interaction. Whereas Klf13 heterozygote mice have no detectable cardiac defects, loss of a Klf13 allele in Tbx5 heterozygote mice significantly increases the penetrance of TBX5-dependent cardiac abnormalities including atrial, atrial-ventricular and ventricular septal defects. The results reveal for the first time combinatorial interaction between a T-box protein and a KLF family member and its importance for heart and possibly other organ development. The data also suggest that, in human, KLF13 may be a genetic modifier of the Holt-Oram Syndrome gene TBX5.


Asunto(s)
Anomalías Múltiples/genética , Proteínas de Ciclo Celular/genética , Atrios Cardíacos/metabolismo , Cardiopatías Congénitas/genética , Defectos del Tabique Interatrial/genética , Factores de Transcripción de Tipo Kruppel/genética , Deformidades Congénitas de las Extremidades Inferiores/genética , Proteínas Represoras/genética , Proteínas de Dominio T Box/genética , Deformidades Congénitas de las Extremidades Superiores/genética , Anomalías Múltiples/metabolismo , Anomalías Múltiples/patología , Animales , Proteínas de Ciclo Celular/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Atrios Cardíacos/patología , Cardiopatías Congénitas/metabolismo , Cardiopatías Congénitas/patología , Defectos del Tabique Interatrial/metabolismo , Defectos del Tabique Interatrial/patología , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Heterocigoto , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo , Deformidades Congénitas de las Extremidades Inferiores/metabolismo , Deformidades Congénitas de las Extremidades Inferiores/patología , Ratones , Mutación , Unión Proteica , Dominios Proteicos/genética , Mapas de Interacción de Proteínas/genética , Proteínas Represoras/metabolismo , Proteínas de Dominio T Box/metabolismo , Activación Transcripcional/genética , Deformidades Congénitas de las Extremidades Superiores/metabolismo , Deformidades Congénitas de las Extremidades Superiores/patología
4.
Cell Tissue Res ; 367(2): 369-385, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27826738

RESUMEN

The phenylephrine-induced complex-1 (PEX1) transcription factor, also known as zinc-finger protein 260 (Zfp260), is an effector of endothelin-1 and α1-adrenergic signaling in cardiac hypertrophy. However, the role of PEX1 in transcriptional regulation of myocardial remodeling remains largely unknown. In the present study, we used PEX1 gain- and loss-of-function to examine the effects of PEX1 on left ventricular remodeling. Adenoviral constructs expressing PEX1, antisense PEX1, or LacZ were delivered by local injection into the anterior wall of the left ventricle in Sprague-Dawley rats. PEX1 overexpression led to induction of hypertrophic gene program and increased fibrosis. In agreement with this, the expression of genes involved in the fibrotic process, such as collagens I and III, matrix metalloproteinases (MMPs), fibronectin-1, transforming growth factor beta-1 and connective tissue growth factor, were significantly up-regulated following PEX1 overexpression, whereas silencing of PEX1 significantly inhibited the expression of pro-fibrotic genes and increased left ventricular ejection fraction and fractional shortening. In vitro luciferase reporter assays showed that PEX1 regulates the expression of MMP-9 by activating promoter. Furthermore, PEX1 gain- and loss-of-function experiments in rat neonatal cardiac fibroblasts and myocytes revealed that MMP-9 gene expression was affected by PEX1 predominantly in fibroblasts. Our results indicate that PEX1 is involved in regulating cardiac fibrosis and extracellular matrix turnover, particularly fibroblasts being responsible for the fibrosis-associated changes in gene expression. Furthermore, PEX1 activation of the MMP-9 promoter triggers the pro-fibrotic response directed by PEX1.


