Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS One ; 16(8): e0252282, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34358226

RESUMEN

Epilepsy is a complex neurological condition characterized by repeated spontaneous seizures and can be induced by initiating seizures known as status epilepticus (SE). Elaborating the critical molecular mechanisms following SE are central to understanding the establishment of chronic seizures. Here, we identify a transient program of molecular and metabolic signaling in the early epileptogenic period, centered on day five following SE in the pre-clinical kainate or pilocarpine models of temporal lobe epilepsy. Our work now elaborates a new molecular mechanism centered around Wnt signaling and a growing network comprised of metabolic reprogramming and mTOR activation. Biochemical, metabolomic, confocal microscopy and mouse genetics experiments all demonstrate coordinated activation of Wnt signaling, predominantly in neurons, and the ensuing induction of an overall aerobic glycolysis (Warburg-like phenomenon) and an altered TCA cycle in early epileptogenesis. A centerpiece of the mechanism is the regulation of pyruvate dehydrogenase (PDH) through its kinase and Wnt target genes PDK4. Intriguingly, PDH is a central gene in certain genetic epilepsies, underscoring the relevance of our elaborated mechanisms. While sharing some features with cancers, the Warburg-like metabolism in early epileptogenesis is uniquely split between neurons and astrocytes to achieve an overall novel metabolic reprogramming. This split Warburg metabolic reprogramming triggers an inhibition of AMPK and subsequent activation of mTOR, which is a signature event of epileptogenesis. Interrogation of the mechanism with the metabolic inhibitor 2-deoxyglucose surprisingly demonstrated that Wnt signaling and the resulting metabolic reprogramming lies upstream of mTOR activation in epileptogenesis. To augment the pre-clinical pilocarpine and kainate models, aspects of the proposed mechanisms were also investigated and correlated in a genetic model of constitutive Wnt signaling (deletion of the transcriptional repressor and Wnt pathway inhibitor HBP1). The results from the HBP1-/- mice provide a genetic evidence that Wnt signaling may set the threshold of acquired seizure susceptibility with a similar molecular framework. Using biochemistry and genetics, this paper outlines a new molecular framework of early epileptogenesis and advances a potential molecular platform for refining therapeutic strategies in attenuating recurrent seizures.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Epilepsia del Lóbulo Temporal/metabolismo , Glucólisis , Hipocampo/metabolismo , Estado Epiléptico/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Vía de Señalización Wnt , Proteínas Quinasas Activadas por AMP/genética , Animales , Astrocitos/metabolismo , Astrocitos/patología , Modelos Animales de Enfermedad , Epilepsia del Lóbulo Temporal/genética , Hipocampo/patología , Masculino , Ratones , Ratones Noqueados , Neuronas/metabolismo , Neuronas/patología , Estado Epiléptico/genética , Serina-Treonina Quinasas TOR/genética
3.
PLoS One ; 8(9): e73464, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24039951

RESUMEN

Inflammatory Breast Cancer (IBC) is a highly aggressive form of cancer characterized by high rates of proliferation, lymphangiogenesis and metastasis, and an overall poor survival. As regular green tea consumption has been associated with improved prognosis of breast cancer patients, including decreased risk of recurrence, here the effects of the green tea polyphenol epigallocatechin-3-gallate (EGCG) were tested on two IBC lines: SUM-149 and SUM-190. EGCG decreased expression of genes that promote proliferation, migration, invasion, and survival. Consistently, growth, invasive properties, and survival of IBC cells were reduced by EGCG treatment. EGCG also reduced lymphangiogenesis-promoting genes, in particular VEGF-D. Conditioned media from EGCG-treated IBC cells displayed decreased VEGF-D secretion and reduced ability to promote lymphangiogenesis in vitro as measured by hTERT-HDLEC lymphatic endothelial cell migration and tube formation. Tumorsphere formation by SUM-149 cells was robustly inhibited by EGCG, suggesting effects on self-renewal ability. Stem-like SUM-149 cells with high aldehyde dehydrogenase (ALDH) activity, previously implicated in poor patient prognosis, were isolated. EGCG treatment reduced growth and induced apoptosis of the stem-like SUM-149 cells in culture. In an orthotopic mouse model, EGCG decreased growth of pre-existing tumors derived from ALDH-positive stem-like SUM-149 cells and their expression of VEGF-D, which correlated with a significant decrease in peritumoral lymphatic vessel density. Thus, EGCG inhibits the overall aggressive IBC phenotype. Reduction of the stem-like cell compartment by EGCG may explain the decreased risk of breast cancer recurrence among green tea drinkers. Recent clinical trials demonstrate the efficacy of green tea polyphenol extracts in treatment of prostate cancer and lymphocytic leukemia with low toxicity. Given the poor prognosis of IBC patients, our findings suggest further exploration of EGCG or green tea in combinatorial treatments against active IBC disease or in maintenance regimens to avoid recurrence is warranted.


Asunto(s)
Anticarcinógenos/uso terapéutico , Mama/efectos de los fármacos , Mama/patología , Catequina/análogos & derivados , Neoplasias Inflamatorias de la Mama/tratamiento farmacológico , Neoplasias Inflamatorias de la Mama/patología , Animales , Anticarcinógenos/farmacología , Mama/metabolismo , Catequina/farmacología , Catequina/uso terapéutico , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Inflamatorias de la Mama/genética , Neoplasias Inflamatorias de la Mama/metabolismo , Linfangiogénesis/efectos de los fármacos , Ratones , Ratones SCID , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Invasividad Neoplásica/prevención & control , Factor D de Crecimiento Endotelial Vascular/metabolismo
4.
Oral Oncol ; 49(2): 129-35, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22944050

RESUMEN

OBJECTIVES: Overexpression of the epidermal growth factor (EGF) receptor (EGFR) gene in the squamous cell carcinomas of the head and neck (SCCHN) is often associated with inauspicious prognosis and poor survival. N-acetylcysteine (NAC), a compound from some vegetables and allium species, appears anti-tumorigenesis, but the underlying mechanism is unclear. The objective of this study is to investigate the role of NAC in EGFR-overexpressing oral cancer. MATERIALS AND METHODS: Both HSC-3 and SCC-4 human tongue squamous carcinoma cell lines and an HSC-3 xenograft mouse model were used to test the anti-growth efficacy of NAC in vitro and in vivo, respectively. RESULTS: NAC treatment suppressed cell growth, with concomitantly increased expression of HMG box-containing protein 1 (HBP1), a transcription suppressor, and decreased EGFR/Akt activation, in EGFR-overexpressing HSC-3 oral cancer cells. HBP1 knockdown attenuated the growth arrest and apoptosis induced by NAC. Lastly, NAC and AG1478, an EGFR inhibitor, additively suppressed colony formation in HSC-3 cells. CONCLUSION: Taken together, our data indicate that NAC exerts its growth-inhibitory function through modulating EGFR/Akt signaling and HBP1 expression in EGFR-overexpressing oral cancer.


Asunto(s)
Acetilcisteína/farmacología , División Celular/efectos de los fármacos , Receptores ErbB/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Neoplasias de la Boca/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Apoptosis , Secuencia de Bases , Ciclo Celular , Cartilla de ADN , Técnicas de Silenciamiento del Gen , Proteínas del Grupo de Alta Movilidad/genética , Humanos , Masculino , Ratones , Neoplasias de la Boca/metabolismo , Invasividad Neoplásica , Proteínas Represoras/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
Mol Cell Biol ; 33(5): 887-903, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23249948

RESUMEN

The activity of DNA methyltransferase 1 (DNMT1) is associated with diverse biological activities, including cell proliferation, senescence, and cancer development. In this study, we demonstrated that the HMG box-containing protein 1 (HBP1) transcription factor is a new repressor of DNMT1 in a complex mechanism during senescence. The DNMT1 gene contains an HBP1-binding site at bp -115 to -134 from the transcriptional start site. HBP1 repressed the endogenous DNMT1 gene through sequence-specific binding, resulting in both gene-specific (e.g., p16(INK4)) and global DNA hypomethylation changes. The HBP1-mediated repression by DNMT1 contributed to replicative and premature senescence, the latter of which could be induced by Ras and HBP1 itself. A detailed investigation unexpectedly revealed that HBP1 has dual and complex transcriptional functions, both of which contribute to premature senescence. HBP1 both repressed the DNMT1 gene and activated the p16 gene in premature senescence. The opposite transcriptional functions proceeded through different DNA sequences and differential protein acetylation. While intricate, the reciprocal partnership between HBP1 and DNMT1 has exceptional importance, since its abrogation compromises senescence and promotes tumorigenesis. Together, our results suggest that the HBP1 transcription factor orchestrates a complex regulation of key genes during cellular senescence, with an impact on overall DNA methylation state.


Asunto(s)
Senescencia Celular , ADN (Citosina-5-)-Metiltransferasas/genética , Regulación hacia Abajo , Proteínas del Grupo de Alta Movilidad/metabolismo , Proteínas Represoras/metabolismo , Acetilación , Línea Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , ADN/genética , ADN/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1 , Metilación de ADN , Genes p16 , Humanos , Regiones Promotoras Genéticas , Unión Proteica , Activación Transcripcional
6.
Cancer Res ; 67(13): 6136-45, 2007 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-17616670

RESUMEN

Invasive breast cancer has a high risk of recurrence to incurable disease and needs improved prognostic and therapeutic tools. Our work combines clinical and molecular analyses to show that the transcriptional repressor HBP1 may be a new target for invasive breast cancer. Previous work indicated that HBP1 regulated proliferation and senescence and inhibited Wnt signaling. Two of these functions have been associated with invasive breast cancer. In 76 breast tumors, we identified 10 HBP1 mutations/variants that were associated with fully invasive breast cancer. In a separate analysis, we found that a subset of invasive breast cancer specimens also had reduced HBP1 mRNA levels. These clinical correlations suggested that mutation or reduction of HBP1 occurs in invasive breast cancer and that HBP1 might regulate the proliferation and invasiveness of this breast cancer type. Analysis of the HBP1 mutants showed they were functionally defective for suppressing Wnt signaling. To test the consequences of reduced HBP1 levels, we used RNA interference to knock down HBP1 and observed increased Wnt signaling, tumorigenic proliferation, and invasiveness in cell and animal breast cancer models. Lastly, statistical analysis of a breast cancer patient database linked reduced HBP1 expression to breast cancer recurrence. In considering two-gene criteria for relapse potential, reduced expression of HBP1 and SFRP1, which is another Wnt inhibitor that was recently linked to invasive breast cancer, strikingly correlated with recurrence. Together, these data indicate that HBP1 may be a molecularly and clinically relevant regulator of breast cancer transitions that eventually lead to poor prognosis.


Asunto(s)
Neoplasias de la Mama/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas del Grupo de Alta Movilidad/biosíntesis , Proteínas del Grupo de Alta Movilidad/genética , Proteínas Represoras/biosíntesis , Proteínas Represoras/genética , Transcripción Genética , Animales , Femenino , Humanos , Ratones , Ratones SCID , Mutación , Células 3T3 NIH , Invasividad Neoplásica , Metástasis de la Neoplasia , Trasplante de Neoplasias , Resultado del Tratamiento
7.
J Cell Biochem ; 102(2): 377-88, 2007 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17348027

RESUMEN

Green tea has been reported as potential dietary protection against numerous cancers and has been shown to have activity in bladder tumor inhibition in different animal models. The goal of this study was to examine the effects of (-)-epigallocatechin gallate (EGCG-the major phytochemical in green tea) on growth inhibition and behavior of human bladder carcinoma cells and to identify the altered signaling pathway(s) underlying the response to EGCG exposure. EGCG inhibited the in vitro growth of invasive bladder carcinoma cells with an IC(50) range of 70-87 microM. At a concentration of 20 microM, EGCG decreased the migratory potential of bladder carcinoma cells with concomitant activation of p42/44 MAPK and STAT3 and inactivation of Akt. Using biochemical inhibitors of MAPK/ERK, and siRNA to knockdown STAT3 and Akt, inhibition of migration was recorded associated with Akt but not MAPK/ERK or STAT3 signaling in bladder cells. In addition, EGCG downregulated N-cadherin in a dose-dependent manner where reduction in N-cadherin expression paralleled declining migratory potential. Continuous feeding of EGCG to mice prior to and during the establishment of bladder carcinoma xenografts in vivo revealed >50% reduction in mean final tumor volume (P

