Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neuroreport ; 32(12): 1035-1040, 2021 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-34232127

RESUMO

Drug addiction is a chronic recurrent brain disease characterized by compulsive drug use and a high tendency to relapse. We previously reported that the Ras-extracellular signal-regulated kinase (ERK)-ΔFosB pathway in the caudate putamen (CPu) was involved in methamphetamine-induced behavioral sensitization. Rap1, as an antagonist of Ras originally, was found to participate in neuronal synaptic plasticity recently, but the role of Rap1 in methamphetamine addiction is unclear. First, in this study, we constructed the acquisition, extinction and reinstatement of methamphetamine-induced conditioned place preference (CPP) in mice, respectively. Then, protein levels of Rap1, Ras and pERK/ERK in the prefrontal cortex (PFc), CPu and hippocampus of CPP mice on three phases were detected. We found that protein levels of Rap1, Ras and pERK/ERK in the CPu were significantly increased after repeated methamphetamine administration, as well as Rap1 and pERK/ERK in the hippocampus. However, protein levels of Rap1 and pERK/ERK in the CPu were decreased on the reinstatement of CPP mice. Therefore, Rap1 and Ras in the CPu and Rap1 in the hippocampus may participate in the regulation of the acquisition of methamphetamine-induced CPP in mice by activating ERK. Moreover, Rap1-ERK cascade in the CPu contributes to both the acquisition and reinstatement of methamphetamine-induced CPP in mice.


Assuntos
Estimulantes do Sistema Nervoso Central/farmacologia , Condicionamento Operante/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Metanfetamina/farmacologia , Proteínas rap1 de Ligação ao GTP/biossíntese , Proteínas ras/biossíntese , Animais , Condicionamento Operante/fisiologia , Expressão Gênica , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Proteínas rap1 de Ligação ao GTP/genética , Proteínas ras/genética
2.
J Exp Clin Cancer Res ; 37(1): 178, 2018 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-30064475

RESUMO

BACKGROUND: Colorectal cancer (CRC) is a commonly diagnosed digestive malignancy worldwide. Ras-related protein 1A (RAP1A) is a member of the Ras superfamily of small GTPases and has been recently identified as a novel oncoprotein in several human malignancies. However, its specific role in CRC remains unclear. METHOD: In this study, we firstly analyzed its expression and clinical significance in a retrospective cohort of 144 CRC patients. Then, cellular assays in vitro and in vivo were performed to clarify its biological role in CRC cells. Finally, microarray analysis was utilized to investigate the molecular mechanisms regulated by RAP1A in CRC progression. RESULTS: Firstly, RAP1A expression was abnormally higher in CRC tissues as compared with adjacent normal tissues, and significantly correlated tumor invasion. High RAP1A expression was an independent unfavourable prognostic factor for CRC patients. Combining RAP1A expression and preoperative CEA level contributed to a more accurate prognostic stratification in CRC patients. Secondly, knockdown of RAP1A dramatically inhibited the growth of CRC cells, while it was opposite for RAP1A overexpression. Finally, the microarray analysis revealed RAP1A promoted CRC growth partly through phosphatase and tensin homolog (PTEN)/forkhead box O3(FOXO3)/cyclin D1(CCND1) signaling pathway. FOXO3 overexpression could partly mimic the inhibitory effect of RAP1A knockdown in CRC growth. Moreover, FOXO3 overexpression inhibited CCND1 expression, but had no impact on RAP1A and PTEN expression. CONCLUSION: RAP1A promotes CRC development partly through PTEN/FOXO3 /CCND1 signaling pathway. It has a great potential to be an effective clinical biomarker and therapeutic target for CRC patients.


Assuntos
Neoplasias Colorretais/metabolismo , Ciclina D1/metabolismo , Proteína Forkhead Box O3/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Proteínas rap1 de Ligação ao GTP/biossíntese , Estudos de Coortes , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fenótipo , Prognóstico , Estudos Retrospectivos , Transdução de Sinais , Proteínas rap1 de Ligação ao GTP/metabolismo
3.
Curr Eye Res ; 43(7): 934-940, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29601231

RESUMO

PURPOSE: Choroidal neovascularization (CNV) is a common vision-threatening complication associated with many  fundus diseases. The retinal pigment epithelial (RPE) cell junction barrier has critical functions in preventing CNV, and oxidative stress can cause compromise of barrier integrity and induce angiogenesis. Rap1, a small guanosine triphosphatase (GTPase), is involved in regulating endothelial and epithelial cell junctions. In this work, we explored the function and mechanism of Rap1 in CNV in vivo. METHODS: A laser-induced rat CNV model was developed. Rap1 was activated through intravitreal injection of the Rap1 activator 8CPT-2'-O-Me-cAMP (8CPT). At 14 days after laser treatment, CNV size in RPE/choroid flat mounts was measured by fluorescein isothiocyanate-dextran staining. Expression of vascular endothelial growth factor (VEGF) and cell junction proteins in RPE/choroid tissues were analyzed by western blots and quantitative real-time PCR assays. Reactive oxygen species (ROS) in RPE cells were detectedbydichloro-dihydro-fluorescein diacetate assays. The antioxidant apocynin was intraperitoneally injected into rats. RESULTS: Activating Rap1 by 8CPT significantly reduced CNV size and VEGF expression in the rat CNV model. Rap1 activation enhanced protein and mRNA levels of ZO-1 and occludin, two tight junction proteins in the RPE barrier. In addition, reducing ROS generation by injection of apocynin, a NADPH oxidase inhibitor, inhibited CNV formation. Rap1 activation reduced ROS generation and expression of NADPH oxidase 4. CONCLUSIONS: Rap1 activation inhibits CNV through regulating barrier integrity and ROS generation of RPE in vivo, and selectively activating Rap1 may be a way to reduce vision loss from CNV.


Assuntos
Neovascularização de Coroide/tratamento farmacológico , AMP Cíclico/análogos & derivados , Regulação da Expressão Gênica , RNA/genética , Espécies Reativas de Oxigênio/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Proteínas rap1 de Ligação ao GTP/genética , Animais , Western Blotting , Neovascularização de Coroide/genética , Neovascularização de Coroide/metabolismo , AMP Cíclico/farmacologia , Modelos Animais de Doenças , Junções Intercelulares , Injeções Intravítreas , Masculino , Estresse Oxidativo , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Epitélio Pigmentado da Retina/patologia , Transdução de Sinais , Proteínas rap1 de Ligação ao GTP/biossíntese
4.
J Gen Virol ; 95(Pt 9): 2010-2018, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24920725

RESUMO

Coxsackievirus B3 (CVB3) infection can trigger myocarditis and can ultimately lead to dilated cardiomyopathy. It is known that CVB3-induced T-cell infiltration into cardiac tissues is one of the pathological factors causing cardiomyocyte injury by inflammation. However, the underlying mechanism for this remains unclear. We investigated the mechanism of T-cell infiltration by two types of CVB3: the H3 WT strain and the YYFF attenuated strain. T-cell activation was confirmed by changes in the distribution of lymphocyte function-associated antigen-1 (LFA-1). Finally, we identified which viral gene was responsible for LFA-1 activation. CVB3 could infect and activate T-cells in vivo and in vitro, and activated T-cells were detected in CVB3-infected mouse hearts. LFA-1 expressed on the surface of these T-cells had been activated through the cAMP/Rap1 pathway. Recombinant lentiviruses expressing VP2 of CVB3 could also induce LFA-1 activation via an increase in cAMP, whilst VP2 of YYFF did not. These results indicated that CVB3 infection increased cAMP levels and then activated Rap1 in T-cells. In particular, VP2, among the CVB3 proteins, might be critical for this activation. This VP2-cAMP-Rap1-LFA-1 axis could be a potential therapeutic target for treating CVB3-induced myocarditis.


Assuntos
AMP Cíclico/metabolismo , Enterovirus/imunologia , Antígeno-1 Associado à Função Linfocitária/biossíntese , Linfócitos T/imunologia , Proteínas rap1 de Ligação ao GTP/metabolismo , Animais , Cardiomiopatias/imunologia , Cardiomiopatias/virologia , Infecções por Coxsackievirus/imunologia , Infecções por Coxsackievirus/virologia , AMP Cíclico/biossíntese , Enterovirus/genética , Células HeLa , Coração/virologia , Humanos , Molécula 1 de Adesão Intercelular , Células Jurkat , Ativação Linfocitária/imunologia , Antígeno-1 Associado à Função Linfocitária/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Miocardite/imunologia , Miocardite/virologia , Miocárdio/citologia , Miocárdio/imunologia , Receptores Virais/metabolismo , Proteínas rap1 de Ligação ao GTP/biossíntese
5.
J Biol Chem ; 289(25): 17689-98, 2014 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-24790104

RESUMO

Rap1 is a Ras family GTPase with a well documented role in ERK/MAP kinase signaling and integrin activation. Stimulation of the G-protein-coupled receptor PAR-1 with thrombin in human 1321N1 glioblastoma cells led to a robust increase in Rap1 activation. This response was sustained for up to 6 h and mediated through RhoA and phospholipase D (PLD). Thrombin treatment also induced a 5-fold increase in cell adhesion to fibronectin, which was blocked by down-regulating PLD or Rap1A or by treatment with a ß1 integrin neutralizing antibody. In addition, thrombin treatment led to increases in phospho-focal adhesion kinase (tyrosine 397), ERK1/2 phosphorylation and cell proliferation, which were significantly inhibited in cells treated with ß1 integrin antibody or Rap1A siRNA. To assess the role of Rap1A in tumor formation in vivo, we compared growth of 1321N1 cells stably expressing control, Rap1A or Rap1B shRNA in a mouse xenograft model. Deletion of Rap1A, but not of Rap1B, reduced tumor mass by >70% relative to control. Similar observations were made with U373MG glioblastoma cells in which Rap1A was down-regulated. Collectively, these findings implicate a Rap1A/ß1 integrin pathway, activated downstream of G-protein-coupled receptor stimulation and RhoA, in glioblastoma cell proliferation. Moreover, our data demonstrate a critical role for Rap1A in glioblastoma tumor growth in vivo.


Assuntos
Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Glioblastoma/metabolismo , Integrina beta1/metabolismo , Proteínas de Neoplasias/metabolismo , Trombina/metabolismo , Proteínas rap1 de Ligação ao GTP/biossíntese , Animais , Linhagem Celular Tumoral , Glioblastoma/genética , Xenoenxertos , Humanos , Integrina beta1/genética , Camundongos , Proteínas de Neoplasias/genética , Transplante de Neoplasias , Trombina/genética , Proteínas rap1 de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo
6.
Breast Cancer Res ; 13(2): R31, 2011 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-21429211

RESUMO

INTRODUCTION: The adhesion protein junctional adhesion molecule-A (JAM-A) regulates epithelial cell morphology and migration, and its over-expression has recently been linked with increased risk of metastasis in breast cancer patients. As cell migration is an early requirement for tumor metastasis, we sought to identify the JAM-A signalling events regulating migration in breast cancer cells. METHODS: MCF7 breast cancer cells (which express high endogenous levels of JAM-A) and primary cultures from breast cancer patients were used for this study. JAM-A was knocked down in MCF7 cells using siRNA to determine the consequences for cell adhesion, cell migration and the protein expression of various integrin subunits. As we had previously demonstrated a link between the expression of JAM-A and ß1-integrin, we examined activation of the ß1-integrin regulator Rap1 GTPase in response to JAM-A knockdown or functional antagonism. To test whether JAM-A, Rap1 and ß1-integrin lie in a linear pathway, we tested functional inhibitors of all three proteins separately or together in migration assays. Finally we performed immunoprecipitations in MCF7 cells and primary breast cells to determine the binding partners connecting JAM-A to Rap1 activation. RESULTS: JAM-A knockdown in MCF7 breast cancer cells reduced adhesion to, and migration through, the ß1-integrin substrate fibronectin. This was accompanied by reduced protein expression of ß1-integrin and its binding partners αV- and α5-integrin. Rap1 activity was reduced in response to JAM-A knockdown or inhibition, and pharmacological inhibition of Rap1 reduced MCF7 cell migration. No additive anti-migratory effect was observed in response to simultaneous inhibition of JAM-A, Rap1 and ß1-integrin, suggesting that they lie in a linear migratory pathway. Finally, in an attempt to elucidate the binding partners putatively linking JAM-A to Rap1 activation, we have demonstrated the formation of a complex between JAM-A, AF-6 and the Rap1 activator PDZ-GEF2 in MCF7 cells and in primary cultures from breast cancer patients. CONCLUSIONS: Our findings provide compelling evidence of a novel role for JAM-A in driving breast cancer cell migration via activation of Rap1 GTPase and ß1-integrin. We speculate that JAM-A over-expression in some breast cancer patients may represent a novel therapeutic target to reduce the likelihood of metastasis.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Moléculas de Adesão Celular/metabolismo , Receptores de Superfície Celular/metabolismo , Proteínas rap1 de Ligação ao GTP/metabolismo , Adesão Celular , Moléculas de Adesão Celular/genética , Linhagem Celular Tumoral , Movimento Celular , Feminino , Fibronectinas/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Integrina beta1/metabolismo , Cinesinas/metabolismo , Miosinas/metabolismo , Metástase Neoplásica , Proteínas do Tecido Nervoso/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Receptores de Superfície Celular/genética , Transdução de Sinais , Proteínas rap1 de Ligação ao GTP/biossíntese
7.
Exp Hematol ; 35(5): 793-801, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17577928

RESUMO

OBJECTIVE: The interactions between cells and extracellular matrices in the bone marrow microenvironment are critical for normal hematopoiesis, controlling cell survival, proliferation, differentiation, and motility. A number of hematopoietic growth factors and cytokines can mediate these interactions by changing expression and/or activity of specific integrins, or by changing cell shape. Thrombopoietin (TPO) has previously been shown to stimulate adhesion. in certain hematopoietic cell types, although the exact mechanisms by which adhesion is promoted remain elusive. MATERIALS AND METHODS: The role of TPO in hematopoietic cell adhesion was determined with fibronectin adhesion and binding assays, flow cytometry, and immunocytochemistry using the hematopoietic cell line UT-7/TPO and bone marrow-derived primary mouse megakaryocytes. The role of Rapl in TPO-mediated adhesion was determined using a RaplGAP overexpressing UT-7/TPO cell line, in which Rapl could not be activated. RESULTS: We found that TPO promoted hematopoietic cell adhesion by causing cytoskeletal reorganization and not by increasing integrin expression, localization, or affinity, as previously hypothesized. Through studies using the UT-7/TPO-RaplGAP cell line, we found that TPO-mediated cell shape change occurred via activation of Rapl. CONCLUSIONS: These data demonstrate an important role for TPO in mediating interactions in the bone marrow microenvironment and make a significant contribution to our understanding of how TPO may affect hematopoiesis.


Assuntos
Células-Tronco Hematopoéticas/fisiologia , Integrina beta1/fisiologia , Trombopoetina/fisiologia , Proteínas rap1 de Ligação ao GTP/metabolismo , Animais , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Linhagem Celular , Relação Dose-Resposta a Droga , Fibronectinas/metabolismo , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/ultraestrutura , Humanos , Integrina beta1/efeitos dos fármacos , Megacariócitos/efeitos dos fármacos , Megacariócitos/fisiologia , Megacariócitos/ultraestrutura , Camundongos , Trombopoetina/farmacologia , Proteínas rap1 de Ligação ao GTP/biossíntese , Proteínas rap1 de Ligação ao GTP/efeitos dos fármacos
8.
J Biol Chem ; 280(6): 5022-31, 2005 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-15569673

RESUMO

Endothelial cell migration is promoted by chemoattractants and is accompanied with microtubule extension toward the leading edge. Cytoskeletal microtubules polarize to function as rails for delivering a variety of molecules by motor proteins during cell migration. It remains, however, unclear how directional migration with polarized extension of microtubules is regulated. Here we report that Rap1 controls the migration of vascular endothelial cells. We found that Rap1-associating molecule, RAPL, which belongs to the Ras association domain family (Rassf), localized on microtubules and that activated Rap1 induced dissociation of RAPL from microtubules. A Rap1 activation-monitoring probe based on the fluorescence resonance energy transfer enabled us to demonstrate that local Rap1 activation occurs at the leading edge of the cells under the two types of cell migration, chemotaxis and wound healing. Time lapse imaging of microtubules marked by enhanced green fluorescent protein-RAPL showed the directional growth of microtubules toward the leading edge of the migrating cells. Using adenovirus, inactivation of Rap1 by expression of rap1GAPII inhibited wound healing. In addition, disconnection of Rap1 and RAPL by expression of a RAPL mutant also perturbed wound healing. Collectively, the locally activated Rap1 and its association with RAPL controls the directional migration of vascular endothelial cells.


Assuntos
Endotélio Vascular/citologia , Microtúbulos/metabolismo , Proteínas rap1 de Ligação ao GTP/biossíntese , Proteínas rap1 de Ligação ao GTP/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Adenoviridae/genética , Proteínas Reguladoras de Apoptose , Linhagem Celular , Movimento Celular , Células Cultivadas , Citoesqueleto/metabolismo , Transferência Ressonante de Energia de Fluorescência , Proteínas de Fluorescência Verde/metabolismo , Humanos , Immunoblotting , Imunoprecipitação , Microscopia Confocal , Microscopia de Fluorescência , Microtúbulos/ultraestrutura , Modelos Biológicos , Proteínas Monoméricas de Ligação ao GTP , Plasmídeos/metabolismo , Ligação Proteica , RNA Interferente Pequeno/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Cicatrização
9.
Neuroscience ; 127(4): 973-81, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15312909

RESUMO

Effects of two small G-proteins, Rap1 and Ras, on the sodium channel activity in NG108-15 cells were studied using sindbis virus-mediated gene transfer. When an activated Rap1A mutant (Rap1-12V, the activated mutant of Rap1 carrying glycine to valine substitution at codon 12) or a dominant-negative H-Ras mutant (Ras-17N, carrying serine to asparagine substitution at codon 17) was expressed in differentiated NG108-15 cells, the proportion of cells generating action potential decreased and the amplitudes of sodium current diminished. This effect was sensitive to an inhibitor of protein kinase A. The effects of a cyclic AMP (cAMP) analog (dibutyl cAMP) on sodium current in these cells were biphasic: inhibitory at lower concentrations (<100 microM) and enhancing at higher concentrations (200-500 microM). The inhibitory phase of cAMP effect was suppressed by an activated Ras mutant (Ras-12V) while the enhancing phase was suppressed by Rap1-12V. These data are consistent with the model that Rap1 and Ras function as counteracting regulators of voltage-gated sodium current through cAMP-dependent mechanisms.


Assuntos
Potenciais de Ação/fisiologia , Proteínas rap1 de Ligação ao GTP/biossíntese , Proteínas ras/biossíntese , Animais , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Camundongos , Proteínas rap1 de Ligação ao GTP/genética , Proteínas ras/genética
10.
Neuroreport ; 14(9): 1203-7, 2003 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-12824760

RESUMO

Rap1A, first identified as a suppressor of transformed phenotype induced by an activated ras oncogene, is abundantly expressed in the brain. Its neurophysiological function, however, is poorly understood. When an activated Rap1A mutant (Rap1-12V) or a dominant negative H-Ras mutant (Ras-17N) was expressed in CA1 neurons in cultured hippocampal slices using the sindbis virus-mediated gene transfer technique, NMDA receptor current in response to Schaffer collateral stimulation was suppressed. Expression of activated H-Ras mutant (Ras-12V) resulted in the elevation of both NMDA receptor current and AMPA receptor current. These results implicate counteracting functions of Ras and Rap1 in the regulation of NMDA receptor-mediated synaptic transmission and a positive regulatory role of Ras in AMPA receptor-mediated synaptic transmission.


Assuntos
Hipocampo/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transmissão Sináptica/fisiologia , Proteínas rap1 de Ligação ao GTP/fisiologia , Proteínas ras/fisiologia , Animais , Genes ras/fisiologia , Humanos , Masculino , Mutação/fisiologia , Técnicas de Cultura de Órgãos , Ratos , Ratos Wistar , Proteínas rap1 de Ligação ao GTP/biossíntese , Proteínas rap1 de Ligação ao GTP/genética , Proteínas ras/biossíntese , Proteínas ras/genética
11.
EMBO J ; 22(4): 859-69, 2003 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-12574122

RESUMO

Epidermal growth factor (EGF) activates Ras and Rap1 at distinct intracellular regions. Here, we explored the mechanism underlying this phenomenon. We originally noticed that in cells expressing Epac, a cAMP-dependent Rap1 GEF (guanine nucleotide exchange factor), cAMP activated Rap1 at the perinuclear region, as did EGF. However, in cells expressing e-GRF, a recombinant cAMP-responsive Ras GEF, cAMP activated Ras at the peripheral plasma membrane. Based on the uniform cytoplasmic expression of Epac and e-GRF, GEF did not appear to account for the non-uniform increase in the activities of Ras and Rap1. In contrast, when we used probes with reduced sensitivity to GTPase-activating proteins (GAPs), both Ras and Rap1 appeared to be activated uniformly in the EGF-stimulated cells. Furthermore, we calculated the local rate constants of GEFs and GAPs from the video images of Ras activation and found that GAP activity was higher at the central plasma membrane than the periphery. Thus we propose that GAP primarily dictates the spatial regulation of Ras family G proteins, whereas GEF primarily determines the timing of Ras activation.


Assuntos
Regulação da Expressão Gênica/fisiologia , Proteínas rap1 de Ligação ao GTP/genética , Proteínas ras/genética , Animais , Células COS , AMP Cíclico/metabolismo , Endocitose/fisiologia , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas rap1 de Ligação ao GTP/biossíntese , Proteínas ras/biossíntese
12.
Int J Oncol ; 22(1): 195-200, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12469204

RESUMO

Increased small GTPase protein mitogenic signaling is common in tumors. We have previously demonstrated that sporadic astrocytomas exhibit high levels of activated Ras important for tumor growth. Individuals with tuberous sclerosis complex (TSC) develop astrocytoma-like tumors resulting from mutations in the TSC2 protein, tuberin, which is hypothesized to function as a Rap1 GTPase activating protein (GAP). Since we have previously reported that high-grade astrocytomas frequently exhibit loss of tuberin expression or increased Rap1 levels, we sought to determine whether there is a correlation between decreased tuberin Rap1-GAP function or Rap1 overexpression and tumor Rap1 activity. In this study, we compared levels of Rap1-GTP, Rap1 and tuberin levels in normal brain tissue and 24 grade II-IV astrocytoma specimens. Whereas Rap1 overexpression was observed in astrocytomas of all malignancy grades, tuberin loss was seen most frequently in the higher-grade astrocytomas. In the grade IV glioblastoma multiforme tumors, Rap1 activity was 2-3-fold higher than in lower grade or non-neoplastic brain. However, there was no correlation between tuberin expression or Rap1 overexpression and the levels of Rap1 activity in the tumors studied, suggesting that the increased Rap1 activation is not the direct result of reduced tuberin Rap1-GAP function or elevated Rap1 protein expression.


Assuntos
Astrocitoma/metabolismo , Proteínas Repressoras/genética , Proteínas rap1 de Ligação ao GTP/biossíntese , Astrocitoma/patologia , Expressão Gênica , Guanosina Trifosfato/metabolismo , Humanos , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor
13.
Physiol Chem Phys Med NMR ; 34(1): 1-15, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12403272

RESUMO

It has been reported that inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase suppress cell proliferation and induce apoptosis. One inhibitor which induces apoptosis is mevastatin. However, the molecular mechanism of apoptosis induction is not well understood so the effects of mevastatin on various functions of HL-60 cells were investigated. We confirmed that mevastatin activated caspase-3 by release of cytochrome c (Cyt. c) from mitochondria through a membrane permeability transition mechanism and also induced typical fragmentation and ladder formation of DNA in HL-60 cells. These effects were inhibited by mevalonate, a metabolic intermediate of cholesterol biosynthesis. Mevalonate and geranylgeraniol (GGOH) inhibited DNA fragmentation whereas farnesol (FOH) did not. Mevastatin also induced cell differentiation to monocytic cells via a mevalonate inhibitable mechanism. Furthermore, mevastatin decreased the amount of an isoprenylated membrane bound Rap1 small GTPase concomitant with an increase in cytosolic Rap1 which occurred before apoptosis and differentiation. On the contrary, both mevastatin and geranylgeranylacetone (GGA), which competes with geranylgeranyl pyrophosphate, induced membrane depolarization of isolated mitochondria without swelling and Cyt. c release. These results suggest that mevastatin-induced apoptosis of HL-60 cells might be caused indirectly by activation of the caspase cascade through the modulation of mitochondrial functions and that some relationship between a certain small GTPase molecule, such as Rap1, and mevastatin-induced apoptosis may exist.


Assuntos
Apoptose/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Lovastatina/análogos & derivados , Lovastatina/administração & dosagem , Mitocôndrias/efeitos dos fármacos , Proteínas rap1 de Ligação ao GTP/biossíntese , Caspase 1/metabolismo , Caspase 3 , Caspases/metabolismo , Grupo dos Citocromos c/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática , Células HL-60/citologia , Células HL-60/efeitos dos fármacos , Células HL-60/fisiologia , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/administração & dosagem , Potenciais da Membrana/efeitos dos fármacos , Mitocôndrias/fisiologia , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
14.
Acta Neuropathol ; 104(3): 231-40, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12172908

RESUMO

Cavernous malformations are vascular anomalies that can cause severe neurological deficits, seizures and hemorrhagic stroke if these lesions are located in the brain. In patients with cavernomas, constitutional mutations of the KRIT1 gene have been identified. The pathogenetic mechanisms leading to cerebral cavernous malformations (CCM) development are poorly understood. CCM development might be induced in utero owing to the underlying KRIT1 defect, and is triggered by environmental factors. Another model suggests that CCM develop according to the two-hit model of tumorigenesis associated with biallelic inactivation of KRIT1. So far, CCM specimens themselves have not been subjected to mutation analysis. We identified two somatic mutations in the cavernoma of a sporadic case, suggesting that pathogenesis is associated with somatic KRIT1 alterations. To gain a better understanding of the role of KRIT1 during morphogenesis, the main goal of this study was to provide a detailed description of the spatio-temporal expression pattern of Krit1 and its interaction partner Rap1A during mouse embryogenesis. We did not observe enhanced expression of either gene in the heart or large vessels; however, their expression in the developing small vessels or capillaries could not be assessed by the methods applied. At early embryonic stages, Krit1 and, to a lesser extent, Rap1A are expressed in the developing nervous system. During later phases of fetal development, specific expression of both genes is observed in regions of ossification, the dermis, tendons and in the meninges. These findings provide evidence of differential Krit1 and Rap1A expression during mouse ontogenesis and suggest a more widespread functional significance of Krit1, not restricted to vascular endothelial cells.


Assuntos
Malformações Vasculares do Sistema Nervoso Central/genética , Desenvolvimento Embrionário e Fetal/genética , Proteínas Associadas aos Microtúbulos/genética , Proteínas Proto-Oncogênicas/genética , Proteínas rap1 de Ligação ao GTP/biossíntese , Proteínas rap1 de Ligação ao GTP/genética , Adulto , Animais , Animais Recém-Nascidos , Humanos , Hibridização In Situ , Proteína KRIT1 , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Especificidade de Órgãos , Mutação Puntual , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
J Neurosci ; 21(22): 8842-53, 2001 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-11698596

RESUMO

The vasoconstrictor peptide endothelin (ET-1) exerts its physiological and pathological effects via activation of ET(A) and ET(B) receptor (ET-R) subtypes. In this study, we demonstrate that both ET-R subtypes are highly expressed in rat astrocytes in vivo, indicating that these cells are potential targets of the biological effects of ET-1 in the brain. In cultured cortical astrocytes, both ET-R subtypes are expressed, and selective stimulation of ET(B)-R with ET-1 induces phosphorylation of cAMP response element-binding protein (CREB). The signal transduction pathway activated by ET-1 includes the Rap1/B-Raf and the Ras/Raf-1 complexes, protein kinase C (PKC) together with extracellular signal-regulated kinases (ERK), and the ribosomal S6 kinase (RSK) isoforms RSK2 and RSK3, two kinases that lie immediately downstream of ERK and are able to phosphorylate CREB. Moreover, ET-1 activates the p38 mitogen-activated protein kinase (MAPK)-dependent, but not the c-jun N-terminal kinase (JNK)-dependent pathway. By using selective protein kinase inhibitors and expression of dominant-negative Rap1 protein, we also found that the Rap1/PKC/ERK-dependent pathway induces the phosphorylation of activating transcription factor-1, CREB, and Elk-1, whereas the p38MAPK-dependent pathway only causes CREB phosphorylation. ET-1-induced transcription of the immediate early gene c-fos requires the concomitant activation of both the PKC/ERK- and p38MAPK-dependent pathways, because inhibitors of either pathway block the ET-1-induced increase of c-fos mRNA. Our findings indicate that changes in the expression of cAMP response element-dependent immediate and delayed response genes could play a pivotal role in the physiological effects elicited by ET-1 in astrocytes.


Assuntos
Astrócitos/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas de Ligação a DNA , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-fos/biossíntese , Receptores de Endotelina/metabolismo , Transdução de Sinais/fisiologia , Fator 1 Ativador da Transcrição , Animais , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Células Cultivadas , Endotelina-1/farmacologia , Fosforilação/efeitos dos fármacos , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas B-raf , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-raf/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor de Endotelina A , Receptor de Endotelina B , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno , Proteínas rap1 de Ligação ao GTP/biossíntese , Proteínas rap1 de Ligação ao GTP/genética
16.
Mol Cell Biol ; 20(3): 779-85, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10629034

RESUMO

Rap1, a small GTPase of the Ras family, is ubiquitously expressed and particularly abundant in platelets. Previously we have shown that Rap1 is rapidly activated after stimulation of human platelets with alpha-thrombin. For this activation, a phospholipase C-mediated increase in intracellular calcium is necessary and sufficient. Here we show that thrombin induces a second phase of Rap1 activation, which is mediated by protein kinase C (PKC). Indeed, the PKC activator phorbol 12-myristate 13-acetate induced Rap1 activation, whereas the PKC-inhibitor bisindolylmaleimide inhibited the second, but not the first, phase of Rap1 activation. Activation of the integrin alpha(IIb)beta(3), a downstream target of PKC, with monoclonal antibody LIBS-6 also induced Rap1 activation. However, studies with alpha(IIb)beta(3)-deficient platelets from patients with Glanzmann's thrombasthenia type 1 show that alpha(IIb)beta(3) is not essential for Rap1 activation. Interestingly, induction of platelet aggregation by thrombin resulted in the inhibition of Rap1 activation. This downregulation correlated with the translocation of Rap1 to the Triton X-100-insoluble, cytoskeletal fraction. We conclude that in platelets, alpha-thrombin induces Rap1 activation first by a calcium-mediated pathway independently of PKC and then by a second activation phase mediated by PKC and, in part, integrin alpha(IIb)beta(3). Inactivation of Rap1 is mediated by an aggregation-dependent process that correlates with the translocation of Rap1 to the cytoskeletal fraction.


Assuntos
Plaquetas/metabolismo , Agregação Plaquetária/fisiologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/fisiologia , Trombastenia/sangue , Proteínas rap1 de Ligação ao GTP/sangue , Anticorpos Monoclonais/farmacologia , Plaquetas/efeitos dos fármacos , Cálcio/sangue , Citoesqueleto/metabolismo , Humanos , Técnicas In Vitro , Indóis/farmacologia , Maleimidas/farmacologia , Fator de Ativação de Plaquetas/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Proteína Quinase C/sangue , Valores de Referência , Acetato de Tetradecanoilforbol/farmacologia , Trombina/farmacologia , Fosfolipases Tipo C/sangue , Proteínas rap1 de Ligação ao GTP/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...