Asunto(s)
Matriz Extracelular/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Proteínas de la Membrana/metabolismo , Factores de Transcripción/metabolismo , Adenoviridae/metabolismo , Envejecimiento/metabolismo , Angiotensina II , Animales , Animales Recién Nacidos , Secuencia de Bases , Fibroblastos/metabolismo , Fibrosis , Regulación de la Expresión Génica , Silenciador del Gen , Técnicas de Transferencia de Gen , Hipertensión/complicaciones , Hipertensión/enzimología , Hipertensión/patología , Hipertensión/fisiopatología , Metaloproteinasa 9 de la Matriz/metabolismo , Modelos Biológicos , Infarto del Miocardio/complicaciones , Infarto del Miocardio/enzimología , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Regiones Promotoras Genéticas/genética , Ratas Sprague-Dawley
5.
PLoS One ; 10(12): e0144145, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26642209

RESUMEN

AIMS: Transcription factor GATA4 is a dosage sensitive regulator of heart development and alterations in its level or activity lead to congenital heart disease (CHD). GATA4 has also been implicated in cardiac regeneration and repair. GATA4 action involves combinatorial interaction with other cofactors such as NKX2-5, another critical cardiac regulator whose mutations also cause CHD. Despite its critical importance to the heart and its evolutionary conservation across species, the structural basis of the GATA4-NKX2-5 interaction remains incompletely understood. METHODS AND RESULTS: A homology model was constructed and used to identify surface amino acids important for the interaction of GATA4 and NKX2-5. These residues were subjected to site-directed mutagenesis, and the mutant proteins were characterized for their ability to bind DNA and to physically and functionally interact with NKX2-5. The studies identify 5 highly conserved amino acids in the second zinc finger (N272, R283, Q274, K299) and its C-terminal extension (R319) that are critical for physical and functional interaction with the third alpha helix of NKX2-5 homeodomain. Integration of the experimental data with computational modeling suggests that the structural arrangement of the zinc finger-homeodomain resembles the architecture of the conserved DNA binding domain of nuclear receptors. CONCLUSIONS: The results provide novel insight into the structural basis for protein-protein interactions between two important classes of transcription factors. The model proposed will help to elucidate the molecular basis for disease causing mutations in GATA4 and NKX2-5 and may be relevant to other members of the GATA and NK classes of transcription factors.


Asunto(s)
ADN/metabolismo , Factor de Transcripción GATA4/metabolismo , Cardiopatías Congénitas/metabolismo , Proteínas de Homeodominio/metabolismo , Modelos Biológicos , Mutación , Receptores Citoplasmáticos y Nucleares/metabolismo , Factores de Transcripción/metabolismo , Animales , Células COS , Chlorocebus aethiops , ADN/genética , Factor de Transcripción GATA4/genética , Cardiopatías Congénitas/genética , Proteína Homeótica Nkx-2.5 , Proteínas de Homeodominio/genética , Ratones , Unión Proteica , Estructura Terciaria de Proteína , Receptores Citoplasmáticos y Nucleares/genética , Factores de Transcripción/genética , Dedos de Zinc
7.
J Biol Chem ; 290(11): 6844-56, 2015 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-25623069

RESUMEN

TBX5 is the gene mutated in Holt-Oram syndrome, an autosomal dominant disorder with complex heart and limb deformities. Its protein product is a member of the T-box family of transcription factors and an evolutionarily conserved dosage-sensitive regulator of heart and limb development. Understanding TBX5 regulation is therefore of paramount importance. Here we uncover the existence of novel exons and provide evidence that TBX5 activity may be extensively regulated through alternative splicing to produce protein isoforms with differing N- and C-terminal domains. These isoforms are also present in human heart, indicative of an evolutionarily conserved regulatory mechanism. The newly identified isoforms have different transcriptional properties and can antagonize TBX5a target gene activation. Droplet Digital PCR as well as immunohistochemistry with isoform-specific antibodies reveal differential as well as overlapping expression domains. In particular, we find that the predominant isoform in skeletal myoblasts is Tbx5c, and we show that it is dramatically up-regulated in differentiating myotubes and is essential for myotube formation. Mechanistically, TBX5c antagonizes TBX5a activation of pro-proliferative signals such as IGF-1, FGF-10, and BMP4. The results provide new insight into Tbx5 regulation and function that will further our understanding of its role in health and disease. The finding of new exons in the Tbx5 locus may also be relevant to mutational screening especially in the 30% of Holt-Oram syndrome patients with no mutations in the known TBX5a exons.


