Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 171
Filtrar
1.
Oncogene ; 33(31): 4021-35, 2014 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-24037532

RESUMEN

The aberrant activity of Ras homologous (Rho) family small GTPases (20 human members) has been implicated in cancer and other human diseases. However, in contrast to the direct mutational activation of Ras found in cancer and developmental disorders, Rho GTPases are activated most commonly in disease by indirect mechanisms. One prevalent mechanism involves aberrant Rho activation via the deregulated expression and/or activity of Rho family guanine nucleotide exchange factors (RhoGEFs). RhoGEFs promote formation of the active GTP-bound state of Rho GTPases. The largest family of RhoGEFs is comprised of the Dbl family RhoGEFs with 70 human members. The multitude of RhoGEFs that activate a single Rho GTPase reflects the very specific role of each RhoGEF in controlling distinct signaling mechanisms involved in Rho activation. In this review, we summarize the role of Dbl RhoGEFs in development and disease, with a focus on Ect2 (epithelial cell transforming squence 2), Tiam1 (T-cell lymphoma invasion and metastasis 1), Vav and P-Rex1/2 (PtdIns(3,4,5)P3 (phosphatidylinositol (3,4,5)-triphosphate)-dependent Rac exchanger).


Asunto(s)
Neoplasias/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Animales , División Celular , Anomalías Congénitas/metabolismo , Regulación Neoplásica de la Expresión Génica , Enfermedades Genéticas Congénitas/metabolismo , Humanos , Proteínas Proto-Oncogénicas c-vav/fisiología
2.
Oncogene ; 29(3): 380-91, 2010 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-19838215

RESUMEN

The Rheb1 and Rheb2 small GTPases and their effector mTOR are aberrantly activated in human cancer and are attractive targets for anti-cancer drug discovery. Rheb is targeted to endomembranes via its C-terminal CAAX (C=cysteine, A=aliphatic, X=terminal amino acid) motif, a substrate for posttranslational modification by a farnesyl isoprenoid. After farnesylation, Rheb undergoes two additional CAAX-signaled processing steps, Ras converting enzyme 1 (Rce1)-catalyzed cleavage of the AAX residues and isoprenylcysteine carboxyl methyltransferase (Icmt)-mediated carboxylmethylation of the farnesylated cysteine. However, whether these postprenylation processing steps are required for Rheb signaling through mTOR is not known. We found that Rheb1 and Rheb2 localize primarily to the endoplasmic reticulum and Golgi apparatus. We determined that Icmt and Rce1 processing is required for Rheb localization, but is dispensable for Rheb-induced activation of the mTOR substrate p70 S6 kinase (S6K). Finally, we evaluated whether farnesylthiosalicylic acid (FTS) blocks Rheb localization and function. Surprisingly, FTS prevented S6K activation induced by a constitutively active mTOR mutant, indicating that FTS inhibits mTOR at a level downstream of Rheb. We conclude that inhibitors of Icmt and Rce1 will not block Rheb function, but FTS could be a promising treatment for Rheb- and mTOR-dependent cancers.


Asunto(s)
Proteínas de Unión al GTP Monoméricas/metabolismo , Neuropéptidos/metabolismo , Procesamiento Proteico-Postraduccional , Transducción de Señal , Animales , Sitios de Unión/genética , Western Blotting , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Retículo Endoplásmico/metabolismo , Farnesol/análogos & derivados , Farnesol/farmacología , Aparato de Golgi/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Microscopía Fluorescente , Proteínas de Unión al GTP Monoméricas/genética , Mutación , Células 3T3 NIH , Neuropéptidos/genética , Fosforilación/efectos de los fármacos , Prenilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína Homóloga de Ras Enriquecida en el Cerebro , Ratas , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Salicilatos/farmacología , Serina-Treonina Quinasas TOR , Transfección
3.
Cancer Metastasis Rev ; 28(1-2): 77-83, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19221866

RESUMEN

DLC-1 was originally identified as a potential tumor suppressor. One of the key biochemical functions of DLC-1 is to serve as a GTPase activating protein (GAP) for members of the Rho family of GTPases, particularly Rho A-C and Cdc 42. Since these GTPases are critically involved in regulation of the cytoskeleton and cell migration, it seems clear that DLC-1 will also influence these processes. In this review we examine basic aspects of the actin cyoskeleton and how it relates to cell motility. We then delineate the characteristics of DLC-1 and other members of its family, and describe how they may have multiple effects on the regulation of cell polarity, actin organization, and cell migration.


Asunto(s)
Citoesqueleto/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Proteínas Supresoras de Tumor/fisiología , Animales , Movimiento Celular , Humanos , Ratones , Modelos Biológicos , Invasividad Neoplásica , Metástasis de la Neoplasia , Transducción de Señal , Proteínas Supresoras de Tumor/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Proteína de Unión al GTP rhoB/metabolismo , Proteína rhoC de Unión a GTP
4.
Oncogene ; 28(11): 1401-9, 2009 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-19151751

RESUMEN

DLC1 (deleted in liver cancer 1), which encodes a Rho GTPase-activating protein (Rho-GAP), is a potent tumor suppressor gene that is frequently inactivated in several human cancers. DLC1 is a multidomain protein that has been shown previously to bind members of the tensin gene family. Here we show that p120Ras-GAP (Ras-GAP; also known as RASA1) interacts and extensively colocalizes with DLC1 in focal adhesions. The binding was mapped to the SH3 domain located in the N terminus of Ras-GAP and to the Rho-GAP catalytic domain located in the C terminus of the DLC1. In vitro analyses with purified proteins determined that the isolated Ras-GAP SH3 domain inhibits DLC1 Rho-GAP activity, suggesting that Ras-GAP is a negative regulator of DLC1 Rho-GAP activity. Consistent with this possibility, we found that ectopic overexpression of Ras-GAP in a Ras-GAP-insensitive tumor line impaired the growth-suppressing activity of DLC1 and increased RhoA activity in vivo. Our observations expand the complexity of proteins that regulate DLC1 function and define a novel mechanism of the cross talk between Ras and Rho GTPases.1R01CA129610


Asunto(s)
Neoplasias/patología , Proteínas Supresoras de Tumor/fisiología , Proteína Activadora de GTPasa p120/fisiología , Proteína de Unión al GTP rhoA/metabolismo , Dominio Catalítico , Línea Celular Tumoral , Proliferación Celular , Proteínas Activadoras de GTPasa , Humanos , Proteínas Supresoras de Tumor/análisis , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/química , Proteína Activadora de GTPasa p120/análisis , Proteína Activadora de GTPasa p120/química , Dominios Homologos src
5.
Oncogene ; 26(22): 3291-310, 2007 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-17496923

RESUMEN

Mitogen-activated protein kinase (MAPK) cascades are key signaling pathways involved in the regulation of normal cell proliferation, survival and differentiation. Aberrant regulation of MAPK cascades contribute to cancer and other human diseases. In particular, the extracellular signal-regulated kinase (ERK) MAPK pathway has been the subject of intense research scrutiny leading to the development of pharmacologic inhibitors for the treatment of cancer. ERK is a downstream component of an evolutionarily conserved signaling module that is activated by the Raf serine/threonine kinases. Raf activates the MAPK/ERK kinase (MEK)1/2 dual-specificity protein kinases, which then activate ERK1/2. The mutational activation of Raf in human cancers supports the important role of this pathway in human oncogenesis. Additionally, the Raf-MEK-ERK pathway is a key downstream effector of the Ras small GTPase, the most frequently mutated oncogene in human cancers. Finally, Ras is a key downstream effector of the epidermal growth factor receptor (EGFR), which is mutationally activated and/or overexpressed in a wide variety of human cancers. ERK activation also promotes upregulated expression of EGFR ligands, promoting an autocrine growth loop critical for tumor growth. Thus, the EGFR-Ras-Raf-MEK-ERK signaling network has been the subject of intense research and pharmaceutical scrutiny to identify novel target-based approaches for cancer treatment. In this review, we summarize the current status of the different approaches and targets that are under evaluation and development for the therapeutic intervention of this key signaling pathway in human disease.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Quinasas raf/antagonistas & inhibidores , Animales , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Humanos , Quinasas Quinasa Quinasa PAM/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Neoplasias/genética , Quinasas raf/genética , Quinasas raf/fisiología
6.
Dev Cell ; 1(1): 63-72, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11703924

RESUMEN

Mutations in the neurofibromatosis type II (NF2) tumor suppressor predispose humans and mice to tumor development. The study of Nf2+/- mice has demonstrated an additional effect of Nf2 loss on tumor metastasis. The NF2-encoded protein, merlin, belongs to the ERM (ezrin, radixin, and moesin) family of cytoskeleton:membrane linkers. However, the molecular basis for the tumor- and metastasis- suppressing activity of merlin is unknown. We have now placed merlin in a signaling pathway downstream of the small GTPase Rac. Expression of activated Rac induces phosphorylation and decreased association of merlin with the cytoskeleton. Furthermore, merlin overexpression inhibits Rac-induced signaling in a phosphorylation-dependent manner. Finally, Nf2-/- cells exhibit characteristics of cells expressing activated alleles of Rac. These studies provide insight into the normal cellular function of merlin and how Nf2 mutation contributes to tumor initiation and progression.


Asunto(s)
Neurofibromina 2/metabolismo , Transducción de Señal/fisiología , Proteínas de Unión al GTP rac/metabolismo , Células 3T3 , Secuencia de Aminoácidos , Animales , Expresión Génica/fisiología , Ratones , Datos de Secuencia Molecular , Neurofibromina 2/genética , Fosforilación , Proteína de Unión al GTP cdc42/metabolismo
7.
Nat Struct Biol ; 8(12): 1037-41, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11685227

RESUMEN

Rho GTPases are activated by a family of guanine nucleotide exchange factors (GEFs) known as Dbl family proteins. The structural basis for how GEFs recognize and activate Rho GTPases is presently ill defined. Here, we utilized the crystal structure of the DH/PH domains of the Rac-specific GEF Tiam1 in complex with Rac1 to determine the structural elements of Rac1 that regulate the specificity of this interaction. We show that residues in the Rac1 beta2-beta3 region are critical for Tiam1 recognition. Additionally, we determined that a single Rac1-to-Cdc42 mutation (W56F) was sufficient to abolish Rac1 sensitivity to Tiam1 and allow recognition by the Cdc42-specific DH/PH domains of Intersectin while not impairing Rac1 downstream activities. Our findings identified unique GEF specificity determinants in Rac1 and provide important insights into the mechanism of DH/PH selection of GTPase targets.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular , Factores de Intercambio de Guanina Nucleótido/química , Factores de Intercambio de Guanina Nucleótido/metabolismo , Mapeo de Interacción de Proteínas , Proteína de Unión al GTP rac1/química , Proteína de Unión al GTP rac1/metabolismo , Células 3T3 , Sustitución de Aminoácidos/genética , Animales , Sitios de Unión , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Transformación Celular Neoplásica , Activación Enzimática , Humanos , Ligandos , Ratones , Modelos Moleculares , Mutagénesis Sitio-Dirigida/genética , Mutación/genética , Unión Proteica , Estructura Terciaria de Proteína , Proteínas/química , Proteínas/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Especificidad por Sustrato , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T , Proteína de Unión al GTP cdc42/química , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/genética
8.
J Biol Chem ; 276(51): 48257-61, 2001 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-11641393

RESUMEN

PLC-epsilon was identified recently as a phosphoinositide-hydrolyzing phospholipase C (PLC) containing catalytic domains (X, Y, and C2) common to all PLC isozymes as well as unique CDC25- and Ras-associating domains. Novel regulation of this PLC isozyme by the Ras oncoprotein and alpha-subunits (Galpha(12)) of heterotrimeric G proteins was illustrated. Sequence analyses of PLC-epsilon revealed previously unrecognized PH and EF-hand domains in the amino terminus. The known interaction of Gbetagamma subunits with the PH domains of other proteins led us to examine the capacity of Gbetagamma to activate PLC-epsilon. Co-expression of Gbeta(1)gamma(2) with PLC-epsilon in COS-7 cells resulted in marked stimulation of phospholipase C activity. Gbeta(2) and Gbeta(4) in combination with Ggamma(1), Ggamma(2), Ggamma(3), or Ggamma(13) also activated PLC-epsilon to levels similar to those observed with Gbeta(1)-containing dimers of these Ggamma-subunits. Gbeta(3) in combination with the same Ggamma-subunits was less active, and Gbeta(5)-containing dimers were essentially inactive. Gbetagamma-promoted activation of PLC-epsilon was blocked by cotransfection with either of two Gbetagamma-interacting proteins, Galpha(i1) or the carboxyl terminus of G protein receptor kinase 2. Pharmacological inhibition of PI3-kinase-gamma had no effect on Gbeta(1)gamma(2)-promoted activation of PLC-epsilon. Similarly, activation of Ras in the action of Gbetagamma is unlikely, because a mutation in the second RA domain of PLC-epsilon that blocks Ras activation of PLC failed to alter the stimulatory activity of Gbeta(1)gamma(2). Taken together, these results reveal the presence of additional functional domains in PLC-epsilon and add a new level of complexity in the regulation of this novel enzyme by heterotrimeric G proteins.


Asunto(s)
Proteínas de Unión al GTP Heterotriméricas/metabolismo , Fosfolipasas de Tipo C/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Dimerización , Activación Enzimática , Proteínas de Unión al GTP Heterotriméricas/química , Datos de Secuencia Molecular , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoinositido Fosfolipasa C , Homología de Secuencia de Aminoácido , Transducción de Señal , Fosfolipasas de Tipo C/química
9.
J Biol Chem ; 276(45): 42259-67, 2001 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-11533059

RESUMEN

Using microarray analysis, we identified a unique ras superfamily gene, termed RERG (ras-related and estrogen-regulated growth inhibitor), whose expression was decreased or lost in a significant percentage of primary human breast tumors that show a poor clinical prognosis. Importantly, high RERG expression correlated with expression of a set of genes that define a breast tumor subtype that is estrogen receptor-positive and associated with a slow rate of tumor cell proliferation and a favorable prognosis for these cancer patients. RERG mRNA expression was induced rapidly in MCF-7 cells stimulated by beta-estradiol and repressed by tamoxifen treatment. Like Ras, RERG protein exhibited intrinsic GDP/GTP binding and GTP hydrolysis activity. Unlike Ras proteins, RERG lacks a known recognition signal for COOH-terminal prenylation and was localized primarily in the cytoplasm. Expression of RERG protein in MCF-7 breast carcinoma cells resulted in a significant inhibition of both anchorage-dependent and anchorage-independent growth in vitro and inhibited tumor formation in nude mice. These features of RERG are strikingly different from most Ras superfamily GTP-binding pro-teins and suggest that the loss of RERG expression may contribute to breast tumorigenesis.


Asunto(s)
Neoplasias de la Mama/genética , Estrógenos/farmacología , Proteínas de Unión al GTP/genética , Genes ras , Inhibidores de Crecimiento/genética , Células 3T3 , Secuencia de Aminoácidos , Animales , Neoplasias de la Mama/patología , Femenino , GTP Fosfohidrolasas/metabolismo , Humanos , Ratones , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Células Tumorales Cultivadas
10.
J Biol Chem ; 276(49): 45868-75, 2001 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-11577097

RESUMEN

Normally, Rho GTPases are activated by the removal of bound GDP and the concomitant loading of GTP catalyzed by members of the Dbl family of guanine nucleotide exchange factors (GEFs). This family of GEFs invariantly contain a Dbl homology (DH) domain adjacent to a pleckstrin homology (PH) domain, and while the DH domain usually is sufficient to catalyze nucleotide exchange, possible roles for the conserved PH domain remain ambiguous. Here we demonstrate that the conserved PH domains of three distinct Dbl family proteins, intersectin, Dbs, and Tiam1, selectively bind lipid vesicles only when phosphoinositides are present. While the PH domains of intersectin and Dbs promiscuously bind several multiphosphorylated phosphoinositides, Tiam1 selectively interacts with phosphatidylinositol 3-phosphate (K(D) approximately 5-10 microm). In addition, and in contrast to recent reports, catalysis of nucleotide exchange on nonprenylated Rac1 provided by various extended portions of Tiam1 is not influenced by (a) soluble phosphoinositide head groups, (b) dibutyl versions of phosphoinositides, or (c) lipid vesicles containing phosphoinositides. Likewise, GEF activity afforded by DH/PH fragments of intersectin and Dbs are also not altered by phosphoinositide interactions. These results strongly suggest that unless all relevant components are localized to a lipid membrane surface, Dbl family GEFs generally are not intrinsically modulated by binding phosphoinositides.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular , Proteínas Portadoras/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Fosfatidilinositoles/metabolismo , Proteínas/metabolismo , Immunoblotting , Unión Proteica , Factores de Intercambio de Guanina Nucleótido Rho , Resonancia por Plasmón de Superficie , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T
11.
Cancer Lett ; 171(1): 1-10, 2001 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-11485822

RESUMEN

The important contribution of aberrant Ras activation in oncogenesis is well established. Our knowledge of the signaling pathways that are regulated by Ras is considerable. However, the number of downstream effectors of Ras continues to increase and our understanding of the role of these effector signaling pathways in mediating oncogenesis is far from complete and continues to evolve. Similarly, our understanding of the components that control mitogen-stimulated cell cycle progression is also very advanced. Where our understanding has lagged has been the delineation of the mechanism by which Ras causes a deregulation of cell cycle progression to promote the uncontrolled proliferation of the cancer cell. In this review, we summarize our current knowledge of how deregulated Ras activation alters the function of cyclin D1, p21(Cip1), and p27(Kip1). The two themes that we have emphasized are the involvement of Rho small GTPases in cell cycle regulation and the cell-type differences in how Ras signaling interfaces with the cell cycle machinery.


Asunto(s)
Ciclo Celular/fisiología , Transformación Celular Neoplásica , Proteínas Supresoras de Tumor , Proteínas ras/fisiología , Proteínas de Unión al GTP rho/fisiología , Células 3T3/citología , Células 3T3/metabolismo , Animales , Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/fisiología , División Celular/fisiología , Ciclina D1/genética , Ciclina D1/fisiología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Quinasas Ciclina-Dependientes/fisiología , Ciclinas/fisiología , Células Epiteliales/citología , Células Epiteliales/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Regulación de la Expresión Génica/fisiología , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Modelos Biológicos , Proteína de Retinoblastoma/fisiología , Transducción de Señal/fisiología
12.
Mol Cell Biol ; 21(16): 5488-99, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11463831

RESUMEN

Activated Ras, but not Raf, causes transformation of RIE-1 rat intestinal epithelial cells, demonstrating the importance of Raf-independent effector signaling in mediating Ras transformation. To further assess the contribution of Raf-dependent and Raf-independent function in oncogenic Ras transformation, we evaluated the mechanism by which oncogenic Ras blocks suspension-induced apoptosis, or anoikis, of RIE-1 cells. We determined that oncogenic versions of H-, K-, and N-Ras, as well as the Ras-related proteins TC21 and R-Ras, protected RIE-1 cells from anoikis. Surprisingly, our analyses of Ras effector domain mutants or constitutively activated effectors indicated that activation of Raf-1, phosphatidylinositol 3-kinase (PI3K), or RalGDS alone is not sufficient to promote Ras inhibition of anoikis. Treatment of Ras-transformed cells with the U0126 MEK inhibitor caused partial reversion to an anoikis-sensitive state, indicating that extracellular signal-regulated kinase activation contributes to inhibition of anoikis. Unexpectedly, oncogenic Ras failed to activate Akt, and treatment of Ras-transformed RIE-1 cells with the LY294002 PI3K inhibitor did not affect anoikis resistance or growth in soft agar. Thus, while important for Ras transformation of fibroblasts, PI3K may not be involved in Ras transformation of RIE-1 cells. Finally, inhibition of epidermal growth factor receptor kinase activity did not overcome Ras inhibition of anoikis, indicating that this autocrine loop essential for transformation is not involved in anoikis protection. We conclude that a PI3K- and RalGEF-independent Ras effector(s) likely cooperates with Raf to confer anoikis resistance upon RIE-1 cells, thus underscoring the complex nature by which Ras transforms cells.


Asunto(s)
Anoicis/genética , Transformación Celular Neoplásica/genética , Genes ras/genética , Fosfatidilinositol 3-Quinasas/genética , Animales , Línea Celular , Activación Enzimática/genética , Regulación de la Expresión Génica , Ratas , Transducción de Señal/genética
14.
Oncogene ; 20(16): 1953-63, 2001 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-11360179

RESUMEN

We utilized a cDNA expression library derived from the B6SutA(1) mouse myeloid progenitor cell line to search for novel oncogenes that promote growth transformation of NIH3T3 cells. A 2.2 kb transforming cDNA was recovered that encodes the wild type thrombin-stimulated G protein-coupled receptor PAR-1. In addition to its potent focus forming activity, constitutive overexpression of PAR-1 in NIH3T3 cells promoted the loss of anchorage- and serum-dependent growth. Although inhibitors of thrombin failed to block PAR-1 transforming activity, a PAR-1 mutant that cannot be cleaved by thrombin was nontransforming. Since the foci of transformed cells induced by PAR-1 bear a striking resemblance to those induced by activated RhoA, we determined if PAR-1 transformation was due to the aberrant activation of a specific Rho family member. Like RhoA, PAR-1 cooperated with activated Raf-1 and caused synergistic enhancement of transforming activity, induced stress fibers when microinjected into porcine aortic endothelial cells, stimulated the activity of the serum response factor and NF-kappaB transcription factors, and PAR-1 transformation was blocked by co-expression of dominant negative RhoA. Finally, PAR-1 transforming activity was blocked by pertussis toxin and by co-expression of the RGS domain of Lsc, implicating Galpha(i) and Galpha(12)/Galpha(13) subunits, respectively, as mediators of PAR-1 transformation. Taken together, these observations suggest that PAR-1 growth transformation is mediated, in part, by activation of RhoA.


Asunto(s)
Transformación Celular Neoplásica , Receptores de Trombina/fisiología , Transducción de Señal/fisiología , Proteína de Unión al GTP rhoA/fisiología , Células 3T3/citología , Células 3T3/metabolismo , Células 3T3/fisiología , Actinas/metabolismo , Animales , Adhesión Celular/fisiología , División Celular/fisiología , Línea Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , ADN Complementario/genética , Proteínas de Unión al ADN/fisiología , Subunidades alfa de la Proteína de Unión al GTP G12-G13 , Proteínas de Unión al GTP Heterotriméricas/fisiología , Ratones , Células Mieloides/fisiología , Receptor PAR-1 , Receptores de Trombina/biosíntesis , Receptores de Trombina/genética , Transfección , Proteínas de Unión al GTP rho/metabolismo
15.
J Biol Chem ; 276(29): 27145-51, 2001 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-11373293

RESUMEN

Leukemia-associated Rho guanine nucleotide exchange factor (LARG) was originally identified as a fusion partner with mixed-lineage leukemia in a patient with acute myeloid leukemia. LARG possesses a tandem Dbl homology and pleckstrin homology domain structure and, consequently, may function as an activator of Rho GTPases. In this study, we demonstrate that LARG is a functional Dbl protein. Expression of LARG in cells caused activation of the serum response factor, a known downstream target of Rho-mediated signaling pathways. Transient overexpression of LARG did not activate the extracellular signal-regulated kinase or c-Jun NH(2)-terminal kinase mitogen-activated protein kinase cascade, suggesting LARG is not an activator of Ras, Rac, or Cdc42. We performed in vitro exchange assays where the isolated Dbl homology (DH) or DH/pleckstrin homology domains of LARG functioned as a strong activator of RhoA, but exhibited no activity toward Rac1 or Cdc42. We found that LARG could complex with RhoA, but not Rac or Cdc42, in vitro, and that expression of LARG caused an increase in the levels of the activated GTP-bound form of RhoA, but not Rac1 or Cdc42, in vivo. Thus, we conclude that LARG is a RhoA-specific guanine nucleotide exchange factor. Finally, like activated RhoA, we determined that LARG cooperated with activated Raf-1 to transform NIH3T3 cells. These data demonstrate that LARG is the first functional Dbl protein mutated in cancer and indicate LARG-mediated activation of RhoA may play a role in the development of human leukemias.


Asunto(s)
Transformación Celular Neoplásica , Factores de Intercambio de Guanina Nucleótido/fisiología , Leucemia Mieloide Aguda/fisiopatología , Proteína de Unión al GTP rhoA/metabolismo , Proteínas de Unión al ADN/metabolismo , Humanos , Masculino , Proteínas Nucleares/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho , Factor de Respuesta Sérica
16.
Methods Enzymol ; 332: 221-32, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11305099

RESUMEN

In conclusion, RDA provides a fast, technically simple, and inexpensive way to characterize genes aberrantly expressed due to Ras transformation. The identification and characterization of these genes may provide insight not only into the mechanism by which Ras causes transformation, but also may identify novel targets for rational drug design and development of anticancer drugs.


Asunto(s)
Genes ras , Técnicas Genéticas , Animales , Secuencia de Bases , Línea Celular , Clonación Molecular , Cartilla de ADN/genética , ADN Complementario/genética , ADN Complementario/aislamiento & purificación , Regulación hacia Abajo , Regulación de la Expresión Génica , Hibridación de Ácido Nucleico , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , ARN Mensajero/aislamiento & purificación , Ratas
18.
Mol Cell Biol ; 21(8): 2847-57, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11283263

RESUMEN

The Rho family of Ras-related proteins, which includes Rac1, RhoA, and Cdc42, is distinguished from other members of the Ras superfamily of small GTPases in that its members possess additional sequences positioned between beta-strand 5 and alpha-helix 4, designated the insert region. Previous studies have established the importance of an intact insert region for the transforming, but not actin cytoskeletal reorganization, activities of Cdc42 and RhoA. Similarly, the insert region was determined to be essential for Rac1-mediated mitogenesis. Additionally, an intact insert region was also determined to be required for the antiapoptotic activity of Rac1 as well as for Rac1 activation of reactive oxygen species and the NF-kappaB transcription factor. However, it has not been determined whether the insert region is important for Rac1-mediated growth transformation. In this study, we assessed the requirement for the insert region in Rac1 transformation and signaling in NIH 3T3 cells. Unexpectedly, we found that a mutant of constitutively activated Rac1 that lacked the insert region retained potent transforming activity. The insert region of Rac1 was dispensable for Rac1 stimulation of transcription from the cyclin D1 promoter and for activation of the c-Jun, NF-kappaB, and E2F-1 transcription factors but was essential for Rac1 induction of serum response factor activity. While an intact insert region was dispensable for inducing reactive oxygen species production in vivo, it was required for Rac1 induction of lamellipodia. When taken together, these results show that the insert region of Rac1 serves roles in regulating actin organization and cell growth that are distinct from those of the analogous regions of Cdc42 and RhoA and support its involvement in regulating specific downstream effector interactions.


Asunto(s)
Proteínas Portadoras , Proteínas de Ciclo Celular , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Transformación Celular Neoplásica , Proteína de Unión al GTP rac1/química , Proteína de Unión al GTP rac1/metabolismo , Células 3T3 , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Ciclina D1/genética , Cartilla de ADN/genética , Proteínas de Unión al ADN/biosíntesis , Factores de Transcripción E2F , Factor de Transcripción E2F1 , Humanos , Ratones , Datos de Secuencia Molecular , Mutagénesis Insercional , FN-kappa B/metabolismo , Proteínas Nucleares/biosíntesis , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-jun/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína 1 de Unión a Retinoblastoma , Factor de Respuesta Sérica , Factor de Transcripción DP1 , Factores de Transcripción/metabolismo , Transcripción Genética , Proteína de Unión al GTP rac1/genética
19.
Oncogene ; 20(13): 1547-55, 2001 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-11313901

RESUMEN

G protein coupled receptors (GPCRs) constitute the largest family of cell surface receptors, with more than 1000 members, and are responsible for converting a diverse array of extracellular stimuli into intracellular signaling events. Most members of the family have defined roles in intermediary metabolism and generally perform these functions in well-differentiated cells. However, there is an increasing awareness that some GPCRs can also regulate proliferative signaling pathways and that chronic stimulation or mutational activation of receptors can lead to oncogenic transformation. Activating mutations in GPCRs are associated with several types of human tumors and some receptors exhibit potent oncogenic activity due to agonist overexpression. Additionally, expression screening analyses for novel oncogenes identified GPCRs whose expression causes the oncogenic transformation of NIH3T3 mouse fibroblasts. These include Mas, G2A, and the PAR-1 thrombin receptor. In this review we summarize the signaling and transforming properties of these GPCR oncoproteins. What has emerged from these studies is the delineation of a GTPase cascade where transforming GPCRs cause aberrant growth regulation via activation of Rho family small GTPases.


Asunto(s)
Transformación Celular Neoplásica , Receptores de Superficie Celular/metabolismo , Receptores Acoplados a Proteínas G , Proteínas de Unión al GTP rho/metabolismo , Proteínas de Ciclo Celular , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Modelos Biológicos , Oncogenes , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas , Receptor PAR-1 , Receptores de Trombina , Transducción de Señal
20.
Mol Med ; 7(12): 816-30, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11844870

RESUMEN

BACKGROUND: Relatively few genes have been shown to directly affect the metastatic phenotype of breast cancer epithelial cells in vivo. The Rho family of proteins, incluing the Rho, Rac and Cdc42 subfamilies, are related to the small GTP binding protein Ras and regulated diverse biological processes including gene transcription, cytoskeletal organization, cell proliferation and transformation. The effects of Cdc42, Rac and Rho on the actin cytoskeleton suggested a possible role for Rho proteins in cellular motility and metastasis; however, a formal analysis of the role of Rho proteins in breast cancer cellular growth and metastasis in vivo had not previously been performed. MATERIALS AND METHODS: We generated a panel of MTLn3 rat mammary adenocarcinoma cells that expressed similar levels of dominant inhibitory mutants of Cdc42-, Rac- and Rho-dependent signaling, to examine the contribution of these GTPases to cell spreading, guided chemotaxis, and metastasis in vivo. The ability of Rho proteins to regulate intravasation into the peripheral blood was determined by implanting MTLn3 cell stable dominant negative lines in nude mice and measuring the formation of breast cancer cell colonies grown from the peripheral blood. Serial sectioning of the lungs was performed to determine the presence of metastasis in mice in which mammary tumors expressing the dominant negative Rho family proteins had grown to a similar size. RESULTS: Cell spreading of MTLn3 cells was selectively abrogated by N17Rac1. N19RhoA and N17Cdc42 reduced the number of focal contacts (FCs) and disrupted the co-localization of vinculin with phosphotyrosine at FCs. While N17Rac1 and N17Cdc42 preferentially inhibited colony formation in soft agar, all three GTPases affected cell growth in vivo. To distinguish effects on tumorigenicity from intravasation into the bloodstream, implanted tumors were grown to the same size in nude mice. Each dominant inhibitory Rho protein reduced intravasation into the peripheral blood. Lung metastasis of MTLn3 cells was also abrogated by the dominant inhibitory Rho proteins, despite the presence of residual CFU. CONCLUSIONS: These studies demonstrate for the first time a critical role for the Rho GTPases involving independent signaling pathways to limit mammary tumor cellular growth and metastasis in vivo.


Asunto(s)
Neoplasias Mamarias Experimentales/fisiopatología , Proteínas de Unión al GTP rho/metabolismo , Adenocarcinoma , Animales , División Celular , Quimiotaxis , Factor de Crecimiento Epidérmico/metabolismo , Epitelio/fisiopatología , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/sangre , Neoplasias Mamarias Experimentales/enzimología , Familia de Multigenes , Ratas , Transducción de Señal , Fibras de Estrés , Transfección , Células Tumorales Cultivadas , Tirosina/metabolismo , Proteína de Unión al GTP cdc42/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA