Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nephron ; 145(6): 748-759, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34438395

RESUMEN

BACKGROUND: We intended to investigate the function of circular RNA RasGEF domain family member 1B (circ_RASGEF1B) in lipopolysaccharide (LPS)-induced septic acute kidney injury (AKI) cell model and its associated mechanism. METHODS: TCMK-1 cells were exposed to 10 µg/mL LPS for 24 h to establish a septic AKI cell model. Mice were intraperitoneally injected with 10 mg/kg LPS to establish a septic AKI mice model. Reverse transcription quantitative polymerase chain reaction (RT-qPCR) and Western blot assay were used to measure RNA and protein expression, respectively. Cell viability and apoptosis were assessed by 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and flow cytometry. Cell inflammatory response was analyzed using enzyme-linked immunosorbent assay. Dual-luciferase reporter assay was conducted to confirm the predicted target relationship between microRNA-146a-5p (miR-146a-5p) and circ_RASGEF1B or pyruvate dehydrogenase kinase 1 (Pdk1). RESULTS: The circ_RASGEF1B level was upregulated in LPS-induced TCMK-1 cells and septic AKI mice models. LPS exposure reduced cell viability and promoted cell apoptosis and inflammatory response partly by upregulating circ_RASGEF1B. Circ_RASGEF1B bound to miR-146a-5p and miR-146a-5p interference partly overturned circ_RASGEF1B silencing-mediated effects in LPS-induced TCMK-1 cells. Pdk1 was a target of miR-146a-5p, and Pdk1 accumulation partly counteracted miR-146a-5p-induced influences in TCMK-1 cells upon LPS stimulation. CONCLUSION: Circ_RASGEF1B promoted LPS-induced apoptosis and inflammatory response in renal tubular epithelial cells partly by upregulating Pdk1 via acting as miR-146a-5p sponge.


Asunto(s)
Lesión Renal Aguda/patología , Apoptosis/efectos de los fármacos , Inflamación/inducido químicamente , Lipopolisacáridos/toxicidad , MicroARNs/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/metabolismo , Sepsis/patología , Factores de Intercambio de Guanina Nucleótido ras/fisiología , Lesión Renal Aguda/complicaciones , Lesión Renal Aguda/metabolismo , Animales , Modelos Animales de Enfermedad , Ratones , Sepsis/complicaciones , Sepsis/metabolismo
2.
J Neurosci ; 39(32): 6325-6338, 2019 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-31182637

RESUMEN

Ras/Raf/MEK/ERK (Ras-ERK) signaling has been implicated in the effects of drugs of abuse. Inhibitors of MEK1/2, the kinases upstream of ERK1/2, have been critical in defining the role of the Ras-ERK cascade in drug-dependent alterations in behavioral plasticity, but the Ras family of small GTPases has not been extensively examined in drug-related behaviors. We examined the role of Ras Guanine Nucleotide Releasing Factor 1 (RasGRF1) and 2 (RasGRF2), upstream regulators of the Ras-ERK signaling cascade, on cocaine self-administration (SA) in male mice. We first established a role for Ras-ERK signaling in cocaine SA, demonstrating that pERK1/2 is upregulated following SA in C57BL/6N mice in striatum. We then compared RasGRF1 and RasGRF2 KO mouse lines, demonstrating that cocaine SA in RasGRF2 KO mice was increased relative to WT controls, whereas RasGRF1 KO and WT mice did not differ. This effect in RasGRF2 mice is likely mediated by the Ras-ERK signaling pathway, as pERK1/2 upregulation following cocaine SA was absent in RasGRF2 KO mice. Interestingly, the lentiviral knockdown of RasGRF2 in the NAc had the opposite effect to that in RasGRF2 KO mice, reducing cocaine SA. We subsequently demonstrated that the MEK inhibitor PD325901 administered peripherally prior to cocaine SA increased cocaine intake, replicating the increase seen in RasGRF2 KO mice, whereas PD325901 administered into the NAc decreased cocaine intake, similar to the effect seen following lentiviral knockdown of RasGRF2. These data indicate a role for RasGRF2 in cocaine SA in mice that is ERK-dependent, and suggest a differential effect of global versus site-specific RasGRF2 inhibition.SIGNIFICANCE STATEMENT Exposure to drugs of abuse activates a variety of intracellular pathways, and following repeated exposure, persistent changes in these pathways contribute to drug dependence. Downstream components of the Ras-ERK signaling cascade are involved in the acute and chronic effects of drugs of abuse, but their upstream mediators have not been extensively characterized. Here we show, using a combination of molecular, pharmacological, and lentiviral techniques, that the guanine nucleotide exchange factor RasGRF2 mediates cocaine self-administration via an ERK-dependent mechanism, whereas RasGRF1 has no effect on responding for cocaine. These data indicate dissociative effects of mediators of Ras activity on cocaine reward and expand the understanding of the contribution of Ras-ERK signaling to drug-taking behavior.


Asunto(s)
Trastornos Relacionados con Cocaína/fisiopatología , Cocaína/farmacología , Cuerpo Estriado/fisiopatología , Sistema de Señalización de MAP Quinasas/fisiología , Recompensa , Factores de Intercambio de Guanina Nucleótido ras/fisiología , Acetilación , Animales , Benzamidas/farmacología , Cocaína/administración & dosificación , Condicionamiento Operante , Cuerpo Estriado/efectos de los fármacos , Difenilamina/análogos & derivados , Difenilamina/farmacología , Técnicas de Silenciamiento del Gen , Vectores Genéticos/genética , Histonas/metabolismo , Lentivirus/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/fisiopatología , Especificidad de Órganos , Fosforilación , Procesamiento Proteico-Postraduccional , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Autoadministración , Factores de Intercambio de Guanina Nucleótido ras/deficiencia , Factores de Intercambio de Guanina Nucleótido ras/genética , ras-GRF1/deficiencia , ras-GRF1/genética , ras-GRF1/fisiología
3.
PLoS One ; 11(3): e0151638, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26982501

RESUMEN

Ras guanine nucleotide-releasing protein-4 (RasGRP4) is an evolutionarily conserved calcium-regulated, guanine nucleotide exchange factor and diacylglycerol/phorbol ester receptor. While an important intracellular signaling protein for CD117+ mast cells (MCs), its roles in other immune cells is less clear. In this study, we identified a subset of in vivo-differentiated splenic CD117+ dendritic cells (DCs) in wild-type (WT) C57BL/6 mice that unexpectedly contained RasGRP4 mRNA and protein. In regard to the biologic significance of these data to innate immunity, LPS-treated splenic CD117+ DCs from WT mice induced natural killer (NK) cells to produce much more interferon-γ (IFN-γ) than comparable DCs from RasGRP4-null mice. The ability of LPS-responsive MCs to cause NK cells to increase their expression of IFN-γ was also dependent on this intracellular signaling protein. The discovery that RasGRP4 is required for CD117+ MCs and DCs to optimally induce acute NK cell-dependent immune responses to LPS helps explain why this signaling protein has been conserved in evolution.


Asunto(s)
Células Dendríticas/inmunología , Células Asesinas Naturales/inmunología , Lipopolisacáridos/farmacología , Mastocitos/inmunología , Proteínas Proto-Oncogénicas c-kit/inmunología , Factores de Intercambio de Guanina Nucleótido ras/fisiología , Animales , Técnicas de Cocultivo , Interferón gamma/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ligando OX40 , Transducción de Señal , Bazo/citología , Bazo/efectos de los fármacos , Bazo/inmunología , Factores de Necrosis Tumoral/metabolismo
4.
J Cell Sci ; 129(4): 729-42, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26743081

RESUMEN

Detailed immunocytochemical analyses comparing wild-type (WT), GRF1-knockout (KO), GRF2-KO and GRF1/2 double-knockout (DKO) mouse retinas uncovered the specific accumulation of misplaced, 'ectopic' cone photoreceptor nuclei in the photoreceptor segment (PS) area of retinas from GRF2-KO and GRF1/2-DKO, but not of WT or GRF1-KO mice. Localization of ectopic nuclei in the PS area of GRF2-depleted retinas occurred postnatally and peaked between postnatal day (P)11 and P15. Mechanistically, the generation of this phenotype involved disruption of the outer limiting membrane and intrusion into the PS layer by cone nuclei displaying significant perinuclear accumulation of signaling molecules known to participate in nuclear migration and cytoskeletal reorganization, such as PAR3, PAR6 and activated, phosphorylated forms of PAK, MLC2 and VASP. Electroretinographic recordings showed specific impairment of cone-mediated retinal function in GRF2-KO and GRF1/2-DKO retinas compared with WT controls. These data identify defective cone nuclear migration as a novel phenotype in mouse retinas lacking GRF2 and support a crucial role of GRF2 in control of the nuclear migration processes required for proper postnatal development and function of retinal cone photoreceptors.


Asunto(s)
Núcleo Celular/fisiología , Células Fotorreceptoras Retinianas Conos/ultraestructura , Factores de Intercambio de Guanina Nucleótido ras/fisiología , Animales , Transporte Biológico , Electrorretinografía , Ratones Endogámicos C57BL , Ratones Noqueados , Retina/citología , Retina/fisiología , Células Fotorreceptoras Retinianas Conos/metabolismo , Transducción de Señal
5.
Nat Commun ; 5: 4657, 2014 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-25118589

RESUMEN

Host immune cells can detect and destruct invading pathogens via pattern-recognition receptors. Small Rap GTPases act as conserved molecular switches coupling extracellular signals to various cellular responses, but their roles as regulators in Toll-like receptor (TLR) signalling have not been fully elucidated. Here we report that Ras guanine nucleotide-releasing protein 3 (RasGRP3), a guanine nucleotide-exchange factor activating Ras and Rap1, limits production of proinflammatory cytokines (especially IL-6) in macrophages by activating Rap1 on activation by low levels of TLR agonists. We demonstrate that RasGRP3, a dominant member of RasGRPs in macrophages, impairs TLR3/4/9-induced IL-6 production and relieves dextrane sulphate sodium-induced colitis and collagen-induced arthritis. In RasGRP3-deficient RAW264.7 cells obtained by CRISPR-Cas9 genome editing, TLR3/4/9-induced activation of Rap1 was inhibited while ERK1/2 activation was enhanced. Our study suggests that RasGRP3 limits inflammatory response by activating Rap1 on low-intensity pathogen infection, setting a threshold for preventing excessive inflammatory response.


Asunto(s)
GTP Fosfohidrolasas/fisiología , Inflamación/fisiopatología , Macrófagos/fisiología , Receptores Toll-Like/fisiología , Proteínas de Unión al GTP rap1/fisiología , Factores de Intercambio de Guanina Nucleótido ras/fisiología , Animales , Artritis Experimental/patología , Artritis Experimental/fisiopatología , Células Cultivadas , Modelos Animales de Enfermedad , Inflamación/patología , Interleucina-6/fisiología , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/fisiología , Receptor Toll-Like 3/fisiología , Receptor Toll-Like 4/fisiología , Receptor Toll-Like 9/fisiología , Factores de Intercambio de Guanina Nucleótido ras/deficiencia , Factores de Intercambio de Guanina Nucleótido ras/genética
6.
Proc Natl Acad Sci U S A ; 109(51): 21128-33, 2012 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-23223532

RESUMEN

The firing of mesolimbic dopamine neurons is important for drug-induced reinforcement, although underlying genetic factors remain poorly understood. In a recent genome-wide association metaanalysis of alcohol intake, we identified a suggestive association of SNP rs26907 in the ras-specific guanine-nucleotide releasing factor 2 (RASGRF2) gene, encoding a protein that mediates Ca(2+)-dependent activation of the ERK pathway. We performed functional characterization of this gene in relation to alcohol-related phenotypes and mesolimbic dopamine function in both mice and adolescent humans. Ethanol intake and preference were decreased in Rasgrf2(-/-) mice relative to WT controls. Accordingly, ethanol-induced dopamine release in the ventral striatum was blunted in Rasgrf2(-/-) mice. Recording of dopamine neurons in the ventral tegmental area revealed reduced excitability in the absence of Ras-GRF2, likely because of lack of inhibition of the I(A) potassium current by ERK. This deficit provided an explanation for the altered dopamine release, presumably linked to impaired activation of dopamine neurons firing. Functional neuroimaging analysis of a monetary incentive-delay task in 663 adolescent boys revealed significant association of ventral striatal activity during reward anticipation with a RASGRF2 haplotype containing rs26907, the SNP associated with alcohol intake in our previous metaanalysis. This finding suggests a link between the RASGRF2 haplotype and reward sensitivity, a known risk factor for alcohol and drug addiction. Indeed, follow-up of these same boys at age 16 y revealed an association between this haplotype and number of drinking episodes. Together, these combined animal and human data indicate a role for RASGRF2 in the regulation of mesolimbic dopamine neuron activity, reward response, and alcohol use and abuse.


Asunto(s)
Dopamina/metabolismo , Neuronas/metabolismo , Factores de Intercambio de Guanina Nucleótido ras/genética , Factores de Intercambio de Guanina Nucleótido ras/fisiología , Adolescente , Animales , Encéfalo/metabolismo , Calcio/metabolismo , Niño , Neuronas Dopaminérgicas/metabolismo , Electrofisiología/métodos , Etanol/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Genotipo , Haplotipos , Humanos , Masculino , Ratones , Ratones Transgénicos , ARN Mensajero/metabolismo , Refuerzo en Psicología , Factores de Tiempo , Área Tegmental Ventral/metabolismo
7.
Carcinogenesis ; 32(12): 1758-72, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21890458

RESUMEN

Chronic myelogenous leukemia (CML) is characterized by a reciprocal chromosomal translocation (9;22) that generates the Bcr-Abl fusion gene. BCR-ABL transforming activity is mediated by critical downstream signaling pathways that are aberrantly activated by tyrosine kinases. However, the mechanisms of BCR-ABL anti-apoptotic effects and the signaling pathways by which BCR-ABL influences apoptosis in BCR-ABL-expressing cells are poorly defined. In this study, we found that treatment with ABL kinase inhibitors or depletion of BCR-ABL induced the expression of RAB45 messenger RNA and protein and induced apoptosis via reduction of mitochondrial membrane potential and p38 activation in CML cell lines and BCR-ABL(+) progenitor cells from CML patients. Overexpressed RAB45 induced the activation of caspases-3 and -9 and reduced the expression of Survivin, XIAP, c-IAP1 and c-IAP2 in CML cells. Moreover, in colony-forming cells derived from CML-aldehyde dehydrogenase(hi)/CD34(+) cells, treatment with ABL kinase inhibitors induced RAB45 expression and reduced mitochondrial membrane potential, resulting in inhibited colony formation of Bcr-Abl(+) progenitor cells. The overexpression of RAB45 significantly decreased colony numbers and induced apoptosis through the activation of caspases-3 and -9. Furthermore, the overexpression of RAB45 increased the phosphorylation levels of p38, resulting in the induction of apoptosis and inhibition of proliferation of CML progenitor cells. Our results identify a new signaling molecule involved in BCR-ABL modulation of apoptosis and suggest that RAB45 induction strategies may have therapeutic utility in patients with CML.


Asunto(s)
Apoptosis/fisiología , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Células Madre Neoplásicas/patología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Factores de Intercambio de Guanina Nucleótido ras/fisiología , Secuencia de Bases , Western Blotting , Caspasa 3/metabolismo , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Cartilla de ADN , Activación Enzimática , Genes abl , Humanos , Inmunoprecipitación , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Potenciales de la Membrana , Fosforilación , Interferencia de ARN , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Intercambio de Guanina Nucleótido ras/genética
8.
Circ Res ; 108(10): 1199-208, 2011 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-21474816

RESUMEN

RATIONALE: Fetuses that develop in diabetic mothers have a higher incidence of birth defects that include cardiovascular defects, but the signaling pathways that mediate these developmental effects are poorly understood. It is reasonable to hypothesize that diabetic maternal effects are mediated by 1 or more pathways activated downstream of aberrant glucose metabolism, because poorly controlled maternal glucose levels correlate with the frequency and severity of the defects. OBJECTIVE: We investigated whether RasGRP3 (Ras guanyl-releasing protein 3), a Ras activator expressed in developing blood vessels, mediates diabetes-induced vascular developmental defects. RasGRP3 is activated by diacylglycerol, and diacylglycerol is overproduced by aberrant glucose metabolism in diabetic individuals. We also investigated the effects of overactivation and loss of function for RasGRP3 in primary endothelial cells and developing vessels. METHODS AND RESULTS: Analysis of mouse embryos from diabetic mothers showed that diabetes-induced developmental defects were dramatically attenuated in embryos that lacked Rasgrp3 function. Endothelial cells that expressed activated RasGRP3 had elevated Ras-ERK signaling and perturbed migration, whereas endothelial cells that lacked Rasgrp3 function had attenuated Ras-ERK signaling and did not migrate in response to endothelin-1. Developing blood vessels exhibited endothelin-stimulated vessel dysmorphogenesis that required Rasgrp3 function. CONCLUSIONS: These findings provide the first evidence that RasGRP3 contributes to developmental defects found in embryos that develop in a diabetic environment. The results also elucidate RasGRP3-mediated signaling in endothelial cells and identify endothelin-1 as an upstream input and Ras/MEK/ERK as a downstream effector pathway. RasGRP3 may be a novel therapeutic target for the fetal complications of diabetes.


Asunto(s)
Movimiento Celular/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Células Madre Embrionarias/metabolismo , Células Endoteliales/metabolismo , Complicaciones Cardiovasculares del Embarazo/metabolismo , Complicaciones Cardiovasculares del Embarazo/patología , Factores de Intercambio de Guanina Nucleótido ras/fisiología , Animales , Células Cultivadas , Diabetes Mellitus Experimental/genética , Técnicas de Cultivo de Embriones , Desarrollo Embrionario/genética , Células Madre Embrionarias/patología , Células Endoteliales/patología , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Embarazo , Complicaciones Cardiovasculares del Embarazo/genética , Factores de Intercambio de Guanina Nucleótido ras/deficiencia , Factores de Intercambio de Guanina Nucleótido ras/genética
9.
Front Biosci (Landmark Ed) ; 16(4): 1445-60, 2011 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-21196241

RESUMEN

The marked increase in skeletal muscle mass during the neonatal period is largely due to a high rate of postprandial protein synthesis that is modulated by an enhanced sensitivity to insulin and amino acids. The amino acid signaling pathway leading to the stimulation of protein synthesis has not been fully elucidated. Among the amino acids, leucine is considered to be a principal anabolic agent that regulates protein synthesis. mTORC1, which controls protein synthesis, has been implicated as a target for leucine. Until recently, there have been few studies exploring the role of amino acids in enhancing muscle protein synthesis in vivo. In this review, we discuss amino acid-induced protein synthesis in muscle in the neonate, focusing on current knowledge of the role of amino acids in the activation of mTORC1 leading to mRNA translation. The role of the amino acid transporters, SNAT2, LAT1, and PAT, in the modulation of mTORC1 activation and the role of amino acids in the activation of putative regulators of mTORC1, i.e., raptor, Rheb, MAP4K3, Vps34, and Rag GTPases, are discussed.


Asunto(s)
Aminoácidos/fisiología , Proteínas Musculares/biosíntesis , Músculo Esquelético/metabolismo , Proteínas Adaptadoras Transductoras de Señales/fisiología , Sistema de Transporte de Aminoácidos A/fisiología , Sistemas de Transporte de Aminoácidos/fisiología , Animales , Animales Recién Nacidos , Fosfatidilinositol 3-Quinasas Clase III/fisiología , Humanos , Recién Nacido , Transportador de Aminoácidos Neutros Grandes 1/fisiología , Leucina/farmacología , Diana Mecanicista del Complejo 1 de la Rapamicina , Proteínas de Unión al GTP Monoméricas/fisiología , Complejos Multiproteicos , Músculo Esquelético/efectos de los fármacos , Neuropéptidos/fisiología , Biosíntesis de Proteínas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas/fisiología , ARN Mensajero/metabolismo , Proteína Homóloga de Ras Enriquecida en el Cerebro , Proteína Reguladora Asociada a mTOR , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Porcinos , Simportadores/fisiología , Serina-Treonina Quinasas TOR , Factores de Intercambio de Guanina Nucleótido ras/fisiología
10.
Dev Cell ; 18(5): 737-49, 2010 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-20493808

RESUMEN

Ras was found to regulate Dictyostelium chemotaxis, but the mechanisms that spatially and temporally control Ras activity during chemotaxis remain largely unknown. We report the discovery of a Ras signaling complex that includes the Ras guanine exchange factor (RasGEF) Aimless, RasGEFH, protein phosphatase 2A (PP2A), and a scaffold designated Sca1. The Sca1/RasGEF/PP2A complex is recruited to the plasma membrane in a chemoattractant- and F-actin-dependent manner and is enriched at the leading edge of chemotaxing cells where it regulates F-actin dynamics and signal relay by controlling the activation of RasC and the downstream target of rapamycin complex 2 (TORC2)-Akt/protein kinase B (PKB) pathway. In addition, PKB and PKB-related PKBR1 phosphorylate Sca1 and regulate the membrane localization of the Sca1/RasGEF/PP2A complex, and thereby RasC activity, in a negative feedback fashion. Thus, our study uncovered a molecular mechanism whereby RasC activity and the spatiotemporal activation of TORC2 are tightly controlled at the leading edge of chemotaxing cells.


Asunto(s)
Movimiento Celular/fisiología , Quimiotaxis/fisiología , Neutrófilos/fisiología , Factores de Transcripción/fisiología , Proteínas ras/fisiología , Animales , Agregación Celular/fisiología , Membrana Celular/fisiología , AMP Cíclico/fisiología , Dictyostelium/fisiología , Eliminación de Gen , Humanos , Neutrófilos/citología , Fosforilación , Proteínas Proto-Oncogénicas p21(ras)/fisiología , Proteínas Protozoarias/fisiología , Factores de Transcripción/genética , Factores de Intercambio de Guanina Nucleótido ras/fisiología , Proteínas ras/genética
11.
Genes Immun ; 11(6): 447-57, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20090772

RESUMEN

Guanine-nucleotide exchange factors (GEFs) stimulate the intrinsic GDP/GTP exchange activity of Ras and promote the formation of active Ras-GTP, which in turn controls diverse signalling networks important for the regulation of cell proliferation, survival, differentiation, vesicular trafficking, and gene expression. RasGEF1b is a GEF, whose expression is induced in macrophages on stimulation with toll-like receptor (TLR) agonists. Here, we showed that in vitro RasGEF1b expression by macrophages is mostly induced by TLR3 (poly I:C) and TLR4 (lipopolysaccharyde) through the MyD88-independent pathway. In vivo infection with the protozoan parasites Trypanosoma cruzi and Plasmodium chabaudi induced RasGEF1b in an MyD88-, TRIF-, and IFN-gamma-dependent manner. Ectopically expressed RasGEF1b was found, mostly, in the heavy membrane fraction of HEK 293T, and by confocal microscopy, it was found to be located at early endosomes. Computational modelling of the RasGEF1b-Ras interaction revealed that RasGEF1b interacts with the binding domain site of Ras, a critical region for interacting with GEFs involved in the activation of Ras-Raf-MEK-ERK pathway. More important, RasGEF1b was found to be closely associated with Ras in live cells and to trigger Ras activity. Altogether, these results indicate that on TLR activation, RasGEF1b may trigger Ras-like proteins and regulate specific biological activities described for this subtype of GTPases.


Asunto(s)
Endosomas/metabolismo , Receptores Toll-Like/fisiología , Factores de Intercambio de Guanina Nucleótido ras/biosíntesis , Animales , Células CHO , Cricetinae , Cricetulus , Endosomas/química , Femenino , Células HEK293 , Humanos , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/fisiología , Lipopolisacáridos/farmacología , Macrófagos Peritoneales/química , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Toll-Like/metabolismo , Factores de Intercambio de Guanina Nucleótido ras/metabolismo , Factores de Intercambio de Guanina Nucleótido ras/fisiología
12.
Int J Dev Biol ; 54(4): 609-15, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-19598105

RESUMEN

Ras guanyl nucleotide-releasing protein 2 (RASGRP2), one of the Ras guanine exchange factors, is implicated as a critical regulator of inside-out integrin activation in human lymphocytes, neutrophils and platelets. However, the activities of this protein in endothelial cells remain unclear. In the current study, we identify a physiological function in blood vessel formation for XRASGRP2, which is the Xenopus ortholog of mammalian RASGRP2. XRASGRP2 over-expression induced ectopic vascular formation, and XRASGRP2-knockdown embryos showed delayed vascular development. We also investigated the upstream signaling of XRASGRP2 in endothelium formation. XRASGRP2 expression was up-regulated in the presence of VEGF-A and down-regulated following VEGF-A depletion. XRASGRP2 knockdown abolished the ectopic induction of endothelial cells by VEGF-A in the posterior ventral blood island. These results suggest that XRASGRP2 is essential for vascular formation during Xenopus development.


Asunto(s)
Vasos Sanguíneos/metabolismo , Proteínas de Xenopus/fisiología , Xenopus/metabolismo , Factores de Intercambio de Guanina Nucleótido ras/fisiología , Animales , Diferenciación Celular/genética , Células Endoteliales/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Xenopus/genética , Proteínas de Xenopus/genética , Factores de Intercambio de Guanina Nucleótido ras/genética
13.
Mol Biol Cell ; 20(20): 4289-302, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19692568

RESUMEN

RasGRF family guanine nucleotide exchange factors (GEFs) promote guanosine diphosphate (GDP)/guanosine triphosphate (GTP) exchange on several Ras GTPases, including H-Ras and TC21. Although the mechanisms controlling RasGRF function as an H-Ras exchange factor are relatively well characterized, little is known about how TC21 activation is regulated. Here, we have studied the structural and spatial requirements involved in RasGRF 1/2 exchange activity on TC21. We show that RasGRF GEFs can activate TC21 in all of its sublocalizations except at the Golgi complex. We also demonstrate that TC21 susceptibility to activation by RasGRF GEFs depends on its posttranslational modifications: farnesylated TC21 can be activated by both RasGRF1 and RasGRF2, whereas geranylgeranylated TC21 is unresponsive to RasGRF2. Importantly, we show that RasGRF GEFs ability to catalyze exchange on farnesylated TC21 resides in its pleckstrin homology 1 domain, by a mechanism independent of localization and of its ability to associate to membranes. Finally, our data indicate that Cdc42-GDP can inhibit TC21 activation by RasGRF GEFs, demonstrating that Cdc42 negatively affects the functions of RasGRF GEFs irrespective of the GTPase being targeted.


Asunto(s)
Proteínas de la Membrana/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Proteína de Unión al GTP cdc42/fisiología , Factores de Intercambio de Guanina Nucleótido ras/fisiología , ras-GRF1/fisiología , Animales , Células COS/metabolismo , Chlorocebus aethiops , Activación Enzimática/efectos de los fármacos , Guanosina Difosfato/metabolismo , Células HeLa/metabolismo , Humanos , Ionomicina/farmacología , Lisofosfolípidos/farmacología , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas de Unión al GTP Monoméricas/química , Proteínas de Unión al GTP Monoméricas/genética , Orgánulos/metabolismo , Prenilación , Mapeo de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , ARN Interferente Pequeño/farmacología , Proteínas Recombinantes de Fusión/fisiología , Relación Estructura-Actividad , Fracciones Subcelulares/metabolismo , Especificidad por Sustrato , Factores de Intercambio de Guanina Nucleótido ras/antagonistas & inhibidores , Factores de Intercambio de Guanina Nucleótido ras/genética , ras-GRF1/genética
14.
Biochem Biophys Res Commun ; 372(4): 886-91, 2008 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-18539143

RESUMEN

Previously, we described the DNA microarray screening of vascular endothelial cells that were formed by treatment of aggregates prepared from Xenopus animal cap cells with activin and angiopoietin-2. One of the genes identified in this screening showed homology to human RASGRP2 which plays a role in the regulation of GTP-GDP exchange of the Ras and Rap proteins, and was named XRASGRP2. In the present study, we analyzed the expression pattern of xrasgrp2 during Xenopus embryogenesis. The xrasgrp2 mRNA was expressed after stage 24, as assessed by stage PCR analysis. Whole-mount in situ hybridization showed that xrasgrp2 mRNA was located in the vascular region of the embryo. Loss-of-function analysis revealed that the formation of blood and endothelial cells in the explants transplanted into Xenopus embryos was inhibited by antisense morpholino oligonucleotides that block xrasgrp2 translation. These results suggest that XRASGRP2 plays a role in angiogenesis in Xenopus embryos.


Asunto(s)
Embrión no Mamífero/irrigación sanguínea , Neovascularización Fisiológica , Proteínas de Xenopus/fisiología , Xenopus laevis/embriología , Factores de Intercambio de Guanina Nucleótido ras/fisiología , Secuencia de Aminoácidos , Animales , Embrión no Mamífero/metabolismo , Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , Datos de Secuencia Molecular , Neovascularización Fisiológica/genética , Oligonucleótidos Antisentido/genética , Filogenia , ARN Mensajero/metabolismo , Proteínas de Xenopus/clasificación , Proteínas de Xenopus/genética , Xenopus laevis/metabolismo , Factores de Intercambio de Guanina Nucleótido ras/clasificación , Factores de Intercambio de Guanina Nucleótido ras/genética
15.
J Cell Biol ; 181(5): 747-60, 2008 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-18504297

RESUMEN

Ras guanine nucleotide exchange factor (GEF) Q, a nucleotide exchange factor from Dictyostelium discoideum, is a 143-kD protein containing RasGEF domains and a DEP domain. We show that RasGEF Q can bind to F-actin, has the potential to form complexes with myosin heavy chain kinase (MHCK) A that contain active RasB, and is the predominant exchange factor for RasB. Overexpression of the RasGEF Q GEF domain activates RasB, causes enhanced recruitment of MHCK A to the cortex, and leads to cytokinesis defects in suspension, phenocopying cells expressing constitutively active RasB, and myosin-null mutants. RasGEF Q(-) mutants have defects in cell sorting and slug migration during later stages of development, in addition to cell polarity defects. Furthermore, RasGEF Q(-) mutants have increased levels of unphosphorylated myosin II, resulting in myosin II overassembly. Collectively, our results suggest that starvation signals through RasGEF Q to activate RasB, which then regulates processes requiring myosin II.


Asunto(s)
Dictyostelium/metabolismo , Miosina Tipo II/química , Miosinas/fisiología , Factores de Intercambio de Guanina Nucleótido ras/fisiología , Proteínas ras/química , Proteínas ras/metabolismo , Proteínas ras/fisiología , Actinas/química , Animales , Quimiotaxis , AMP Cíclico/metabolismo , Técnicas Genéticas , Luz , Modelos Biológicos , Miosinas/química , Fosforilación , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/química , Factores de Intercambio de Guanina Nucleótido ras/metabolismo
16.
Clin Cancer Res ; 12(22): 6611-6, 2006 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17121879

RESUMEN

PURPOSE AND EXPERIMENTAL DESIGN: To identify novel molecular targets for the treatment of intrahepatic cholangiocarcinoma (ICC), the second most common type of primary hepatobiliary cancer, we earlier analyzed genome-wide expression profiles of genes in 25 ICCs. Among the genes whose expression levels were commonly elevated in the tumors, we identified a novel gene termed RASGEF1A that encodes a putative Ras guanine nucleotide exchange factor domain-containing protein. RESULTS: We showed in this article that RASGEF1A protein has a guanine nucleotide exchange activity to K-RAS, H-RAS, and N-RAS proteins in vitro. Consistently, exogenous RASGEF1A expression increased the activity of Ras. In addition, suppression of RASGEF1A by small interfering RNA retarded the growth of cholangiocarcinoma cells. Interestingly, COS7 cells expressing exogenous RASGEF1A showed enhanced cellular motility in Transwell and wound-healing assays. CONCLUSIONS: These data suggest that elevated expression of RASGEF1A may play an essential role for proliferation and progression of ICC. Our data indicate that RASGEF1A may be a promising therapeutic target for the majority of ICCs.


Asunto(s)
Neoplasias de los Conductos Biliares/metabolismo , Neoplasias de los Conductos Biliares/patología , Movimiento Celular , Proliferación Celular , Colangiocarcinoma/metabolismo , Colangiocarcinoma/patología , Factores de Intercambio de Guanina Nucleótido ras/metabolismo , Factores de Intercambio de Guanina Nucleótido ras/fisiología , Animales , Conductos Biliares Intrahepáticos/citología , Conductos Biliares Intrahepáticos/metabolismo , Células COS , Chlorocebus aethiops , Regulación Neoplásica de la Expresión Génica , Guanosina Difosfato/metabolismo , Humanos , Ratones , Células 3T3 NIH , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/fisiología , Proteína Oncogénica p21(ras)/metabolismo
17.
Biochem Soc Trans ; 34(Pt 5): 858-61, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17052215

RESUMEN

RasGRPs (guanine nucleotide releasing proteins) are a family of four GEFs (guanine nucleotide-exchange factors) (Ras GEFs) that positively regulate Ras and related small GTPases. RasGRP1 possesses a catalytic region consisting of a REM (Ras exchange motif) and a CDC25 (cell division cycle 25) domain. RasGRP1 also possesses a DAG (diacylglycerol)-binding C1 domain and a pair of EF hands that bind calcium. RasGRP1 is selectively expressed in lymphocytes as well as in some cells of the brain, kidney and skin. Functional analysis supports the hypothesis that RasGRP1 serves to couple TCR (T-cell receptor) stimulation and phospholipase C activation with Ras signalling. In B-cells, both RasGRP1 and RasGRP3 play a similar role downstream of the B-cell receptor. RasGRP2 acts on the Ras-related protein Rap and functions in platelet adhesion. RasGRP4 is expressed in mast cells and certain myeloid leukaemia cells. Membrane DAG regulates RasGRPs directly by recruitment to cellular membranes, as well as indirectly by protein kinase C-mediated phosphorylation. The properties of RasGRPs provide a novel view of Ras regulation in lymphocytes and explain several earlier observations. Many experimental results obtained with DAG analogues could be reviewed in light of these findings.


Asunto(s)
Linfocitos/fisiología , Factores de Intercambio de Guanina Nucleótido ras/fisiología , Proteínas de Unión al ADN/fisiología , Factores de Intercambio de Guanina Nucleótido/fisiología , Humanos , Procesamiento Proteico-Postraduccional , Receptores de Antígenos de Linfocitos T/fisiología , Transducción de Señal
18.
Biochem Biophys Res Commun ; 348(3): 908-15, 2006 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-16899220

RESUMEN

Mg2+ is essential for guanosine triphosphatase activity and plays key roles in guanine nucleotide binding and preserving the structural integrity of GTP-binding proteins. To understand the structural basis for Mg2+ function during the GDP/GTP exchange process, we determined the crystal structure of Delta9-Sar1-GDP at low Mg2+ concentration at 1.8A. Two Sar1-GDP molecules in the crystal form a dimer with Mg2+ presenting only in molecule B but not in molecule A. The absence of Mg2+ induces significant conformational changes in the switch I region in molecule A that shows similarities with those of Ha-Ras bound to Sos. The current structure reveals an important regulatory role for Mg2+. We suggest that guanine nucleotide exchange factor may utilize this feature to generate an open conformation for GDP/GTP exchange. Furthermore, we propose a mechanism for COPII assembly and disassembly in which dimerization of Sar1 plays an important role.


Asunto(s)
Guanosina Difosfato/química , Magnesio/química , Factores de Intercambio de Guanina Nucleótido ras/química , Proteínas ras/química , Animales , Vesículas Cubiertas por Proteínas de Revestimiento/metabolismo , Cricetinae , Cricetulus , Cristalización , Cristalografía por Rayos X , Dimerización , Guanosina Difosfato/metabolismo , Magnesio/fisiología , Conformación Proteica , Pliegue de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Factores de Intercambio de Guanina Nucleótido ras/fisiología , Proteínas ras/fisiología
19.
J Biol Chem ; 281(33): 23367-76, 2006 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-16769729

RESUMEN

The regulation of cell polarity plays an important role in chemotaxis. GbpD, a putative nucleotide exchange factor for small G-proteins of the Ras family, has been implicated in adhesion, cell polarity, and chemotaxis in Dictyostelium. Cells overexpressing GbpD are flat, exhibit strongly increased cell-substrate attachment, and extend many bifurcated and lateral pseudopodia. These cells overexpressing GbpD are severely impaired in chemotaxis, most likely due to the induction of many protrusions rather than an enhanced adhesion. The GbpD-overexpression phenotype is similar to that of cells overexpressing Rap1. Here we demonstrate that GbpD activates Rap1 both in vivo and in vitro but not any of the five other characterized Ras proteins. In a screen for Rap1 effectors, we overexpressed GbpD in several mutants defective in adhesion or cell polarity and identified Phg2 as Rap1 effector necessary for adhesion, but not cell polarity. Phg2, a serine/threonine-specific kinase, directly interacts with Rap1 via its Ras association domain.


Asunto(s)
Polaridad Celular/fisiología , Dictyostelium/química , Transducción de Señal/fisiología , Proteínas de Unión al GTP rap1/química , Proteínas de Unión al GTP rap1/fisiología , Factores de Intercambio de Guanina Nucleótido ras/química , Animales , Adhesión Celular/genética , Adhesión Celular/fisiología , Línea Celular , Polaridad Celular/genética , Quimiotaxis/genética , Quimiotaxis/fisiología , Dictyostelium/genética , Dictyostelium/fisiología , Seudópodos/metabolismo , Seudópodos/fisiología , Transducción de Señal/genética , Proteínas de Unión al GTP rap1/genética , Factores de Intercambio de Guanina Nucleótido ras/genética , Factores de Intercambio de Guanina Nucleótido ras/fisiología
20.
J Biol Chem ; 281(15): 10598-609, 2006 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-16423831

RESUMEN

E-cadherin is a key cell-cell adhesion molecule at adherens junctions (AJs) and undergoes endocytosis when AJs are disrupted by the action of an extracellular signal, such as hepatocyte growth factor (HGF)/scatter factor. Rab5 small G protein has been implicated in the HGF-induced endocytosis of E-cadherin, but the molecular mechanism for the regulation of Rab5 activity remains unknown. We first studied this mechanism by using the cell-free assay system for the endocytosis of E-cadherin of the AJ-enriched fraction from rat livers. HGF induced activation of Ras small G protein, which then bound to RIN2, a Rab5 GDP/GTP exchange factor with the Vps9p-like guanine nucleotide exchange factor and Ras association domains, and activated it. Activated RIN2 then activated Rab5, eventually inducing the endocytosis of E-cadherin. We then studied whether RIN2 was involved in the HGF-induced endocytosis of E-cadherin in intact Madin-Darby canine kidney cells. RIN2 localized at the cell-cell adhesion sites, and its guanine nucleotide exchange factor activity was required for the HGF-induced endocytosis of E-cadherin in Madin-Darby canine kidney cells. These results indicate that RIN2 connects Ras to Rab5 in the HGF-induced endocytosis of E-cadherin.


Asunto(s)
Cadherinas/química , Proteínas Portadoras/química , Factores de Intercambio de Guanina Nucleótido/química , Factor de Crecimiento de Hepatocito/metabolismo , Proteínas de Unión al GTP rab5/química , Factores de Intercambio de Guanina Nucleótido ras/química , Factores de Intercambio de Guanina Nucleótido ras/fisiología , Animales , Cadherinas/metabolismo , Adhesión Celular , Línea Celular , Citosol/metabolismo , Perros , Relación Dosis-Respuesta a Droga , Endocitosis , Vectores Genéticos , Glutatión Transferasa/metabolismo , Modelos Biológicos , Unión Proteica , Estructura Terciaria de Proteína , Interferencia de ARN , Ratas , Proteínas de Saccharomyces cerevisiae/metabolismo , Temperatura , Factores de Tiempo , Transfección , Proteínas de Transporte Vesicular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...