Asunto(s)
Exones , Proteínas de Dominio T Box/análisis , Proteínas de Dominio T Box/genética , Anomalías Múltiples/genética , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , Expresión Génica , Cardiopatías Congénitas/genética , Defectos del Tabique Interatrial/genética , Humanos , Deformidades Congénitas de las Extremidades Inferiores/genética , Ratones , Datos de Secuencia Molecular , Células Musculares/citología , Células Musculares/metabolismo , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/metabolismo , Mutación , Miocardio/metabolismo , Miocardio/ultraestructura , Isoformas de Proteínas/análisis , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Alineación de Secuencia , Proteínas de Dominio T Box/metabolismo , Deformidades Congénitas de las Extremidades Superiores/genética
8.
Cell Mol Life Sci ; 72(10): 2005-22, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25504289

RESUMEN

The regulation of cardiac differentiation is critical for maintaining normal cardiac development and function. The precise mechanisms whereby cardiac differentiation is regulated remain uncertain. Here, we have identified a GATA-4 target, EGF, which is essential for cardiogenesis and regulates cardiac differentiation in a dose- and time-dependent manner. Moreover, EGF demonstrates functional interaction with GATA-4 in inducing the cardiac differentiation of P19CL6 cells in a time- and dose-dependent manner. Biochemically, GATA-4 forms a complex with STAT3 to bind to the EGF promoter in response to EGF stimulation and cooperatively activate the EGF promoter. Functionally, the cooperation during EGF activation results in the subsequent activation of cyclin D1 expression, which partly accounts for the lack of additional induction of cardiac differentiation by the GATA-4/STAT3 complex. Thus, we propose a model in which the regulatory cascade of cardiac differentiation involves GATA-4, EGF, and cyclin D1.


Asunto(s)
Diferenciación Celular/fisiología , Factor de Crecimiento Epidérmico/metabolismo , Factor de Transcripción GATA4/metabolismo , Corazón/embriología , Modelos Biológicos , Miocardio/citología , Transducción de Señal/fisiología , Animales , Western Blotting , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Técnicas Histológicas , Inmunoprecipitación , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Tiempo
9.
Eur J Heart Fail ; 16(4): 367-76, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24504921

RESUMEN

AIMS: Chemotherapy-induced heart failure is increasingly recognized as a major clinical challenge. Cardiotoxicity of imatinib mesylate, a highly selective and effective anticancer drug belonging to the new class of tyrosine kinase inhibitors, is being reported in patients, some progressing to congestive heart failure. This represents an unanticipated challenge that could limit effective drug use. Understanding the mechanisms and risk factors of imatinib mesylate cardiotoxicity is crucial for prevention of cardiovascular complications in cancer patients. METHODS AND RESULTS: We used genetically engineered mice and primary rat neonatal cardiomyocytes to analyse the action of imatinib on the heart. We found that treatment with imatinib (200 mg/kg/day for 5 weeks) leads to mitochondrial-dependent myocyte loss and cardiac dysfunction, as confirmed by electron microscopy, RNA analysis, and echocardiography. Imatinib cardiotoxicity was more severe in older mice, in part due to an age-dependent increase in oxidative stress. Mechanistically, depletion of the transcription factor GATA4 resulting in decreased levels of its prosurvival targets Bcl-2 and Bcl-XL was an underlying cause of imatinib toxicity. Consistent with this, GATA4 haploinsufficient mice were more susceptible to imatinib, and myocyte-specific up-regulation of GATA4 or Bcl-2 protected against drug-induced cardiotoxicity. CONCLUSION: The results indicate that imatinib action on the heart targets cardiomyocytes and involves mitochondrial impairment and cell death that can be further aggravated by oxidative stress. This in turn offers a possible explanation for the current conflicting data regarding imatinib cardiotoxicity in cancer patients and suggests that cardiac monitoring of older patients receiving imatinib therapy may be especially warranted.


Asunto(s)
Envejecimiento/fisiología , Benzamidas/toxicidad , Miocitos Cardíacos/efectos de los fármacos , Piperazinas/toxicidad , Inhibidores de Proteínas Quinasas/toxicidad , Pirimidinas/toxicidad , Disfunción Ventricular Izquierda/inducido químicamente , Animales , Cardiotoxicidad , Ecocardiografía , Factor de Transcripción GATA4/metabolismo , Mesilato de Imatinib , Etiquetado Corte-Fin in Situ , Ratones , Ratones Transgénicos , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/ultraestructura , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/ultraestructura , Estrés Oxidativo/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Factores de Riesgo , Disfunción Ventricular Izquierda/diagnóstico por imagen , Disfunción Ventricular Izquierda/metabolismo , Disfunción Ventricular Izquierda/patología , Proteína bcl-X/metabolismo
10.
Mol Cell Biol ; 32(12): 2214-23, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22473995

RESUMEN

Transcription factor GATA4 is a critical regulator of the embryonic and postnatal heart, but the mechanisms and cofactors required for its diverse functions are not fully understood. Here, we show that whereas the N-terminal domain of GATA4 is required for inducing cardiogenesis and for promoting postnatal cardiomyocyte survival, distinct residues and domains therein are necessary to mediate these effects. Cardiogenic activity of GATA4 requires a 24-amino-acid (aa) region (aa 129 to 152) which is needed for transcriptional synergy and physical interaction with BAF60c. The same region is not essential for induction of endoderm or blood cell markers by GATA4, suggesting that it acts as a cell-type-specific transcriptional activation domain. On the other hand, a serine residue at position 105, which is a known target for mitogen-activated protein kinase (MAPK) phosphorylation, is necessary for GATA4-dependent cardiac myocyte survival and hypertrophy but is entirely dispensable for GATA4-induced cardiogenesis. We find that S105 is differentially required for transcriptional synergy between GATA4 and serum response factor (SRF) but not other cardiac cofactors such as TBX5 and NKX2.5. The findings provide new insight into GATA4 mechanisms of action and suggest that distinct regulatory pathways regulate activities of GATA4 in embryonic development and postnatal hearts.


Asunto(s)
Factor de Transcripción GATA4 , Corazón/embriología , Miocitos Cardíacos , Proteínas de Xenopus , Animales , Aumento de la Célula , Supervivencia Celular , Células Cultivadas , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA4/metabolismo , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodominio/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Organogénesis , Ratas , Análisis de Secuencia , Serina , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriología , Xenopus laevis/metabolismo , Proteínas de Pez Cebra
11.
J Biol Chem ; 286(2): 1508-16, 2011 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-21051538

RESUMEN

Pressure and volume overload induce hypertrophic growth of postnatal cardiomyocytes and genetic reprogramming characterized by reactivation of a subset of fetal genes. Despite intense efforts, the nuclear effectors of cardiomyocyte hypertrophy remain incompletely defined. Endothelin-1 (ET-1) plays an important role in cardiomyocyte growth and is involved in mediating the neurohormonal effects of mechanical stress. Here, we show that the phenylephrine-induced complex-1 (PEX1), also known as zinc finger transcription factor ZFP260, is essential for cardiomyocyte response to ET-1 as evidenced in cardiomyocytes with PEX1 knockdown. We found that ET-1 enhances PEX1 transcriptional activity via a PKC-dependent pathway which phosphorylates the protein and further potentiates its synergy with GATA4. Consistent with a role for PEX1 in cardiomyocyte hypertrophy, overexpression of PEX1 is sufficient to induce cardiomyocyte hypertrophy in vitro and in vivo. Importantly, transgenic mice with inducible PEX1 expression in the adult heart develop cardiac hypertrophy with preserved heart function. Together, the results identify a novel nuclear effector of ET-1 signaling and suggest that PEX1 may be a regulator of the early stages of cardiac hypertrophy.


Asunto(s)
Cardiomegalia , Endotelina-1/metabolismo , Transducción de Señal/fisiología , Transactivadores/genética , Transactivadores/metabolismo , Factores de Edad , Animales , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatología , Células Cultivadas , Ratones , Ratones Transgénicos , Miocitos Cardíacos/citología , Fosforilación/fisiología , Procesamiento Proteico-Postraduccional/fisiología , Ratas , Transfección
12.
J Cell Sci ; 120(Pt 3): 407-16, 2007 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17213336

RESUMEN

In L6 skeletal myoblasts induced to differentiate by Arg8-vasopressin treatment, a short-lived lowering of ceramide levels was observed, followed by a long-lasting elevation that was prevented by inhibitors of the de novo synthesis pathway, fumonisin B1 and myriocin. Both inhibitors increased the expression of myogenic differentiation markers and cell fusion rate, whereas short-chain ceramides inhibited these responses. Similar drug effects were observed on primary mouse satellite cell differentiation. Furthermore, bacterial sphingomyelinase overexpression suppressed myogenin nuclear accumulation in L6 cells. These data suggested that endogenous ceramide mediates a negative feedback mechanism limiting myogenic differentiation, and that inhibitors of ceramide synthesis promoted myogenesis by removing this control. Phospholipase D (PLD), a recognized target of ceramide, is required for myogenesis, as shown by the negative effects of PLD1 isoform depletion obtained by siRNA treatment. Fumonisin induced an increase in PLD activity of L6 cells, whereas C6-ceramide decreased it. The expression of PLD1 mRNA transcripts was selectively decreased by C6-ceramide, and increased by ceramide synthesis inhibitors. An early step of myogenic response is the PLD1-dependent formation of actin stress fiber-like structures. C6-ceramide addition or overexpression of sphingomyelinase impaired actin fiber formation. Ceramide might thus regulate myogenesis through downregulation of PLD1 expression and activity.


Asunto(s)
Ceramidas/antagonistas & inhibidores , Músculo Esquelético/metabolismo , Fosfolipasa D/fisiología , Animales , Diferenciación Celular , Línea Celular , Ceramidas/biosíntesis , Ceramidas/fisiología , Células Clonales , Fosfolipasa D/metabolismo , ARN Mensajero/metabolismo , Ratas , Regulación hacia Arriba
13.
FEBS Lett ; 580(26): 6224-32, 2006 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-17069807

RESUMEN

Tumor necrosis factor alpha (TNFalpha), a pleiotropic cytokine, activates both apoptotic and pro-survival signals depending on the cell model. Using ECV304 cells, which can be made TNFalpha-sensitive by cycloheximide (CHX) co-treatment, we evaluated the potential roles of ceramide and phospholipase D (PLD) in TNFalpha-induced apoptosis. TNFalpha/CHX induced a robust increase in ceramide levels after 16 h of treatment when cell death was maximal. PLD activity was increased at early time point (1h) whereas both PLD activity and PLD1 protein were strongly decreased after 24h. TNFalpha/CHX-induced cell death was significantly lowered by exogenous bacterial PLD and phoshatidic acid, and in cells overexpressing PLD1. Conversely, cells depleted in PLD proteins by small interference RNA (siRNA) treatment exhibited higher susceptibility to apoptosis. These results show that PLD exerts a protective role against TNFalpha-induced cell death.


Asunto(s)
Apoptosis/efectos de los fármacos , Endotelio Vascular/citología , Fosfolipasa D/fisiología , Factor de Necrosis Tumoral alfa/farmacología , Línea Celular , Ceramidas/genética , Cicloheximida/farmacología , Humanos , Fosfolipasa D/análisis , Fosfolipasa D/genética , Sustancias Protectoras , ARN Interferente Pequeño/farmacología , Transfección
14.
Mol Cell Biol ; 25(22): 9829-44, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16260600

RESUMEN

Angiotensin II (AII), a potent vasoactive hormone, acts on numerous organs via G-protein-coupled receptors and elicits cell-specific responses. At the level of the heart, AII stimulation alters gene transcription and leads to cardiomyocyte hypertrophy. Numerous intracellular signaling pathways are activated in this process; however, which of these directly link receptor activation to transcriptional regulation remains undefined. We used the atrial natriuretic factor (ANF) gene (NPPA) as a marker to elucidate the signaling cascades involved in AII transcriptional responses. We show that ANF transcription is activated directly by the AII type 1 receptor and precedes the development of myocyte hypertrophy. This response maps to STAT and GATA binding sites, and the two elements transcriptionally cooperate to mediate signaling through the JAK-STAT and protein kinase C (PKC)-GATA-4 pathways. PKC phosphorylation enhances GATA-4 DNA binding activity, and STAT-1 functionally and physically interacts with GATA-4 to synergistically activate AII and other growth factor-inducible promoters. Moreover, GATA factors are able to recruit STAT proteins to target promoters via GATA binding sites, which are sufficient to support synergy. Thus, STAT proteins can act as growth factor-inducible coactivators of tissue-specific transcription factors. Interactions between STAT and GATA proteins may provide a general paradigm for understanding cell specificity of cytokine and growth factor signaling.


Asunto(s)
Factor de Transcripción GATA4/fisiología , Regulación de la Expresión Génica , Proteína Quinasa C/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Factores de Transcripción STAT/metabolismo , Adenoviridae/genética , Angiotensina II/metabolismo , Animales , Factor Natriurético Atrial/metabolismo , Sitios de Unión , Northern Blotting , Línea Celular , Núcleo Celular/metabolismo , Proliferación Celular , Inmunoprecipitación de Cromatina , Citocinas/metabolismo , ADN/metabolismo , Inhibidores Enzimáticos/farmacología , Humanos , Inmunoprecipitación , Ratones , Modelos Genéticos , Miocitos Cardíacos/metabolismo , Células 3T3 NIH , Fosforilación , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Ratas , Transducción de Señal , Transcripción Genética , Activación Transcripcional , Fosfolipasas de Tipo C/metabolismo
15.
Mol Biol Cell ; 16(3): 1232-44, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15616193

RESUMEN

We investigated the role of phospholipase D (PLD) and its product phosphatidic acid (PA) in myogenic differentiation of cultured L6 rat skeletal myoblasts. Arginine-vasopressin (AVP), a differentiation inducer, rapidly activated PLD in a Rho-dependent way, as shown by almost total suppression of activation by C3 exotoxin pretreatment. Addition of 1-butanol, which selectively inhibits PA production by PLD, markedly decreased AVP-induced myogenesis. Conversely, myogenesis was potentiated by PLD1b isoform overexpression but not by PLD2 overexpression, establishing that PLD1 is involved in this process. The expression of the PLD isoforms was differentially regulated during differentiation. AVP stimulation of myoblasts induced the rapid formation of stress fiber-like actin structures (SFLSs). 1-Butanol selectively inhibited this response, whereas PLD1b overexpression induced SFLS formation, showing that it was PLD dependent. Endogenous PLD1 was located at the level of SFLSs, and by means of an intracellularly expressed fluorescent probe, PA was shown to be accumulated along these structures in response to AVP. In addition, AVP induced a PLD-dependent neosynthesis of phosphatidylinositol 4,5-bisphosphate (PIP2), which also was accumulated along actin fibers. These data support the hypothesis that PLD participates in myogenesis through PA- and PIP2-dependent actin fiber formation.


Asunto(s)
Actinas/química , Citoesqueleto/metabolismo , Músculos/citología , Fosfolipasa D/fisiología , 1-Butanol/química , Actinas/metabolismo , Animales , Arginina Vasopresina/química , Diferenciación Celular , Línea Celular , Núcleo Celular/metabolismo , Células Cultivadas , Regulación hacia Abajo , Immunoblotting , Microscopía Fluorescente , Músculo Esquelético/citología , Miogenina/metabolismo , Faloidina/farmacología , Fenotipo , Ácidos Fosfatidicos/química , Fosfatidilinositol 4,5-Difosfato/química , Fosfolipasa D/metabolismo , Plásmidos/metabolismo , Isoformas de Proteínas , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Transfección , Proteína de Unión al GTP rhoA/metabolismo
16.
FEBS Lett ; 577(3): 409-14, 2004 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-15556619

RESUMEN

TPA, a potent PKC activator, inhibits myogenic differentiation and activates phospholipase D (PLD). We evaluated the involvement of PLD in the TPA effects on L6 myoblasts differentiation. TPA, at concentrations inhibiting differentiation of L6 cells, induced a strong, though transient, PLD activation. Surprisingly, at nanomolar concentration, TPA induced both myogenic differentiation and sustained activation of PLD. Differential effect of TPA can be ascribed to PKC downregulation induced by highest TPA concentrations. TPA-induced differentiation was inhibited by 1-butanol, confirming the involvement of PLD in this effect. These data suggest that prolonged elevation of PLD activity is required for myogenic differentiation.


Asunto(s)
Carcinógenos/farmacología , Diferenciación Celular/efectos de los fármacos , Mioblastos/efectos de los fármacos , Ésteres del Forbol/farmacología , Fosfolipasa D/metabolismo , 1-Butanol/farmacología , Actinas/metabolismo , Animales , Línea Celular , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Activación Enzimática , Cinética , Miogenina/metabolismo , Miosinas/metabolismo , Proteína Quinasa C/metabolismo , Ratas , Acetato de Tetradecanoilforbol/farmacología
17.
J Biol Chem ; 278(49): 49308-15, 2003 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-14506285

RESUMEN

We have previously shown that myogenesis induction by Arg8-vasopressin (AVP) in L6 rat myoblasts involves a sustained stimulation of type 4 cAMP-phosphodiesterase. In this model, we observed that a transient cAMP generation occurs in the minutes following AVP addition. Evidence suggests that cAMP generation is due to the prostaglandins produced in response to AVP binding to V1a receptors and subsequent activation of phospholipase A2. The early cAMP increase was effective in activating cAMP-dependent protein kinase (PKA) and increasing phosphorylation of CREB transcription factor. Inhibition of PKA by compound H89 prior to AVP addition led to a significant reduction of expression of the differentiation marker creatine kinase, whereas H89 added 1-5 h after AVP had no significant effect. Furthermore, PKA inhibition 24 h after the beginning of AVP treatment potentiated differentiation. This shows that both an early activation and a later down-regulation of the cAMP pathway are required for AVP induction of myogenesis. Because phosphodiesterase PDE4D3 overexpressed in L6 cells lost its ability to potentiate AVP-induced differentiation when mutated and rendered insensitive to PKA phosphorylation and activation, we hypothesize that the early cAMP increase is required to trigger the down-regulation of cAMP pathway through stimulation of phosphodiesterase.


Asunto(s)
Diferenciación Celular , AMP Cíclico/metabolismo , Mioblastos/citología , Sulfonamidas , Animales , Arginina Vasopresina/farmacología , Secuencia de Bases , Línea Celular , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Cartilla de ADN , Inhibidores Enzimáticos/farmacología , Isoquinolinas/farmacología , Mioblastos/enzimología , Mioblastos/metabolismo , Ratas , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA