Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Neurochem Int ; 147: 105051, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33979572

RESUMEN

Glioblastoma (GBM) is a highly aggressive brain tumor. During screening work, we found a new compound named phragmunis A (PGA), which is derived from the fruitbody of Trogia venenata, exhibits a potential cytotoxic effect on patient-derived recurrent GBM cells and temozolomide (TMZ)-resistant cell lines. The present study was designed to investigate the potential molecular mechanism of the anti-glioma effects of PGA in vitro and in vivo. Studies investigating the mechanism revealed that PGA diminished the binding efficiency of ETS family of transcription factor (ELK1) and Serum response factor (SRF), and suppressed ELK1-SRF complex-dependent transcription, which decreased the transcriptional levels of downstream genes Early growth response protein 1 (EGR1)-Polycomb ring finger (BMI1), thus inducing the imbalanced regulation between Myeloid cell leukaemia-1 (MCL1) and F-Box and WD repeat domain containing 7 (FBXW7). Finally, orthotopic xenograft models were established to confirm the anti-glioma effect of PGA on tumour growth. We showed, for the first time, that the cytotoxic effects of PGA occurred by inducing MCL1 inhibition and FBXW7 activation by blocking ELK1-SRF complex-dependent transcription. The blockage of ELK1-mediated transcription resulted in the suppression of EGR1-BMI1, which led to the upregulation of FBXW7 expression and downregulation of MCL1. These findings suggested that PGA could be a therapeutic drug candidate for the treatment of recurrent GBM by targeting the ELK1-SRF complex.


Asunto(s)
Proteína 7 que Contiene Repeticiones F-Box-WD/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Glioblastoma/tratamiento farmacológico , Extractos Vegetales/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Proteína 7 que Contiene Repeticiones F-Box-WD/metabolismo , Regulación de la Expresión Génica/fisiología , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/efectos de los fármacos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Factor de Respuesta Sérica/efectos de los fármacos , Factor de Respuesta Sérica/metabolismo , Proteína Elk-1 con Dominio ets/efectos de los fármacos , Proteína Elk-1 con Dominio ets/metabolismo
2.
J Cachexia Sarcopenia Muscle ; 11(1): 169-194, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31647200

RESUMEN

BACKGROUND: Myopalladin (MYPN) is a striated muscle-specific, immunoglobulin-containing protein located in the Z-line and I-band of the sarcomere as well as the nucleus. Heterozygous MYPN gene mutations are associated with hypertrophic, dilated, and restrictive cardiomyopathy, and homozygous loss-of-function truncating mutations have recently been identified in patients with cap myopathy, nemaline myopathy, and congenital myopathy with hanging big toe. METHODS: Constitutive MYPN knockout (MKO) mice were generated, and the role of MYPN in skeletal muscle was studied through molecular, cellular, biochemical, structural, biomechanical, and physiological studies in vivo and in vitro. RESULTS: MKO mice were 13% smaller compared with wild-type controls and exhibited a 48% reduction in myofibre cross-sectional area (CSA) and significantly increased fibre number. Similarly, reduced myotube width was observed in MKO primary myoblast cultures. Biomechanical studies showed reduced isometric force and power output in MKO mice as a result of the reduced CSA, whereas the force developed by each myosin molecular motor was unaffected. While the performance by treadmill running was similar in MKO and wild-type mice, MKO mice showed progressively decreased exercise capability, Z-line damage, and signs of muscle regeneration following consecutive days of downhill running. Additionally, MKO muscle exhibited progressive Z-line widening starting from 8 months of age. RNA-sequencing analysis revealed down-regulation of serum response factor (SRF)-target genes in muscles from postnatal MKO mice, important for muscle growth and differentiation. The SRF pathway is regulated by actin dynamics as binding of globular actin to the SRF-cofactor myocardin-related transcription factor A (MRTF-A) prevents its translocation to the nucleus where it binds and activates SRF. MYPN was found to bind and bundle filamentous actin as well as interact with MRTF-A. In particular, while MYPN reduced actin polymerization, it strongly inhibited actin depolymerization and consequently increased MRTF-A-mediated activation of SRF signalling in myogenic cells. Reduced myotube width in MKO primary myoblast cultures was rescued by transduction with constitutive active SRF, demonstrating that MYPN promotes skeletal muscle growth through activation of the SRF pathway. CONCLUSIONS: Myopalladin plays a critical role in the control of skeletal muscle growth through its effect on actin dynamics and consequently the SRF pathway. In addition, MYPN is important for the maintenance of Z-line integrity during exercise and aging. These results suggest that muscle weakness in patients with biallelic MYPN mutations may be associated with reduced myofibre CSA and SRF signalling and that the disease phenotype may be aggravated by exercise.