Asunto(s)
Anticarcinógenos/farmacología , Cadherinas/biosíntesis , Catequina/análogos & derivados , Movimiento Celular/efectos de los fármacos , , Neoplasias de la Vejiga Urinaria/metabolismo , Animales , Catequina/farmacología , Cateninas/biosíntesis , Línea Celular Tumoral , Regulación hacia Abajo , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Invasividad Neoplásica , Trasplante de Neoplasias , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Trasplante Heterólogo , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/patología
8.
Mol Cell Biol ; 26(22): 8252-66, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16966377

RESUMEN

Oncogene-mediated premature senescence has emerged as a potential tumor-suppressive mechanism in early cancer transitions. Previous work shows that RAS and p38 MAPK participate in premature senescence, but transcriptional effectors have not been identified. Here, we demonstrate that the HBP1 transcriptional repressor participates in RAS- and p38 MAPK-induced premature senescence. In cell lines, we had previously isolated HBP1 as a retinoblastoma (RB) target but have determined that it functions as a proliferation regulator by inhibiting oncogenic pathways as a transcriptional repressor. In primary cells, the results indicate that HBP1 is a necessary component of premature senescence by RAS and p38 MAPK. Similarly, a knockdown of WIP1 (a p38 MAPK phosphatase) induced premature senescence that also required HBP1. Furthermore, HBP1 requires regulation by RB, in which few transcriptional regulators for premature senescence have been shown. Together, the data suggest a model in which RAS and p38 MAPK signaling engage HBP1 and RB to trigger premature senescence. As an initial step toward clinical relevance, a bioinformatics approach shows that the relative expression levels of HBP1 and WIP1 correlated with decreased relapse-free survival in breast cancer patients. Together, these studies highlight p38 MAPK, HBP1, and RB as important components for a premature-senescence pathway with possible clinical relevance to breast cancer.


Asunto(s)
Neoplasias de la Mama/diagnóstico , Senescencia Celular , Proteínas del Grupo de Alta Movilidad/fisiología , Proteína Oncogénica p21(ras)/fisiología , Proteínas Represoras/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Sitios de Unión , Línea Celular , Replicación del ADN , Bases de Datos Genéticas , Humanos , Modelos Biológicos , Fosfoproteínas Fosfatasas/metabolismo , Proteína Fosfatasa 2C , Proteína de Retinoblastoma/metabolismo , Proteína de Retinoblastoma/fisiología , Transducción de Señal , Transfección
9.
J Biol Chem ; 281(16): 10865-75, 2006 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-16495219

RESUMEN

Genetic and biochemical de-regulation of Wnt signaling is correlated with breast and other cancers. Our goal was to identify compounds that block Wnt signaling as a first step toward investigating new strategies for suppression of invasive and other breast cancers. In a limited phytonutrient screen, EGCG ((-)-epigallocatechin 3-gallate), the major phytochemical in green tea, emerged as an intriguing candidate. Epidemiological studies have associated green tea consumption with reduced recurrence of invasive and other breast cancers. Wnt signaling was inhibited by EGCG in a dose-dependent manner in breast cancer cells. The apparent mechanism targeted the HBP1 transcriptional repressor, which we had previously characterized as a suppressor of Wnt signaling. EGCG treatment induced HBP1 transcriptional repressor levels through an increase in HBP1 mRNA stability, but not transcriptional initiation. To test functionality, DNA-based short hairpin RNA (shRNA) was used to knockdown the endogenous HBP1 gene. Consistently, the HBP1 knockdown lines had reduced sensitivity to EGCG in the suppression of Wnt signaling and of a target gene (c-MYC). Because our ongoing studies clinically link abrogation of HBP1 with invasive breast cancer, we tested if EGCG also regulated biological functions associated with de-regulated Wnt signaling and with invasive breast cancer. EGCG reduced both breast cancer cell tumorigenic proliferation and invasiveness in an HBP1-dependent manner. Together, the emerging mechanism is that EGCG blocks Wnt signaling by inducing the HBP1 transcriptional repressor and inhibits aspects of invasive breast cancer. These studies provide a framework for considering future studies in breast cancer treatment and prevention.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Catequina/análogos & derivados , Regulación Neoplásica de la Expresión Génica , Proteínas del Grupo de Alta Movilidad/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Wnt/biosíntesis , Proteínas Wnt/metabolismo , Western Blotting , Neoplasias de la Mama/metabolismo , Catequina/farmacología , Línea Celular , Línea Celular Tumoral , Movimiento Celular , Dactinomicina/farmacología , Relación Dosis-Respuesta a Droga , Genes Reporteros , Humanos , Modelos Biológicos , Fosforilación , Plásmidos/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN/metabolismo , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , , Factores de Tiempo , Transcripción Genética , Transfección
10.
Gene ; 336(1): 1-13, 2004 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-15225871

RESUMEN

Mechanisms that inhibit cell cycle progression and establish growth arrest are fundamental to tumor suppression and to normal cell differentiation. A complete understanding of these mechanisms should provide new diagnostic and therapeutic targets for future clinical applications related to cancer-specific pathways. This review will focus on the HMG-box protein 1 (HBP1) transcriptional repressor and its roles in cell cycle progression and tumor suppression. The work of several labs now suggests a new pathway for inhibiting G1 progression with exciting possible implications for tumor suppression. Our recent work suggests that the two previously unassociated proteins-the HBP1 transcription factor and the p38 MAP kinase pathway-may now participate together in a G1 regulatory network. Several recent papers collectively highlight an unexpected role and connection of the p38 MAP kinase-signaling pathway in cell cycle control, senescence, and tumor suppression. Together, these initially divergent observations may provide clues into a new tumor suppressive network and spur further investigations that may contribute to new diagnostic and therapeutic targets for cancer.


Asunto(s)
Proteínas del Grupo de Alta Movilidad/fisiología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Represoras/fisiología , Transducción de Señal , Secuencia de Aminoácidos , Animales , Fase G1/genética , Fase G1/fisiología , Proteínas del Grupo de Alta Movilidad/genética , Humanos , Modelos Biológicos , Datos de Secuencia Molecular , Neoplasias/genética , Neoplasias/fisiopatología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/genética , Homología de Secuencia de Aminoácido , Proteínas Wnt , Proteínas Quinasas p38 Activadas por Mitógenos
11.
Mol Cell Biol ; 24(7): 3011-24, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15024088

RESUMEN

Several studies have linked the production of reactive oxygen species (ROS) by the NADPH oxidase to cellular growth control. In many cases, activation of the NADPH oxidase and subsequent ROS generation is required for growth factor signaling and mitogenesis in nonimmune cells. In this study, we demonstrate that the transcriptional repressor HBP1 (HMG box-containing protein 1) regulates the gene for the p47phox regulatory subunit of the NADPH oxidase. HBP1 represses growth regulatory genes (e.g., N-Myc, c-Myc, and cyclin D1) and is an inhibitor of G(1) progression. The promoter of the p47phox gene contains six tandem high-affinity HBP1 DNA-binding elements at positions -1243 to -1318 bp from the transcriptional start site which were required for repression. Furthermore, HBP1 repressed the expression of the endogenous p47phox gene through sequence-specific binding. With HBP1 expression and the subsequent reduction in p47phox gene expression, intracellular superoxide production was correspondingly reduced. Using both the wild type and a dominant-negative mutant of HBP1, we demonstrated that the repression of superoxide production through the NADPH oxidase contributed to the observed cell cycle inhibition by HBP1. Together, these results indicate that HBP1 may contribute to the regulation of NADPH oxidase-dependent superoxide production through transcriptional repression of the p47phox gene. This study defines a transcriptional mechanism for regulating intracellular ROS levels and has implications in cell cycle regulation.


Asunto(s)
Ciclo Celular/fisiología , Regulación de la Expresión Génica , Proteínas del Grupo de Alta Movilidad/metabolismo , NADPH Oxidasas/metabolismo , Fosfoproteínas/genética , Proteínas Represoras/metabolismo , Animales , Línea Celular , Proteínas del Grupo de Alta Movilidad/genética , Humanos , Ratones , Modelos Biológicos , Regiones Promotoras Genéticas , Especies Reactivas de Oxígeno/metabolismo , Proteínas Represoras/genética , Superóxidos/metabolismo
12.
Mol Cell Biol ; 23(23): 8890-901, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14612426

RESUMEN

The p38 mitogen-activated protein (MAP) kinase signaling pathway participates in both apoptosis and G1 arrest. In contrast to the established role in apoptosis, the documented induction of G1 arrest by activation of the p38 MAP kinase pathway has attracted recent attention with reports of substrates that are linked to cell cycle regulation. Here, we identify the high-mobility group box protein HBP1 transcriptional repressor as a new substrate for p38 MAP kinase. Our previous work had shown that HBP1 inhibits G1 progression in cell and animal models, and thus indicated that HBP1 could be a relevant substrate for p38 MAP kinase in cell cycle regulation. In the present work, a p38 MAP kinase docking site (amino acids [aa] 81 to 125) and a p38 MAP kinase phosphorylation site (serine 401) were identified in the HBP1 protein. Furthermore, the docking and phosphorylation sites on HBP1 were specific for p38 MAP kinase. In defining the role of p38 MAP kinase regulation, the inhibition of p38 MAP kinase activity was shown to decrease HBP1 protein levels by triggering protein instability, as manifested by a decrease in protein half-life. Consistently, a decrease in protein levels was accompanied by a decrease in overall DNA binding activity. A mutation of the p38 MAP kinase phosphorylation site at aa 401 [(S-A)401HBP1] also triggered HBP1 protein instability. While protein stability was compromised by mutation, the specific activities of (S-A)401HBP1 and of wild-type HBP1 appeared comparable for transcriptional repression. This comparison of transcription-specific activity highlighted that p38 MAP kinase regulated HBP1 protein levels but not the intrinsic activity for DNA binding or for transcriptional repression. Finally, p38 MAP kinase-mediated regulation of the HBP1 protein also contributed to the regulation of G1 progression. Together, our work supports a molecular framework in which p38 MAP kinase activity contributes to cell cycle inhibition by increasing HBP1 and other G1 inhibitory factors by regulating protein stability.


Asunto(s)
Ciclo Celular/fisiología , Proteínas del Grupo de Alta Movilidad/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Represoras/metabolismo , Animales , Secuencia de Bases , ADN Complementario/genética , Fase G1 , Proteínas del Grupo de Alta Movilidad/química , Proteínas del Grupo de Alta Movilidad/genética , Técnicas In Vitro , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Quinasas Activadas por Mitógenos/genética , Modelos Biológicos , Mutagénesis Sitio-Dirigida , Fosforilación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Represoras/química , Proteínas Represoras/genética , Proteínas Quinasas p38 Activadas por Mitógenos
13.
Free Radic Biol Med ; 32(6): 551-61, 2002 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-11958956

RESUMEN

The nitrone spin trap PBN has been shown to protect neuronal cells from reactive oxygen species both in culture and in vivo. As an approach to understanding the molecular mechanisms by which PBN may function to protect cells, we examined whether PBN alters the cellular response to reactive oxygen species. H(2)O(2) stimulation of PC-12 cells results in weak activation of both the ERK and JNK signal transduction pathways. PBN pretreatment of PC-12 cells, followed by H(2)O(2) stimulation, results in strong and selective activation of the pro-survival ERK pathway. H(2)O(2) induction of ERK activity in PBN-pretreated cells was shown to be dependent on extracellular Ca(+2) influx. Further analysis of the ERK pathway showed that in PBN-pretreated cells, EGF receptor and the adapter protein SHC were phosphorylated in a Ca(+2)-dependent, ligand-independent manner following H(2)O(2) stimulation. Interestingly, H(2)O(2) stimulation of PBN-pretreated cells results in only 30% of the increase in intracellular Ca(+2) as compared to untreated cells following H(2)O(2) stimulation. These data suggest a model in which PBN attenuates H(2)O(2)-induced Ca(+2) entry, yet magnifies or alters Ca(+2) action, resulting in the activation of the EGF receptor/ERK pathway.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas Adaptadoras del Transporte Vesicular , Receptores ErbB/metabolismo , Depuradores de Radicales Libres/farmacología , Peróxido de Hidrógeno/farmacología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Óxidos de Nitrógeno/farmacología , Animales , Calcio/metabolismo , Óxidos N-Cíclicos , Interacciones Farmacológicas , Activación Enzimática , Células PC12 , Fosforilación/efectos de los fármacos , Proteínas/metabolismo , Ratas , Proteínas Adaptadoras de la Señalización Shc , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...