Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 117(50): 31993-32004, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-33262282

RESUMO

Effective cancer prevention requires the discovery and intervention of a factor critical to cancer development. Here we show that ovarian progesterone is a crucial endogenous factor inducing the development of primary tumors progressing to metastatic ovarian cancer in a mouse model of high-grade serous carcinoma (HGSC), the most common and deadliest ovarian cancer type. Blocking progesterone signaling by the pharmacologic inhibitor mifepristone or by genetic deletion of the progesterone receptor (PR) effectively suppressed HGSC development and its peritoneal metastases. Strikingly, mifepristone treatment profoundly improved mouse survival (∼18 human years). Hence, targeting progesterone/PR signaling could offer an effective chemopreventive strategy, particularly in high-risk populations of women carrying a deleterious mutation in the BRCA gene.


Assuntos
Proteína BRCA1/genética , Cistadenocarcinoma Seroso/prevenção & controle , Mifepristona/farmacologia , Neoplasias Ovarianas/prevenção & controle , Progesterona/antagonistas & inibidores , Adulto , Animais , Mama/patologia , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Neoplasias da Mama/prevenção & controle , Cistadenocarcinoma Seroso/química , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/patologia , Modelos Animais de Doenças , Estradiol/administração & dosagem , Feminino , Humanos , Camundongos , Pessoa de Meia-Idade , Mifepristona/uso terapêutico , Mutação , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Neoplasias Experimentais/prevenção & controle , Neoplasias Ovarianas/induzido quimicamente , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Ovário/patologia , Ovário/cirurgia , Progesterona/administração & dosagem , Progesterona/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Salpingo-Ooforectomia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
2.
Biochim Biophys Acta ; 1852(5): 778-91, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25545047

RESUMO

Protective effects of prostacyclin (PC) or its stable analog beraprost against agonist-induced lung vascular inflammation have been associated with elevation of intracellular cAMP and Rac GTPase signaling which inhibited the RhoA GTPase-dependent pathway of endothelial barrier dysfunction. This study investigated a distinct mechanism of PC-stimulated lung vascular endothelial (EC) barrier recovery and resolution of LPS-induced inflammation mediated by small GTPase Rap1. Efficient barrier recovery was observed in LPS-challenged pulmonary EC after prostacyclin administration even after 15 h of initial inflammatory insult and was accompanied by the significant attenuation of p38 MAP kinase and NFκB signaling and decreased production of IL-8 and soluble ICAM1. These effects were reproduced in cells post-treated with 8CPT, a small molecule activator of Rap1-specific nucleotide exchange factor Epac. By contrast, pharmacologic Epac inhibitor, Rap1 knockdown, or knockdown of cell junction-associated Rap1 effector afadin attenuated EC recovery caused by PC or 8CPT post-treatment. The key role of Rap1 in lung barrier restoration was further confirmed in the murine model of LPS-induced acute lung injury. Lung injury was monitored by measurements of bronchoalveolar lavage protein content, cell count, and Evans blue extravasation and live imaging of vascular leak over 6 days using a fluorescent tracer. The data showed significant acceleration of lung recovery by PC and 8CPT post-treatment, which was abrogated in Rap1a(-/-) mice. These results suggest that post-treatment with PC triggers the Epac/Rap1/afadin-dependent mechanism of endothelial barrier restoration and downregulation of p38MAPK and NFκB inflammatory cascades, altogether leading to accelerated lung recovery.


Assuntos
Lesão Pulmonar Aguda/prevenção & controle , Endotélio Vascular/efeitos dos fármacos , Epoprostenol/farmacologia , Proteínas rap1 de Ligação ao GTP/metabolismo , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/genética , Animais , Permeabilidade da Membrana Celular/efeitos dos fármacos , Células Cultivadas , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Epoprostenol/análogos & derivados , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Immunoblotting , Molécula 1 de Adesão Intercelular/metabolismo , Interleucina-8/metabolismo , Lipopolissacarídeos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , NF-kappa B/metabolismo , Inibidores da Agregação Plaquetária/farmacologia , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas rap1 de Ligação ao GTP/genética
3.
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
4.
FASEB J ; 28(1): 265-74, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24043260

RESUMO

Activation of Rap1 GTPase can improve the integrity of the barrier of the retina pigment epithelium (RPE) and reduce choroidal neovascularization (CNV). Inhibition of NADPH oxidase activation also reduces CNV. We hypothesize that Rap1 inhibits NADPH oxidase-generated ROS and thereby reduces CNV formation. Using a murine model of laser-induced CNV, we determined that reduced Rap1 activity in RPE/choroid occurred with CNV formation and that activation of Rap1 by 2'-O-Me-cAMP (8CPT)-reduced laser-induced CNV via inhibiting NADPH oxidase-generated ROS. In RPE, inhibition of Rap1 by Rap1 GTPase-activating protein (Rap1GAP) increased ROS generation, whereas activation of Rap1 by 8CPT reduced ROS by interfering with the assembly of NADPH oxidase membrane subunit p22phox with NOX4 or cytoplasmic subunit p47phox. Activation of NADPH oxidase with Rap1GAP reduced RPE barrier integrity via cadherin phosphorylation and facilitated choroidal EC migration across the RPE monolayer. Rap1GAP-induced ROS generation was inhibited by active Rap1a, but not Rap1b, and activation of Rap1a by 8CPT in Rap1b(-/-) mice reduced laser-induced CNV, in correlation with decreased ROS generation in RPE/choroid. These findings provide evidence that active Rap1 reduces CNV by interfering with the assembly of NADPH oxidase subunits and increasing the integrity of the RPE barrier.


Assuntos
Neovascularização de Coroide/metabolismo , NADPH Oxidases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Proteínas rap1 de Ligação ao GTP/metabolismo , Animais , Camundongos , Camundongos Knockout , Oxirredução
5.
J Biol Chem ; 288(22): 15687-98, 2013 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-23612979

RESUMO

Many factors, including duration and intensity of the unfolded protein response (UPR), dictate whether cells will adapt to endoplasmic reticulum stress or undergo apoptosis. In tuberous sclerosis (TSC), elevation of mammalian target of rapamycin complex 1 (mTORC1) activity has been proposed to compound the induction of UPR transcription factors ATF4 and CHOP, suggesting that the UPR could be targeted to eradicate TSC1/2-null cells during patient therapy. Here we report that control of c-MYC translation by mTORC1 plays a key role in determining whether TSC2-null Elt3 rat leiomyoma cells apoptose in response to UPR induction by the proteasome inhibitor bortezomib. Although bortezomib induces eukaryotic initiating factor 2α phosphorylation, mTORC1 activity was also required for downstream induction of the UPR transcription factors ATF4 and CHOP by a mechanism involving increased expression of c-MYC. Although bortezomib-induced c-MYC transcription was resistant to rapamycin treatment, mTORC1 activity was required for efficient c-MYC translation. c-MYC subsequently bound to the ATF4 promoter, suggesting direct involvement of an mTORC1/c-MYC-driven signaling pathway in the activation of the UPR. Consistent with this notion, exogenously expressed c-MYC reversed the ability of rapamycin to prevent bortezomib-induced CHOP and ATF4 expression as well as apoptosis. These findings indicate that the induction of ATF4/CHOP expression occurs via mTORC1 regulation of c-MYC and that this signaling pathway is a major determinant in the ability of bortezomib to induce apoptosis.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Ácidos Borônicos/farmacologia , Biossíntese de Proteínas/fisiologia , Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Pirazinas/farmacologia , Proteínas Supressoras de Tumor , Resposta a Proteínas não Dobradas/efeitos dos fármacos , Fator 4 Ativador da Transcrição/genética , Fator 4 Ativador da Transcrição/metabolismo , Animais , Apoptose/fisiologia , Bortezomib , Linhagem Celular Tumoral , Regulação da Expressão Gênica , Humanos , Imunossupressores/farmacologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Complexos Multiproteicos , Proteínas/genética , Proteínas Proto-Oncogênicas c-myc/genética , Ratos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Sirolimo/farmacologia , Serina-Treonina Quinases TOR , Fator de Transcrição CHOP/genética , Fator de Transcrição CHOP/metabolismo , Proteína 1 do Complexo Esclerose Tuberosa , Proteína 2 do Complexo Esclerose Tuberosa , Resposta a Proteínas não Dobradas/fisiologia
6.
J Immunol ; 189(8): 3800-4, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22984083

RESUMO

Phagocyte NADPH oxidase plays a key role in pathogen clearance via reactive oxygen species (ROS) production. Defects in oxidase function result in chronic granulomatous disease with hallmark recurrent microbial infections and inflammation. The oxidase's role in the adaptive immune response is not well understood. Class II presentation of cytoplasmic and exogenous Ag to CD4(+) T cells was impaired in human B cells with reduced oxidase p40(phox) subunit expression. Naturally arising mutations, which compromise p40(phox) function in a chronic granulomatous disease patient, also perturbed class II Ag presentation and intracellular ROS production. Reconstitution of patient B cells with a wild-type, but not a mutant, p40(phox) allele restored exogenous Ag presentation and intracellular ROS generation. Remarkably, class II presentation of epitopes from membrane Ag was robust in p40(phox)-deficient B cells. These studies reveal a role for NADPH oxidase and p40(phox) in skewing epitope selection and T cell recognition of self Ag.


Assuntos
Apresentação de Antígeno/imunologia , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Antígenos HLA-DR/metabolismo , NADPH Oxidases/fisiologia , Apresentação de Antígeno/genética , Subpopulações de Linfócitos B/enzimologia , Linhagem Celular Transformada , Humanos , Líquido Intracelular/enzimologia , Líquido Intracelular/imunologia , Líquido Intracelular/metabolismo , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Fosfoproteínas/biossíntese , Fosfoproteínas/deficiência , Fosfoproteínas/genética , Espécies Reativas de Oxigênio/metabolismo , Regulação para Cima/genética , Regulação para Cima/imunologia
7.
Am J Physiol Cell Physiol ; 303(5): C499-511, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22621783

RESUMO

Intracellular signaling by the second messenger cyclic AMP (cAMP) activates the Ras-related small GTPase Rap1 through the guanine exchange factor Epac. This activation leads to effector protein interactions, activation, and biological responses in the vasculature, including vasorelaxation. In vascular smooth muscle cells derived from human dermal arterioles (microVSM), Rap1 selectively regulates expression of G protein-coupled α(2C)-adrenoceptors (α(2C)-ARs) through JNK-c-jun nuclear signaling. The α(2C)-ARs are generally retained in the trans-Golgi compartment and mobilize to the cell surface and elicit vasoconstriction in response to cellular stress. The present study used human microVSM to examine the role of Rap1 in receptor localization. Complementary approaches included murine microVSM derived from tail arteries of C57BL6 mice that express functional α(2C)-ARs and mice deficient in Rap1A (Rap1A-null). In human microVSM, increasing intracellular cAMP by direct activation of adenylyl cyclase by forskolin (10 µM) or selectively activating Epac-Rap signaling by the cAMP analog 8-pCPT-2'-O-Me-cAMP (100 µM) activated RhoA, increased α(2C)-AR expression, and reorganized the actin cytoskeleton, increasing F-actin. The α(2C)-ARs mobilized from the perinuclear region to intracellular filamentous structures and to the plasma membrane. Similar results were obtained in murine wild-type microVSM, coupling Rap1-Rho-actin dynamics to receptor relocalization. This signaling was impaired in Rap1A-null murine microVSM and was rescued by delivery of constitutively active (CA) mutant of Rap1A. When tested in heterologous HEK293 cells, Rap1A-CA or Rho-kinase (ROCK-CA) caused translocation of functional α(2C)-ARs to the cell surface (~4- to 6-fold increase, respectively). Together, these studies support vascular bed-specific physiological role of Rap1 and suggest a role in vasoconstriction in microVSM.


Assuntos
AMP Cíclico/metabolismo , Miócitos de Músculo Liso/metabolismo , Transporte Proteico/fisiologia , Receptores Adrenérgicos alfa 2/metabolismo , Proteínas rap1 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Arteríolas/citologia , Células Cultivadas , AMP Cíclico/genética , Regulação da Expressão Gênica/fisiologia , Humanos , Camundongos , Camundongos Knockout , Ligação Proteica , Receptores Adrenérgicos alfa 2/genética , Proteínas rap1 de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/genética
8.
J Biol Chem ; 286(49): 42316-42324, 2011 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-22009749

RESUMO

Phosphatases of the regenerating liver (PRL) play oncogenic roles in cancer development and metastasis. Although previous studies indicate that PRL-1 promotes cell growth and migration by activating both the ERK1/2 and RhoA pathways, the mechanism by which it activates these signaling events remains unclear. We have identified a PRL-1-binding peptide (Peptide 1) that shares high sequence identity with a conserved motif in the Src homology 3 (SH3) domain of p115 Rho GTPase-activating protein (GAP). p115 RhoGAP directly binds PRL-1 in vitro and in cells via its SH3 domain. Structural analyses of the PRL-1·Peptide 1 complex revealed a novel protein-protein interaction whereby a sequence motif within the PxxP ligand-binding site of the p115 RhoGAP SH3 domain occupies a folded groove within PRL-1. This prevents the canonical interaction between the SH3 domain of p115 RhoGAP and MEKK1 and results in activation of ERK1/2. Furthermore, PRL-1 binding activates RhoA signaling by inhibiting the catalytic activity of p115 RhoGAP. The results demonstrate that PRL-1 binding to p115 RhoGAP provides a coordinated mechanism underlying ERK1/2 and RhoA activation.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Regulação Enzimológica da Expressão Gênica , Fatores de Troca do Nucleotídeo Guanina/química , Proteínas Imediatamente Precoces/metabolismo , Proteínas de Membrana/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Quinases da Família src/metabolismo , Motivos de Aminoácidos , Animais , Fibroblastos/metabolismo , Células HEK293 , Humanos , Ligantes , Camundongos , Ligação Proteica , Fatores de Troca de Nucleotídeo Guanina Rho , Transdução de Sinais , Proteínas rho de Ligação ao GTP/metabolismo
9.
J Cell Biochem ; 113(4): 1253-64, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22121046

RESUMO

Constitutive activation of M-Ras has previously been reported to cause morphologic and growth transformation of murine cells, suggesting that M-Ras plays a role in tumorigenesis. Cell transformation by M-Ras correlated with weak activation of the Raf/MEK/ERK pathway, although contributions from other downstream effectors were suggested. Recent studies indicate that signaling events distinct from the Raf/MEK/ERK cascade are critical for human tumorigenesis. However, it is unknown what signaling events M-Ras triggers in human cells. Using constitutively active M-Ras (Q71L) containing additional mutations within its effector-binding loop, we found that M-Ras induces MEK/ERK-dependent and -independent Elk1 activation as well as phosphatidylinositol 3 kinase (PI3K)/Akt and JNK/cJun activation in human MCF-7 breast cancer cells. Among several human cell lines examined, M-Ras-induced MEK/ERK-independent Elk1 activation was only detected in MCF-7 cells, and correlated with Rlf/M-Ras interaction and Ral/JNK activation. Supporting a role for M-Ras signaling in breast cancer, EGF activated M-Ras and promoted its interaction with endogenous Rlf. In addition, constitutive activation of M-Ras induced estrogen-independent growth of MCF-7 cells that was dependent on PI3K/Akt, MEK/ERK, and JNK activation. Thus, our studies demonstrate that M-Ras signaling activity differs between human cells, highlighting the importance of defining Ras protein signaling within each cell type, especially when designing treatments for Ras-induced cancer. These findings also demonstrate that M-Ras activity may be important for progression of EGFR-dependent tumors.


Assuntos
Regulação Enzimológica da Expressão Gênica , MAP Quinase Quinase 4/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Monoméricas de Ligação ao GTP/fisiologia , Proteínas ral de Ligação ao GTP/metabolismo , Linhagem Celular Tumoral , Ativação Enzimática , Feminino , Humanos , Imunoprecipitação , Transdução de Sinais
10.
Am J Physiol Renal Physiol ; 303(6): F831-44, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22791333

RESUMO

Tuberous sclerosis complex (TSC), an inherited tumor predisposition syndrome associated with mutations in TSC1 or TSC2, affects ∼1 in 6,000 individuals. Eighty percent of TSC patients develop renal angiomyolipomas, and renal involvement is a major contributor to patient morbidity and mortality. Recent work has shown that mammalian target of rapamycin complex 1 (mTORC1) inhibition caused angiomyolipoma shrinkage but that this treatment may cause cytostatic not a cytotoxic effect. Endoplasmic reticulum (ER) stress can develop in TSC-associated cells due to mTORC1-driven protein translation. We hypothesized that renal angiomyolipoma cells experience ER stress that can be leveraged to result in targeted cytotoxicity. We used immortalized human angiomyolipoma cells stably transfected with empty vector or TSC2 (encoding tuberin). Using cell number quantification and cell death assays, we found that mTORC1 inhibition with RAD001 suppressed angiomyolipoma cell proliferation in a cytostatic manner. Angiomyolipoma cells exhibited enhanced sensitivity to proteasome inhibitor-induced ER stress compared with TSC2-rescued cells. After proteasome inhibition with MG-132, Western blot analyses showed greater induction of C/EBP-homologous protein (CHOP) and more poly (ADP-ribose) polymerase (PARP) and caspase-3 cleavage, supporting ER stress-induced apoptosis. Live cell numbers also were decreased and cell death increased by MG-132 in angiomyolipoma cells compared with TSC2 rescued. Intriguingly, while pretreatment of angiomyolipoma cells with RAD001 attenuated CHOP and BiP induction, apoptotic markers cleaved PARP and caspase-3 and eukaryotic translation initiation factor 2α phosphorylation were increased, along with evidence of increased autophagy. These results suggest that human angiomyolipoma cells are uniquely susceptible to agents that exacerbate ER stress and that additional synergy may be achievable with targeted combination therapy.


Assuntos
Angiomiolipoma/metabolismo , Estresse do Retículo Endoplasmático , Neoplasias Renais/metabolismo , Esclerose Tuberosa/complicações , Proteínas Supressoras de Tumor/metabolismo , Angiomiolipoma/etiologia , Angiomiolipoma/genética , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Caspase 3/metabolismo , Linhagem Celular Tumoral , Fator de Iniciação 2 em Eucariotos/metabolismo , Everolimo , Humanos , Imunossupressores/farmacologia , Neoplasias Renais/etiologia , Neoplasias Renais/genética , Leupeptinas/farmacologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Complexos Multiproteicos , Fosforilação , Poli(ADP-Ribose) Polimerases/metabolismo , Inibidores de Proteassoma/farmacologia , Proteínas/metabolismo , Sirolimo/análogos & derivados , Sirolimo/farmacologia , Serina-Treonina Quinases TOR , Fator de Transcrição CHOP/metabolismo , Transfecção , Esclerose Tuberosa/genética , Esclerose Tuberosa/metabolismo , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/genética
11.
Am J Physiol Endocrinol Metab ; 301(6): E1072-80, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21828338

RESUMO

Second-phase insulin release requires the sustained mobilization of insulin granules from internal storage pools to the cell surface for fusion with the plasma membrane. However, the detailed mechanisms underlying this process remain largely unknown. GTP-loading of the small GTPase Cdc42 is the first glucose-specific activation step in the process, although how glucose triggers Cdc42 activation is entirely unknown. In a directed candidate screen for guanine nucleotide exchange factors (GEFs), which directly activate small GTPases, Cool-1/ßPix was identified in pancreatic islet beta cells. In support of its role as the beta cell Cdc42 GEF, ßPix coimmunoprecipitated with Cdc42 in human islets and MIN6 beta cells in a glucose-dependent manner, peaking just prior to Cdc42 activation. Furthermore, RNAi-mediated ßPix reduction by 50% corresponded to full ablation of glucose-induced Cdc42 activation and significant attenuation of basal and glucose-stimulated insulin secretion. Of the two Cdc42 guanine nucleotide dissociation inhibitor (GDI) proteins identified in beta cells, ßPix competed selectively with caveolin-1 (Cav-1) but not RhoGDI in coimmunoprecipitation and GST-Cdc42-GDP interaction assays. However, a phospho-deficient Cav-1-Y14F mutant failed to compete with ßPix; Cav-1(Tyr14) is an established phosphorylation site for Src kinase. Taken together, these data support a new model, wherein glucose stimulates Cav-1 and induces its dissociation from Cdc42, possibly via Src kinase activation to phosphorylate Cav-1(Tyr14), to promote Cdc42-ßPix binding and Cdc42 activation, and to trigger downstream signaling and ultimately sustain insulin release.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/fisiologia , Insulina/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo , Animais , Caveolina 1/metabolismo , Células Cultivadas , Técnicas de Silenciamento de Genes , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/genética , RNA Interferente Pequeno/farmacologia , Fatores de Troca de Nucleotídeo Guanina Rho , Estudos de Validação como Assunto , Proteína cdc42 de Ligação ao GTP/genética
12.
Mol Cell Biol ; 27(1): 312-23, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17060455

RESUMO

The phagocyte NADPH oxidase catalyzes the reduction of molecular oxygen to superoxide and is essential for microbial defense. Electron transport through the oxidase flavocytochrome is activated by the Rac effector p67(phox). Previous studies suggest that Vav1 regulates NADPH oxidase activity elicited by the chemoattractant formyl-Met-Leu-Phe (fMLP). We show that Vav1 associates with p67(phox) and Rac2, but not Rac1, in fMLP-stimulated human neutrophils, correlating with superoxide production. The interaction of p67(phox) with Vav1 is direct and activates nucleotide exchange on Rac, which enhances the interaction between p67(phox) and Vav1. This provides new molecular insights into regulation of the neutrophil NADPH oxidase, suggesting that chemoattractant-stimulated superoxide production can be amplified by a positive feedback loop in which p67(phox) targets Vav1-mediated Rac activation.


Assuntos
Regulação Enzimológica da Expressão Gênica , Regulação da Expressão Gênica , NADPH Oxidases/metabolismo , Fagócitos/metabolismo , Fosfoproteínas/fisiologia , Proteínas Proto-Oncogênicas c-vav/fisiologia , Proteínas rac de Ligação ao GTP/metabolismo , Animais , Células COS , Chlorocebus aethiops , Ativação Enzimática , Granulócitos/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Neutrófilos/metabolismo , Fosfoproteínas/metabolismo
13.
J Cell Biol ; 167(1): 111-22, 2004 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-15479739

RESUMO

The Ras-related GTPase Rap1 stimulates integrin-mediated adhesion and spreading in various mammalian cell types. Here, we demonstrate that Rap1 regulates cell spreading by localizing guanine nucleotide exchange factors (GEFs) that act via the Rho family GTPase Rac1. Rap1a activates Rac1 and requires Rac1 to enhance spreading, whereas Rac1 induces spreading independently of Rap1. Active Rap1a binds to a subset of Rac GEFs, including VAV2 and Tiam1 but not others such as SWAP-70 or COOL-1. Overexpressed VAV2 and Tiam1 specifically require Rap1 to promote spreading, even though Rac1 is activated independently of Rap1. Rap1 is necessary for the accumulation of VAV2 in membrane protrusions at the cell periphery. In addition, if VAV2 is artificially localized to the cell edge with the subcellular targeting domain of Rap1a, it increases cell spreading independently of Rap1. These results lead us to propose that Rap1 promotes cell spreading by localizing a subset of Rac GEFs to sites of active lamellipodia extension.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas rap1 de Ligação ao GTP/metabolismo , Adesão Celular , Proteínas de Ciclo Celular/metabolismo , Membrana Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Vetores Genéticos , Glutationa Transferase/metabolismo , Células HeLa , Humanos , Imunoprecipitação , Microscopia de Fluorescência , Antígenos de Histocompatibilidade Menor , Modelos Biológicos , Proteínas Nucleares/metabolismo , Proteínas Oncogênicas/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-vav , Pseudópodes/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho , Proteína 1 Indutora de Invasão e Metástase de Linfoma de Células T , Transfecção
14.
J Cell Biochem ; 105(4): 1027-37, 2008 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-18767072

RESUMO

E-cadherin based adherens junctions are finely regulated by multiple cellular signaling events. Here we show that the Ras-related Rap1 GTPase is enriched in regions of nascent cell-cell contacts and strengthens E-cadherin junctions: constitutively active Rap1 expressing MDCK cells exhibit increased junctional contact and resisted calcium depletion-induced cell-cell junction disruption. E-cadherin disengagement activated Rap1 and this correlated with E-cadherin association with the Rap GEFs, C3G and PDZ-GEF I. PDZ-GEF I associated with E-cadherin and beta-catenin whereas C3G interaction with E-cadherin did not involve beta-catenin. Knockdown of PDZ-GEF I in MDCK cells decreased Rap1 activity following E-cadherin junction disruption. We hereby show that Rap1 plays a role in the maintenance and repair of E-cadherin junctions and is activated via an "outside-in" signaling pathway initiated by E-cadherin and mediated at least in part by PDZ-GEF I.


Assuntos
Caderinas/metabolismo , Transdução de Sinais , Proteínas rap1 de Ligação ao GTP/metabolismo , Animais , Caderinas/fisiologia , Linhagem Celular , Cães , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Fator 2 de Liberação do Nucleotídeo Guanina/metabolismo , beta Catenina/metabolismo
15.
Sci STKE ; 2007(414): pe67, 2007 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-18042941

RESUMO

The activation of the small guanosine triphosphatase Ras is critical for many biological events. It is therefore not surprising that the ubiquitously expressed Ras guanine nucleotide exchange factor (GEF) SOS (Son of Sevenless), which couples protein tyrosine kinases to Ras activation, is under tight autoinhibitory control. Several studies have revealed how multiple regulatory domains might affect SOS activity. Most notably, a second Ras-binding site on SOS allosterically regulates the duration and amplitude of Ras activation. This allosteric Ras-GTP is produced by another GEF, Ras guanine nucleotide-releasing protein 1 (RasGRP1). SOS and RasGRP1 are both activated downstream of phospholipase D(2), and gain-of-function mutants of SOS contribute to inherited diseases. These studies not only enable us to better appreciate the complexity of the regulation of GEFs but also prompt us to reevaluate our current understanding of pathways that lead to Ras activation.


Assuntos
Proteína Son Of Sevenless de Drosófila/metabolismo , Sítio Alostérico , Animais , Domínio Catalítico , Proteínas de Ligação a DNA/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Conformação Proteica , Proteína Son Of Sevenless de Drosófila/química
16.
Elife ; 72018 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-29360039

RESUMO

Specific cell shapes are fundamental to the organization and function of multicellular organisms. Fibroblast Growth Factor (FGF) signaling induces the elongation of lens fiber cells during vertebrate lens development. Nonetheless, exactly how this extracellular FGF signal is transmitted to the cytoskeletal network has previously not been determined. Here, we show that the Crk family of adaptor proteins, Crk and Crkl, are required for mouse lens morphogenesis but not differentiation. Genetic ablation and epistasis experiments demonstrated that Crk and Crkl play overlapping roles downstream of FGF signaling in order to regulate lens fiber cell elongation. Upon FGF stimulation, Crk proteins were found to interact with Frs2, Shp2 and Grb2. The loss of Crk proteins was partially compensated for by the activation of Ras and Rac signaling. These results reveal that Crk proteins are important partners of the Frs2/Shp2/Grb2 complex in mediating FGF signaling, specifically promoting cell shape changes.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Forma Celular , Fatores de Crescimento de Fibroblastos/metabolismo , Fibroblastos/fisiologia , Cristalino/embriologia , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas c-crk/metabolismo , Transdução de Sinais , Animais , Fibroblastos/efeitos dos fármacos , Proteína Adaptadora GRB2/metabolismo , Camundongos , Morfogênese , Ligação Proteica , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo
17.
Mol Cell Biol ; 22(21): 7645-57, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12370311

RESUMO

Rho family GTPases activate intracellular kinase cascades to modulate transcription of multiple genes. Previous studies have examined the roles of the ubiquitously expressed Rho GTPase, Rac1, in regulation of gene expression in cell lines and implicated NF-kappaB, serum response factor, and kinase signaling pathways in this regulation. To understand the role of the closely related but hematopoiesis-specific Rho GTPase, Rac2, in regulation of gene transcription, we compared the gene expression profiles between wild-type and Rac2(-/-) bone marrow-derived mast cells. Our data demonstrate remarkable specificity in the regulation of gene expression by Rac2 versus Rac1. Microarray analysis demonstrated that expression of 38 known genes was significantly altered in Rac2(-/-) mast cells after cytokine stimulation compared with those in wild-type cells. Of these, the expression of the mouse mast cell protease 7 (MMCP-7) gene in wild-type cells was highly induced at the transcriptional level after stimulation with stem cell factor (SCF). In spite of compensatorily increased expression of Rac1 in Rac2-deficient cells, SCF-induced MMCP-7 transcription did not occur. Surprisingly, the loss of MMCP-7 induction was not due to decreased activation of NF-kappaB, a transcription factor postulated to lie downstream of Rac1 and known to play a critical role in hematopoietic cell differentiation and proliferation. However, the activities of c-Jun N-terminal kinases (JNKs) were markedly decreased in Rac2(-/-) mast cells. Our results suggest that cytokine-stimulated activation of MMCP-7 gene transcription is selectively regulated by a Rac2-dependent JNK signaling pathway in primary mast cells and imply a remarkable specificity in the regulation of transcriptional activity by these two highly related Rho GTPases.


Assuntos
Regulação Enzimológica da Expressão Gênica , Mastócitos/enzimologia , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas rac de Ligação ao GTP/fisiologia , Proteínas rho de Ligação ao GTP/metabolismo , Motivos de Aminoácidos , Animais , Northern Blotting , Western Blotting , Células da Medula Óssea/metabolismo , Núcleo Celular/metabolismo , Dactinomicina/farmacologia , Ativação Enzimática , Immunoblotting , Proteínas Quinases JNK Ativadas por Mitógeno , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Plasmídeos/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Serina Endopeptidases/metabolismo , Transdução de Sinais , Fator de Células-Tronco/metabolismo , Fatores de Tempo , Transcrição Gênica , Triptases , Regulação para Cima , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteína RAC2 de Ligação ao GTP
18.
Mol Cancer Res ; 3(1): 32-41, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15671247

RESUMO

AND-34, a 95-kDa protein with modest homology to Ras GDP exchange factors, associates with the focal adhesion protein p130Cas. Overexpression of AND-34 confers anti-estrogen resistance in breast cancer cell lines, a property linked to its ability to activate Rac. Here, we show that both the GDP exchange factor-like domain and the SH2 domain of AND-34 are required for Rac activation and for resistance to the estrogen receptor (ER) antagonist ICI 182,780. As phosphatidylinositol 3-kinase (PI3K) signaling can regulate Rac activation, we examined the effects of AND-34 on PI3K. Overexpression of AND-34 in MCF-7 cells increased PI3K activity and augmented Akt Ser(473) phosphorylation and kinase activity. Inhibition of PI3K with LY294002 or a dominant-negative p85 construct blocked AND-34-mediated Rac and Akt activation. Although R-Ras can activate PI3K, transfection with constitutively active R-Ras failed to induce Rac activation and AND-34 overexpression failed to induce R-Ras activation. Treatment of either vector-only or AND-34-transfected ZR-75-1 cells with ICI 182,780 markedly diminished ERalpha levels, suggesting that AND-34-induced anti-estrogen resistance is likely to occur by an ERalpha-independent mechanism. Treatment of a ZR-75-1 breast cancer cell line stably transfected with AND-34 plus 2 micromol/L LY294002 or 10 micromol/L NSC23766, a Rac-specific inhibitor, abrogated AND-34-induced resistance to ICI 182,780. Our studies suggest that AND-34-mediated PI3K activation induces Rac activation and anti-estrogen resistance in human breast cancer cell lines.


Assuntos
Estradiol/análogos & derivados , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células , Cromonas/farmacologia , Proteína Substrato Associada a Crk , Relação Dose-Resposta a Droga , Estradiol/farmacologia , Antagonistas de Estrogênios/metabolismo , Estrogênios/metabolismo , Adesões Focais , Fulvestranto , GTP Fosfo-Hidrolases/metabolismo , Genes Dominantes , Vetores Genéticos , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Humanos , Morfolinas/farmacologia , Fosforilação , Plasmídeos/metabolismo , Ligação Proteica , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Terciária de Proteína , Proteínas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Receptores de Estrogênio/metabolismo , Proteína p130 Retinoblastoma-Like , Serina/química , Transdução de Sinais , Transfecção , Proteínas rac de Ligação ao GTP/metabolismo , Domínios de Homologia de src
19.
Methods Enzymol ; 407: 108-14, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16757318

RESUMO

Ral proteins regulate a variety of biological processes and are major downstream targets of Ras because of association of the RalGDS family of exchange factors with Ras-GTP. However, a second, less-characterized family of Ral GEFs has also been isolated. Described here are methods used to determine the substrate specificity of this RalGPS family of GEFs, measure their membrane localization, interactions of with SH3 domains and phospholipids, as well as to measure their ability to activate Ral in vivo.


Assuntos
Fator ral de Troca do Nucleotídeo Guanina/biossíntese , Fracionamento Celular , Linhagem Celular , Humanos , Fosfolipídeos/metabolismo , Especificidade por Substrato , Transfecção , Fator ral de Troca do Nucleotídeo Guanina/metabolismo , Domínios de Homologia de src/fisiologia
20.
Methods Enzymol ; 407: 55-63, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16757314

RESUMO

AND-34 is a member of a novel family of proteins (NSP1, NSP2, and NSP3) that have an amino-terminal SH2 domain but bind by a carboxy-terminal GEF (Cdc25)-like domain to the carboxy-terminus of the focal adhesion adapter protein p130Cas. Direct GEF activity of AND-34 toward Ras subfamily members has not been demonstrated with purified protein. Overexpression of AND-34 in epithelial breast cancer cells leads to activation of Rac and Cdc42 by a PI3K-dependent mechanism. This chapter will describe the techniques we used to examine AND-34-induced Rac, Cdc42, Akt, and PAK1 activation in human breast cancer cell lines and in murine lymphoid cell lines. In addition, we summarize techniques used to determine that AND-34 overexpression does not activate R-Ras in MCF-7 cells.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/fisiologia , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Linfócitos B , Linhagem Celular Tumoral , Humanos , Camundongos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quinases Ativadas por p21/metabolismo , Proteínas ras/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA