Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 10(1): 3667, 2019 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-31413255

RESUMO

Receptor type protein tyrosine phosphatase-sigma (PTPσ) is primarily expressed by adult neurons and regulates neural regeneration. We recently discovered that PTPσ is also expressed by hematopoietic stem cells (HSCs). Here, we describe small molecule inhibitors of PTPσ that promote HSC regeneration in vivo. Systemic administration of the PTPσ inhibitor, DJ001, or its analog, to irradiated mice promotes HSC regeneration, accelerates hematologic recovery, and improves survival. Similarly, DJ001 administration accelerates hematologic recovery in mice treated with 5-fluorouracil chemotherapy. DJ001 displays high specificity for PTPσ and antagonizes PTPσ via unique non-competitive, allosteric binding. Mechanistically, DJ001 suppresses radiation-induced HSC apoptosis via activation of the RhoGTPase, RAC1, and induction of BCL-XL. Furthermore, treatment of irradiated human HSCs with DJ001 promotes the regeneration of human HSCs capable of multilineage in vivo repopulation. These studies demonstrate the therapeutic potential of selective, small-molecule PTPσ inhibitors for human hematopoietic regeneration.


Assuntos
Apoptose/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Proteínas Tirosina Fosfatases Classe 2 Semelhantes a Receptores/antagonistas & inibidores , Regeneração/efeitos dos fármacos , Regulação Alostérica , Animais , Antimetabólitos Antineoplásicos/farmacologia , Apoptose/efeitos da radiação , Fluoruracila/farmacologia , Células-Tronco Hematopoéticas/efeitos da radiação , Humanos , Camundongos , Radiação , Regeneração/efeitos da radiação , Proteína bcl-X/efeitos dos fármacos , Proteína bcl-X/metabolismo , Proteínas rac1 de Ligação ao GTP/efeitos dos fármacos , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/efeitos dos fármacos , Proteínas rho de Ligação ao GTP/metabolismo
2.
Ann Rheum Dis ; 76(8): 1440-1448, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28416516

RESUMO

OBJECTIVES: Inappropriate activation of neutrophils plays a pathological role in antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV). The aim of this study was to investigate the functions of semaphorin 4D (SEMA4D) in regulation of neutrophil activation, and its involvement in AAV pathogenesis. METHODS: Serum levels of soluble SEMA4D were evaluated by ELISA. Blood cell-surface expression of membrane SEMA4D was evaluated by flow cytometry. To determine the functional interactions between neutrophil membrane SEMA4D and endothelial plexin B2, wild-type and SEMA4D-/- mice neutrophils were cultured with an endothelial cell line (MS1) stained with SYTOX green, and subjected to neutrophil extracellular trap (NET) formation assays. The efficacy of treating human neutrophils with recombinant plexin B2 was assessed by measuring the kinetic oxidative burst and NET formation assays. RESULTS: Serum levels of soluble SEMA4D were elevated in patients with AAV and correlated with disease activity scores. Cell-surface expression of SEMA4D was downregulated in neutrophils from patients with AAV, a consequence of proteolytic cleavage of membrane SEMA4D. Soluble SEMA4D exerted pro-inflammatory effects on endothelial cells. Membranous SEMA4D on neutrophils bound to plexin B2 on endothelial cells, and this interaction decreased NET formation. Recombinant plexin B2 suppressed neutrophil Rac1 activation through SEMA4D's intracellular domain, and inhibited pathogen-induced or ANCA-induced oxidative burst and NET formation. CONCLUSIONS: Neutrophil surface SEMA4D functions as a negative regulator of neutrophil activation. Proteolytic cleavage of SEMA4D as observed in patients with AAV may amplify neutrophil-mediated inflammatory responses. SEMA4D is a promising biomarker and potential therapeutic target for AAV.


Assuntos
Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos/imunologia , Antígenos CD/imunologia , Células Endoteliais/imunologia , Armadilhas Extracelulares/imunologia , Proteínas do Tecido Nervoso/imunologia , Neutrófilos/imunologia , Semaforinas/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Antígenos CD/genética , Ensaio de Imunoadsorção Enzimática , Armadilhas Extracelulares/efeitos dos fármacos , Feminino , Citometria de Fluxo , Humanos , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/farmacologia , Neutrófilos/efeitos dos fármacos , Espécies Reativas de Oxigênio/imunologia , Explosão Respiratória/efeitos dos fármacos , Semaforinas/genética , Proteínas rac1 de Ligação ao GTP/efeitos dos fármacos , Proteínas rac1 de Ligação ao GTP/imunologia
3.
J Craniofac Surg ; 27(2): 516-20, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26963302

RESUMO

UNLABELLED: Angiogenesis is the development of new capillaries from existing blood vessels and is a prerequisite for the wound-healing process. Many lines of scientific evidences have shown that complicated roles of small guanosine triphosphatases (GTPases) (ras-related C3 botulinum toxin substrate 1 [Rac1], cell division control protein 42 [Cdc42], and ras homolog gene family, member A [RhoA]) in regulation of signal transduction pathways exist to transmit distinct cellular effects on the modulation of actin cytoskeleton remodeling such as cell cycle progression, cell survival, and cell motility. In addition, these small GTPases activate mitogen-activated protein kinase kinase kinases (MAP3Ks) leading to activated mitogen-activated protein kinase kinases (MAPKK), mitogen-activated protein kinase (MAPK), and various transcription factors such as vascular endothelial growth factor with involvement of MAPK signaling pathways.In this study, the authors hypothesized that botulinum toxin A increases angiogenesis via the expression of small GTPases in vivo and in vitro studies.In vivo experiment, 24 Sprague-Dawley rats were randomly divided into 2 groups: a control group and a botulinum toxin A group. Five days prior to superiorly based transverse rectus abdominis myocutaneous flap elevation, the botulinum toxin A (BoTA) group was pretreated with BoTA, while the control group was pretreated with normal saline. quantitative real-time polymerase chain reaction was performed to evaluate the expression of Rac1, RhoA, and Cdc42.The angiogenic effects of botulinum toxin A on human dermal fibroblasts were measured in vitro experiment. To understand the mechanism of botulinum toxin A on small GTPases production of fibroblasts, Rac1, Cdc42, and RhoA were measured using qRT-PCR.The relative messenger ribonucleic acid expression of Rac1, RhoA, and Cdc42 was significantly higher in the BoTA group than in the control group, in every zone and pedicle muscle, on postoperative days 1, 3, and 5. Levels of these molecules increased significantly in human dermal fibroblasts grown in the presence of BoTA compared with control group over 5 IU.Our in vivo and in vitro studies suggest that administration of BoTA upregulates the expression of RhoA, Rac1, and Cdc42 in a dose-dependent manner. MAPK signaling pathway might be involved in BoTA-induced angiogenesis mechanism. LEVEL OF EVIDENCE: N/A.


Assuntos
Toxinas Botulínicas Tipo A/farmacologia , Proteína cdc42 de Ligação ao GTP/efeitos dos fármacos , Proteínas rac1 de Ligação ao GTP/efeitos dos fármacos , Proteína rhoA de Ligação ao GTP/efeitos dos fármacos , Indutores da Angiogênese/farmacologia , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica/efeitos dos fármacos , Sobrevivência de Enxerto/efeitos dos fármacos , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Retalho Miocutâneo/irrigação sanguínea , Retalho Miocutâneo/cirurgia , Neovascularização Fisiológica/efeitos dos fármacos , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Reto do Abdome/irrigação sanguínea , Reto do Abdome/cirurgia , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
4.
Toxins (Basel) ; 7(11): 4610-21, 2015 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-26556375

RESUMO

Eukaryotic cells secrete extracellular vesicles (EVs), either constitutively or in a regulated manner, which represent an important mode of intercellular communication. EVs serve as vehicles for transfer between cells of membrane and cytosolic proteins, lipids and RNA. Furthermore, certain bacterial protein toxins, or possibly their derived messages, can be transferred cell to cell via EVs. We have herein demonstrated that eukaryotic EVs represent an additional route of cell-to-cell propagation for the Escherichia coli protein toxin cytotoxic necrotizing factor 1 (CNF1). Our results prove that EVs from CNF1 pre-infected epithelial cells can induce cytoskeleton changes, Rac1 and NF-κB activation comparable to that triggered by CNF1. The observation that the toxin is detectable inside EVs derived from CNF1-intoxicated cells strongly supports the hypothesis that extracellular vesicles can offer to the toxin a novel route to travel from cell to cell. Since anthrax and tetanus toxins have also been reported to engage in the same process, we can hypothesize that EVs represent a common mechanism exploited by bacterial toxins to enhance their pathogenicity.


Assuntos
Toxinas Bacterianas/farmacologia , Toxinas Bacterianas/uso terapêutico , Proteínas de Escherichia coli/farmacologia , Proteínas de Escherichia coli/uso terapêutico , Vesículas Extracelulares/efeitos dos fármacos , Comunicação Celular/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Tumoral , Citoesqueleto/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Humanos , NF-kappa B/efeitos dos fármacos , Proteínas rac1 de Ligação ao GTP/biossíntese , Proteínas rac1 de Ligação ao GTP/efeitos dos fármacos
5.
Hypertension ; 59(2): 500-6, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22232135

RESUMO

Overactivation of the mineralocorticoid receptor signaling is implicated in cardiovascular disease, including hypertensive heart disease. Oxidative stress is suggested to augment mineralocorticoid receptor signal transduction, but the precise mechanisms remain unclear. Mineralocorticoid receptor activity is regulated by multiple factors, in addition to plasma ligand levels. We previously identified Rac1 GTPase as a modulator of mineralocorticoid receptor activity. Here we show that oxidative stress induces mineralocorticoid receptor activation in a ligand-independent, Rac1-depenent manner in cardiomyocytes. Oxidant stress was induced in rat cultured cardiomyocytes (H9c2) by l-buthionine sulfoximine (BSO), an inhibitor of glutathione synthesis. BSO depleted intracellular glutathione and concomitantly increased reactive oxygen species (199%; P<0.01). BSO significantly enhanced the corticosterone-induced, mineralocorticoid receptor-dependent luciferase reporter activity (186%; P<0.01) and basal luciferase activity without ligand stimulation. These effects were inhibited by the antioxidant N-acetylcysteine. The ligand independency of BSO action was indicated using a mutant mineralocorticoid receptor that does not bind ligands. With this mutant mineralocorticoid receptor, BSO-evoked mineralocorticoid receptor activation remained intact, whereas ligand-induced mineralocorticoid receptor activation was abolished. We next examined the involvement of Rac1. BSO increased active Rac1 in a redox-dependent fashion, and Rac inhibition suppressed the enhancing effect of BSO. Constitutively active Rac1, indeed, potentiated mineralocorticoid receptor transactivation. Furthermore, mineralocorticoid receptor transactivation by BSO was accompanied by enhanced nuclear accumulation of mineralocorticoid receptor. We conclude that alteration of redox state modulates mineralocorticoid receptor-dependent transcriptional activity via Rac1 in the heart. This redox-sensitive, ligand-independent mineralocorticoid receptor activation may contribute to the processes by which oxidant stress promotes cardiac injury.


Assuntos
Miócitos Cardíacos/metabolismo , Estresse Oxidativo/fisiologia , Receptores de Mineralocorticoides/metabolismo , Transdução de Sinais/fisiologia , Proteínas rac1 de Ligação ao GTP/fisiologia , Animais , Butionina Sulfoximina/farmacologia , Linhagem Celular , Núcleo Celular/metabolismo , Inibidores Enzimáticos/farmacologia , Glutationa/antagonistas & inibidores , Glutationa/metabolismo , Modelos Animais , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Ratos , Receptores de Mineralocorticoides/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Proteínas rac1 de Ligação ao GTP/efeitos dos fármacos
6.
Cell Adh Migr ; 5(5): 382-6, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21975546

RESUMO

Previous studies from our lab have shown that both boric (BA) and phenylboronic- acid (PBA) inhibit the migration of prostate cancer cell lines, as well as non-tumorigenic prostate cells. Our results indicate that PBA is more potent than BA in targeting metastatic and proliferative properties of cancer cells. Here we focus on the impact of BA and PBA on Rho family of GTP-binding proteins and their downstream targets. Treatment with 1mM PBA and BA decreases activities of RhoA, Rac1, and Cdc42 in DU-145 metastatic prostate cancer cells, but not in normal RWPE-1 prostate cells. Furthermore, ROCKII activity and phosphorylation of myosin light chain kinase decrease as a result of either PBA or BA treatment in DU-145 cells, suggesting these compounds target actomyosin-based contractility.


Assuntos
Ácidos Bóricos/administração & dosagem , Ácidos Borônicos/administração & dosagem , Movimento Celular/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Proteínas rho de Ligação ao GTP/metabolismo , Quinases Associadas a rho/antagonistas & inibidores , Actinas/efeitos dos fármacos , Actinas/metabolismo , Linhagem Celular Tumoral , Humanos , Masculino , Quinase de Cadeia Leve de Miosina/química , Quinase de Cadeia Leve de Miosina/efeitos dos fármacos , Quinase de Cadeia Leve de Miosina/metabolismo , Fosforilação/efeitos dos fármacos , Neoplasias da Próstata/patologia , Proteína cdc42 de Ligação ao GTP/efeitos dos fármacos , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/efeitos dos fármacos , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/efeitos dos fármacos , Quinases Associadas a rho/efeitos dos fármacos , Quinases Associadas a rho/metabolismo
7.
J Periodontol ; 82(11): 1623-31, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21417583

RESUMO

BACKGROUND: Recent studies have shown that the 15-member macrolide antibiotic azithromycin (AZM) not only has antibacterial activity, but also results in the role of immunomodulator. Interleukin (IL)-8 is an important inflammatory mediator in periodontal disease. However, there have been no reports on the effects of AZM on IL-8 production from human oral epithelium. Therefore, we investigated the effects of AZM on IL-8 production in an oral epithelial cell line. METHODS: KB cells were stimulated by Escherichia coli or Aggregatibacter actinomycetemcomitans (previously Actinobacillus actinomycetemcomitans) lipopolysaccharide (LPS) with or without AZM. IL-8 mRNA and protein expression and production in response to LPS were analyzed by quantitative polymerase chain reaction, flow cytometry, and enzyme-linked immunosorbent assay. The activation of nuclear factor-kappa B (NF-κB) and Rac1, which is important for IL-8 expression, was analyzed by enzyme-linked immunosorbent assay and Western blotting, respectively. RESULTS: IL-8 mRNA expression, IL-8 production, and NF-κB activation in LPS-stimulated KB cells were inhibited by the addition of AZM. LPS-induced Rac1 activation was also suppressed by AZM. CONCLUSIONS: This study suggests that AZM inhibits LPS-induced IL-8 production in an oral epithelial cell line, in part caused by the suppression of Rac1 and NF-κB activation. The use of AZM might provide possible benefits in periodontal therapy, with respect to both its antibacterial action and apparent anti-inflammatory effect.


Assuntos
Antibacterianos/farmacologia , Azitromicina/farmacologia , Células Epiteliais/efeitos dos fármacos , Interleucina-8/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Células Cultivadas , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Lipopolissacarídeos/farmacologia , NF-kappa B/efeitos dos fármacos , NF-kappa B/metabolismo , Proteínas rac1 de Ligação ao GTP/efeitos dos fármacos , Proteínas rac1 de Ligação ao GTP/metabolismo
8.
Dig Liver Dis ; 43(5): 395-403, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21334995

RESUMO

BACKGROUND: Simvastatin is a cholesterol-lowering drug that is widely used to prevent and treat atherosclerotic cardiovascular disease. Simvastatin exhibits numerous pleiotropic effects including anti-cancer activity. However, the effect of simvastatin on cholangiocarcinoma has not been evaluated. AIM: The aim of our study was to determine the effect of simvastatin on cholangiocarcinoma proliferation. METHODS: The effect of simvastatin was evaluated in five human cholangiocarcinoma cell lines (Mz-ChA-1, HuH-28, TFK-1, SG231, and HuCCT1) and normal cholangiocyte cell line (HiBEpiC). RESULTS: We found that simvastatin stimulates a reduction in cell viability and apoptosis of cholangiocarcinoma cell lines, whilst in normal human cholangiocytes, HiBEpiC, simvastatin inhibits proliferation with no effect on apoptosis. Simvastatin-induced reduction of cell viability was partially blocked by pre-treatment with metabolites of the mevalonate pathway. In Mz-ChA-1 cells, pre-treatment with cholesterol alone stimulated an increase in the number of viable cells and fully restored cell viability following simvastatin treatment. Treatment with simvastatin triggered the loss of lipid raft localised Rac1 and reduction of Rac1 activity in Mz-ChA-1 cells. This effect was prevented by pre-treatment with cholesterol. CONCLUSION: Collectively, our results demonstrate that simvastatin induces cholangiocarcinoma cancer cell death by disrupting Rac1/lipid raft colocalisation and depression of Rac1 activity.


Assuntos
Anticolesterolemiantes/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias dos Ductos Biliares/patologia , Ductos Biliares Intra-Hepáticos/patologia , Colangiocarcinoma/patologia , Células Epiteliais/efeitos dos fármacos , Sinvastatina/farmacologia , Proteínas rac1 de Ligação ao GTP/metabolismo , Neoplasias dos Ductos Biliares/metabolismo , Ductos Biliares Intra-Hepáticos/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Colangiocarcinoma/metabolismo , Humanos , Transdução de Sinais/efeitos dos fármacos , Estatísticas não Paramétricas , Proteínas rac1 de Ligação ao GTP/efeitos dos fármacos
9.
Cell Signal ; 22(7): 1124-31, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20211724

RESUMO

To activate the GTPase Rac in rat basophilic leukemia (RBL) cells and mouse bone marrow-derived mast cells (BMMC) a TAT fusion toxin of Bordetella dermonecrotic toxin (DNT-TAT) was constructed. The fusion toxin activated Rac1 and RhoA in vitro but only Rac in RBL cells and BMMC. DNT-TAT caused an increase in inositol phosphate formation, calcium mobilization, ERK activation and degranulation of mast cells. All these effects were inhibited by the Rho GTPase-inactivating Clostridium difficile toxin B and Clostridium sordellii lethal toxin. Also the calcium ionophore A23187 caused mast cell activation, including ERK phosphorylation, by processes involving an activation of Rac. The data indicate pleiotropic functions of Rac in mast cell activation.


Assuntos
Mastócitos/efeitos dos fármacos , Transglutaminases/farmacologia , Fatores de Virulência de Bordetella/farmacologia , Proteínas rac1 de Ligação ao GTP/fisiologia , Animais , Cálcio/metabolismo , Degranulação Celular , Linhagem Celular , Permeabilidade da Membrana Celular , Células Cultivadas , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Mastócitos/metabolismo , Mastócitos/fisiologia , Camundongos , Peptídeos/genética , Ratos , Proteínas Recombinantes de Fusão/farmacologia , Transglutaminases/genética , Fatores de Virulência de Bordetella/genética , Proteínas rac1 de Ligação ao GTP/efeitos dos fármacos , Proteínas rho de Ligação ao GTP/metabolismo
10.
Stem Cells ; 28(3): 564-72, 2010 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-20082286

RESUMO

Human mesenchymal stem cells (hMSCs) are multipotent cells that can differentiate into many cell types. Chondrogenesis is induced in hMSCs cultured as a micromass pellet to mimic cellular condensation during cartilage development, and exposed to transforming growth factor beta (TGFbeta). Interestingly, TGFbeta can also induce hMSC differentiation to smooth-muscle-like cell types, but it remains unclear what directs commitment between these two lineages. Our previous work revealed that cell shape regulates hMSC commitment between osteoblasts and adipocytes through RhoA signaling. Here we show that cell shape also confers a switch between chondrogenic and smooth muscle cell (SMC) fates. Adherent and well-spread hMSCs stimulated with TGF beta 3 upregulated SMC genes, whereas cells allowed to attach onto micropatterned substrates, but prevented from spreading and flattening, upregulated chondrogenic genes. Interestingly, cells undergoing SMC differentiation exhibited little change in RhoA, but significantly higher Rac1 activity than chondrogenic cells. Rac1 activation inhibited chondrogenesis and was necessary and sufficient for inducing SMC differentiation. Furthermore, TGF beta 3 and Rac1 signaling upregulated N-cadherin, which was required for SMC differentiation. These results demonstrate a chondrogenic-SMC fate decision mediated by cell shape, Rac1, and N-cadherin, and highlight the tight coupling between lineage commitment and the many changes in cell shape, cell-matrix adhesion, and cell-cell adhesion that occur during morphogenesis.


Assuntos
Antígenos CD/metabolismo , Caderinas/metabolismo , Linhagem da Célula/fisiologia , Forma Celular/fisiologia , Condrócitos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Miócitos de Músculo Liso/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Antígenos CD/efeitos dos fármacos , Antígenos CD/genética , Caderinas/efeitos dos fármacos , Caderinas/genética , Adesão Celular/efeitos dos fármacos , Adesão Celular/genética , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem da Célula/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Células Cultivadas , Condrócitos/citologia , Condrócitos/efeitos dos fármacos , Condrogênese/efeitos dos fármacos , Condrogênese/fisiologia , Matriz Extracelular/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Desenvolvimento Muscular/efeitos dos fármacos , Desenvolvimento Muscular/fisiologia , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Fator de Crescimento Transformador beta3/metabolismo , Fator de Crescimento Transformador beta3/farmacologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética , Proteínas rac1 de Ligação ao GTP/efeitos dos fármacos , Proteínas rac1 de Ligação ao GTP/genética
11.
Int J Oncol ; 35(3): 593-9, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19639179

RESUMO

Benzyl isothiocyanate can exert anti-tumor effect by arrest of cell cycle progression and induction of apoptosis in human pancreatic cancer cells. Among them, the dissection of the molecular mechanism of induction of apoptosis is important because the knowledge may be exploited for both cancer prevention and treatment. Our studies reported here indicate that BITC-mediated apoptosis involves the disappearance of intact 21-kDa Bid protein, cytochrome c release and predominant procaspase-3 cleavage. Using adenocarcinoma and metastatic pancreatic cancer cells, we investigated whether this dietary isothiocyanate induces apoptosis by converging two major pathways: the death receptor-mediated extrinsic and the mitochondrial intrinsic pathway. Indeed, cell surface receptor analysis by flow cytometry demonstrates the up-regulation of DR4, a member of death receptor family in BITC exposed pancreatic cancer cells. Since BITC is able to trigger death receptor signaling, we were interested in examining the effects of BITC and death receptor ligand TRAIL together on pancreatic cancer cell death. Interestingly, BITC augments TRAIL-induced apoptosis in both metastatic and adenocarcinoma cells. Moreover, we report for the first time that the sensitivity of metastatic pancreatic cancer cells to this isothiocyanate might be due to down-modulation of the proangiogenic molecule small GTPase Rac1 and caspase-3 substrate RasGAP, a regulator of Rho GTPase family.


Assuntos
Adenocarcinoma/metabolismo , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Isotiocianatos/farmacologia , Neoplasias Pancreáticas/metabolismo , Western Blotting , Linhagem Celular Tumoral , Regulação para Baixo , Citometria de Fluxo , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Receptores do Fator de Necrose Tumoral/efeitos dos fármacos , Receptores do Fator de Necrose Tumoral/metabolismo , Proteínas rac1 de Ligação ao GTP/efeitos dos fármacos , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteínas Ativadoras de ras GTPase/efeitos dos fármacos , Proteínas Ativadoras de ras GTPase/metabolismo
12.
Cell Struct Funct ; 33(1): 123-32, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18388398

RESUMO

Porphyromonas gingivalis, a periodontal pathogen, was previously suggested to exploit alpha5beta1 integrin and lipid rafts to invade host cells. However, it is unknown if the functional roles of these host components are distinct from one another during bacterial invasion. In the present study, we analyzed the mechanisms underlying P. gingivalis invasion, using fluorescent beads coated with bacterial membrane vesicles (MV beads). Cholesterol depletion reagents including methyl-beta-cyclodextrin (MbetaCD) drastically inhibited the entry of MV beads into epithelial cells, while they were less effective on bead adhesion to the cells. Bead entry was also abolished in CHO cells deficient in sphingolipids, components of lipid rafts, whereas adhesion was negligibly influenced. Following MbetaCD treatment, downstream events leading to actin polymerization were abolished; however, alpha5beta1 integrin was recruited to beads attached to the cell surface. Dominant-negative Rho GTPase Rac1 abolished cellular engulfment of the beads, whereas dominant-negative Cdc42 did not. Following cellular interaction with the beads, Rac1 was found to be translocated to the lipid rafts fraction, which was inhibited by MbetaCD. These results suggest that alpha5beta1 integrin, independent of lipid rafts, promotes P. gingivalis adhesion to epithelial cells, while the subsequent uptake process requires lipid raft components for actin organization, with Rho GTPase Rac1.


Assuntos
Aderência Bacteriana/fisiologia , Células Epiteliais/fisiologia , Corantes Fluorescentes/química , Integrina alfa5beta1/metabolismo , Microdomínios da Membrana/metabolismo , Porphyromonas gingivalis/fisiologia , Actinas/efeitos dos fármacos , Actinas/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/fisiologia , Células Cultivadas , Colesterol/metabolismo , Corantes Fluorescentes/metabolismo , Células HeLa , Humanos , Microscopia de Fluorescência , Microesferas , Porphyromonas gingivalis/citologia , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Esfingolipídeos/metabolismo , Propriedades de Superfície , beta-Ciclodextrinas/farmacologia , Proteína cdc42 de Ligação ao GTP/efeitos dos fármacos , Proteína cdc42 de Ligação ao GTP/fisiologia , Proteínas rac1 de Ligação ao GTP/efeitos dos fármacos , Proteínas rac1 de Ligação ao GTP/fisiologia
13.
J Neurosci Res ; 86(9): 2062-70, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18335519

RESUMO

Paraquat (1,1'-dimethyl-4,4'-bipyridinium) is structurally similar to the neurotoxin 1-methyl-4-phenyl-4-phenylpyridium ion (MPP+), the active metabolite of the parkinsonism-inducing agent 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), which can induce the parkinsonism property in rodents, nonhuman primates, and human. In contrast to the neurotoxic effects of paraquat, little is known about its effects on glial cells. Here, we examined the mechanisms of paraquat toxicity in glial cells in culture. Paraquat treatment also reduced the viability of C6 glial cells in primary astrocyte cultures, and cell death was mostly apoptotic in nature. PKCdelta played a central role in the paraquat-induced glial cell death: (1) the PKCdelta-specific inhibitor rottlerin blocked paraquat-induced glial cell death; (2) paraquat induced tyrosine and threonine phosphorylation of PKCdelta; and (3) transfection of the dominant-negative mutant of PKCdelta attenuated paraquat toxicity. PKCdelta was also involved in the generation of reactive oxygen species (ROS), which mediated the paraquat toxicity. The nicotinamide adenine dinucleotide phosphate (reduced form) oxidase (NADPH oxidase) inhibitor diphenyleneiodonium blocked the paraquat-induced ROS production and subsequent cell death, indicating the involvement of NADPH oxidase in the cytotoxic action of paraquat in glia. PKCdelta was also important in glial cell death induced by MPP+ but not in that induced by rotenone. Last, Rac1 appeared to antagonize paraquat toxicity in glia. These results indicate a gliotoxic effect of paraquat and an opposing role of PKCdelta and Rac1 in paraquat-induced glial cell death.


Assuntos
Astrócitos/enzimologia , Morte Celular/efeitos dos fármacos , Neuroglia/enzimologia , Paraquat/toxicidade , Proteína Quinase C-delta/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Acetofenonas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Benzopiranos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Glioma , Neuroglia/citologia , Neuroglia/efeitos dos fármacos , Proteína Quinase C-delta/efeitos dos fármacos , Proteína Quinase C-delta/genética , Ratos , Proteínas Recombinantes/efeitos dos fármacos , Proteínas Recombinantes/metabolismo , Transfecção , Proteínas rac1 de Ligação ao GTP/efeitos dos fármacos , Proteínas rac1 de Ligação ao GTP/genética
14.
Neurochem Int ; 51(2-4): 216-26, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17561310

RESUMO

Two signaling pathways, phosphoinositide 3-kinase (PI-3k)/Akt and Ras/MAPK, are major effectors triggered by nerve growth factor (NGF). Rac1, Cdc42 and GSK-3beta are reported to be targets of PI-3k in the signal transduction for neurite outgrowth. Immediately after NGF was added, broad ruffles were observed temporarily around the periphery of PC12 cells prior to neurite growth. As PC12D cells are characterized by a very rapid extension of neurites in response to various agents, the signaling pathways described above were studied in relation to the NGF-induced formation of ruffles and outgrowth of neurites. Wortmannin, an Akt inhibitor (V), and GSK-3beta inhibitor (SB425286) suppressed the neurite growth in NGF-treated cells, but not in dbcAMP-treated cells. The outgrowth of neurites induced by NGF but not by dbcAMP was inhibited with the expression of mutant Ras. But upon the expression of dominant-negative Rac1, cells often extended protrusions, incomplete neurites, lacking F-actin. Intact neurites were observed in cells with dominant-negative Cdc42. These results suggest that NGF-dependent neurite outgrowth occurs via a mechanism involving activation of the Ras/PI-3K/Akt/GSK-3beta pathway, while dbcAMP-dependent neurite growth might be induced in a distinct manner. However, inhibitors for GSK-3beta and PI-3k (wortmannin) did not suppress the NGF-dependent formation of ruffles. In addition, the formation of ruffles was not inhibited by the expression of mutant Ras. On the other hand, it was suppressed by the expression of dominant-negative Rac1 or Cdc42. These results suggest that the NGF-induced ruffling requires activation of Rac1 and Cdc42, but does not require Ras, PI-3k, Akt and GSK-3beta. Taken together, the NGF-dependent formation of ruffles might not require Ras/PI-3k/Akt/GSK-3beta, but these pathways might contribute to the formation of intact neurites due to combined actions including Rac1.


Assuntos
Diferenciação Celular/fisiologia , Extensões da Superfície Celular/metabolismo , Fator de Crescimento Neural/metabolismo , Neuritos/fisiologia , Transdução de Sinais/fisiologia , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Extensões da Superfície Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Quinase 3 da Glicogênio Sintase/efeitos dos fármacos , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Mutação/genética , Fator de Crescimento Neural/farmacologia , Neuritos/efeitos dos fármacos , Células PC12 , Fosfatidilinositol 3-Quinases/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Transdução de Sinais/efeitos dos fármacos , Proteína cdc42 de Ligação ao GTP/efeitos dos fármacos , Proteína cdc42 de Ligação ao GTP/genética , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/efeitos dos fármacos , Proteínas rac1 de Ligação ao GTP/genética , Proteínas ras/efeitos dos fármacos , Proteínas ras/metabolismo
15.
J Mol Cell Cardiol ; 40(1): 87-95, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16332375

RESUMO

Estrogens confer atheroprotective effects that remain poorly understood. We hypothesised that estrogens directly target monocytes, and investigated the pathways via which estrogens might impact on monocyte adhesion. In an in vitro model of the vasculature (parallel plate laminar flow chamber, 2 dynes/cm2), 17beta-estradiol (24 h, 0.1-1 microM) potently inhibits monocyte adhesion. In parallel, 17beta-estradiol down-regulates Rac1 GTPase activity in monocytes. Transfection of monocytic cells with dominant-negative Rac1N17 significantly decreases adhesion to human endothelial cells, while constitutively-active Rac1L61 augments adhesion. As determined by pull-down assays, Rac1 is rapidly activated by the chemokine stromal-derived factor-1 (SDF-1) in human monocytes (100 nM, 30 s). Within the same time period, SDF-1 mediates both ICAM-1/beta2- and VCAM-1/beta1-integrin-dependent monocyte adhesion, which is significantly decreased in cells overexpressing dominant-negative Rac1N17. Inhibitor studies revealed that Rac1-triggered monocyte adhesion is dependent upon actin rearrangement, while production of reactive oxygen species via Rac1 is not involved. Estrogen directly inhibits monocyte adhesion via down-regulation of Rac1, which is both necessary and sufficient to enhance monocyte adhesion under physiological flow conditions. These studies extend current knowledge about the mechanisms responsible for the vascular recruitment of pro-inflammatory cells, and potentially open up new avenues for the therapy of atherosclerosis.


Assuntos
Adesão Celular/efeitos dos fármacos , Estradiol/farmacologia , Monócitos/citologia , Monócitos/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Actinas/metabolismo , Técnicas de Cultura de Células/métodos , Células Cultivadas , Quimiocina CXCL12 , Quimiocinas CXC/metabolismo , Quimiocinas CXC/farmacologia , Regulação para Baixo , Estradiol/metabolismo , Humanos , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Monócitos/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Receptores CXCR4/efeitos dos fármacos , Receptores CXCR4/metabolismo , Transdução de Sinais , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/metabolismo , Proteínas rac1 de Ligação ao GTP/efeitos dos fármacos , Proteínas rac1 de Ligação ao GTP/genética
16.
Am J Physiol Cell Physiol ; 288(2): C450-7, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15469954

RESUMO

Primary cultures of guinea pig gastric mucosal cells express NADPH oxidase 1 (Nox1), a homolog of gp91(phox), and produce superoxide anion (O2-) at a rate of approximately 100 nmol.mg protein(-1).h(-1) in response to Helicobacter pylori (H. pylori) lipopolysaccharide (LPS) from virulent type I strains. The upregulated O2- production also enhances H. pylori LPS-stimulated tumor necrosis factor-alpha or cyclooxygenase-2 mRNA expression, which suggests a potential role for Nox1 in the pathogenesis of H. pylori-associated diseases. The H. pylori LPS-stimulated O2- production in cultured gastric mucosal cells was inhibited by actinomycin D as well as cycloheximide, suggesting that the induction is regulated at the transcriptional level. The LPS treatment not only increased the Nox1 mRNA to a greater extent but also induced expression of the message-encoding, Nox-organizing protein 1 (NOXO1), a novel p47phox homolog required for Nox1 activity. In addition, H. pylori LPS activated Rac1; i.e., it converted Rac1 to the GTP-bound state. A phosphoinositide 3-kinase inhibitor, LY-294002, blocked H. pylori LPS-induced Rac1 activation and O2- generation without interfering with the expression of Nox1 and NOXO1 mRNA. O2- production inhibited by LY-294002 was completely restored by transfection of an adenoviral vector encoding a constitutively active Rac1 but not an inactive Rac1 or a constitutively active Cdc42. These findings indicate that Rac1 plays a crucial role in Nox1 activation. Thus the H. pylori LPS-stimulated O2- production in gastric mucosal cells appears to require two distinct events: 1) transcriptional upregulation of Nox1 and NOXO1 and 2) activation of Rac1.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/biossíntese , Mucosa Gástrica/metabolismo , Lipopolissacarídeos/farmacologia , NADH NADPH Oxirredutases/biossíntese , Proteínas rac1 de Ligação ao GTP/efeitos dos fármacos , Proteínas Adaptadoras de Transporte Vesicular/efeitos dos fármacos , Animais , Northern Blotting , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Mucosa Gástrica/efeitos dos fármacos , Cobaias , Peptídeos e Proteínas de Sinalização Intracelular/farmacologia , Masculino , NADH NADPH Oxirredutases/efeitos dos fármacos , NADPH Oxidase 1 , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Superóxidos/metabolismo , Transcrição Gênica , Proteínas rac1 de Ligação ao GTP/metabolismo
17.
J Neurochem ; 91(2): 501-12, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15447683

RESUMO

G-protein-coupled receptors signal through Rho to induce actin cytoskeletal rearrangement. We previously demonstrated that thrombin stimulates Rho-dependent process retraction and rounding of 1321N1 astrocytoma cells. Surprisingly, while lysophosphatidic acid (LPA) activated RhoA in 1321N1 cells, it failed to produce cell rounding. Thrombin, unlike LPA, decreased Rac1 activity, and activated (GTPase-deficient) Rac1 inhibited thrombin-stimulated cell rounding, while expression of dominant-negative Rac1 promoted LPA-induced rounding. LPA and thrombin receptors appear to differ in coupling to Gi, as LPA but not thrombin-stimulated 1321N1 cell proliferation was pertussis toxin-sensitive. Blocking Gi with pertussis toxin enabled LPA to induce cell rounding and to decrease activated Rac1. These data support the hypothesis that Rac1 and Gi activation antagonize cell rounding. Thrombin and LPA receptors also differentially activated Gq pathways as thrombin but not LPA increased InsP3 formation and reduced phosphatidylinositol 4,5-bisphosphate (PIP2) levels. Microinjection of the plekstrin homology domain of phospholipase C (PLC)delta1, which binds PIP2, enabled LPA to elicit cell rounding, consistent with a requirement for PIP2 reduction. We suggest that Rho-mediated cytoskeletal responses are enhanced by concomitant reductions in cellular levels of PIP2 and Rac1 activation and thus effected only by G-protein-coupled receptors with appropriate subsets of G protein activation.


Assuntos
Astrocitoma/metabolismo , Citoesqueleto/metabolismo , Lisofosfolipídeos/farmacologia , Fosfatidilinositol 4,5-Difosfato/fisiologia , Proteínas rac1 de Ligação ao GTP/fisiologia , Proteínas rho de Ligação ao GTP/metabolismo , Astrocitoma/tratamento farmacológico , Linhagem Celular Tumoral , Tamanho Celular/efeitos dos fármacos , Tamanho Celular/fisiologia , Citoesqueleto/efeitos dos fármacos , DNA/biossíntese , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Humanos , Microinjeções , Fosfatidilinositol 4,5-Difosfato/biossíntese , Fosfatidilinositol 4,5-Difosfato/genética , Estrutura Terciária de Proteína/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Trombina/farmacologia , Fosfolipases Tipo C/efeitos dos fármacos , Fosfolipases Tipo C/metabolismo , Proteínas rac1 de Ligação ao GTP/efeitos dos fármacos , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rho de Ligação ao GTP/efeitos dos fármacos
18.
J Biol Chem ; 279(42): 44197-210, 2004 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-15304504

RESUMO

We observed evolutionary conservation of canonical nuclear localization signal sequences (K(K/R)X(K/R)) in the C-terminal polybasic regions (PBRs) of some Rac and Rho isoforms. Canonical D-box sequences (RXXL), which target proteins for proteasome-mediated degradation, are also evolutionarily conserved near the PBRs of these small GTPases. We show that the Rac1 PBR (PVKKRKRK) promotes Rac1 nuclear accumulation, whereas the RhoA PBR (RRGKKKSG) keeps RhoA in the cytoplasm. A mutant Rac1 protein named Rac1 (pbrRhoA), in which the RhoA PBR replaces the Rac1 PBR, has greater cytoplasmic localization, enhanced resistance to proteasome-mediated degradation, and higher protein levels than Rac1. Mutating the D-box by substituting alanines at amino acids 174 and 177 significantly increases the protein levels of Rac1 but not Rac1(pbrRhoA). These results suggest that Rac1 (pbrRhoA) is more resistant than Rac1 to proteasome-mediated degradative pathways involving the D-box. The cytoplasmic localization of Rac1(pbrRhoA) provides the most obvious reason for its resistance to proteasome-mediated degradation, because we show that Rac1(pbrRhoA) does not greatly differ from Rac1 in its ability to stimulate membrane ruffling or to interact with SmgGDS and IQGAP1-calmodulin complexes. These findings support the model that nuclear localization signal sequences in the PBR direct Rac1 to the nucleus, where Rac1 participates in signaling pathways that ultimately target it for degradation.


Assuntos
Núcleo Celular/metabolismo , Proteínas rac1 de Ligação ao GTP/química , Proteínas rac1 de Ligação ao GTP/metabolismo , Alanina , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Células CHO , Cricetinae , Inibidores de Cisteína Proteinase/farmacologia , Humanos , Leupeptinas/farmacologia , Mutagênese Sítio-Dirigida , Fragmentos de Peptídeos , Biossíntese de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas rac1 de Ligação ao GTP/efeitos dos fármacos , Proteína rhoA de Ligação ao GTP/química , Proteína rhoA de Ligação ao GTP/metabolismo
19.
Exp Cell Res ; 295(2): 497-511, 2004 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15093747

RESUMO

Ras family small GTPases play a critical role in malignant transformation, and Rho subfamily members contribute significantly to this process. Anchorage-independent growth and the ability to avoid detachment-induced apoptosis (anoikis) are hallmarks of transformed epithelial cells. In this study, we have demonstrated that constitutive activation of Cdc42 inhibits anoikis in Madin-Darby canine kidney (MDCK) epithelial cells. We showed that activated Cdc42 stimulates the ERK, JNK, and p38 MAPK pathways in suspension condition; however, inhibition of these signaling does not affect Cdc42-stimulated cell survival. However, we demonstrated that inhibition of phosphatidylinositol 3-kinase (PI3K) pathway abolishes the protective effect of Cdc42 on anoikis. Taking advantage of a double regulatory expression system, we also showed that Cdc42-stimulated cell survival in suspension condition is, at least in part, mediated by Rac1. We also provide evidence for a positive feedback loop involving Rac1 and PI3K. In addition, we show that the survival functions of both constitutively active Cdc42 and Rac1 GTPases are abrogated by Latrunculin B, an actin filament-depolymerizing agent, implying an important role for the actin cytoskeleton in mediating survival signaling activated by Cdc42 and Rac1. Together, our results indicate a role for Cdc42 in anchorage-independent survival of epithelial cells. We also propose that this survival function depends on a positive feedback loop involving Rac1 and PI3K.


Assuntos
Anoikis , Células Epiteliais/metabolismo , Proteínas Serina-Treonina Quinases , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Actinas/metabolismo , Animais , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Linhagem Celular , Sobrevivência Celular , Cães , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Fator de Crescimento Epidérmico/metabolismo , Células Epiteliais/enzimologia , Retroalimentação Fisiológica , Proteínas Quinases JNK Ativadas por Mitógeno , Rim/citologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais , Tiazóis/farmacologia , Tiazolidinas , Proteína cdc42 de Ligação ao GTP/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno , Proteínas rac1 de Ligação ao GTP/efeitos dos fármacos
20.
Brain Res Mol Brain Res ; 123(1-2): 18-26, 2004 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-15046862

RESUMO

The mechanisms linked to the neuritogenic effect of PACAP acting in synergy with NGF were analyzed in PC12 cells. Recently, we have shown that PACAP synergizes with NGF to stimulate PACAP gene transcription and neurite outgrowth, differentially dependent on both the ERK1/2 and p38 MAP kinase pathways in PC12 cells. This suggests that PACAP modulates mitogen signaling pathways governing cell differentiation, in part through MAP kinase activation and an autocrine mechanism. Here, we studied the mechanism of the underlying neuritogenic actions of PACAP. PACAP induced transient activation of Rac1, a small GTPase involved in neurite outgrowth, in a PI3-kinase-independent manner, and stimulated accumulation of active Rac1 at filamentous actin-rich protrusions on the cell surface to induce subsequent neurite formation. PACAP had no additional effect on the activity of Rac1 beyond the effect of NGF and failed to activate Ras or Cdc42. By contrast, simultaneous treatment with PACAP and NGF acts in synergy to induce prolonged activation of ERK1/2. These results indicate for the first time that PACAP induces activation of Rac1 associated with neurite outgrowth and suggest that the synergistic effect of PACAP and NGF on neurite extension is due to enhanced activation of ERK1/2.


Assuntos
Diferenciação Celular/fisiologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fator de Crescimento Neural/metabolismo , Neuritos/metabolismo , Neuropeptídeos/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Citoesqueleto de Actina/efeitos dos fármacos , Citoesqueleto de Actina/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Extensões da Superfície Celular/efeitos dos fármacos , Extensões da Superfície Celular/metabolismo , Extensões da Superfície Celular/ultraestrutura , Sinergismo Farmacológico , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Fator de Crescimento Neural/farmacologia , Neuritos/efeitos dos fármacos , Neuritos/ultraestrutura , Neuropeptídeos/farmacologia , Células PC12 , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase , Ratos , Proteínas rac1 de Ligação ao GTP/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA