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










Base de datos
Intervalo de año de publicación
3.
Eur J Cancer ; 36(10): 1269-74, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10882865

RESUMEN

Cell migration and the regulation of cadherin-mediated homotypic cell-cell interactions are critical events during development, morphogenesis and wound healing. Aberrations in signalling pathways involved in the regulation of cell migration and cadherin-mediated cell-cell adhesion contribute to tumour invasion and metastasis. The rho family proteins, including cdc42, rac1 and rhoA, regulate signalling pathways that mediate the distinct actin cytoskeleton changes required for both cellular motility and cell-cell adhesion. Recent studies indicate that rac directly influences rho activity at the GTPase level and that the reciprocal balance between rac and rho activity can determine epithelial or mesenchymal cell morphology and migratory behaviour of epithelial (tumour) cells.


Asunto(s)
Adhesión Celular/fisiología , Movimiento Celular/fisiología , Neoplasias/patología , Proteínas de Unión al GTP rho/fisiología , Cadherinas/fisiología , Comunicación Celular , Humanos , Invasividad Neoplásica/fisiopatología
4.
J Cell Biol ; 149(4): 775-82, 2000 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-10811819

RESUMEN

Proteins of the Rho family regulate cytoskeletal rearrangements in response to receptor stimulation and are involved in the establishment and maintenance of epithelial cell morphology. We recently showed that Rac is able to downregulate Rho activity and that the reciprocal balance between Rac and Rho activity is a major determinant of cellular morphology and motility in NIH3T3 fibroblasts. Using biochemical pull-down assays, we analyzed the effect of transient and sustained oncogenic Ras signaling on the activation state of Rac and Rho in epithelial MDCK cells. In contrast to the activation of Rac by growth factor-induced Ras signaling, we found that sustained signaling by oncogenic RasV12 permanently downregulates Rac activity, which leads to upregulation of Rho activity and epithelial-mesenchymal transition. Oncogenic Ras decreases Rac activity through sustained Raf/MAP kinase signaling, which causes transcriptional downregulation of Tiam1, an activator of Rac in epithelial cells. Reconstitution of Rac activity by expression of Tiam1 or RacV12 leads to downregulation of Rho activity and restores an epithelial phenotype in mesenchymal RasV12- or RafCAAX-transformed cells. The present data reveal a novel mechanism by which oncogenic Ras is able to interfere with the balance between Rac and Rho activity to achieve morphological transformation of epithelial cells.


Asunto(s)
Células Epiteliales/fisiología , Mesodermo/fisiología , Proteínas de Unión al GTP rac/metabolismo , Proteínas ras/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Animales , Transformación Celular Neoplásica , Células Cultivadas , Perros , Regulación hacia Abajo , Células Epiteliales/citología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Riñón/citología , Mesodermo/citología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Modelos Biológicos , Morfogénesis , Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-raf/metabolismo , Transducción de Señal
5.
J Cell Biol ; 147(5): 1009-22, 1999 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-10579721

RESUMEN

Using biochemical assays to determine the activation state of Rho-like GTPases, we show that the guanine nucleotide exchange factor Tiam1 functions as a specific activator of Rac but not Cdc42 or Rho in NIH3T3 fibroblasts. Activation of Rac by Tiam1 induces an epithelial-like morphology with functional cadherin-based adhesions and inhibits migration of fibroblasts. This epithelial phenotype is characterized by Rac-mediated effects on Rho activity. Transient PDGF-induced as well as sustained Rac activation by Tiam1 or V12Rac downregulate Rho activity. We found that Cdc42 also downregulates Rho activity. Neither V14Rho or N19Rho affects Rac activity, suggesting unidirectional signaling from Rac towards Rho. Downregulation of Rho activity occurs independently of Rac- induced cytoskeletal changes and cell spreading. Moreover, Rac effector mutants that are defective in mediating cytoskeleton changes or Jun kinase activation both downregulate Rho activity, suggesting that neither of these Rac signaling pathways are involved in the regulation of Rho. Restoration of Rho activity in Tiam1-expressing cells by expression of V14Rho results in reversion of the epithelioid phenotype towards a migratory, fibroblastoid morphology. We conclude that Rac signaling is able to antagonize Rho activity directly at the GTPase level, and that the reciprocal balance between Rac and Rho activity determines cellular morphology and migratory behavior in NIH3T3 fibroblasts.


Asunto(s)
Movimiento Celular/fisiología , Regulación hacia Abajo/fisiología , GTP Fosfohidrolasas/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Proteínas , Proteínas de Unión al GTP rac/metabolismo , Células 3T3 , Animales , Cadherinas/metabolismo , Adhesión Celular/fisiología , Línea Celular , Citoesqueleto/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Factores de Intercambio de Guanina Nucleótido , Ratones , Fenotipo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Biosíntesis de Proteínas , Transducción de Señal/fisiología , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T , Proteína de Unión al GTP cdc42/metabolismo
6.
Nat Cell Biol ; 1(4): 242-8, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10559923

RESUMEN

GTPases of the Rho family regulate actinomyosin-based contraction in non-muscle cells. Activation of Rho increases contractility, leading to cell rounding and neurite retraction in neuronal cell lines. Activation of Rac promotes cell spreading and interferes with Rho-mediated cell rounding. Here we show that activation of Rac may antagonize Rho by regulating phosphorylation of the myosin-II heavy chain. Stimulation of PC12 cells or N1E-115 neuroblastoma cells with bradykinin induces phosphorylation of threonine residues in the myosin-II heavy chain; this phosphorylation is Ca2+ dependent and regulated by Rac. Both bradykinin-mediated and constitutive activation of Rac promote cell spreading, accompanied by a loss of cortical myosin II. Our results identify the myosin-II heavy chain as a new target of Rac-regulated kinase pathways, and implicate Rac as a Rho antagonist during myosin-II-dependent cell-shape changes.


Asunto(s)
Actomiosina/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Animales , Bradiquinina/farmacología , Calcio/metabolismo , Línea Celular , Tamaño de la Célula/fisiología , Ratones , Células PC12 , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas , Transducción de Señal , Proteínas de Unión al GTP rho/metabolismo
7.
J Cell Biol ; 143(5): 1385-98, 1998 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-9832565

RESUMEN

We previously demonstrated that both Tiam1, an activator of Rac, and constitutively active V12Rac promote E-cadherin-mediated cell-cell adhesion in epithelial Madin Darby canine kidney (MDCK) cells. Moreover, Tiam1 and V12Rac inhibit invasion of Ras-transformed, fibroblastoid MDCK-f3 cells by restoring E-cadherin-mediated cell-cell adhesion. Here we show that the Tiam1/Rac-induced cellular response is dependent on the cell substrate. On fibronectin and laminin 1, Tiam1/Rac signaling inhibits migration of MDCK-f3 cells by restoring E-cadherin-mediated cell- cell adhesion. On different collagens, however, expression of Tiam1 and V12Rac promotes motile behavior, under conditions that prevent formation of E-cadherin adhesions. In nonmotile cells, Tiam1 is present in adherens junctions, whereas Tiam1 localizes to lamellae of migrating cells. The level of Rac activation by Tiam1, as determined by binding to a glutathione-S-transferase- PAK protein, is similar on fibronectin or collagen I, suggesting that rather the localization of the Tiam1/Rac signaling complex determines the substrate-dependent cellular responses. Rac activation by Tiam1 requires PI3-kinase activity. Moreover, Tiam1- but not V12Rac-induced migration as well as E-cadherin-mediated cell- cell adhesion are dependent on PI3-kinase, indicating that PI3-kinase acts upstream of Tiam1 and Rac.


Asunto(s)
Adhesión Celular/fisiología , Movimiento Celular/fisiología , Proteínas de Unión al GTP/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas/fisiología , Animales , Secuencia de Bases , Cadherinas/fisiología , Línea Celular , Colágeno/fisiología , Cartilla de ADN/genética , Perros , Células Epiteliales/citología , Células Epiteliales/fisiología , Matriz Extracelular/fisiología , Fibronectinas/fisiología , Proteínas de Unión al GTP/genética , Fenotipo , Proteínas/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Transducción Genética , Proteínas de Unión al GTP rac
8.
EMBO J ; 17(14): 4066-74, 1998 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-9670021

RESUMEN

Rho-like GTPases orchestrate distinct cytoskeletal changes in response to receptor stimulation. Invasion of T-lymphoma cells into a fibroblast monolayer is induced by Tiam1, an activator of the Rho-like GTPase Rac, and by constitutively active V12Rac1. Here we show that activated V12Cdc42 can also induce invasion of T-lymphoma cells. Activated RhoA potentiates invasion, but fails by itself to mimic Rac and Cdc42. However, invasion is inhibited by the Rho-inactivating C3 transferase. Thus, RhoA is required but not sufficient for invasion. Invasion of T-lymphoma cells is critically dependent on the presence of serum. Serum can be replaced by the serum-borne lipids lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P) (10(-7)-10(-6) M), which act on distinct G protein-linked receptors to activate RhoA and phospholipase C (PLC)-Ca2+ signaling. LPA- and S1P-induced invasion is preceded by Rho-dependent F-actin redistribution and pseudopodia formation. However, expression of both V14RhoA and V12Rac1 does not bypass the LPA/S1P requirement for invasion, indicating involvement of an additional signaling pathway independent of RhoA. The PLC inhibitor U-73122, but not the inactive analog U-73343, abolishes invasion. Our results indicate that T-lymphoma invasion is driven by Tiam1/Rac or Cdc42 activation, and is dependent on LPA/S1P receptor-mediated RhoA and PLC signaling pathways which lead to pseudopod formation and enhanced infiltration.


Asunto(s)
Toxinas Botulínicas , GTP Fosfohidrolasas/fisiología , Proteínas de Unión al GTP/fisiología , Linfoma de Células T/patología , Receptores de Superficie Celular/fisiología , Receptores Acoplados a Proteínas G , Transducción de Señal/fisiología , ADP Ribosa Transferasas/farmacología , Actinas , Animales , Sangre , Calcio , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/fisiología , Línea Celular , Tamaño de la Célula , Citoesqueleto , Fibroblastos , GTP Fosfohidrolasas/genética , Proteínas de Unión al GTP/genética , Factores de Intercambio de Guanina Nucleótido , Lisofosfolípidos/farmacología , Invasividad Neoplásica , Proteínas de Neoplasias , Proteína Quinasa C/fisiología , Proteínas/genética , Proteínas/fisiología , Seudópodos , Ratas , Receptores del Ácido Lisofosfatídico , Esfingosina/análogos & derivados , Esfingosina/farmacología , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T , Células Tumorales Cultivadas , Fosfolipasas de Tipo C/metabolismo , Proteína de Unión al GTP cdc42 , Proteínas de Unión al GTP rac , Proteína de Unión al GTP rhoA
9.
Science ; 278(5342): 1464-6, 1997 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-9367959

RESUMEN

Tiam1 encodes an exchange factor for the Rho-like guanosine triphosphatase Rac. Both Tiam1 and activated RacV12 promote invasiveness of T lymphoma cells. In epithelial Madin-Darby canine kidney (MDCK) cells, Tiam1 localized to adherens junctions. Ectopic expression of Tiam1 or RacV12 inhibited hepatocyte growth factor-induced scattering by increasing E-cadherin-mediated cell-cell adhesion accompanied by actin polymerization at cell-cell contacts. In Ras-transformed MDCK cells, expression of Tiam1 or RacV12 restored E-cadherin-mediated adhesion, resulting in phenotypic reversion and loss of invasiveness. These data suggest an invasion-suppressor role for Tiam1 and Rac in epithelial cells.


Asunto(s)
Adhesión Celular , Células Epiteliales/citología , Proteínas de Unión al GTP/metabolismo , Uniones Intercelulares/metabolismo , Invasividad Neoplásica , Proteínas/metabolismo , Actinas/metabolismo , Animales , Cadherinas/metabolismo , Línea Celular , Línea Celular Transformada , Movimiento Celular , Transformación Celular Neoplásica , Citoplasma/metabolismo , Células Epiteliales/metabolismo , Factor de Crecimiento de Hepatocito/farmacología , Fenotipo , Proteínas/genética , Transducción de Señal , Proteínas de Unión al GTP rac
10.
J Biol Chem ; 272(45): 28447-54, 1997 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-9353304

RESUMEN

The Rho-like GTPases Cdc42, Rac, and Rho play key roles in the regulation of the actin cytoskeleton and are implicated in transcriptional activation and cell transformation. We have previously identified the invasion-inducing Tiam1 gene, which encodes an activator of Rac. In fibroblasts, Tiam1 induces Rac-mediated membrane ruffling, which requires the N-terminal pleckstrin homology (PHn) domain. Here we show that this PHn domain is part of a protein interaction domain, which mediates membrane localization of Tiam1. After subcellular fractionation, up to 50% of Tiam1 is recovered in the Triton X-100-insoluble high speed pellet that contains small protein complexes. The regions in Tiam1 that are responsible for these protein interactions comprise the PHn domain, an adjacent putative coiled coil region (CC), and an additional flanking region (Ex). Deletions in each of these regions abolish membrane localization of Tiam1 and membrane ruffling, suggesting that they function cooperatively. Indeed, only polypeptides encompassing the PHn-CC-Ex region, and not the PHn-CC or the Ex region, localize at the membrane. These results indicate that the N-terminal PH domain is part of a larger functional Tiam1 domain that mediates protein complex formation and membrane localization of Tiam1.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Fosfoproteínas , Proteínas/metabolismo , Células 3T3 , Animales , Sitios de Unión , Proteínas Sanguíneas/química , Células COS , Membrana Celular/metabolismo , Centrifugación por Gradiente de Densidad , Factores de Intercambio de Guanina Nucleótido , Ratones , Microscopía Inmunoelectrónica , Mutagénesis Sitio-Dirigida , Unión Proteica , Proteínas/genética , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T , Células Tumorales Cultivadas
11.
J Cell Biol ; 137(2): 387-98, 1997 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-9128250

RESUMEN

Rho-like GTPases, including Cdc42, Rac, and Rho, regulate signaling pathways that control actin cytoskeletal structures and transcriptional activation. The Tiam1 gene encodes an activator of Rac1, and similarly to constitutively activated (V12)Rac1, overexpression of Tiam1 in fibroblasts induces the formation of membrane ruffles. Tiam1 contains a Dbl homology (DH) domain and adjacent pleckstrin homology (PH) domain, hallmarks for activators of Rho-like GTPases. Unique for Tiam1 are an additional PH domain and a Discs-large homology region in the NH2-terminal part of the protein. Here we show that both in fibroblasts and COS cells, membrane localization of Tiam1 is required for the induction of membrane ruffling. A detailed mutational analysis, in combination with confocal laser scanning microscopy and immunoelectron microscopy, demonstrates that the NH2-terminal PH domain of Tiam1, but not the DH-adjacent PH domain, is essential for membrane association. This NH2-terminal PH domain of Tiam1 can be functionally replaced by the myristoylated membrane localization domain of c-Src, indicating that the primary function of this PH domain is to localize the protein at the membrane. After serum starvation, both membrane association of Tiam1 and ruffling can be induced by serum, suggesting that receptor stimulation induces membrane translocation of Tiam1. Similar to V12Rac1, Tiam1 stimulates the activity of the c-Jun NH2-terminal kinase (JNK). This Rac-dependent stimulation of JNK also requires membrane association of Tiam1. We conclude that the regulated membrane localization of Tiam1 through its NH2-terminal PH domain determines the activation of distinct Rac-mediated signaling pathways.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Membrana Celular/química , Proteínas de Drosophila , Proteínas de Unión al GTP/fisiología , Proteínas Quinasas Activadas por Mitógenos , Fosfoproteínas , Proteínas/análisis , Homología de Secuencia de Aminoácido , Proteínas Supresoras de Tumor , Células 3T3 , Animales , Proteínas Sanguíneas/genética , Células COS , Activación Enzimática , Factores de Intercambio de Guanina Nucleótido , Proteínas de Insectos/genética , Proteínas Quinasas JNK Activadas por Mitógenos , Ratones , Proteínas/genética , Proteínas Proto-Oncogénicas pp60(c-src)/genética , Proteínas Recombinantes de Fusión , Eliminación de Secuencia , Transducción de Señal/fisiología , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T , Proteínas de Unión al GTP rac
13.
Oncogene ; 11(11): 2215-21, 1995 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-8570171

RESUMEN

We have recently identified the invasion-inducing Tiam1 gene by proviral insertional mutagenesis. The Tiam1 protein shares a Dbl homology (DH) domain with an increasing number of oncoproteins, some of which have been shown to function as GDP dissociation stimulators (GDS) for small GTPases of the Rho family. In vitro and in vivo analyses indicate that Tiam1 activates the Rho like GTPase Rac1. Here we have analysed the consequences of overexpression of several mutant Tiam1 proteins in NIH3T3 fibroblasts. Similar to other proteins containing a DH domain, N-terminal truncation of the Tiam1 protein activates its oncogenic potential, establishing Tiam1 as a proto-oncogene. In addition, we show the sequences N-terminal of the catalytic DH domain are required for morphological transformation accompanied by the formation of membrane ruffling, but not for the induction of an oncogenic phenotype. Overexpression of constitutively active Rac1 (V12Rac1) in NIH3T3 cells produces a similar oncogenic phenotype, suggesting that the oncogenic effects of Tiam1 are a consequence of Rac activation.


Asunto(s)
Transformación Celular Neoplásica/genética , Proteínas de Unión al GTP/genética , Proteínas/genética , Células 3T3 , Animales , ADN Complementario , Proteínas de Unión al GTP/metabolismo , Factores de Intercambio de Guanina Nucleótido , Ratones , Ratones Desnudos , Proteínas/metabolismo , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T , Transfección , Proteínas de Unión al GTP rac
14.
Nature ; 375(6529): 338-40, 1995 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-7753201

RESUMEN

Rho-like GTPases have been implicated in the regulation of the actin cytoskeleton which controls the morphology, adhesion and motility of cells. Like Ras proteins, they become activated when bound GDP is exchanged for GTP, a process catalysed by GDP-dissociation stimulator (GDS) proteins. Several GDS proteins specific for Rho-like GTPases have been identified. Most of these contain a conserved catalytic domain, the DBL-homology (DH) domain, and activate Cdc42 or Rho but not Rac. We have isolated the invasion-inducing Tiam1 gene, which also encodes a protein with a DH domain. Here we show that Tiam1 is a GDS protein for Rho-like GTPases in vitro. In fibroblasts, Tiam1 induces a similar phenotype as constitutively activated (V12)Rac1, including membrane ruffling, and this is inhibited by dominant negative (N17)Rac1. Moreover, T-lymphoma cells expressing V12Rac1 become invasive, indicating that the Tiam1-Rac signalling pathway could be operating in the invasion and metastasis of tumour cells.


Asunto(s)
Proteínas de Unión al GTP/fisiología , Invasividad Neoplásica , Proteínas/fisiología , Células 3T3 , Animales , Línea Celular , Membrana Celular/fisiología , Transformación Celular Neoplásica , Fibroblastos/fisiología , GTP Fosfohidrolasas/fisiología , Glutatión Transferasa/metabolismo , Factores de Intercambio de Guanina Nucleótido , Guanosina 5'-O-(3-Tiotrifosfato)/fisiología , Guanosina Difosfato/fisiología , Ratones , Metástasis de la Neoplasia , Proteínas Recombinantes de Fusión , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T , Transfección , Células Tumorales Cultivadas , Proteínas de Unión al GTP rac , Proteínas de Unión al GTP rap , Proteína de Unión al GTP rhoA
15.
Oncogene ; 10(7): 1371-6, 1995 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-7731688

RESUMEN

By means of proviral tagging in combination with in vitro selection for invasive T-lymphoma variants, we have previously identified the murine invasion- and metastasis-inducing Tiam1 gene. Tiam1 encodes a novel protein which shares a Dbl-homology (DH) domain with GDP dissociation stimulator-(GDS) proteins that activate Rho-like GTPases. We have cloned the human TIAM1 coding sequence and studied its evolutionary conservation and expression pattern. TIAM1 is highly conserved among vertebrates. The close similarity between human TIAM1 and the mouse homologue is indicated by 88% and 95% identity of nucleotides and predicted sequences, respectively. The murine gene is highly expressed in brain and testis and at low or moderate levels in almost all other normal tissues. Interestingly, Tiam1 transcripts were found in virtually all analysed tumor cell lines of human and rodent origin including B- and T-lymphomas, neuroblastomas, melanomas and carcinomas. The evolutionary conservation as well as the broad expression pattern of Tiam1 in most normal tissues, suggests a general function in cellular signaling processes presumably by activation of a Rho-like GTPase that regulates the cytoskeletal organization.


Asunto(s)
Proteínas/genética , Secuencia de Aminoácidos , Evolución Biológica , Clonación Molecular , ADN Complementario/genética , Regulación Neoplásica de la Expresión Génica , Genes , Factores de Intercambio de Guanina Nucleótido , Humanos , Datos de Secuencia Molecular , Invasividad Neoplásica , ARN Mensajero/genética , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T , Distribución Tisular
16.
Cytogenet Cell Genet ; 70(1-2): 48-51, 1995.
Artículo en Inglés | MEDLINE | ID: mdl-7736788

RESUMEN

The murine invasion-inducing Tiam1 gene maps to the distal end of chromosome 16, 3.8 cM centromeric of the Ets2 gene. TIAM1, the human homolog of Tiam1, maps to the syntenic region (q22) on human chromosome 21. The gene order in 21q22 is cen-TIAM1-AML1-ERG-ETS2-tel.


Asunto(s)
Cromosomas Humanos Par 21 , Proteínas/genética , Animales , Mapeo Cromosómico , Femenino , Factores de Intercambio de Guanina Nucleótido , Humanos , Hibridación Fluorescente in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T
17.
Cell ; 77(4): 537-49, 1994 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-7999144

RESUMEN

Using proviral tagging in combination with in vitro selection for invasiveness, we have identified a gene, designated Tiam-1, that affects invasion. In the selected invasive T lymphoma variants, proviral insertions were found within coding exons of the Tiam-1 gene, resulting in both truncated 5'-end and 3'-end transcripts that give rise to N- and C-terminal Tiam-1 protein fragments. In one invasive variant, amplification of the Tiam-1 locus was observed with concomitant increase in the amount of normal Tiam-1 protein. Cell clones that were invasive in vitro produced experimental metastases in nude mice, and transfection of truncated Tiam-1 cDNAs into noninvasive cells made these cells invasive. The predicted Tiam-1 protein harbors a Dbl- and Pleckstrin-homologous domain, which it shares with GDP-GTP exchangers for Rho-like proteins that have been implicated in cytoskeletal organization.


Asunto(s)
Linfoma de Células T/genética , Invasividad Neoplásica/genética , Proteínas/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , ADN Complementario/análisis , ADN de Neoplasias/análisis , Proteínas Fúngicas/genética , Proteínas de Unión al GTP/metabolismo , Factores de Intercambio de Guanina Nucleótido , Metástasis Linfática , Linfoma de Células T/microbiología , Linfoma de Células T/patología , Ratones , Ratones Endogámicos C3H , Datos de Secuencia Molecular , Virus de la Leucemia Murina de Moloney/genética , Mutagénesis Insercional , Proteínas Oncogénicas/genética , Proteínas/química , Proteínas Proto-Oncogénicas/genética , Mapeo Restrictivo , Alineación de Secuencia , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T , Células Tumorales Cultivadas , Integración Viral/genética , Proteínas de Unión al GTP rap
18.
Cytogenet Cell Genet ; 60(3-4): 200-5, 1992.
Artículo en Inglés | MEDLINE | ID: mdl-1505215

RESUMEN

By somatic cell fusion studies between noninvasive mouse T-lymphoma cells and invasive human activated normal T-cells we have previously shown that the genetic information responsible for the induction of invasive and metastatic potential in interspecies T-cell hybrids is located on human chromosome 7. Apparently, genes derived from normal activated T-cells are dominantly expressed in the hybrids and control the invasive and, as a consequence, metastatic potential of these T-lymphoma cells. To sublocalize the invasion-inducing locus on chromosome 7 we have generated hybrids that harbor only specific regions of human chromosome 7 with or without a small fragment of human chromosome 21. Analysis of these hybrids revealed that the invasion-inducing locus maps to 7p12----cen. The human DNA complement of the hybrids was confirmed by Southern blot analysis using a large panel of chromosome 7-specific DNA probes. Several of these genes could be further sublocalized. These included: ARAF2 to 7p12----cen, D7S21 to 7pter----p12, ACTB to 7p15----p12, EGFR to 7p12, MDH2 to 7cen----q22, and PDGFA to 7pter----p15.


Asunto(s)
Cromosomas Humanos Par 7 , Invasividad Neoplásica/genética , Southern Blotting , Sondas de ADN , Fluorescencia , Humanos , Células Híbridas , Hibridación de Ácido Nucleico
19.
Clin Exp Metastasis ; 8(6): 567-77, 1990.
Artículo en Inglés | MEDLINE | ID: mdl-1699692

RESUMEN

Non-invasive, non-metastatic mouse BW5147 T-lymphoma cells were treated with non-mutagenic concentrations of the hypomethylating agent 5-azacytidine (5-aza-C). Subsequently, invasive variants were selected on monolayers of rat embryo fibroblasts. The estimated frequency of induction of invasive variants was smaller than 1 in 10(6) cells. We obtained several independent clones that were stable in the expression of the invasive phenotype. In contrast to the parental cell line, the highly invasive clones produced widespread metastases upon tail vein injection in all the syngeneic AKR mice tested, whereas clones with an intermediate level of invasiveness formed metastases only in part of the mice tested. DNA analysis using the methylation-sensitive and insensitive restriction enzymes, Hpa-II and Msp-I, respectively, showed that the DNA of the invasive variants remained hypomethylated, up to 6 months after 5-aza-C treatment. 5-aza-C is thus able to induce invasive and metastatic potential in the BW5147 T-lymphoma cells, similar to the activated human c-Ha-ras oncogene or human chromosome 7, as studied previously. The acquisition of invasive and metastatic potential is presumably caused by DNA hypomethylation and thus activation of one or more silent invasion controlling genes.


Asunto(s)
Azacitidina/farmacología , Linfoma de Células T/patología , Invasividad Neoplásica , Metástasis de la Neoplasia , Animales , ADN/metabolismo , Femenino , Metilación , Ratones , Ratones Endogámicos AKR , Oncogenes
20.
Biochim Biophys Acta ; 950(3): 282-95, 1988 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-3167054

RESUMEN

Insulin-like growth factor II (IGF-II) is a polypeptide of 67 amino acids which is thought to play an important role in fetal growth and development. The human IGF-II gene is situated on chromosome 11, very close to the insulin gene. It extends over 30 kb of chromosomal DNA and consists of five noncoding exons (exons 1-4 and 4B) followed by three protein encoding exons (exons 5-7), one of which (exon 7) contains a long 3'-untranslated region. Here we show that differential initiation of transcription can occur at three distinct promoter sites, resulting in the appearance of mRNA species of different lengths. These promoters show a tissue-specific and a development-specific regulation of expression. Furthermore, we have determined the entire nucleotide sequence of the 3'-terminal exon, exon 7, which is about 4 kb long and contains 3.8 kb of 3'-untranslated sequences. This completes the elucidation of the human IGF-II gene structure. Surprisingly, Northern blot analysis of fetal and adult RNA with a probe derived from the 3'-nontranslated region of exon 7 detects a novel 1.8 kb mRNA which appears to be coordinately expressed with the IGF-II mRNAs. In vitro translation of this 1.8 kb mRNA results in the formation of a translation product of 8.3 kDa, which compares well with the size of a predicted translation product from a 252-nucleotides-long open reading frame.


Asunto(s)
Genes , Factor II del Crecimiento Similar a la Insulina/genética , ARN Mensajero/genética , Somatomedinas/genética , Transcripción Genética , Secuencia de Aminoácidos , Secuencia de Bases , Clonación Molecular , ADN/genética , Exones , Humanos , Datos de Secuencia Molecular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...