Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 150
Filtrar
1.
Br J Cancer ; 110(5): 1307-15, 2014 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-24518591

RESUMEN

BACKGROUND: Glioblastoma multiforme (GBM), a highly invasive primary brain tumour, remains an incurable disease. Rho GTPases and their activators, guanine nucleotide exchange factors (GEFs), have central roles in GBM invasion. Anti-angiogenic therapies may stimulate GBM invasion via HGF/c-Met signalling. We aim to identify mediators of HGF-induced GBM invasion that may represent targets in a combination anti-angiogenic/anti-invasion therapeutic paradigm. METHODS: Guanine nucleotide exchange factor expression was measured by microarray analysis and western blotting. Specific depletion of proteins was accomplished using siRNA. Cell invasion was determined using matrigel and brain slice assays. Cell proliferation and survival were monitored using sulforhodamine B and colony formation assays. Guanine nucleotide exchange factor and GTPase activities were determined using specific affinity precipitation assays. RESULTS: We found that expression of Dock7, a GEF, is elevated in human GBM tissue in comparison with non-neoplastic brain. We showed that Dock7 mediates serum- and HGF-induced glioblastoma cell invasion. We also showed that Dock7 co-immunoprecipitates with c-Met and that this interaction is enhanced upon HGF stimulation in a manner that is dependent on the adaptor protein Gab1. Dock7 and Gab1 also co-immunoprecipitate in an HGF-dependent manner. Furthermore, Gab1 is required for HGF-induced Dock7 and Rac1 activation and glioblastoma cell invasion. CONCLUSIONS: Dock7 mediates HGF-induced GBM invasion. Targeting Dock7 in GBM may inhibit c-MET-mediated invasion in tumours treated with anti-angiogenic regimens.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Proteínas Activadoras de GTPasa/genética , Glioblastoma/metabolismo , Glioblastoma/patología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Inhibidores de la Angiogénesis/genética , Inhibidores de la Angiogénesis/metabolismo , Neoplasias Encefálicas/genética , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Supervivencia Celular/fisiología , Glioblastoma/genética , Factores de Intercambio de Guanina Nucleótido/genética , Factor de Crecimiento de Hepatocito/genética , Humanos , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo
2.
Angiogenesis ; 14(4): 457-66, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21789714

RESUMEN

Angiogenesis, the sprouting of new blood vessels from the pre-existing vasculature, is a well established target in anti-cancer therapy. It is thought that the Rho GTPase Rac1 is required during vascular endothelial growth factor (VEGF)-mediated angiogenesis. In the present study, we have used a clinically relevant RNA interference approach to silence Rac1 expression. Human umbilical vein endothelial cells were transiently transfected with non-specific control siRNA (siNS) or Rac1 siRNA (siRac1) using electroporation or Lipofectamine 2000. Functional assays with transfected endothelial cells were performed to determine the effect of Rac1 knockdown on angiogenesis in vitro. Silencing of Rac1 inhibited VEGF-mediated tube formation, cell migration, invasion and proliferation. In addition, treatment with Rac1 siRNA inhibited angiogenesis in an in vivo Matrigel plug assay. Intratumoral injections of siRac1 almost completely inhibited the growth of grafted Neuro2a tumors and reduced tumor angiogenesis. Together, these data indicate that Rac1 is an important regulator of VEGF-mediated angiogenesis. Knockdown of Rac1 may represent an attractive approach to inhibit tumor angiogenesis and growth.


Asunto(s)
Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Proteína de Unión al GTP rac1/metabolismo , Análisis de Varianza , Secuencia de Bases , Western Blotting , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Colágeno , Combinación de Medicamentos , Electroporación , Humanos , Laminina , Datos de Secuencia Molecular , Proteoglicanos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ensayo de Radioinmunoprecipitación , Transfección , Venas Umbilicales/citología , Proteína de Unión al GTP rac1/genética
3.
Int J Popul Data Sci ; 5(3): 1359, 2020 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-34036178

RESUMEN

INTRODUCTION: Fetal Alcohol Spectrum Disorder (FASD) is a neurodevelopmental disorder caused by prenatal alcohol exposure (PAE). FASD research is a rapidly growing field that crosses multiple disciplines. To ensure research is relevant and meaningful for people living with FASD, their families, and the broader public there is a need to engage community members in setting priorities for research. OBJECTIVES: Our primary objective was to formally identify the views of people living with FASD, their parents/caregivers, service providers, and the general community on the research priorities for FASD and alcohol use in pregnancy in Australia. Our secondary objective was to provide an overview of current research in the highest priority areas identified. METHODS: The approach for this study involved two community surveys and a consensus workshop, followed by a rapid literature review. Survey responses (n = 146) were collected and grouped using qualitative thematic analysis. The themes identified were then ranked in a second survey (n = 45). The 22 highest ranked themes were considered in a workshop with 21 community members, and consensus on the top ten priority areas was sought. The priority areas were grouped into conceptually similar topics and rapid literature reviews were undertaken on each. RESULTS: A diverse range of priorities was identified within key areas of prevention, diagnosis, and therapy. On request from participants, separate priority lists were developed by Aboriginal and non-Aboriginal participants. CONCLUSION: There is need for a national network of researchers to take forward the research commenced by the Centre of Research Excellence, FASD Research Australia, in addressing community priorities. KEY WORDS: Community, priorities, FASD, rapid review, Australia.

4.
Trends Cell Biol ; 10(10): 415-9, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10998597

RESUMEN

Rho family GTPases control a large variety of biological processes. Cycling of Rho proteins between the GDP-bound and the GTP-bound state is controlled by several classes of regulatory proteins. In this review, we discuss the signal-transduction mechanisms that control these regulators. We will emphasize the subcellular localization of Rho GTPases and their regulatory proteins and the role of GTP hydrolysis in signal transmission.


Asunto(s)
Transducción de Señal/fisiología , Proteínas de Unión al GTP rho/metabolismo , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo
5.
J Cell Biol ; 118(5): 1235-44, 1992 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-1512294

RESUMEN

Cell-substrate adhesion is crucial at various stages of development and for the maintenance of normal tissues. Little is known about the regulation of these adhesive interactions. To investigate the role of GTPases in the control of cell morphology and cell-substrate adhesion we have injected guanine nucleotide analogs into Xenopus XTC fibroblasts. Injection of GTP gamma S inhibited ruffling and increased spreading, suggesting an increase in adhesion. To further investigate this, we made use of GRGDSP, a peptide which inhibits binding of integrins to vitronectin and fibronectin. XTC fibroblasts injected with non-hydrolyzable analogs of GTP took much more time to round up than mock-injected cells in response to treatment with GRGDSP, while GDP beta S-injected cells rounded up in less time than controls. Injection with GTP gamma S did not inhibit cell rounding induced by trypsin however, showing that cell contractility is not significantly affected by the activation of GTPases. These data provide evidence for the existence of a GTPase which can control cell-substrate adhesion from the cytoplasm. Treatment of XTC fibroblasts with the phorbol ester 12-o-tetradecanoylphorbol-13-acetate reduced cell spreading and accelerated cell rounding in response to GRGDSP, which is essentially opposite to the effect exerted by non-hydrolyzable GTP analogs. These results suggest the existence of at least two distinct pathways controlling cell-substrate adhesion in XTC fibroblasts, one depending on a GTPase and another one involving protein kinase C.


Asunto(s)
Adhesión Celular , GTP Fosfohidrolasas/metabolismo , Secuencia de Aminoácidos , Animales , Adhesión Celular/efectos de los fármacos , Línea Celular , Citoesqueleto/efectos de los fármacos , Citoesqueleto/ultraestructura , Dimetilsulfóxido/farmacología , Fibroblastos , Guanosina 5'-O-(3-Tiotrifosfato)/farmacología , Guanosina Trifosfato/análogos & derivados , Guanosina Trifosfato/farmacología , Datos de Secuencia Molecular , Oligopéptidos/farmacología , Seudópodos/efectos de los fármacos , Seudópodos/ultraestructura , Acetato de Tetradecanoilforbol/farmacología , Xenopus
6.
J Cell Biol ; 114(3): 503-13, 1991 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-1860882

RESUMEN

We have investigated the spatial control of actin polymerization in fibroblasts using rhodamine-labeled muscle actin in; (a) microinjection experiments to follow actin dynamics in intact cells, and (b) incubation with permeabilized cells to study incorporation sites. Rhodamine-actin was microinjected into NIH-3T3 cells which were then fixed and stained with fluorescein-phalloidin to visualize total actin filaments. The incorporation of newly polymerized actin was assayed using rhodamine/fluorescein ratio-imaging. The results indicated initial incorporation of the injected actin near the tip and subsequent transport towards the base of lamellipodia at rates greater than 4.5 microns/min. Furthermore, both fluorescein- and rhodamine-intensity profiles across lamellipodia revealed a decreasing density of actin filaments from tip to base. From this observation and the presence of centripetal flux of polymerized actin we infer that the actin cytoskeleton partially disassembles before it reaches the base of the lamellipodium. In permeabilized cells we found that, in agreement with the injection studies, rhodamine-actin incorporated predominantly in a narrow strip of less than 1-microns wide, located at the tip of lamellipodia. The critical concentration for the rhodamine-actin incorporation (0.15 microM) and its inhibition by CapZ, a barbed-end capping protein, indicated that the nucleation sites for actin polymerization most likely consist of free barbed ends of actin filaments. Because any potential monomer-sequestering system is bypassed by addition of exogenous rhodamine-actin to the permeabilized cells, these observations indicate that the localization of actin incorporation in intact cells is determined, at least in part, by the presence of specific elongation and/or nucleation sites at the tips of lamellipodia and not solely by localized desequestration of subunits. We propose that the availability of the incorporation sites at the tips of lamellipodia is because of capping activities which preferentially inhibit barbed-end incorporation elsewhere in the cell, but leave barbed ends at the tips of lamellipodia free to add subunits.


Asunto(s)
Actinas/metabolismo , Fibroblastos/metabolismo , Animales , Línea Celular , Membrana Celular/metabolismo , Permeabilidad de la Membrana Celular , Procesamiento de Imagen Asistido por Computador , Cinética , Ratones , Microinyecciones , Microscopía Fluorescente , Polímeros , Conejos , Rodaminas/metabolismo , Saponinas/farmacología
7.
J Cell Biol ; 142(2): 573-86, 1998 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-9679153

RESUMEN

The organization of the actin cytoskeleton can be regulated by soluble factors that trigger signal transduction events involving the Rho family of GTPases. Since adhesive interactions are also capable of organizing the actin-based cytoskeleton, we examined the role of Cdc42-, Rac-, and Rho-dependent signaling pathways in regulating the cytoskeleton during integrin-mediated adhesion and cell spreading using dominant-inhibitory mutants of these GTPases. When Rat1 cells initially adhere to the extracellular matrix protein fibronectin, punctate focal complexes form at the cell periphery. Concomitant with focal complex formation, we observed some phosphorylation of the focal adhesion kinase (FAK) and Src, which occurred independently of Rho family GTPases. However, subsequent phosphorylation of FAK and paxillin occurs in a Rho-dependent manner. Moreover, we found Rho dependence of the assembly of large focal adhesions from which actin stress fibers radiate. Initial adhesion to fibronectin also stimulates membrane ruffling; we show that this ruffling is independent of Rho but is dependent on both Cdc42 and Rac. Furthermore, we observed that Cdc42 controls the integrin-dependent activation of extracellular signal-regulated kinase 2 and of Akt, a kinase whose activity has been demonstrated to be dependent on phosphatidylinositol (PI) 3-kinase. Since Rac-dependent membrane ruffling can be stimulated by PI 3-kinase, it appears that Cdc42, PI 3-kinase, and Rac lie on a distinct pathway that regulates adhesion-induced membrane ruffling. In contrast to the differential regulation of integrin-mediated signaling by Cdc42, Rac, and Rho, we observed that all three GTPases regulate cell spreading, an event that may indirectly control cellular architecture. Therefore, several separable signaling pathways regulated by different members of the Rho family of GTPases converge to control adhesion-dependent changes in the organization of the cytoskeleton, changes that regulate cell morphology and behavior.


Asunto(s)
GTP Fosfohidrolasas/metabolismo , Proteínas de Unión al GTP/metabolismo , Proteínas Activadoras de GTPasa , Integrinas/metabolismo , Actinas/metabolismo , Animales , Adhesión Celular , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Citoesqueleto/metabolismo , Fibronectinas/metabolismo , GTP Fosfohidrolasas/genética , Proteínas de Unión al GTP/genética , Mutación , Ratas , Transducción de Señal , Proteína de Unión al GTP cdc42 , Proteína de Unión al GTP rhoA
8.
J Cell Biol ; 143(4): 1087-99, 1998 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-9817764

RESUMEN

Human squamous cell carcinomas (SCC) frequently express elevated levels of epidermal growth factor receptor (EGFR). EGFR overexpression in SCC-derived cell lines correlates with their ability to invade in an in vitro invasion assay in response to EGF, whereas benign epidermal cells, which express low levels of EGFR, do not invade. EGF-induced invasion of SCC-derived A431 cells is inhibited by sustained expression of the dominant negative mutant of c-Jun, TAM67, suggesting a role for the transcription factor AP-1 (activator protein-1) in regulating invasion. Significantly, we establish that sustained TAM67 expression inhibits growth factor-induced cell motility and the reorganization of the cytoskeleton and cell-shape changes essential for this process: TAM67 expression inhibits EGF-induced membrane ruffling, lamellipodia formation, cortical actin polymerization and cell rounding. Introduction of a dominant negative mutant of Rac and of the Rho inhibitor C3 transferase into A431 cells indicates that EGF-induced membrane ruffling and lamellipodia formation are regulated by Rac, whereas EGF-induced cortical actin polymerization and cell rounding are controlled by Rho. Constitutively activated mutants of Rac or Rho introduced into A431 or A431 cells expressing TAM67 (TA cells) induce equivalent actin cytoskeletal rearrangements, suggesting that the effector pathways downstream of Rac and Rho required for these responses are unimpaired by sustained TAM67 expression. However, EGF-induced translocation of Rac to the cell membrane, which is associated with its activation, is defective in TA cells. Our data establish a novel link between AP-1 activity and EGFR activation of Rac and Rho, which in turn mediate the actin cytoskeletal rearrangements required for cell motility and invasion.


Asunto(s)
Factor de Crecimiento Epidérmico/metabolismo , GTP Fosfohidrolasas/metabolismo , Proteínas/metabolismo , Factor de Transcripción AP-1/metabolismo , Actinas/metabolismo , Línea Celular Transformada , Tamaño de la Célula/fisiología , Citoesqueleto/química , Citoesqueleto/metabolismo , Proteínas Activadoras de GTPasa , Expresión Génica/fisiología , Queratinocitos/química , Queratinocitos/citología , Queratinocitos/enzimología , Riñón/citología , Mutagénesis/fisiología , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Transducción de Señal/fisiología , Transfección
9.
Behav Genet ; 39(2): 183-91, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19107586

RESUMEN

C57BL/6J and DBA/2J inbred mouse strains have been extensively studied for the genetic dissection of alcohol-related phenotypes. We have previously found Syntaxin 12 to be associated with alcohol preference in C57BL/6J and DBA/2J due to its strain-specific and ethanol responsive expression in the male brain. In the current study, we combined genetic and expression analyses to assess the segregation of Syntaxin 12 c.*1370G>A polymorphism with its strain-specific expression and alcohol preference in an F (2) population (N = 427) derived from C57BL/6J and DBA/2J strains. Syntaxin 12 c.*1370G>A polymorphism was found to segregate with alcohol preference in the B6D2F2 population and a correlation was identified between Syntaxin 12 expression and alcohol preference in the selected B6D2F2 males (r = -0.473, r (2) = 0.22). We followed up our analysis in the BXD RI lines using resources from WebQTL and the Mouse Phenome Database. Our study detected significant associations of Syntaxin 12 molecular variants with its level of expression and alcohol preference in B6D2F2 males. Overall, our findings support a role for Syntaxin 12 as a potential contributor to alcohol preference in mice.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Alcoholismo/genética , Polimorfismo Genético , Proteínas Qa-SNARE/biosíntesis , Proteínas Qa-SNARE/fisiología , Animales , Encéfalo/metabolismo , Femenino , Regulación de la Expresión Génica , Genotipo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Fenotipo , Síndrome de Abstinencia a Sustancias/genética
10.
Science ; 280(5365): 898-902, 1998 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-9572733

RESUMEN

Integrin-mediated reorganization of cell shape leads to an altered cellular phenotype. Disruption of the actin cytoskeleton, initiated by binding of soluble antibody to alpha5beta1 integrin, led to increased expression of the collagenase-1 gene in rabbit synovial fibroblasts. Activation of the guanosine triphosphate-binding protein Rac1, which was downstream of the integrin, was necessary for this process, and expression of activated Rac1 was sufficient to increase expression of collagenase-1. Rac1 activation generated reactive oxygen species that were essential for nuclear factor kappa B-dependent transcriptional regulation of interleukin-1alpha, which, in an autocrine manner, induced collagenase-1 gene expression. Remodeling of the extracellular matrix and consequent alterations of integrin-mediated adhesion and cytoarchitecture are central to development, wound healing, inflammation, and malignant disease. The resulting activation of Rac1 may lead to altered gene regulation and alterations in cellular morphogenesis, migration, and invasion.


Asunto(s)
Tamaño de la Célula , Colagenasas/genética , GTP Fosfohidrolasas/metabolismo , Proteínas de Unión al GTP/metabolismo , Regulación Enzimológica de la Expresión Génica , Especies Reactivas de Oxígeno/metabolismo , Animales , Línea Celular , Citocalasina D/farmacología , Activación Enzimática , Fibroblastos , Radicales Libres , GTP Fosfohidrolasas/genética , Proteínas de Unión al GTP/genética , Genes Reporteros , Peróxido de Hidrógeno , Interleucina-1/genética , Interleucina-1/metabolismo , Metaloproteinasa 1 de la Matriz , FN-kappa B/metabolismo , Conejos , Receptores de Fibronectina/fisiología , Transcripción Genética , Proteínas de Unión al GTP rac
11.
Science ; 264(5164): 1463-7, 1994 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-7811320

RESUMEN

The small guanine nucleotide binding protein Ras participates in a growth promoting signal transduction pathway. The mechanism by which interaction of Ras with the protein kinase Raf leads to activation of Raf was studied. Raf was targeted to the plasma membrane by addition of the COOH-terminal localization signals of K-ras. This modified form of Raf (RafCAAX) was activated to the same extent as Raf coexpressed with oncogenic mutant Ras. Plasma membrane localization rather than farnesylation or the presence of the additional COOH-terminal sequence accounted for the activation of RafCAAX. The activation of RafCAAX was completely independent of Ras; it was neither potentiated by oncogenic mutant Ras nor abrogated by dominant negative Ras. Raf, once recruited to the plasma membrane, was not anchored there by Ras; most activated Raf in cells was associated with plasma membrane cytoskeletal elements, not the lipid bilayer. Thus, Ras functions in the activation of Raf by recruiting Raf to the plasma membrane where a separate, Ras-independent, activation of Raf occurs.


Asunto(s)
Membrana Celular/enzimología , Proteínas de Unión al GTP/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Citosol/enzimología , Activación Enzimática , MAP Quinasa Quinasa 1 , Proteína Quinasa 1 Activada por Mitógenos , Modelos Biológicos , Datos de Secuencia Molecular , Proteínas Serina-Treonina Quinasas/química , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas c-raf , Proteínas Recombinantes de Fusión/metabolismo
12.
Science ; 265(5179): 1713-6, 1994 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-8085158

RESUMEN

To identify proteins that may participate in the activation of the protein kinase Raf, proteins that interact with Raf were selected in a two-hybrid screen. Two members of the 14-3-3 protein family were isolated that interacted with both the amino terminal regulatory regions of Raf and the kinase domain of Raf, but did not compete with the guanine nucleotide-binding protein Ras for binding to Raf. 14-3-3 proteins associated with Raf in mammalian cells and accompanied Raf to the membrane in the presence of activated Ras. In yeast cells expressing Raf and MEK, mammalian 14-3-3 beta or 14-3-3 zeta activated Raf to a similar extent as did expression of Ras. Therefore, 14-3-3 proteins may participate in or be required for the regulation of Raf function. These findings suggest a role for 14-3-3 proteins in Raf-mediated signal transduction.


Asunto(s)
Quinasas de Proteína Quinasa Activadas por Mitógenos , Proteínas del Tejido Nervioso/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Tirosina 3-Monooxigenasa , Proteínas 14-3-3 , Secuencia de Aminoácidos , Animales , Línea Celular , Membrana Celular/enzimología , Citosol/enzimología , Activación Enzimática , Proteínas de Unión al GTP/metabolismo , Células HeLa , Humanos , MAP Quinasa Quinasa 1 , Datos de Secuencia Molecular , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas c-raf , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crecimiento & desarrollo , Transducción de Señal , Dedos de Zinc
13.
Trends Biochem Sci ; 21(5): 178-81, 1996 May.
Artículo en Inglés | MEDLINE | ID: mdl-8871402

RESUMEN

Until recently, members of the Rho family of GTPases were considered primarily to be regulators of the distinct structures making up the actin cytoskeleton. Several Rho GTPases have now also been shown to play an important role in cell transformation. In addition, Cdc42, Rac and Rho activate transcription, providing a possible mechanism for their role in growth control.


Asunto(s)
Ciclo Celular , Citoesqueleto/metabolismo , Proteínas de Unión al GTP/metabolismo , Actinas/metabolismo , Adhesión Celular , Proteínas de Unión al GTP/clasificación , Modelos Biológicos
14.
Curr Biol ; 10(14): R535-7, 2000 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-10898998

RESUMEN

Interaction of cells with the extracellular matrix influences various aspects of cellular behavior. A recent study shows that cell-substrate adhesion is necessary for effective coupling of the small GTPase Rac to its effector PAK.


Asunto(s)
Adhesión Celular/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas de Unión al GTP rac/fisiología , Animales , Membrana Celular/fisiología , Modelos Biológicos , Transducción de Señal , Quinasas p21 Activadas
15.
Curr Biol ; 10(21): 1383-6, 2000 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-11084340

RESUMEN

The small GTPase Rac has been implicated in a wide range of cellular processes, including the organization of the actin cytoskeleton, transcriptional control and endocytic vesicle trafficking [1-3]. The signaling components that mediate these functions downstream of Rac largely remain to be identified. In this study, we have identified synaptojanin 2, a polyphosphoinositide phosphatase as a novel Rac1 effector. Synaptojanin 2 directly and specifically interacts with Rac1 in a GTP-dependent manner. Expression of constitutively active Rac1 caused the translocation of synaptojanin 2 from the cytoplasm to the plasma membrane. Both activated Rac1 and a membrane-targeted version of synaptojanin 2 inhibited endocytosis of the epidermal growth factor (EGF) and transferrin receptors, a process that is known to be dependent on polyphosphoinositide lipids. Endocytosis of growth factor receptors is thought to play an important role in the regulation of cell proliferation. Thus, these results suggest that synaptojanin 2 may mediate the inhibitory effect of Rac1 on endocytosis and could contribute to Rac1-mediated control of cell growth.


Asunto(s)
Membrana Celular/metabolismo , Endocitosis , Proteínas del Tejido Nervioso/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Actinas/metabolismo , Clatrina/metabolismo , Medio de Cultivo Libre de Suero , Inhibidores Enzimáticos/metabolismo , Receptores ErbB/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/farmacología , Células HeLa , Humanos , Microscopía Fluorescente , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Transporte de Proteínas , Seudópodos/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Células Tumorales Cultivadas , Técnicas del Sistema de Dos Híbridos , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
16.
Curr Biol ; 5(11): 1296-302, 1995 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-8574587

RESUMEN

BACKGROUND: In insulin-sensitive cells, such as adipocytes and skeletal muscle, the activation of phosphoinositide 3-kinase (PI 3-kinase) is thought to be critical in allowing insulin to stimulate both the uptake of glucose and the translocation of a specialized glucose transporter, GLUT4, to the plasma membrane. However, the downstream mediators that couple PI 3-kinase to GLUT4 translocation are still not known. Recent studies have shown that the GTP-binding protein Rac mediates some of the biological effects of PI 3-kinase, and these findings have led to the suggestion that Rac may be a common mediator for a variety of responses mediated by PI 3-kinase. To determine whether Rac couples PI 3-kinase to glucose uptake in adipocytes, we produced 3T3-L1 cells expressing either a constitutively active Rac1 (V12 Rac1, containing a valine residue at position 12) or a dominant-inhibitory Rac1 (N17 Rac1, containing an asparagine residue at position 17). RESULTS: The stable expression of both V12 Rac1 and N17 Rac1 led to observable phenotypes in 3T3-L1 cells; expression of V12 Rac1 resulted in constitutive formation of lamellipodia and constitutive activation of the cJun-N-terminal kinase (JNK), whereas expression of N17 Rac1 inhibited the insulin-stimulated formation of lamellipodia. However, neither basal glucose uptake nor insulin-stimulated glucose uptake was affected by the expression of either mutant Rac protein. In addition, expression of V12 Rac1 did not reverse the inhibition of insulin-stimulated glucose uptake caused by the PI 3-kinase inhibitor wortmannin. CONCLUSIONS: These findings provide direct evidence that PI 3-kinase does not use Rac to couple the insulin receptor to glucose uptake in adipocytes. Furthermore, the finding that Rac does not mediate glucose uptake in response to insulin is consistent with the idea that PI 3-kinase couples to a variety of different effector molecules in cells, and suggests that some of the specificity in the biological responses elicited by PI 3-kinase may be mediated by the activation of different effector molecules.


Asunto(s)
Adipocitos/metabolismo , Proteínas de Unión al GTP/metabolismo , Glucosa/metabolismo , Proteínas Quinasas Activadas por Mitógenos , Proteínas Musculares , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Células 3T3 , Androstadienos/farmacología , Animales , Transporte Biológico , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Medio de Cultivo Libre de Suero , Proteínas de Unión al GTP/genética , Expresión Génica , Transportador de Glucosa de Tipo 4 , Insulina/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos , Ratones , Proteínas de Transporte de Monosacáridos/metabolismo , Fosfatidilinositol 3-Quinasas , Wortmanina , Proteínas de Unión al GTP rac
17.
Curr Biol ; 5(4): 393-403, 1995 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-7627555

RESUMEN

BACKGROUND: Phosphoinositide 3-kinases (PI 3-kinases) are thought to play an important role in coordinating the responses elicited by a variety of growth factors, oncogene products and inflammatory stimuli. These responses include activation of membrane ruffling, chemotaxis, glucose transport, superoxide production, neurite outgrowth and pp70 S6 kinase. Some of these responses are also known to be regulated by Rac, a small GTP-binding protein related to Ras. Neither the transducing elements upstream of Rac, nor those downstream of PI 3-kinase, have been defined. RESULTS: We show here that platelet-derived growth factor (PDGF) can stimulate an increase in the level of GTP-Rac by at least two distinct mechanisms: firstly, by increased guanine nucleotide exchange; and secondly, by inhibition of a Rac GTPase activity. The first of these mechanisms is essential for the activation of Rac, and we show that it is dependent upon PDGR-stimulated synthesis of phosphatidylinositol (3,4,5)-trisphosphate. CONCLUSIONS: These results suggest that Rac activation lies downstream of PI 3-kinase activation on a PDGF-stimulated signalling pathway. Furthermore, as Rac has been implicated in at least two diverse cellular responses that are also though to require activation of PI 3-kinase--a reorganization of the actin cytoskeleton known as membrane ruffling and the neutrophil oxidative burst--these results suggest that Rac may be a major effector protein for the PI 3-kinase signalling pathway in many cell types.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Línea Celular , Permeabilidad de la Membrana Celular , Activación Enzimática , Guanosina Trifosfato/metabolismo , Humanos , Fosfatidilinositol 3-Quinasas , Proteínas de Unión al GTP rac
18.
Mol Cell Biol ; 17(6): 3449-58, 1997 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9154844

RESUMEN

The Rho family members Cdc42, Rac, and Rho play a central role in the organization of the actin cytoskeleton and regulate transcription. Whereas Rac and Rho have been implicated in transformation by oncogenic Ras, the role of Cdc42 in this process remains unknown. In this study, we found that Rat1 fibroblasts expressing constitutively active V12-Cdc42 were anchorage independent and proliferated in nude mice but failed to show enhanced growth in low serum. Similar to V12-Rac1-expressing Rat1 fibroblasts, V12-Cdc42 lines displayed a high frequency of multinucleated cells. Interestingly, coexpression of dominant negative N17-Rac1 blocked the V12-Cdc42-induced multinucleated phenotype but not growth in soft agar, indicating that Cdc42 controls anchorage independence in a Rac-independent fashion. We also showed that dominant negative N17-Cdc42 inhibited Ras focus formation and anchorage-independent growth and caused reversion of the transformed morphology, indicating that Cdc42 is necessary for Ras transformation. N17-Cdc42 caused only partial inhibition of Ras-induced low-serum growth, however. In contrast, whereas N17-Rac1 also effectively inhibited Ras-induced anchorage independence, it did not revert the morphology of Ras-transformed cells. N17-Rac1 strongly inhibited low-serum growth of Ras-transformed cells, however. Together, these data provide a novel function for Cdc42 in cell proliferation and indicate that Cdc42 and Rac play distinct roles in growth control and Ras transformation.


Asunto(s)
Adhesión Celular/genética , Proteínas de Ciclo Celular/metabolismo , Transformación Celular Neoplásica , GTP Fosfohidrolasas/metabolismo , Proteínas de Unión al GTP/metabolismo , Proteínas ras/metabolismo , Células 3T3 , Animales , División Celular , Fibroblastos/metabolismo , Ratones , Microscopía Fluorescente , Ratas , Proteína de Unión al GTP cdc42 de Saccharomyces cerevisiae , Proteínas de Unión al GTP rac
19.
Mol Cell Biol ; 18(3): 1225-35, 1998 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9488437

RESUMEN

The Mas oncogene encodes a novel G-protein-coupled receptor that was identified originally as a transforming protein when overexpressed in NIH 3T3 cells. The mechanism and signaling pathways that mediate Mas transformation have not been determined. We observed that the foci of transformed NIH 3T3 cells caused by Mas were similar to those caused by activated Rho and Rac proteins. Therefore, we determined if Mas signaling and transformation are mediated through activation of a specific Rho family protein. First, we observed that, like activated Rac1, Mas cooperated with activated Raf and caused synergistic transformation of NIH 3T3 cells. Second, both Mas- and Rac1-transformed NIH 3T3 cells retained actin stress fibers and showed enhanced membrane ruffling. Third, like Rac, Mas induced lamellipodium formation in porcine aortic endothelial cells. Fourth, Mas and Rac1 strongly activated the JNK and p38, but not ERK, mitogen-activated protein kinases. Fifth, Mas and Rac1 stimulated transcription from common DNA promoter elements: NF-kappaB, serum response factor (SRF), Jun/ATF-2, and the cyclin D1 promoter. Finally, Mas transformation and some of Mas signaling (SRF and cyclin D1 but not NF-kappaB activation) were blocked by dominant negative Rac1. Taken together, these observations suggest that Mas transformation is mediated in part by activation of Rac-dependent signaling pathways. Thus, Rho family proteins are common mediators of transformation by a diverse variety of oncogene proteins that include Ras, Dbl family, and G-protein-coupled oncogene proteins.


Asunto(s)
Transformación Celular Neoplásica , GTP Fosfohidrolasas/metabolismo , Proteínas de Unión al GTP/metabolismo , Proteínas Proto-Oncogénicas c-raf , Proteínas Proto-Oncogénicas/genética , Transducción de Señal , Células 3T3 , Actinas/fisiología , Animales , Citoesqueleto/fisiología , GTP Fosfohidrolasas/genética , Proteínas de Unión al GTP/genética , Ratones , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/biosíntesis , Receptores Acoplados a Proteínas G , Proteínas Oncogénicas de Retroviridae/biosíntesis , Proteínas Oncogénicas de Retroviridae/genética , Proteínas de Unión al GTP rac , Proteínas ras/genética , Proteínas ras/metabolismo , Proteína de Unión al GTP rhoA
20.
Mol Cell Biol ; 17(3): 1324-35, 1997 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9032259

RESUMEN

Rac1 and RhoA are members of the Rho family of Ras-related proteins and function as regulators of actin cytoskeletal organization, gene expression, and cell cycle progression. Constitutive activation of Rac1 and RhoA causes tumorigenic transformation of NIH 3T3 cells, and their functions may be required for full Ras transformation. The effectors by which Rac1 and RhoA mediate these diverse activities, as well as the interrelationship between these events, remain poorly understood. Rac1 is distinct from RhoA in its ability to bind and activate the p65 PAK serine/threonine kinase, to induce lamellipodia and membrane ruffling, and to activate the c-Jun NH2-terminal kinase (JNK). To assess the role of PAK in Rac1 function, we identified effector domain mutants of Rac1 and Rac1-RhoA chimeric proteins that no longer bound PAK. Surprisingly, PAK binding was dispensable for Rac1-induced transformation and lamellipodium formation, as well as activation of JNK, p38, and serum response factor (SRF). However, the ability of Rac1 to bind to and activate PAK correlated with its ability to stimulate transcription from the cyclin D1 promoter. Furthermore, Rac1 activation of JNK or SRF, or induction of lamellipodia, was neither necessary nor sufficient for Rac1 transforming activity. Finally, the signaling pathways that mediate Rac1 activation of SRF or JNK were distinct from those that mediate Rac1 induction of lamellipodia. Taken together, these observations suggest that Rac1 regulates at least four distinct effector-mediated functions and that multiple pathways may contribute to Rac1-induced cellular transformation.


Asunto(s)
Actinas/fisiología , Transformación Celular Neoplásica , Proteínas de Unión al GTP/fisiología , Regulación de la Expresión Génica/fisiología , Proteínas Quinasas Activadas por Mitógenos , Proteínas Serina-Treonina Quinasas/fisiología , Células 3T3 , Secuencia de Aminoácidos , Animales , Células COS , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Línea Celular , Ciclina D1 , Ciclinas/genética , Proteínas de Unión al ADN/metabolismo , Endotelio Vascular , Activación Enzimática , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos , Ratones , Datos de Secuencia Molecular , Proteínas Nucleares/metabolismo , Proteínas Oncogénicas/genética , Regiones Promotoras Genéticas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Seudópodos , Proteínas Recombinantes de Fusión , Factor de Respuesta Sérica , Transducción de Señal/fisiología , Porcinos , Activación Transcripcional , eIF-2 Quinasa , Proteínas de Unión al GTP rac , Proteína de Unión al GTP rhoA
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA