Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Nat Cell Biol ; 3(9): 785-92, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11533657

RESUMEN

Both ErbB1 and ErbB2 are overexpressed or amplified in breast tumours. To examine the effects of activating ErbB receptors in a context that mimics polarized epithelial cells in vivo, we activated ErbB1 and ErbB2 homodimers in preformed, growth-arrested mammary acini cultured in three-dimensional basement membrane gels. Activation of ErbB2, but not that of ErbB1, led to a reinitiation of cell proliferation and altered the properties of mammary acinar structures. These altered structures share several properties with early-stage tumours, including a loss of proliferative suppression, an absence of lumen, retention of the basement membrane and a lack of invasive properties. ErbB2 activation also disrupted tight junctions and the cell polarity of polarized epithelia, whereas ErbB1 activation did not have any effect. Our results indicate that ErbB receptors differ in their ability to induce early stages of mammary carcinogenesis in vitro and this three-dimensional model system can reveal biological activities of oncogenes that cannot be examined in vitro in standard transformation assays.


Asunto(s)
Células Epiteliales/citología , Receptores ErbB/metabolismo , Receptor ErbB-2/metabolismo , Animales , Mama , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , División Celular/efectos de los fármacos , División Celular/fisiología , Línea Celular , Polaridad Celular , Dimerización , Perros , Factor de Crecimiento Epidérmico/farmacología , Células Epiteliales/fisiología , Células Epiteliales/ultraestructura , Femenino , Humanos , Riñón , Proteínas Recombinantes de Fusión/metabolismo
2.
Mol Cell Biol ; 19(10): 6845-57, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10490623

RESUMEN

The four members of the ErbB family of receptor tyrosine kinases are involved in a complex array of combinatorial interactions involving homo- and heterodimers. Since most cell types express more than one member of the ErbB family, it is difficult to distinguish the biological activities of different homo- and heterodimers. Here we describe a method for inducing homo- or heterodimerization of ErbB receptors by using synthetic ligands without interference from the endogenous receptors. ErbB receptor chimeras containing synthetic ligand binding domains (FK506-binding protein [FKBP] or FKBP-rapamycin-binding domain [FRB]) were homodimerized with the bivalent FKBP ligand AP1510 and heterodimerized with the bifunctional FKBP-FRB ligand rapamycin. AP1510 treatment induced tyrosine phosphorylation of ErbB1 and ErbB2 homodimers and recruitment of Src homology 2 domain-containing proteins (Shc and Grb2). In addition, ErbB1 and ErbB2 homodimers activated downstream signaling pathways leading to Erk2 and Akt phosphorylation. However, only ErbB1 homodimers were internalized upon AP1510 stimulation, and only ErbB1 homodimers were able to associate with and induce phosphorylation of c-Cbl. Cells expressing AP1510-induced ErbB1 homodimers were able to associate with and induce phosphorylation of c-Cbl. Cells expressing AP1510-induced ErbB1 homodimers were able to form foci; however, cells expressing ErbB2 homodimers displayed a five- to sevenfold higher focus-forming ability. Using rapamycin-inducible heterodimerization we show that c-Cbl is unable to associate with ErbB1 in a ErbB1-ErbB2 heterodimer most likely because ErbB2 is unable to phosphorylate the c-Cbl binding site on ErbB1. Thus, we demonstrate that ErbB1 and ErbB2 homodimers differ in their abilities to transform fibroblasts and provide evidence for differential signaling by ErbB homodimers and heterodimers. These observations also validate the use of synthetic ligands to study the signaling and biological specificity of selected ErbB dimers in any cell type.


