Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
J Biol Chem ; 276(36): 33419-27, 2001 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-11445579

RESUMO

In this study, we report on the isolation of a PDZ domain protein, here designated as IIP-1, insulin-like growth factor-1 (IGF-1) receptor-interacting protein-1, which binds to the IGF-1 receptor, but not to the related insulin receptor, and which is involved in the regulation of cell motility. The interaction between the IGF-1 receptor and IIP-1 as well as a splice variant IIP-1/p26 was demonstrated in the yeast two-hybrid system. Using co-precipitation experiments, we confirmed the interaction in transfected cells as well as in vitro. Analysis of deletion mutants indicates that the PDZ domain of IIP-1 mediates interaction with the C-terminal tail of the IGF-1 receptor (serine-threonine-cysteine). This finding demonstrates that the C terminus of the IGF-1 receptor acts as novel PDZ domain binding site. Immunofluorescence analysis revealed an overlapping localization of IIP-1 and the IGF-1 receptor in the breast cancer cell line MCF-7. A functional connection between IIP-1 and the IGF-1 receptor is further supported by the finding that the level of expression of IIP-1 and the IGF-1 receptor strongly correlates in different normal and cancer cells. Furthermore, overexpression of IIP-1 resulted in an attenuation of migration of MCF-7 cells, which is one of the biological activities mediated by the IGF-1 signaling system.


Assuntos
Proteínas de Transporte/química , Receptor IGF Tipo 1/química , Receptor de Insulina/química , Células 3T3 , Processamento Alternativo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Proteínas de Transporte/metabolismo , Divisão Celular , Movimento Celular , Clonagem Molecular , Cisteína/química , DNA Complementar/metabolismo , Deleção de Genes , Biblioteca Gênica , Glutationa Transferase/metabolismo , Humanos , Immunoblotting , Células Jurkat , Camundongos , Microscopia de Fluorescência , Dados de Sequência Molecular , Mutagênese , Fosforilação , Testes de Precipitina , Ligação Proteica , Estrutura Terciária de Proteína , Serina/química , Transdução de Sinais , Treonina/química , Transfecção , Células Tumorais Cultivadas , Técnicas do Sistema de Duplo-Híbrido
2.
J Cell Biol ; 152(2): 325-34, 2001 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-11266449

RESUMO

Male "viable motheaten" (me(v)) mice, with a naturally occurring mutation in the gene of the SH2 domain protein tyrosine phosphatase SHP-1, are sterile. Known defects in sperm maturation in these mice correlate with an impaired differentiation of the epididymis, which has similarities to the phenotype of mice with a targeted inactivation of the Ros receptor tyrosine kinase. Ros and SHP-1 are coexpressed in epididymal epithelium, and elevated phosphorylation of Ros in the epididymis of me(v) mice suggests that Ros signaling is under control of SHP-1 in vivo. Phosphorylated Ros strongly and directly associates with SHP-1 in yeast two-hybrid, glutathione S-transferase pull-down, and coimmunoprecipitation experiments. Strong binding of SHP-1 to Ros is selective compared to six other receptor tyrosine kinases. The interaction is mediated by the SHP-1 NH(2)-terminal SH2 domain and Ros phosphotyrosine 2267. Overexpression of SHP-1 results in Ros dephosphorylation and effectively downregulates Ros-dependent proliferation and transformation. We propose that SHP-1 is an important downstream regulator of Ros signaling.


Assuntos
Células Epiteliais/fisiologia , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases , Receptor trkA/fisiologia , Transdução de Sinais/fisiologia , Células 3T3 , Animais , Linhagem Celular , Epididimo/citologia , Células Epiteliais/citologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Camundongos , Camundongos Knockout , Camundongos Mutantes , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Proteínas Tirosina Fosfatases/química , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Receptor trkA/genética , Proteínas Recombinantes de Fusão/metabolismo , Transfecção , Domínios de Homologia de src
3.
J Biol Chem ; 276(5): 3017-23, 2001 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-11069926

RESUMO

Hepatocyte growth factor (HGF)/scatter factor is a multifunctional cytokine that induces mitogenesis, motility, and morphogenesis in epithelial, endothelial, and neuronal cells. The receptor for HGF/scatter factor was identified as c-Met tyrosine kinase, and activation of the receptor induces multiple signaling cascades. To gain further insight into c-Met-mediated multiple events at a molecular level, we isolated several signaling molecules including a novel binding partner of c-Met, SH2 domain-containing inositol 5-phosphatase 1 (SHIP-1). Western blot analysis revealed that SHIP-1 is expressed in the epithelial cell line, Madin-Darby canine kidney (MDCK) cells. SHIP-1 binds at phosphotyrosine 1356 at the multifunctional docking site. Because a number of signaling molecules such as Grb2, phosphatidylinositol 3-kinase, and Gab1 bind to the multifunctional docking site, we further performed an in vitro competition study using glutathione S-transferase- or His-tagged signaling molecules with c-Met tyrosine kinase. Our binding study revealed that SHIP-1, Grb2, and Gab1 bound preferentially over phosphatidylinositol 3-kinase. Surprisingly, MDCK cells that overexpress SHIP-1 demonstrated branching tubulogenesis within 2 days after HGF treatment, whereas wild-type MDCK cells showed tubulogenesis only after 6 days following treatment without altering cell scattering or cell growth potency. Furthermore, overexpression of a mutant SHIP-1 lacking catalytic activity impaired HGF-mediated branching tubulogenesis.


Assuntos
Fator de Crescimento de Hepatócito/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Animais , Sítios de Ligação , Divisão Celular/fisiologia , Células Cultivadas , Cães , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases , Fosforilação , Proteínas Proto-Oncogênicas c-kit/metabolismo , Receptor de Insulina/metabolismo , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Receptor trkA/metabolismo , Domínios de Homologia de src
4.
FEBS Lett ; 467(1): 91-6, 2000 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-10664463

RESUMO

Grb7 is a member of a family of molecular adapters which are able to contribute positively but also negatively to signal transduction and whose precise roles remain obscure. Rnd1 is a member of the Rho family, but, as opposed to usual GTPases, it is constitutively bound to GTP. We show here that Rnd1 and Grb7 interact, in two-hybrid assays, in vitro, and in pull-down experiments performed with SK-BR3, a breast cancer cell line that overexpresses Grb7. This interaction involves switch II loop of Rnd1, a region crucial for guanine nucleotide exchange in all GTPases, and a Grb7 SH2 domain, a region crucial for Grb7 interaction with several activated receptors. The contribution of the interaction between Rnd1 and Grb7 to their respective functions and properties is discussed.


Assuntos
Proteínas/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Anticorpos/imunologia , Sítios de Ligação , Escherichia coli/genética , Proteína Adaptadora GRB7 , Células HeLa , Humanos , Peso Molecular , Fosforilação , Fosfotirosina/imunologia , Fosfotirosina/metabolismo , Testes de Precipitina , Ligação Proteica , Proteínas/química , Proteínas/genética , Proteínas/isolamento & purificação , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Deleção de Sequência/genética , Homologia de Sequência de Aminoácidos , Transfecção , Células Tumorais Cultivadas , Técnicas do Sistema de Duplo-Híbrido , Proteínas rho de Ligação ao GTP/química , Proteínas rho de Ligação ao GTP/genética , Proteínas rho de Ligação ao GTP/isolamento & purificação , Domínios de Homologia de src
5.
Oncogene ; 18(47): 6488-95, 1999 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-10597251

RESUMO

Hematopoietic cell growth, differentiation, and commitment to a restricted lineage are guided by a set of cytokines acting exclusively on cells expressing the corresponding cytokine receptor. The macrophage colony stimulating factor (M-CSF, also termed CSF-1) and its cognate receptor, the tyrosine kinase c-Fms, are essential for monocyte and macrophage development. The underlying molecular mechanism, however, is poorly understood. Here we identified a novel Fms-interacting protein (FMIP, MW 78 kDa) which binds transiently via its N-terminal 144 residues to the cytoplasmic domain of activated Fms-molecules. Binding of FMIP was paralleled by rapid tyrosine phosphorylation within the binding domain which drastically reduced its ability to associate with Fms. Binding was specific as evidenced by co-immunoprecipitation and association with recombinant GST-Fms fusion proteins. No binding was observed with the tyrosine phosphorylated cytoplasmic domains of c-Kit, TrkA, c-Met, and the insulin receptor. The role of FMIP in hematopoietic differentiation was studied in the bipotential myeloid progenitor cell line, FDC-P1Mac11. Overexpression of FMIP prevented M-CSF induced macrophage differentiation. Instead, cells differentiated into granulocytes. Our data suggest that the level of FMIP expression could form a threshold that decides about differentiation either into macrophages or into granulocytes.


Assuntos
Proteínas de Transporte/metabolismo , Diferenciação Celular , Granulócitos/citologia , Peptídeos e Proteínas de Sinalização Intracelular , Macrófagos/citologia , Sequência de Aminoácidos , Proteínas de Transporte/química , Proteínas de Transporte/genética , Linhagem Celular , Fator Estimulador de Colônias de Macrófagos/fisiologia , Dados de Sequência Molecular , Fosforilação , Ligação Proteica , Receptores Proteína Tirosina Quinases/metabolismo , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Especificidade por Substrato
6.
Mol Biol Cell ; 10(3): 551-65, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10069803

RESUMO

The scatter factor/hepatocyte growth factor regulates scattering and morphogenesis of epithelial cells through activation of the MET tyrosine kinase receptor. In particular, the noncatalytic C-terminal tail of MET contains two autophosphorylation tyrosine residues, which form a multisubstrate-binding site for several cytoplasmic effectors and are thought to be essential for signal transduction. We show here that a MET receptor mutated on the four C-terminal tyrosine residues, Y1311F, Y1347F, Y1354F, and Y1363F, can induce efficiently a transcriptional response and cell scattering, whereas it cannot induce cell morphogenesis. Although the mutated receptor had lost its ability to recruit and/or activate known signaling molecules, such as GRB2, SHC, GAB1, and PI3K, by using a sensitive association-kinase assay we found that the mutated receptor can still associate and phosphorylate a approximately 250-kDa protein. By further examining signal transduction mediated by the mutated MET receptor, we established that it can transmit efficient RAS signaling and that cell scattering by the mutated MET receptor could be inhibited by a pharmacological inhibitor of the MEK-ERK (MAP kinase kinase-extracellular signal-regulated kinase) pathway. We propose that signal transduction by autophosphorylation of the C-terminal tyrosine residues is not the sole mechanism by which the activated MET receptor can transmit RAS signaling and cell scattering.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas Adaptadoras de Transporte Vesicular , Movimento Celular/fisiologia , Proteínas Proto-Oncogênicas c-met/genética , Proteínas Proto-Oncogênicas c-met/metabolismo , Transdução de Sinais , Proteínas ras/metabolismo , Animais , Sítios de Ligação , Linhagem Celular , Cães , Proteína Adaptadora GRB10 , Fator de Crescimento de Hepatócito/metabolismo , Fator de Crescimento de Hepatócito/farmacologia , Mutação , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/metabolismo , Fosforilação , Proteínas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-ets , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Adaptadoras da Sinalização Shc , Fator de Transcrição AP-1/metabolismo , Fatores de Transcrição/metabolismo , Ativação Transcricional , Transfecção , Tirosina/metabolismo , Proteínas ras/genética
7.
Clin Exp Metastasis ; 17(8): 641-8, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10919708

RESUMO

Interactions of tumor cells with the endothelium and tissue stroma are considered to be critical steps in metastasis formation and progression of cancer. To identify cellular receptors that mediate the binding of tumor cells to endothelium, a murine T cell lymphoma-derived expression library was screened for adhesion-inducing cDNA clones. We identified a novel cell adhesion-promoting molecule, termed ARM-1 (adhesion regulating molecule-1), which is homologous to a human Mr 110,000 tumor-associated antigen. The ARM-1 cDNA codes for a type I transmembrane protein of 407 amino acids with potential O- and N-glycosylation sites that does not belong to any of the known families of cell adhesion molecules. Overexpression of ARM-1 in 293T human embryonic kidney cells significantly increased adhesion to different endothelial cells. ARM-1 expression in 293T cells did not alter integrin expression or beta1-integrin-mediated cell adhesion. Northern blot analysis of human breast cancer cell lines revealed 3- to 5-fold elevated ARM-1 mRNA levels in metastatic as compared to non-metastatic cells. In conclusion, we have identified ARM-1 as a novel cell adhesion-promoting receptor that is upregulated in metastatic cancer cells.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Moléculas de Adesão Celular/biossíntese , Moléculas de Adesão Celular/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Neoplasias da Mama/metabolismo , Adesão Celular/fisiologia , Moléculas de Adesão Celular/metabolismo , Linhagem Celular , Clonagem Molecular , DNA de Neoplasias/genética , Endotélio/metabolismo , Humanos , Integrinas/biossíntese , Integrinas/genética , Integrinas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Rim/citologia , Linfoma de Células T/genética , Linfoma de Células T/metabolismo , Linfoma de Células T/patologia , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Metástase Neoplásica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Homologia de Sequência do Ácido Nucleico , Transfecção , Células Tumorais Cultivadas , Regulação para Cima
8.
Oncogene ; 17(8): 941-8, 1998 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-9747873

RESUMO

Bcr-Abl is an oncogenic tyrosine kinase expressed in tumor cells of CML and a subset of ALL which in its unregulated and activated state is thought to cause cell transformation and leukemia. Bcr-Abl contains several autophosphorylation sites which serve as potential docking sites for SH2-containing signaling molecules. Mutational analysis has indicated that these autophosphorylation sites play a critical role in the transforming capability of Bcr-Abl. It has been shown that the SH2-containing adapter protein Grb2 binds to the autophosphorylation site Tyr(p)177 whereby it couples Bcr-Abl to the Ras pathway. The biological consequences of this interaction, however, are presently unclear. A Tyr177-mutated Bcr-Abl which lacks the ability to interact with the Grb2-SH2 domain still transforms myeloid cells and generates tumors in nude mice. We performed a yeast two-hybrid screen to identify signaling proteins which bind to distinct Bcr-Abl autophosphorylation sites. Autophosphorylation of Bcr-Abl in yeast was accomplished by using the DNA binding protein LexA which permits dimerization and crossphosphorylation of the fused bait. Using a LexA-Bcr-Abl full length fusion protein as bait, we identified several SH2-containing proteins. Among them we confirmed molecules already shown by others to interact with Bcr-Abl, in vivo, including Grb2, PI-3-kinase and Crk indicating that dimerization in yeast leads to autophosphorylation of tyrosine residues crucial for Bcr-Abl signaling in vivo. More importantly, we identified the SH2-containing protein Grb10 as a new binding partner for Bcr-Abl. This binding occurs in a phosphotyrosine-dependent manner at Bcr sites of Bcr-Abl. Both Abl and Bcr alone, as well as a kinase-defective Bcr-Abl, failed to interact with Grb10 in yeast. Mutational analysis uncovered a new SH2 binding site in Bcr-Abl located between Bcr aa242-446, which is different from the Grb2 binding site. Binding could be demonstrated in vitro and also in vivo as shown by co-immunoprecipitation analysis in CML cells. Using a temperature sensitive Bcr-Abl stably overexpressed in hematopoetic cells, we demonstrated that complex formation of Grb10 with Bcr-Abl was kinase activation-dependent in vivo. Notably, a Bcr-Abl mutant protein (Bcr/1-242-Abl) which lacks the ability to interact with Grb10 partially alleviated IL-3 dependence of Ba/F3 cells, indicating that the Grb10/Bcr-Abl interaction is important for Bcr-Abl-induced IL-3 independence of Ba/F3 cells. In addition, the Bcr/1-242-Abl mutant has a reduced capacity to induce focus formation in fibroblasts.


Assuntos
Proteínas de Fusão bcr-abl/metabolismo , Proteínas/metabolismo , Domínios de Homologia de src , Absorção , Linhagem Celular , Transformação Celular Neoplásica/genética , Ativação Enzimática , Receptores ErbB/metabolismo , Proteínas de Fusão bcr-abl/genética , Proteína Adaptadora GRB10 , Vetores Genéticos/metabolismo , Humanos , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Mutação , Ligação Proteica , Proteínas Tirosina Quinases/metabolismo , Saccharomyces cerevisiae/genética , Transfecção , Células Tumorais Cultivadas
9.
Mol Cell Biol ; 17(2): 799-808, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9001234

RESUMO

The Met tyrosine kinase receptor is a widely expressed molecule which mediates pleiotropic cellular responses following activation by its ligand, hepatocyte growth factor/scatter factor (HGF/SF). In this communication we demonstrate that significant Met degradation is induced by HGF/SF and that this degradation can be blocked by lactacystin, an inhibitor of proteasome activity. We also show that Met is rapidly polyubiquitinated in response to ligand and that polyubiquitinated Met molecules, which are normally unstable, are stabilized by lactacystin. Both HGF/SF-induced degradation and polyubiquitination of Met were shown to be dependent on the receptor possessing intact tyrosine kinase activity. Finally, we found that a normally highly labile 55-kDa fragment of the Met receptor is stabilized by lactacystin and demonstrate that it represents a cell-associated remnant that is generated following the ligand-independent proteolytic cleavage of the Met receptor in its extracellular domain. This truncated Met molecule encompasses the kinase domain of the receptor and is itself tyrosine phosphorylated. We conclude that the ubiquitin-proteasome pathway plays a significant role in the degradation of the Met tyrosine kinase receptor as directed by ligand-dependent and -independent signals. We propose that this proteolytic pathway may be important for averting cellular transformation by desensitizing Met signaling following ligand stimulation and by eliminating potentially oncogenic fragments generated via extracellular cleavage of the Met receptor.


Assuntos
Cisteína Endopeptidases/metabolismo , Fator de Crescimento de Hepatócito/farmacologia , Complexos Multienzimáticos/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Ubiquitinas/metabolismo , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacologia , Animais , Brefeldina A , Linhagem Celular , Membrana Celular/metabolismo , Ciclopentanos/farmacologia , Inibidores de Cisteína Proteinase/farmacologia , Humanos , Ligantes , Peso Molecular , Fatores de Crescimento Neural/farmacologia , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Fosforilação , Complexo de Endopeptidases do Proteassoma , Inibidores da Síntese de Proteínas/farmacologia , Proteínas Proto-Oncogênicas c-met , Receptores Proteína Tirosina Quinases/genética , Proteínas Recombinantes de Fusão , Acetato de Tetradecanoilforbol/farmacologia , Transfecção
10.
Nature ; 384(6605): 173-6, 1996 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-8906793

RESUMO

The proteins Gab1 and the related DOS (for 'daughter of sevenless') each bind to substrates of tyrosine kinases like Grb2 or Corkscrew, and act in signalling pathways downstream of tyrosine kinase receptors. Here we show that Gab1 interacts directly with the c-met-encoded receptor tyrosine kinase but not with a number of other tyrosine kinases from different subfamilies. A newly identified proline-rich domain of Gab1 is responsible for the binding of this protein to the tyrosine-phosphorylated bidentate docking site in c-Met. Expression of Gab1 in epithelial cells is sufficient to induce the c-Met-specific activities, including branching morphogenesis. Thus we have discovered a new phosphotyrosine interaction domain in Gab1 and shown that Gab1 is the substrate of the c-Met receptor tyrosine kinase that mediates epithelial morphogenesis.


Assuntos
Morfogênese/fisiologia , Fosfoproteínas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Sequência de Aminoácidos , Animais , Sítios de Ligação , Linhagem Celular , Epitélio/embriologia , Humanos , Camundongos , Dados de Sequência Molecular , Fosfoproteínas/genética , Testes de Precipitina , Prolina/metabolismo , Ligação Proteica , Proteínas Proto-Oncogênicas c-met , Receptores Proteína Tirosina Quinases/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Saccharomyces cerevisiae , Transdução de Sinais , Especificidade por Substrato
11.
J Cell Biol ; 133(5): 1095-1107, 1996 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8655582

RESUMO

Receptor tyrosine kinases play essential roles in morphogenesis and differentiation of epithelia. Here we examined various tyrosine kinase receptors, which are preferentially expressed in epithelia (c-met, c-ros, c-neu, and the keratin growth factor [KGF] receptor), for their capacity to induce cell motility and branching morphogenesis of epithelial cells. We exchanged the ligand-binding domain of these receptors by the ectodomain of trkA and could thus control signaling by the new ligand, NGF. We demonstrate here that the tyrosine kinases of c-met, c-ros, c-neu, the KGF receptor, and trkA, but not the insulin receptor, induced scattering and increased motility of kidney epithelial cells in tissue culture. Mutational analysis suggests that SHC binding is essential for scattering and increased cell motility induced by trkA. The induction of motility in epithelial cells is thus an important feature of various receptor tyrosine kinases, which in vivo play a role in embryogenesis and metastasis. In contrast, only the c-met receptor promoted branching morphogenesis of kidney epithelial cells in three-dimensional matrices, which resemble the formation of tubular epithelia in development. Interestingly, the ability of c-met to induce morphogenesis could be transferred to trkA, when in a novel receptor hybrid COOH-terminal sequences of c-met (including Y14 to Y16) were fused to the trkA kinase domain. These data demonstrate that tubulogenesis of epithelia is a restricted activity of tyrosine kinases, as yet only demonstrated for the c-met receptor. We predict the existence of specific substrates that mediate this morphogenesis signal.


Assuntos
Movimento Celular/fisiologia , Morfogênese/fisiologia , Receptores Proteína Tirosina Quinases/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Primers do DNA/genética , DNA Recombinante/genética , Cães , Células Epiteliais , Epitélio/efeitos dos fármacos , Epitélio/enzimologia , Humanos , Dados de Sequência Molecular , Estrutura Molecular , Morfogênese/efeitos dos fármacos , Fatores de Crescimento Neural/farmacologia , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-met , Receptores Proteína Tirosina Quinases/química , Receptores Proteína Tirosina Quinases/genética , Receptor trkA , Receptores de Fator de Crescimento Neural/química , Receptores de Fator de Crescimento Neural/genética , Receptores de Fator de Crescimento Neural/fisiologia , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transfecção
12.
J Biol Chem ; 271(8): 3959-62, 1996 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-8626723

RESUMO

Stimulation of the insulin receptor (IR) results in tyrosine phosphorylation of the intermediate molecules insulin receptor substrate-1 (IRS-1), IRS-2, and Shc, which then couple the IR to downstream signaling pathways by serving as binding sites for signaling molecules with SH2 domains. It has been proposed that direct binding of IRS-1, IRS-2, and Shc to an NPX-Tyr(P) motif in the juxtamembrane region of the IR is required for tyrosine phosphorylation of these molecules by the IR. In this regard, Shc and IRS-1 contain domains that are distinct from SH2 domains, referred to as the phosphotyrosine binding (PTB) or phosphotyrosine interaction (PI) domains, which bind phosphotyrosine in the context of an NPX-Tyr(P) motif. To further clarify the role of the Shc PTB/PI domain, we identified a mutation in this domain that abrogated binding of Shc to the IR in vitro. Interestingly, this mutation completely abolished Shc phosphorylation by the IR in vivo whereas mutation of the arginine in the FLVRES motif of the Shc SH2 domain did not affect Shc phosphorylation by insulin. In addition, we identified specific amino acids on the IR that are required for the IR to stimulate Shc but not IRS-1 phosphorylation in vivo. As with the PTB/PI domain Shc mutant, the ability of these mutant receptors to phosphorylate Shc correlates with the binding of the PTB/PI domain of Shc to similar sequences in vitro. These findings support a model in which binding of the PTB/PI domain of Shc directly to the NPX-Tyr(P) motif on the IR mediates Shc phosphorylation by insulin.


Assuntos
Fosfoproteínas/metabolismo , Fosfotirosina/metabolismo , Receptor trkA/imunologia , Domínios de Homologia de src , Células 3T3 , Sequência de Aminoácidos , Animais , Membrana Celular/metabolismo , Clonagem Molecular , Humanos , Proteínas Substratos do Receptor de Insulina , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação Puntual , Receptor trkA/biossíntese , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais
14.
J Cell Biol ; 131(6 Pt 1): 1573-86, 1995 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8522613

RESUMO

Hepatocyte growth factor/scatter factor (HGF/SF) is the mesenchymal ligand of the epithelial tyrosine kinase receptor c-Met. In vitro, HGF/SF has morphogenic properties, e.g., induces kidney epithelial cells to form branching ducts in collagen gels. Mutation of the HGF/SF gene in mice results in embryonic lethality due to severe liver and placenta defects. Here, we have evaluated the morphogenic activity of HGF/SF with a large variety of epithelial cells grown in three-dimensional collagen matrices. We found that HGF/SF induces SW 1222 colon carcinoma cells to form crypt-like structures. In these organoids, cells exhibit apical/basolateral polarity and build a well-developed brush border towards the lumen. Capan 2 pancreas carcinoma cells, upon addition of HGF/SF, develop large hollow spheroids lined with a tight layer of polarized cells. Collagen inside the cysts is digested and the cells show features of pancreatic ducts. HGF/SF induces EpH4 mammary epithelial cells to form long branches with end-buds that resemble developing mammary ducts. pRNS-1-1 prostate epithelial cells in the presence of HGF/SF develop long ducts with distal branching as found in the prostate. Finally, HGF/SF simulates alveolar differentiation in LX-1 lung carcinoma cells. Expression of transfected HGF/SF cDNA in LX-1 lung carcinoma and EpH4 mammary epithelial cells induce morphogenesis in an autocrine manner. In the cell lines tested, HGF/SF activated the Met receptor by phosphorylation of tyrosine residues. These data show that HGF/SF induces intrinsic, tissue-specific morphogenic activities in a wide variety of epithelial cells. Apparently, HGF/SF triggers respective endogenous programs and is thus an inductive, not an instructive, mesenchymal effector for epithelial morphogenesis.


Assuntos
Fator de Crescimento de Hepatócito/fisiologia , Fígado/citologia , Fígado/crescimento & desenvolvimento , Adenocarcinoma , Animais , Carcinoma , Tamanho Celular/fisiologia , Neoplasias do Colo , Cães , Desenvolvimento Embrionário e Fetal/fisiologia , Células Epiteliais , Epitélio/fisiologia , Epitélio/ultraestrutura , Humanos , Rim/citologia , Pulmão/citologia , Masculino , Camundongos , Microscopia Eletrônica , Morfogênese/fisiologia , Pâncreas/citologia , Fosforilação , Próstata/citologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-met , Receptores Proteína Tirosina Quinases/metabolismo , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/fisiologia , Células Tumorais Cultivadas/ultraestrutura
15.
J Cell Biol ; 131(1): 215-26, 1995 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-7559778

RESUMO

We have examined the role of two mesenchymal ligands of epithelial tyrosine kinase receptors in mouse mammary gland morphogenesis. In organ cultures of mammary glands, hepatocyte growth factor (HGF, scatter factor) promoted branching of the ductal trees but inhibited the production of secretory proteins. Neuregulin (NRG, neu differentiation factor) stimulated lobulo-alveolar budding and the production of milk proteins. These functional effects are paralleled by the expression of the two factors in vivo: HGF is produced in mesenchymal cells during ductal branching in the virgin animal; NRG is expressed in the mesenchyme during lobulo-alveolar development at pregnancy. The receptors of HGF and NRG (c-met, c-erbB3, and c-erbB4), which are expressed in the epithelial cells, are not regulated. In organ culture, branching morphogenesis and lobulo-alveolar differentiation of the mammary gland could be abolished by blocking expression of endogenous HGF and NRG by the respective antisense oligonucleotides; in antisense oligonucleotide-treated glands, morphogenesis could again be induced by the addition of recombinant HGF and NRG. We thus show that two major postnatal morphogenic periods of mammary gland development are dependent on sequential mesenchymal-epithelial interactions mediated by HGF and NRG.


Assuntos
Glicoproteínas/fisiologia , Fator de Crescimento de Hepatócito/fisiologia , Glândulas Mamárias Animais/citologia , Animais , Sequência de Bases , Diferenciação Celular/fisiologia , Células Epiteliais , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Hibridização In Situ , Glândulas Mamárias Animais/embriologia , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Morfogênese/fisiologia , Neurregulinas , Oligonucleotídeos Antissenso/farmacologia , Técnicas de Cultura de Órgãos
16.
Proc Natl Acad Sci U S A ; 92(7): 2597-601, 1995 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-7708691

RESUMO

Signals transduced by the met tyrosine kinase, which is the receptor for scatter factor/hepatocyte growth factor, are of major importance for the regulation of epithelial cell motility, morphogenesis, and proliferation. We report here that different sets of tyrosine residues in the cytoplasmic domain of the met receptor affect signal transduction in epithelial cells in a positive or negative fashion: mutation of the C-terminal tyrosine residues 13-16 (Y1311, Y1347, Y1354, and Y1363) reduced or abolished ligand-induced cell motility and branching morphogenesis. In contrast, mutation of the juxtamembrane tyrosine residue 2 (Y1001) produced constitutively mobile, fibroblastoid cells. Furthermore, the gain-of-function mutation of tyrosine residue 2 suppressed the loss-of-function mutations of tyrosine residue 15 or 16. The opposite roles of the juxtamembrane and C-terminal tyrosine residues may explain the suggested dual function of the met receptor in both epithelial-mesenchymal interactions and tumor progression.


Assuntos
Mutação Puntual , Receptores Proteína Tirosina Quinases/metabolismo , Tirosina , Sequência de Aminoácidos , Animais , Western Blotting , Divisão Celular , Linhagem Celular , Membrana Celular/metabolismo , Movimento Celular , Análise Mutacional de DNA , Cães , Epitélio/metabolismo , Fator de Crescimento de Hepatócito/farmacologia , Rim , Morfogênese , Mutagênese Sítio-Dirigida , Fenótipo , Fosforilação , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-met , Receptores Proteína Tirosina Quinases/biossíntese , Receptores Proteína Tirosina Quinases/isolamento & purificação , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo
17.
J Biol Chem ; 269(35): 21936-9, 1994 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-8071312

RESUMO

Scatter factor/hepatocyte growth factor (SF/HGF) has various biological effects upon different cells, i.e. induces increased motility and proliferation as well as invasiveness and morphogenesis. The signals given to epithelial cells by SF/HGF are all mediated through the Met receptor tyrosine kinase (Weidner, K. M., Sachs, M., and Birchmeier, W. (1993) J. Cell Biol. 111, 145-154) suggesting that signal diversity is due to the interplay of different downstream pathways. It has also been shown that SF/HGF activates the protooncogene product Ras, i.e. stimulates guanine nucleotide exchange. In order to examine whether Ras is involved in mediating the dissociation and motility signal of SF/HGF to epithelial cells, we have expressed in Madin-Darby canine kidney cells the dominant-negative N17Ras under the control of a modified metallothionein promoter. Induced expression of N17Ras by the addition of Zn2+ clearly prevented dissociation of the cells by SF/HGF. These data indicate that the Ras pathway is indeed essential to mediate the motility signal of SF/HGF-Met to the cell-cell adhesion system and the cytoskeleton of epithelial cells.


Assuntos
Fator de Crescimento de Hepatócito/metabolismo , Proteína Oncogênica p21(ras)/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais , Animais , Movimento Celular , Células Cultivadas , Cães , Camundongos , Proteínas Proto-Oncogênicas c-met
18.
J Cell Biol ; 123(1): 223-35, 1993 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8408200

RESUMO

Scatter factor/hepatocyte growth factor (SF/HGF) has potent motogenic, mitogenic, and morphogenetic activities on epithelial cells in vitro. The cell surface receptor for this factor was recently identified: it is the product of the c-met protooncogene, a receptor-type tyrosine kinase. We report here the novel and distinct expression patterns of SF/HGF and its receptor during mouse development, which was determined by a combination of in situ hybridization and RNase protection experiments. Predominantly, we detect transcripts of c-met in epithelial cells of various developing organs, whereas the ligand is expressed in distinct mesenchymal cells in close vicinity. In addition, transient SF/HGF and c-met expression is found at certain sites of muscle formation; transient expression of the c-met gene is also detected in developing motoneurons. SF/HGF and the c-met receptor might thus play multiple developmental roles, most notably, mediate a signal given by mesenchyme and received by epithelial. Mesenchymal signals are known to govern differentiation and morphogenesis of many epithelia, but the molecular nature of the signals has remained poorly understood. Therefore, the known biological activities of SF/HGF in vitro and the embryonal expression pattern reported here indicate that this mesenchymal factor can transmit morphogenetic signals in epithelial development and suggest a molecular mechanism for mesenchymal epithelial interactions.


Assuntos
Comunicação Celular , Epitélio/embriologia , Fator de Crescimento de Hepatócito/metabolismo , Mesoderma/fisiologia , Camundongos Endogâmicos/embriologia , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Animais , Fator de Crescimento de Hepatócito/genética , Fator de Crescimento de Hepatócito/isolamento & purificação , Hibridização In Situ , Rim/embriologia , Rim/ultraestrutura , Pulmão/embriologia , Pulmão/ultraestrutura , Camundongos , Músculos/embriologia , Músculos/ultraestrutura , Tecido Nervoso/embriologia , Neurônios , Pâncreas/embriologia , Pâncreas/ultraestrutura , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/isolamento & purificação , Proteínas Proto-Oncogênicas c-met , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/isolamento & purificação , Glândulas Salivares/embriologia , Glândulas Salivares/ultraestrutura , Distribuição Tecidual
19.
Semin Cancer Biol ; 4(4): 231-9, 1993 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8400145

RESUMO

It has been realized for some time that the loss of epithelial differentiation in carcinomas, which is accompanied by higher mobility and invasiveness of the tumor cells, is a consequence of reduced intercellular adhesion. A variety of recent reports have indicated that the primary cause for the 'scattering' of the cells in invasive carcinomas is a loss of the integrity of intercellular adherens junctions often involving loss of a functional cell-cell adhesion molecule E-cadherin. In the present review, permanent and transient molecular mechanisms are discussed which lead to the impairment of junctional integrity of cells and thus the progression of carcinomas toward a more metastatic state.


Assuntos
Caderinas/fisiologia , Junções Intercelulares/fisiologia , Neoplasias/patologia , Animais , Sequência de Bases , Caderinas/genética , Adesão Celular/fisiologia , Movimento Celular/fisiologia , Galinhas , DNA de Neoplasias/química , Regulação Neoplásica da Expressão Gênica , Fator de Crescimento de Hepatócito/química , Fator de Crescimento de Hepatócito/fisiologia , Humanos , Camundongos , Dados de Sequência Molecular , Metástase Neoplásica/patologia , Regiões Promotoras Genéticas , Ratos , Células Tumorais Cultivadas
20.
Mol Immunol ; 30(11): 1003-11, 1993 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8350870

RESUMO

Armenian hamsters were immunized with non-liganded, partially liganded or affinity-labeled anti-fluorescein Mab 4-4-20. Seventeen hybridoma producing monoclonal anti-4-4-20 antibodies were characterized from chemically-mediated fusions of immune hamster lymphocytes with murine Sp2/O-Ag14 myeloma cells. Distinct populations of anti-4-4-20 monoclonal antibodies were isolated from hamsters receiving immunizations with partially liganded Mab 4-4-20 relative to those receiving affinity-labeled 4-4-20. Two of the three monoclonal antibodies produced in response to partially liganded 4-4-20 were inhibited in their interaction with 4-4-20 by fluorescyl ligand. These two clones, 1F4 and 1B7, recognized unique epitopes on the 4-4-20 molecules, as demonstrated by non-reactivity with members of the 4-4-20 idiotype family. Additionally, 1F4 and 1B7 demonstrated the ability to delay the association of fluorescein with Mab 4-4-20. The 14 characterized non-ligand-inhibitable Mabs elicited to affinity-labeled 4-4-20 were classified into four separate groups based on various binding properties with members of the 4-4-20 idiotype family and binding to resolved H- and L-chains in a western blot. Members of three of the four groups showed strong reactivity with both 04-01 Ig and 04-01 SCA, which utilizes the same L-chain as Mab 4-4-20. Six non-ligand-inhibitable Mabs, 4A6, P1E11, 3A5-1, 2C3, 2C4, and 1A4, delayed the dissociation rate of ligand from Mab 4-4-20 and mutant 4-4-20 SCA L32phe.


Assuntos
Anticorpos Anti-Idiotípicos/imunologia , Anticorpos Monoclonais/imunologia , Fluoresceína-5-Isotiocianato , Região Variável de Imunoglobulina/imunologia , Animais , Ligação Competitiva , Western Blotting , Cricetinae , Cricetulus , Imunização
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...