Asunto(s)
Proteínas Musculares/uso terapéutico , Músculo Esquelético/efectos de los fármacos , Factor de Respuesta Sérica/efectos de los fármacos , Animales , Femenino , Humanos , Ratones , Ratones Noqueados , Proteínas Musculares/farmacología
3.
Mol Psychiatry ; 23(6): 1474-1486, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-28555077

RESUMEN

The nucleus accumbens (NAc) is a primary brain reward region composed predominantly of medium spiny neurons (MSNs). In response to early withdrawal from repeated cocaine administration, de novo dendritic spine formation occurs in NAc MSNs. Much evidence indicates that this new spine formation facilitates the rewarding properties of cocaine. Early withdrawal from repeated cocaine also produces dramatic alterations in the transcriptome of NAc MSNs, but how such alterations influence cocaine's effects on dendritic spine formation remain unclear. Studies in non-neuronal cells indicate that actin cytoskeletal regulatory pathways in nuclei have a direct role in the regulation of gene transcription in part by controlling the access of co-activators to their transcription factor partners. In particular, actin state dictates the interaction between the serum response factor (SRF) transcription factor and one of its principal co-activators, MAL. Here we show that cocaine induces alterations in nuclear F-actin signaling pathways in the NAc with associated changes in the nuclear subcellular localization of SRF and MAL. Using in vivo optogenetics, the brain region-specific inputs to the NAc that mediate these nuclear changes are investigated. Finally, we demonstrate that regulated SRF expression, in turn, is critical for the effects of cocaine on dendritic spine formation and for cocaine-mediated behavioral sensitization. Collectively, these findings reveal a mechanism by which nuclear-based changes influence the structure of NAc MSNs in response to cocaine.


Asunto(s)
Trastornos Relacionados con Cocaína/metabolismo , Espinas Dendríticas/efectos de los fármacos , Factor de Respuesta Sérica/efectos de los fármacos , Actinas/efectos de los fármacos , Animales , Cocaína/efectos adversos , Cocaína/farmacología , Dendritas/efectos de los fármacos , Dendritas/metabolismo , Espinas Dendríticas/metabolismo , Inhibidores de Captación de Dopamina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs , Proteínas Proteolipídicas Asociadas a Mielina y Linfocito/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Neuronas/metabolismo , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Recompensa , Transducción de Señal/efectos de los fármacos
4.
Am J Physiol Heart Circ Physiol ; 311(4): H904-H912, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27521420

RESUMEN

Homeostatic control of vascular smooth muscle cell (VSMC) differentiation is critical for contractile activity and regulation of blood flow. Recently, we reported that precontracted blood vessels are relaxed and the phenotype of VSMC is regulated from a synthetic to contractile state by glucose-6-phosphate dehydrogenase (G6PD) inhibition. In the current study, we investigated whether the increase in the expression of VSMC contractile proteins by inhibition and knockdown of G6PD is mediated through a protein kinase G (PKG)-dependent pathway and whether it regulates blood pressure. We found that the expression of VSMC-restricted contractile proteins, myocardin (MYOCD), and miR-1 and miR-143 are increased by G6PD inhibition or knockdown. Importantly, RNA-sequence analysis of aortic tissue from G6PD-deficient mice revealed uniform increases in VSMC-restricted genes, particularly those regulated by the MYOCD-serum response factor (SRF) switch. Conversely, expression of Krüppel-like factor 4 (KLF4) is decreased by G6PD inhibition. Interestingly, the G6PD inhibition-induced expression of miR-1 and contractile proteins was blocked by Rp-ß-phenyl-1,N2-etheno-8-bromo-guanosine-3',5'-cyclic monophosphorothioate, a PKG inhibitor. On the other hand, MYOCD and miR-143 levels are increased by G6PD inhibition through a PKG-independent manner. Furthermore, blood pressure was lower in the G6PD-deficient compared with wild-type mice. Therefore, our results suggest that the expression of VSMC contractile proteins induced by G6PD inhibition occurs via PKG1α-dependent and -independent pathways.


Asunto(s)
Aorta/metabolismo , Proteínas Contráctiles/genética , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , Glucosafosfato Deshidrogenasa/antagonistas & inhibidores , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Animales , Aorta/efectos de los fármacos , Western Blotting , Bovinos , Cromatografía Liquida , Proteínas Contráctiles/efectos de los fármacos , Proteínas Contráctiles/metabolismo , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/antagonistas & inhibidores , Proteínas Quinasas Dependientes de GMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Técnicas de Silenciamiento del Gen , Glucosafosfato Deshidrogenasa/genética , Inmunoprecipitación , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/efectos de los fármacos , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , MicroARNs/efectos de los fármacos , MicroARNs/genética , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Proteínas Nucleares/efectos de los fármacos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Reacción en Cadena de la Polimerasa , Ratas , Factor de Respuesta Sérica/efectos de los fármacos , Factor de Respuesta Sérica/genética , Factor de Respuesta Sérica/metabolismo , Espectrometría de Masas en Tándem , Transactivadores/efectos de los fármacos , Transactivadores/genética , Transactivadores/metabolismo
5.
Diab Vasc Dis Res ; 13(1): 81-92, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26408645

RESUMEN

PURPOSE: To investigate the expression and function of serum response factor in podocyte epithelial-mesenchymal transition of diabetic nephropathy. METHODS: The expression of serum response factor, epithelial markers and mesenchymal markers was examined in podocytes or renal cortex tissues following high glucose. Serum response factor was upregulated by its plasmids and downregulated by CCG-1423 to investigate how it influenced podocyte epithelial-mesenchymal transition in diabetic nephropathy. Streptozotocin was used to generate diabetes mellitus in rats. RESULTS: In podocytes after high glucose treatment, serum response factor and mesenchymal markers increased, while epithelial markers declined. Similar changes were observed in vivo. Serum response factor overexpression in podocytes induced expression of Snail, an important transcription factor mediating epithelial-mesenchymal transition. Blockade of serum response factor reduced Snail induction, protected podocytes from epithelial-mesenchymal transition and ameliorated proteinuria. CONCLUSION: Together, increased serum response factor activity provokes podocytes' epithelial-mesenchymal transition and dysfunction in diabetic nephropathy. Targeting serum response factor by small-molecule inhibitor may be an attractive therapeutic strategy for diabetic nephropathy.


Asunto(s)
Proteínas de Arabidopsis/efectos de los fármacos , Diabetes Mellitus Experimental/metabolismo , Nefropatías Diabéticas/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Glucosa/farmacología , Transferasas Intramoleculares/efectos de los fármacos , Riñón/patología , Podocitos/efectos de los fármacos , Factor de Respuesta Sérica/efectos de los fármacos , Edulcorantes/farmacología , Actinas/efectos de los fármacos , Actinas/genética , Anilidas/farmacología , Animales , Proteínas de Arabidopsis/metabolismo , Benzamidas/farmacología , Línea Celular , Nefropatías Diabéticas/patología , Fibronectinas , Inmunohistoquímica , Transferasas Intramoleculares/metabolismo , Masculino , Ratones , Podocitos/metabolismo , Podocitos/patología , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína de Unión al Calcio S100A4 , Proteínas S100/efectos de los fármacos , Proteínas S100/genética , Factor de Respuesta Sérica/genética , Factor de Respuesta Sérica/metabolismo , Factores de Transcripción de la Familia Snail , Transactivadores/efectos de los fármacos , Transactivadores/genética , Factores de Transcripción/metabolismo , Proteína de la Zonula Occludens-1/efectos de los fármacos , Proteína de la Zonula Occludens-1/genética
6.
Cell Signal ; 21(4): 551-8, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19135527

RESUMEN

The mitogenic Pasteurella multocida toxin (PMT) is a major virulence factor of P. multocida, which causes Pasteurellosis in man and animals. The toxin activates the small GTPase RhoA, the MAP kinase ERK and STAT proteins via the stimulation of members of two G protein families, G(q) and G(12/13). PMT action also results in an increase in inositol phosphates, which is due to the stimulation of PLCbeta via Galpha(q). Recent studies indicate that PMT additionally activates Galpha(i) to inhibit adenylyl cyclase. Here we show that PMT acts not only via Galpha but also through Gbetagamma signaling. Activation of Gbetagamma by PMT causes stimulation of phosphoinositide 3-kinase (PI3K) gamma and formation of phosphatidylinositol-3,4,5-trisphosphate (PIP(3)) as indicated by the recruitment of a PIP(3)-binding pleckstrin homology (PH) domain-containing protein to the plasma membrane. Moreover, it is demonstrated that Gbetagamma is necessary for PMT-induced signaling via Galpha. Mutants of Galpha(q) incapable of binding or releasing Gbetagamma are not activated by PMT. Similarly, sequestration of Gbetagamma inhibits PMT-induced Galpha-signaling.


Asunto(s)
Proteínas Bacterianas/farmacología , Toxinas Bacterianas/farmacología , Proteínas de Unión al GTP Heterotriméricas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Sustitución de Aminoácidos , Carbacol/farmacología , Línea Celular , Activación Enzimática/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Proteínas del Ojo/farmacología , Reguladores de Proteínas de Unión al GTP/farmacología , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/antagonistas & inhibidores , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Humanos , Mutagénesis Sitio-Dirigida , Mutación Missense , Péptidos Cíclicos/farmacología , Toxina del Pertussis/farmacología , Fosfatos de Fosfatidilinositol/biosíntesis , Fosfoproteínas/farmacología , Mutación Puntual , Transporte de Proteínas/efectos de los fármacos , Receptor Muscarínico M3/efectos de los fármacos , Receptor Muscarínico M3/metabolismo , Factor de Respuesta Sérica/efectos de los fármacos
7.
Exp Cell Res ; 287(1): 106-15, 2003 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-12799187

RESUMEN

Rho proteins (Rho, Rac, Cdc 42) are known to control the organization of the actin cytoskeleton as well as gene expression. Inhibition of Rho proteins by Clostridium difficile toxin B disrupted the F-actin cytoskeleton and enhanced cytokine-induced inducible nitric oxide synthase (iNOS) expression in human epithelial cells. Also specific inhibition by Y-27632 of p160ROCK, which mediates Rho effects on actin fibers, caused a disruption of the actin cytoskeleton and a superinduction of cytokine-induced iNOS expression. Accordingly, direct disruption of the actin cytoskeleton by cytochalasin D, latrunculin B, or jasplakinolide enhanced cytokine-induced iNOS expression. The transcription factor serum response factor (SRF) has been described as mediating actin cytoskeleton-dependent regulation of gene expression. Direct targets of SRF are activating protein 1 (AP1)-dependent genes. All compounds used inhibited SRF- and AP1-dependent reporter gene expression in DLD-1 cells. However, the enhancing effect of the actin cytoskeleton-disrupting compounds on human iNOS promoter activity was much less pronounced than the effect on iNOS mRNA expression. Therefore, besides transcriptional mechanisms, posttranscriptional effects seem to be involved in the regulation of iNOS expression by the above compounds. In conclusion, our data suggest that Rho protein-mediated changes of the actin cytoskeleton negatively modulate the expression of human iNOS.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Proteínas Bacterianas , Citoesqueleto/metabolismo , Depsipéptidos , Células Eucariotas/enzimología , Regulación Enzimológica de la Expresión Génica/genética , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Citoesqueleto de Actina/efectos de los fármacos , Toxinas Bacterianas/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Citocalasina D/farmacología , Citocinas/metabolismo , Citocinas/farmacología , Citoesqueleto/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intracelular , Óxido Nítrico Sintasa/efectos de los fármacos , Óxido Nítrico Sintasa/genética , Péptidos Cíclicos/farmacología , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Factor de Respuesta Sérica/efectos de los fármacos , Factor de Respuesta Sérica/genética , Factor de Respuesta Sérica/metabolismo , Tiazoles/farmacología , Tiazolidinas , Factor de Transcripción AP-1/efectos de los fármacos , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Transcripción Genética/efectos de los fármacos , Transcripción Genética/genética , Células Tumorales Cultivadas , Proteínas de Unión al GTP rho/efectos de los fármacos , Quinasas Asociadas a rho
8.
Am J Physiol Cell Physiol ; 285(2): C370-6, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12711593

RESUMEN

Emerging evidence suggests that tumor necrosis factor (TNF)-alpha plays a role in muscle repair. To determine whether TNF-alpha modulates satellite cell proliferation, the current study evaluated TNF-alpha effects on DNA synthesis in primary myoblasts and on satellite cell activation in adult mouse muscle. Exposure to recombinant TNF-alpha increased total DNA content in rat primary myoblasts dose-dependently over a 24-h period and increased the number of primary myoblasts incorporating 5-bromo-2'-deoxyuridine (BrdU) during a 30-min pulse labeling. Systemic injection of TNF-alpha stimulated BrdU incorporation by satellite cells in muscles of adult mice, whereas no BrdU was incorporated by satellite cells in control mice. TNF-alpha stimulated serum response factor (SRF) binding to the serum response element (SRE) present in the c-fos gene promoter and stimulated reporter gene expression controlled by the same element. Our data suggest that TNF-alpha activates satellite cells to enter the cell cycle and accelerates G1-to-S phase transition, and these actions may involve activation of early response genes via SRF.


Asunto(s)
División Celular/fisiología , Músculo Esquelético/metabolismo , Mioblastos/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Factor de Respuesta Sérica/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Animales Recién Nacidos , División Celular/efectos de los fármacos , Células Cultivadas , ADN/biosíntesis , ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Genes Reporteros/efectos de los fármacos , Genes Reporteros/genética , Masculino , Ratones , Ratones Endogámicos ICR , Músculo Esquelético/efectos de los fármacos , Mioblastos/efectos de los fármacos , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-fos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/genética , Ratas , Regeneración/efectos de los fármacos , Regeneración/fisiología , Células Satélite del Músculo Esquelético/efectos de los fármacos , Elemento de Respuesta al Suero/efectos de los fármacos , Elemento de Respuesta al Suero/genética , Factor de Respuesta Sérica/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología
9.
Am J Respir Cell Mol Biol ; 29(1): 39-47, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12600823

RESUMEN

RhoA and its downstream target Rho kinase regulate serum response factor (SRF)-dependent skeletal and smooth muscle gene expression. We previously reported that long-term serum deprivation reduces transcription of smooth muscle contractile apparatus encoding genes, by redistributing SRF out of the nucleus. Because serum components stimulate RhoA activity, these observations suggest the hypothesis that the RhoA/Rho kinase pathway regulates SRF-dependent smooth muscle gene transcription in part by controlling SRF subcellular localization. Our present results support this hypothesis: cotransfection of cultured airway myocytes with a plasmid expressing constitutively active RhoAV14 selectively enhanced transcription from the SM22 and smooth muscle myosin heavy chain promoters and from a purely SRF-dependent promoter, but had no effect on transcription from the MSV-LTR promoter or from an AP2-dependent promoter. Conversely, inhibition of the RhoA/Rho kinase pathway by cotransfection with a plasmid expressing dominant negative RhoAN19, by cotransfection with a plasmid expressing Clostridial C3 toxin, or by incubation with the Rho kinase inhibitor, Y-27632, all selectively reduced SRF-dependent smooth muscle promoter activity. Furthermore, treatment with Y-27632 selectively reduced binding of SRF from nuclear extracts to its consensus DNA target, selectively reduced nuclear SRF protein content, and partially redistributed SRF from nucleus to cytoplasm, as revealed by quantitative immunocytochemistry. Treatment of cultured airway myocytes with latrunculin B, which reduces actin polymerization, also caused partial redistribution of SRF into the cytoplasm. Together, these results demonstrate for the first time that the RhoA/Rho kinase pathway controls smooth muscle gene transcription in differentiated smooth muscle cells, in part by regulating the subcellular localization of SRF. It is conceivable that the RhoA/Rho kinase pathway influences SRF localization through its effect on actin polymerization dynamics.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Factor de Respuesta Sérica/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Amidas/farmacología , Animales , Toxinas Bacterianas/genética , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Núcleo Celular/efectos de los fármacos , Células Cultivadas , Perros , Inhibidores Enzimáticos/farmacología , Péptidos y Proteínas de Señalización Intracelular , Células Musculares/citología , Células Musculares/metabolismo , Músculo Liso/fisiología , Cadenas Pesadas de Miosina/genética , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Piridinas/farmacología , Factor de Respuesta Sérica/efectos de los fármacos , Factor de Respuesta Sérica/genética , Transducción de Señal , Tiazoles/farmacología , Tiazolidinas , Tráquea/citología , Transcripción Genética , Quinasas Asociadas a rho , Proteína de Unión al GTP rhoA/genética
10.
Mol Endocrinol ; 17(4): 520-33, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12554779

RESUMEN

Early growth response factor (Egr-1) is an inducible zinc finger transcription factor that binds specific GC-rich enhancer elements and impacts female reproduction. These studies document for the first time that FSH rapidly induces Egr-1 expression in granulosa cells of small growing follicles. This response is transient but is reinitiated in preovulatory follicles exposed to the LH analog, human chorionic gonadotropin. Immunohistochemical analysis also showed gonadotropin induced Egr-1 in theca cells. The Egr-1 gene regulatory region responsive to gonadotropin signaling was localized within -164 bp of the transcription initiation site. Binding of Sp1/Sp3 to a proximal GC-box at -64/-46 bp was enhanced by FSH in immature granulosa cells but reduced after human chorionic gonadotropin stimulation of preovulatory follicles despite constant protein expression. This dynamic regulation of Sp1 binding was dependent on gonadotropin-regulated mechanisms that modulate Sp1/3-DNA binding activity. Serum response factor was active in granulosa cells and bound a consensus CArG-box/serum response element site, whereas two putative cAMP response elements within the -164-bp region bound cAMP regulatory element (CRE) binding protein (CREB) and a second cAMP-inducible protein immunologically related to CREB. Transient transfection analyses using Egr-1 promoter-luciferase constructs and site-specific mutations show that the serum response element, GC-box, and CRE-131 are involved in gonadotropin regulation of Egr-1 expression in granulosa cells. Specific kinase inhibitors of Erk or protein kinase A antagonized this induction while exogenously expressed Egr-1 enhanced reporter expression. These observations indicate that the Egr-1 gene is a target of both FSH and LH action that may mediate molecular programs of proliferation and/or differentiation during follicle growth, ovulation, and luteinization.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Hormona Folículo Estimulante/farmacología , Células de la Granulosa/fisiología , Proteínas Inmediatas-Precoces , Hormona Luteinizante/farmacología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales , Sitios de Unión , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Células Cultivadas , AMP Cíclico/farmacología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/efectos de los fármacos , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas de Unión al ADN/efectos de los fármacos , Proteína 1 de la Respuesta de Crecimiento Precoz , Femenino , Hormona Folículo Estimulante/metabolismo , Células de la Granulosa/efectos de los fármacos , Hipofisectomía , Hormona Luteinizante/metabolismo , Ratones , Ratones Mutantes , Ovario/efectos de los fármacos , Ovario/fisiología , Regiones Promotoras Genéticas , Ratas , Elementos de Respuesta , Factor de Respuesta Sérica/efectos de los fármacos , Factor de Respuesta Sérica/genética , Factor de Respuesta Sérica/metabolismo , Factor de Transcripción Sp1/efectos de los fármacos , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo , Factor de Transcripción Sp3 , Acetato de Tetradecanoilforbol/farmacología , Factores de Transcripción/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...