Asunto(s)
Receptores ErbB/metabolismo , Receptor ErbB-2/metabolismo , Ubiquitina-Proteína Ligasas , Animales , Ciclo Celular , Transformación Celular Neoplásica , Dimerización , Relación Dosis-Respuesta a Droga , Endocitosis , Factor de Crecimiento Epidérmico/metabolismo , Ligandos , Fosforilación , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-cbl , Ratas , Proteínas Recombinantes de Fusión , Transducción de Señal , Tacrolimus/análogos & derivados , Tacrolimus/metabolismo
3.
Mol Cell Biol ; 14(1): 735-43, 1994 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-7903421

RESUMEN

Amplification and overexpression of the neu (c-erbB2) proto-oncogene has been implicated in the pathogenesis of 20 to 30% of human breast cancers. Although the activation of Neu receptor tyrosine kinase appears to be a pivotal step during mammary tumorigenesis, the mechanism by which Neu signals cell proliferation is unclear. Molecules bearing a domain shared by the c-Src proto-oncogene (Src homology 2) are thought to be involved in signal transduction from activated receptor tyrosine kinases such as Neu. To test whether c-Src was implicated in Neu-mediated signal transduction, we measured the activity of the c-Src tyrosine kinase in tissue extracts from either mammary tumors or adjacent mammary epithelium derived from transgenic mice expressing a mouse mammary tumor virus promoter/enhancer/unactivated neu fusion gene. The Neu-induced mammary tumors possessed six- to eightfold-higher c-Src kinase activity than the adjacent epithelium. The increase in c-Src tyrosine kinase activity was not due to an increase in the levels of c-Src but rather was a result of the elevation of its specific activity. Moreover, activation of c-Src was correlated with its ability to complex tyrosine-phosphorylated Neu both in vitro and in vivo. Together, these observations suggest that activation of the c-Src tyrosine kinase during mammary tumorigenesis may occur through a direct interaction with activated Neu.


Asunto(s)
Neoplasias Mamarias Experimentales/enzimología , Neoplasias Mamarias Experimentales/genética , Proteínas Tirosina Quinasas/genética , Proto-Oncogenes , Animales , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/etiología , Neoplasias de la Mama/genética , Proteína Tirosina Quinasa CSK , Activación Enzimática/genética , Receptores ErbB/genética , Femenino , Expresión Génica , Genes myc , Humanos , Neoplasias Mamarias Experimentales/etiología , Virus del Tumor Mamario del Ratón/genética , Ratones , Ratones Transgénicos , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-myc/genética , Receptor ErbB-2 , Transducción de Señal/genética , Transducción de Señal/fisiología , Familia-src Quinasas
4.
Mol Cell Biol ; 18(4): 2344-59, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9528804

RESUMEN

Transgenic mice expressing the polyomavirus (PyV) middle T antigen (MT) develop multifocal mammary tumors which frequently metastasize to the lung. The potent transforming activity of PyV MT is correlated with its capacity to activate and associate with a number of signaling molecules, including the Src family tyrosine kinases, the 85-kDa Src homology 2 subunit of the phosphatidylinositol 3' (PI-3') kinase, and the Shc adapter protein. To uncover the role of these signaling proteins in MT-mediated mammary tumorigenesis, we have generated transgenic mice that express mutant PyV MT antigens decoupled from either the Shc or the PI-3' kinase signaling pathway. In contrast to the rapid induction of metastatic mammary tumors observed in the strains expressing wild-type PyV MT, mammary epithelial cell-specific expression of either mutant PyV MT resulted in the induction of extensive mammary epithelial hyperplasias. The mammary epithelial hyperplasias expressing the mutant PyV MT defective in recruiting the PI-3' kinase were highly apoptotic, suggesting that recruitment of PI-3' kinase by MT affects cell survival. Whereas the initial phenotypes observed in both strains were global mammary epithelial hyperplasias, focal mammary tumors eventually arose in all female transgenic mice. Genetic and biochemical analyses of tumorigenesis in the transgenic strains expressing the PyV MT mutant lacking the Shc binding site revealed that a proportion of the metastatic tumors arising in these mice displayed evidence of reversion of the mutant Shc binding site. In contrast, no evidence of reversion of the PI-3' kinase binding site was noted in tumors derived from the strains expressing the PI-3' kinase binding site MT mutant. Tumor progression in both mutant strains was further correlated with upregulation of the epidermal growth factor receptor family members which are known to couple to the PI-3' kinase and Shc signaling pathways. Taken together, these observations suggest that PyV MT-mediated tumorigenesis requires activation of both Shc and PI-3' kinase, which appear to be required for stimulation of cell proliferation and survival signaling pathways, respectively.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas Adaptadoras del Transporte Vesicular , Antígenos Transformadores de Poliomavirus/fisiología , Transformación Celular Neoplásica , Transformación Celular Viral , Neoplasias Mamarias Experimentales/virología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas/metabolismo , Transducción de Señal , Secuencia de Aminoácidos , Animales , Apoptosis , Secuencia de Bases , Sitios de Unión , Mama/patología , ADN , Progresión de la Enfermedad , Activación Enzimática , Receptores ErbB/biosíntesis , Receptores ErbB/genética , Femenino , Hiperplasia , Masculino , Neoplasias Mamarias Experimentales/química , Neoplasias Mamarias Experimentales/etiología , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Mutagénesis , Poliomavirus/fisiología , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/genética , Receptor ErbB-2/biosíntesis , Receptor ErbB-2/genética , Receptor ErbB-3 , Proteínas Adaptadoras de la Señalización Shc , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Distribución Tisular , Regulación hacia Arriba
5.
Mol Cell Biol ; 16(10): 5726-36, 1996 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8816486

RESUMEN

Transgenic mice expressing either the neu proto-oncogene or transforming growth factor (TGF-alpha) in the mammary epithelium develop spontaneous focal mammary tumors that occur after a long latency. Since the epidermal growth factor receptor (EGFR) and Neu are capable of forming heterodimers that are responsive to EGFR ligands such as TGF-alpha, we examined whether coexpression of TGF-alpha and Neu in mammary epithelium could cooperate to accelerate the onset of mammary tumors. To test this hypothesis, we interbred separate transgenic strains harboring either a mouse mammary tumor virus/TGF-alpha or a mouse mammary tumor virus/neu transgene to generate bitransgenic mice that coexpress TGF-alpha and neu in the mammary epithelium. Female mice coexpressing TGF-alpha and neu developed multifocal mammary tumors which arose after a significantly shorter latency period than either parental strain alone. The development of these mammary tumors was correlated with the tyrosine phosphorylation of Neu and the recruitment of c-Src to the Neu complex. Immunoprecipitation and immunoblot analyses with EGFR- and Neu-specific antisera, however, failed to detect physical complexes of these two receptors. Taken together, these observations suggest that Neu and TGF-alpha cooperate in mammary tumorigenesis through a mechanism involving Neu and EGFR transactivation.


Asunto(s)
Transformación Celular Neoplásica , Glándulas Mamarias Animales/metabolismo , Neoplasias Mamarias Experimentales/genética , Receptor ErbB-2/metabolismo , Factor de Crecimiento Transformador alfa/metabolismo , Envejecimiento , Animales , Dimerización , Epitelio/metabolismo , Epitelio/patología , Femenino , Genes erbB-2 , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/fisiopatología , Virus del Tumor Mamario del Ratón , Ratones , Ratones Transgénicos , Fosforilación , Receptor ErbB-2/biosíntesis , Receptor ErbB-2/genética , Transcripción Genética , Factor de Crecimiento Transformador alfa/biosíntesis , Factor de Crecimiento Transformador alfa/genética
6.
Cancer Res ; 61(24): 8887-95, 2001 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-11751413

RESUMEN

Aberrrant signaling by the epidermal growth factor receptor [EGFR (HER1, erbB1)] and/or HER2/neu tyrosine kinases is present in a cohort of breast carcinomas. Because HER2 is constitutively phosphorylated in some breast tumors, we speculated that, in these cancers, transmodulation of HER2 may occur via EGFR signaling. To test this possibility, we examined the effect of EGFR-specific kinase inhibitors against the HER2-overexpressing human breast tumor lines BT-474, SKBR-3, MDA-361, and MDA-453. ZD1839 (Iressa) is an ATP-mimetic that inhibits the purified EGFR and HER2 kinases in vitro with an IC(50) of 0.033 and >3.7 microM, respectively. The specificity of ZD1839 against EGFR was confirmed in Rat1 fibroblasts transfected with EGFR or HER2 chimeric receptors activated by synthetic ligands without the interference of endogenous receptors. Treatment of all breast cancer cell lines (except MDA-453) with 1 microM ZD1839 almost completely eliminated HER2 phosphorylation. In contrast, the incorporation of [gamma-(32)P]ATP in vitro onto HER2 receptors isolated from BT-474 cells was unaffected by 1 microM ZD1839. EGFR is expressed by BT-474, SKBR-3, and MDA-361 but not by MDA-453 cells, suggesting that ZD1839-mediated inhibition of the EGFR kinase explained the inhibition of HER2 phosphorylation in vivo. In SKBR-3 cells, ZD1839 exhibited a greater growth-inhibitory effect than Herceptin, a monoclonal antibody against the HER2 ectodomain. In both SKBR-3 and BT-474 cells, treatment with ZD1839 plus Herceptin induced a greater apoptotic effect than either inhibitor alone. Finally, ZD1839 completely prevented growth of BT-474 xenografts established in nude mice and enhanced the antitumor effect of Herceptin. These data imply that EGFR tyrosine kinase inhibitors will be effective against HER2-overexpressing breast tumor cells that also express EGFR and support their use in combination with HER2 antibodies, such as Herceptin, against mammary carcinomas with high levels of the HER2 proto-oncogene.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Receptores ErbB/antagonistas & inhibidores , Quinazolinas/farmacología , Receptor ErbB-2/biosíntesis , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Dominio Catalítico/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Inhibidores Enzimáticos/administración & dosificación , Femenino , Gefitinib , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Fosforilación/efectos de los fármacos , Proto-Oncogenes Mas , Quinazolinas/administración & dosificación , Receptor ErbB-2/metabolismo , Transducción de Señal/efectos de los fármacos , Especificidad por Sustrato , Factor de Crecimiento Transformador alfa/antagonistas & inhibidores , Factor de Crecimiento Transformador alfa/farmacología , Trastuzumab , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Oncogene ; 11(9): 1801-10, 1995 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-7478608

RESUMEN

Overexpression and amplification of the erbB-2 (neu) is thought to play a major role in mammary cancer. Although studies suggest that Neu is directly involved in the genesis of mammary tumors, the molecular mechanism by which Neu induces tumors is not well understood. Recently, we have demonstrated that the activity of c-Src tyrosine kinase is elevated in Neu-induced mammary tumors and this elevated activity correlates with its capacity to physically associate with Neu. To explore whether other members of the c-Src family are activated in these mammary tumors, we measured the in vitro kinase activity of the c-Yes and Fyn kinases in protein extracts derived from mammary tumor tissue and morphological normal adjacent tissue. These analyses revealed that c-Yes kinase activity was elevated in Neu-induced tumors by comparison to the adjacent tissue. By contrast, no significant activation of the Fyn kinase was noted in these tumors. Activation of c-Yes tyrosine kinase correlated with the capacity of c-Yes to associate with Neu in vivo in lysates derived from primary tumor samples. Studies with Rat.2 fibroblasts overexpressing activated Neu revealed that c-Src requires the presence of tyrosine phosphorylated Neu for its ability to interact with Neu in vivo. Moreover, analyses using radiolabeled c-Yes SH2 fusion protein revealed that this interaction is likely occurring in a direct fashion. Although both c-Src and c-Yes kinase associate with Neu in vivo, a tyrosine phosphorylated protein of 89 kd (p89) was found associated with c-Src but not with c-Yes in cell lysates derived from mammary epithelial cells transformed by either Neu or PyV middle T antigen. Furthermore, this tyrosine phosphorylated protein was not detected in c-Src complexes derived from fibroblasts transformed by either Neu or PyV middle T. These observations suggest that p89 associates with c-Src only in mammary epithelial cells and not in fibroblasts.


Asunto(s)
Genes erbB-2 , Neoplasias Mamarias Experimentales/metabolismo , Proteínas de Neoplasias , Fosfoproteínas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Familia-src Quinasas/metabolismo , Animales , Línea Celular , Electroforesis en Gel de Poliacrilamida , Activación Enzimática , Epitelio , Femenino , Glutatión Transferasa/biosíntesis , Glutatión Transferasa/metabolismo , Immunoblotting , Glándulas Mamarias Animales/metabolismo , Neoplasias Mamarias Experimentales/virología , Virus del Tumor Mamario del Ratón , Ratones , Fosfoproteínas/análisis , Fosfotirosina/análisis , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-yes , Ratas , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/metabolismo , Transfección
8.
Oncogene ; 11(2): 271-9, 1995 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-7542762

RESUMEN

Transgenic mice expressing either the activated or wild type neu oncogene heritably develop metastatic mammary tumors. Tumor development in this transgenic mouse model correlates with activation of the Neu tyrosine kinase. Recently, we have shown that these Neu-induced mammary tumors possess elevated c-Src tyrosine kinase activity. Here, we demonstrate that c-Src requires tyrosine phosphorylated Neu for its ability to associate with Neu in vivo and this association is likely the result of a direct physical binding of c-Src SH2 domain to the tyrosine phosphorylated Neu. By contrast, the c-Src SH2 domain did not interact directly with tyrosine phosphorylated EGFR. Moreover, in established cell lines expressing elevated levels of EGFR, EGF stimulation results in transphosphorylation of Neu and formation of complexes between c-Src and tyrosine phosphorylated Neu. Taken together, these observations suggest that activation of c-Src by these two closely related EGFR family members results from a direct and specific interaction of c-Src with tyrosine phosphorylated Neu.


Asunto(s)
Receptores ErbB/fisiología , Genes src , Proteínas Proto-Oncogénicas/fisiología , Receptor ErbB-2 , Transducción de Señal/fisiología , Animales , Anticuerpos , Activación Enzimática , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/inmunología , Immunoblotting , Neoplasias Mamarias Animales/tratamiento farmacológico , Neoplasias Mamarias Animales/enzimología , Neoplasias Mamarias Animales/patología , Ratones , Fosforilación , Fosfotirosina , Pruebas de Precipitina , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/inmunología , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/inmunología , Células Tumorales Cultivadas/efectos de los fármacos , Tirosina/análogos & derivados , Tirosina/inmunología , Tirosina/metabolismo
10.
Oncogene ; 29(2): 174-87, 2010 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-19826412

RESUMEN

Amplification and overexpression of ErbB2 strongly correlates with aggressive breast cancers. A deeper understanding of pathways downstream of ErbB2 signaling that are required for the transformation of human mammary epithelial cells will identify novel strategies for therapeutic intervention in breast cancer. Using an inducible activation of ErbB2 autophosphorylation qsite mutants and the MCF-10A three-dimensional (3D) culture system, we investigated pathways used by ErbB2 to transform the epithelia. We report that ErbB2 induces cell proliferation and loss of 3D organization by redundant mechanisms, whereas it disrupts apical basal polarity and inhibits apoptosis using Tyr 1201 and Tyr 1226/7, respectively. Signals downstream of Tyr 1226/7 were also sufficient to confer paclitaxel resistance. The Tyr 1226/7 binds Shc, and the knockdown of Shc blocks the ability of ErbB2 to inhibit apoptosis and mediate paclitaxel resistance. Tyr 1226/7 is known to activate the Ras/Erk pathway; however, paclitaxel resistance did not correlate with the activation of Erk or Akt, suggesting the presence of a novel mechanism. Thus, our results show that targeting pathways used by ErbB2 to inhibit cell death is a better option than targeting cell proliferation pathways. Furthermore, we identify a novel function for Shc as a regulator of apoptosis and drug resistance in human mammary epithelial cells transformed by ErbB2.


Asunto(s)
Apoptosis/fisiología , Polaridad Celular/fisiología , Proliferación Celular , Receptor ErbB-2/metabolismo , Proteínas Adaptadoras de la Señalización Shc/metabolismo , Animales , Antineoplásicos Fitogénicos/farmacología , Apoptosis/genética , Sitios de Unión/genética , Western Blotting , Técnicas de Cultivo de Célula/métodos , Línea Celular , Polaridad Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Transformación Celular Neoplásica , Resistencia a Medicamentos/genética , Fibroblastos/citología , Fibroblastos/metabolismo , Citometría de Flujo , Humanos , Mutación , Paclitaxel/farmacología , Fosforilación , Receptor ErbB-2/genética , Proteínas Adaptadoras de la Señalización Shc/genética , Tirosina/genética , Tirosina/metabolismo
11.
Oncogene ; 29(43): 5839-49, 2010 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-20711231

RESUMEN

The activation of receptor tyrosine kinases, particularly ErbB2, has an important role in the genesis of breast cancer. ErbB2 kinase activity promotes Ras-mediated stimulation of downstream protein kinase cascades, including the Ras/Raf-1/MAPK/ERK kinase (Mek)/extracellular signal-regulated kinase (Erk) pathway, leading to tumor cell growth and migration. Signaling through the Ras-Erk pathway can be influenced by p21-activated kinase-1 (Pak1), an effector of the Rho family GTPases Rac and Cdc42. In this study, we asked if ErbB2 expression correlates with Pak1 and Erk activity in human breast cancer specimens, and if Pak1 signaling is required for ErbB2 transformation in a three-dimensional (3D) in vitro setting and in xenografts. We found a correlation between ErbB2 expression and activation of Pak in estrogen receptor-positive human breast tumor samples and observed that in 3D cultures, activation of Rac-Pak1 pathway by ErbB2 homodimers induced growth factor-independent proliferation and promoted disruption of 3D mammary acinar-like structures through activation of the Erk and Akt pathways. Further, we found that inhibition of Pak1 by small molecules compromised activation of Erk and Akt, resulting in reversion of the malignant phenotype and restoration of normal acinar architecture. Finally, ErbB2-amplified breast cancer cells expressing a specific Pak inhibitor showed delayed tumor formation and downregulation of Erk and Akt signaling in vivo. These data imply that the Rac-Pak pathway is vital to ErbB2-mediated transformation and that Pak inhibitors represent plausible drug targets in breast cancers in which ErbB2 signaling is activated.


Asunto(s)
Neoplasias de la Mama/metabolismo , Transformación Celular Neoplásica/metabolismo , Receptor ErbB-2/metabolismo , Transducción de Señal/fisiología , Quinasas p21 Activadas/metabolismo , Animales , Western Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Inhibidores Enzimáticos/farmacología , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Ratones , Ratones SCID , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor ErbB-2/genética , Transducción de Señal/efectos de los fármacos , Análisis de Matrices Tisulares , Ensayos Antitumor por Modelo de Xenoinjerto , Quinasas p21 Activadas/genética
12.
Oncogene ; 27(55): 6878-87, 2008 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-19029931

RESUMEN

In the past 20 years, the discovery and characterization of the molecular machinery that controls cellular polarization have enabled us to achieve a better understanding of many biological processes. Spatial asymmetry or establishment of cell polarity during embryogenesis, epithelial morphogenesis, neuronal differentiation, and migration of fibroblasts and T cells are thought to rely on a small number of evolutionarily conserved proteins and pathways. Correct polarization is crucial for normal cell physiology and tissue homeostasis, and is lost in cancer. Thus, cell polarity signaling is likely to have an important function in tumor progression. Recent findings have identified a regulator of cell polarity, the Par complex, as an important signaling node in tumorigenesis. In normal cell types, the Par complex is part of the molecular machinery that regulates cell polarity and maintains normal cell homeostasis. As such, the polarity regulators are proposed to have a tumor suppressor function, consistent with the loss of polarity genes associated with hyperproliferation in Drosophila melanogaster. However, recent studies showing that some members of this complex also display pro-oncogenic activities suggest a more complex regulation of the polarity machinery during cellular transformation. Here, we examine the existing data about the different functions of the Par complex. We discuss how spatial restriction, binding partners and substrate specificity determine the signaling properties of Par complex proteins. A better understanding of these processes will very likely shed some light on how the Par complex can switch from a normal polarity regulation function to promotion of transformation downstream of oncogenes.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Polaridad Celular/genética , Proteínas de la Membrana/fisiología , Neoplasias/etiología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/fisiología , Animales , Proteínas de Ciclo Celular/genética , Movimiento Celular/genética , Movimiento Celular/fisiología , Polaridad Celular/fisiología , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor/fisiología , Humanos , Proteínas de la Membrana/genética , Modelos Biológicos , Complejos Multiproteicos/fisiología , Neoplasias/genética , Transducción de Señal/genética , Transducción de Señal/fisiología
13.
Genes Dev ; 8(1): 23-32, 1994 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-7507074

RESUMEN

Transgenic mice expressing the polyomavirus (PyV) middle T oncogene in the mammary epithelium develop multifocal mammary tumors that metastasize with high frequency. The potent transforming activity of PyV middle T antigen can, in part, be attributed to its ability to associate with and to activate a number of c-Src family tyrosine kinases (c-Src, c-Yes, and Fyn). As a first step toward assessing the role of individual c-Src family tyrosine kinases in PyV middle T antigen-induced mammary tumorigenesis, we have crossed transgenic mice carrying the mouse mammary tumor virus (MMTV)/PyV middle T antigen fusion gene with mice bearing a disrupted c-src proto-oncogene. In contrast to the rapid tumor progression seen in the original MMTV/PyV middle T antigen strains, mice expressing the transgene in the absence of functional c-Src rarely developed mammary tumors. After long latency, these mice did eventually develop abnormal hyperplastic mammary tissue. This growth disturbance was correlated with elevated expression of the PyV middle T antigen and the activation of the PyV middle T antigen-associated c-Yes tyrosine kinase. However, transgenic mice expressing the PyV middle T antigen in the mammary epithelium of wild-type or Yes-deficient mice developed multifocal mammary tumors with comparable kinetics. Taken together, these findings suggest that c-Src tyrosine kinase activity is required for PyV middle T antigen-induced mammary tumorigenesis and also illustrate an in vivo genetic approach to the dissection of mitogenic signal transduction pathways.


Asunto(s)
Neoplasias Mamarias Experimentales/etiología , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Familia-src Quinasas , Animales , Antígenos Transformadores de Poliomavirus/genética , Activación Enzimática , Neoplasias Mamarias Experimentales/genética , Ratones , Ratones Transgénicos , Oncogenes , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas c-yes , Proteínas Proto-Oncogénicas pp60(c-src)/fisiología
14.
Biochem Cell Biol ; 70(10-11): 1268-76, 1992.
Artículo en Inglés | MEDLINE | ID: mdl-1338413

RESUMEN

Mouse BC3H1 myoblasts were stably transfected with the adenovirus 5 E1A gene. One clonal line, BC3E7, was found to differ in some important respects from those previously reported for E1A-transformed myoblasts. In contrast to BC3H1 cells which differentiate when confluent in medium containing 0.5% fetal calf serum (FCS), BC3E7 cells failed to elongate and align, to express acetylcholine receptor and creatine kinase, and to down-regulate expression of beta- and gamma-actins and tropomyosin isoform (TM) 1. However, increased synthesis of TMs 2, 3, and 4, and myosin light chain 1 associated with differentiation in BC3H1 still occurred in BC3E7 cells, and most surprisingly, alpha-actin was produced at a significant level in both proliferating and confluent BC3E7 cells. Interestingly, myogenin was expressed in confluent BC3E7 cells in 0.5% FCS, but not in 20%. The level of E1A expression in BC3E7 cells was found to be very low by analysis of mRNA, by immunoprecipitation of E1A protein, and by the ability of BC3E7 cells to complement the E1A-deficient adenovirus mutant dl312. These results suggest that different levels of E1A may be needed to repress different promoters and that E1A does not block myogenic differentiation by repressing myogenin expression, but represses each muscle gene independently.


Asunto(s)
Proteínas E1A de Adenovirus/fisiología , Adenovirus Humanos/fisiología , Proteínas Musculares/biosíntesis , Proteínas E1A de Adenovirus/genética , Adenovirus Humanos/genética , Animales , Diferenciación Celular , Electroforesis en Gel Bidimensional , Regulación de la Expresión Génica/fisiología , Ratones , Proteínas Musculares/genética , Músculos/citología , Miogenina , Proteínas Recombinantes de Fusión/metabolismo , Transfección , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA