Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancer Cell ; 36(1): 68-83.e9, 2019 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-31257073

RESUMEN

RAC1 P29 is the third most commonly mutated codon in human cutaneous melanoma, after BRAF V600 and NRAS Q61. Here, we study the role of RAC1P29S in melanoma development and reveal that RAC1P29S activates PAK, AKT, and a gene expression program initiated by the SRF/MRTF transcriptional pathway, which results in a melanocytic to mesenchymal phenotypic switch. Mice with ubiquitous expression of RAC1P29S from the endogenous locus develop lymphoma. When expressed only in melanocytes, RAC1P29S cooperates with oncogenic BRAF or with NF1-loss to promote tumorigenesis. RAC1P29S also drives resistance to BRAF inhibitors, which is reversed by SRF/MRTF inhibitors. These findings establish RAC1P29S as a promoter of melanoma initiation and mediator of therapy resistance, while identifying SRF/MRTF as a potential therapeutic target.


Asunto(s)
Transformación Celular Neoplásica/genética , Resistencia a Antineoplásicos/genética , Transición Epitelial-Mesenquimal/genética , Melanoma/etiología , Melanoma/patología , Mutación , Proteína de Unión al GTP rac1/genética , Alelos , Sustitución de Aminoácidos , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Humanos , Masculino , Melanocitos/metabolismo , Melanoma/mortalidad , Melanoma/terapia , Ratones , Ratones Transgénicos , Modelos Biológicos , Pronóstico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/genética , Factor de Respuesta Sérica , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Cell Rep ; 25(13): 3545-3553.e2, 2018 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-30590030

RESUMEN

RAS family GTPases contribute directly to the regulation of type I phosphoinositide 3-kinases (PI3Ks) via RAS-binding domains in the PI3K catalytic p110 subunits. Disruption of this domain of p110α impairs RAS-mutant-oncogene-driven tumor formation and maintenance. Here, we test the effect of blocking the interaction of RAS with p110α on epidermal growth factor receptor (EGFR)-mutant-driven lung tumorigenesis. Disrupting the RAS-PI3K interaction inhibits activation of both AKT and RAC1 in EGFR-mutant lung cancer cells, leading to reduced growth and survival, and inhibits EGFR-mutant-induced tumor onset and promotes major regression of established tumors in an autochthonous mouse model of EGFR-mutant-induced lung adenocarcinoma. The RAS-PI3K interaction is thus an important signaling node and potential therapeutic target in EGFR-mutant lung cancer, even though RAS oncogenes are not themselves mutated in this setting, suggesting different strategies for tackling tyrosine kinase inhibitor resistance in lung cancer.


Asunto(s)
Receptores ErbB/genética , Neoplasias Pulmonares/genética , Mutación/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas ras/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/metabolismo , Humanos , Neoplasias Pulmonares/patología , Ratones Endogámicos C57BL , Fosfatidilinositol 3-Quinasas/química , Unión Proteica/efectos de los fármacos , Dominios Proteicos
3.
Cancer Cell ; 24(5): 617-30, 2013 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-24229709

RESUMEN

RAS proteins directly activate PI3-kinases. Mice bearing a germline mutation in the RAS binding domain of the p110α subunit of PI3-kinse are resistant to the development of RAS-driven tumors. However, it is unknown whether interaction of RAS with PI3-kinase is required in established tumors. The need for RAS interaction with p110α in the maintenance of mutant Kras-driven lung tumors was explored using an inducible mouse model. In established tumors, removal of the ability of p110α to interact with RAS causes long-term tumor stasis and partial regression. This is a tumor cell-autonomous effect, which is improved significantly by combination with MEK inhibition. Total removal of p110α expression or activity has comparable effects, albeit with greater toxicities.


Asunto(s)
Adenocarcinoma/enzimología , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Neoplasias Pulmonares/enzimología , Proteínas Proto-Oncogénicas p21(ras)/genética , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/patología , Animales , Antineoplásicos Hormonales/farmacología , Fosfatidilinositol 3-Quinasa Clase I/química , Fosfatidilinositol 3-Quinasa Clase I/genética , Progresión de la Enfermedad , Humanos , Pulmón/patología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Mutación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Piridonas/farmacología , Pirimidinonas/farmacología , Tamoxifeno/farmacología , Carga Tumoral
4.
J Cell Sci ; 123(Pt 20): 3467-77, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20930141

RESUMEN

Although TGF-ß suppresses early stages of tumour development, it later contributes to tumour progression when cells become resistant to its suppressive effects. In addition to circumventing TGF-ß-induced growth arrest and apoptosis, malignant tumour cells become capable of undergoing epithelial-to-mesenchymal transition (EMT), favouring invasion and metastasis. Therefore, defining the mechanisms that allow cancer cells to escape from the suppressive effects of TGF-ß is fundamental to understand tumour progression and to design specific therapies. Here, we have examined the role of Snail1 as a suppressor of TGF-ß-induced apoptosis in murine non-transformed hepatocytes, rat and human hepatocarcinoma cell lines and transgenic mice. We show that Snail1 confers resistance to TGF-ß-induced cell death and that it is sufficient to induce EMT in adult hepatocytes, cells otherwise refractory to this transition upon exposure to TGF-ß. Furthermore, we show that Snail1 silencing prevents EMT and restores the cell death response induced by TGF-ß. As Snail1 is a known target of TGF-ß signalling, our data indicate that Snail1 might transduce the tumour-promoting effects of TGF-ß, namely the EMT concomitant with the resistance to cell death.


Asunto(s)
Apoptosis/efectos de los fármacos , Transición Epitelial-Mesenquimal/fisiología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Animales , Western Blotting , Línea Celular , Células Cultivadas , Ensayo de Cambio de Movilidad Electroforética , Transición Epitelial-Mesenquimal/genética , Hepatocitos/citología , Humanos , Inmunohistoquímica , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Ratas , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética
5.
Cell Signal ; 21(11): 1595-606, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19586611

RESUMEN

Treatment of FaO rat hepatoma cells with TGF-beta selects cells that survive to its apoptotic effect and undergo epithelial-mesenchymal transitions (EMT). We have established a cell line (T beta T-FaO, from TGF-beta-treated FaO) that shows a mesenchymal, de-differentiated, phenotype in the presence of TGF-beta and is refractory to its suppressor effects. In the absence of this cytokine, cells revert to an epithelial phenotype in 3-4 weeks and recover the response to TGF-beta. T beta T-FaO show higher capacity to migrate than that observed in the parental FaO cells. We found that FaO cells express low levels of CXCR4 and do not respond to SDF-1 alpha. However, TGF-beta up-regulates CXCR4, through a NF kappaB-dependent mechanism, and T beta T-FaO cells show elevated levels of CXCR4, which is located in the presumptive migration front. A specific CXCR4 antagonist (AMD3100) attenuates the migratory capacity of T beta T-FaO cells on collagen gels. Extracellular SDF-1 alpha activates the ERKs pathway in T beta T-FaO, but not in FaO cells, increasing cell scattering and protecting cells from apoptosis induced by serum deprivation. Targeted knock-down of CXCR4 with specific siRNA blocks the T beta T-FaO response to SDF-1 alpha. Thus, the SDF-1/CXCR4 axis might play an important role in mediating cell migration and survival after a TGF-beta-induced EMT in hepatoma cells.


Asunto(s)
Quimiocina CXCL12/fisiología , Neoplasias Hepáticas Experimentales/metabolismo , Receptores CXCR4/fisiología , Factor de Crecimiento Transformador beta/farmacología , Animales , Apoptosis , Bencilaminas , Diferenciación Celular , Línea Celular Tumoral , Movimiento Celular , Quimiocina CXCL12/metabolismo , Ciclamas , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Técnicas de Silenciamiento del Gen , Compuestos Heterocíclicos/farmacología , Neoplasias Hepáticas Experimentales/patología , Mesodermo/patología , FN-kappa B/metabolismo , Fenotipo , ARN Interferente Pequeño/metabolismo , Ratas , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Transducción de Señal
6.
Mol Cell Biol ; 29(15): 4235-49, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19451229

RESUMEN

Integrin signaling promotes, through p21-activated kinase, phosphorylation and inactivation of the tumor suppressor merlin, thus removing a block to mitogenesis in normal cells. However, the biochemical function of merlin and the effector pathways critical for the pathogenesis of malignant mesothelioma and other NF2-related malignancies are not known. We report that integrin-specific signaling promotes activation of mTORC1 and cap-dependent mRNA translation. Depletion of merlin rescues mTORC1 signaling in cells deprived of anchorage to a permissive extracellular matrix, suggesting that integrin signaling controls mTORC1 through inactivation of merlin. This signaling pathway controls translation of the cyclin D1 mRNA and, thereby, cell cycle progression. In addition, it promotes cell survival. Analysis of a panel of malignant mesothelioma cell lines reveals a strong correlation between loss of merlin and activation of mTORC1. Merlin-negative lines are sensitive to the growth-inhibitory effect of rapamycin, and the expression of recombinant merlin renders them partially resistant to rapamycin. Conversely, depletion of merlin restores rapamycin sensitivity in merlin-positive lines. These results indicate that integrin-mediated adhesion promotes mTORC1 signaling through the inactivation of merlin. Furthermore, they reveal that merlin-negative mesotheliomas display unregulated mTORC1 signaling and are sensitive to rapamycin, thus providing a preclinical rationale for prospective, biomarker-driven clinical studies of mTORC1 inhibitors in these tumors.


Asunto(s)
Integrinas/metabolismo , Neurofibromina 2/genética , Transducción de Señal , Factores de Transcripción/metabolismo , Antibióticos Antineoplásicos/farmacología , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ciclina D1/genética , Fase G1 , Humanos , Immunoblotting , Integrinas/genética , Diana Mecanicista del Complejo 1 de la Rapamicina , Mesotelioma/genética , Mesotelioma/metabolismo , Mesotelioma/patología , Complejos Multiproteicos , Neurofibromina 2/metabolismo , Biosíntesis de Proteínas , Proteínas , Caperuzas de ARN , Interferencia de ARN , ARN Mensajero/genética , Sirolimus/farmacología , Serina-Treonina Quinasas TOR , Factores de Transcripción/genética , Transfección
7.
Biochim Biophys Acta ; 1793(2): 253-63, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18848961

RESUMEN

Transforming growth factor-beta (TGF-beta) induces apoptosis in hepatocytes, through a mechanism mediated by reactive oxygen species (ROS) production. Numerous tumoral cells develop mechanisms to escape from the TGF-beta-induced tumor suppressor effects. In this work we show that in FaO rat hepatoma cells inhibition of the epidermal growth factor receptor (EGFR) with the tyrphostin AG1478 enhances TGF-beta-induced cell death, coincident with an elevated increase in ROS production and GSH depletion. These events correlate with down-regulation of genes involved in the maintenance of redox homeostasis, such as gamma-GCS and MnSOD, and elevated mitochondrial ROS. Nonetheless, not all the ROS proceed from the mitochondria. Emerging evidences indicate that ROS production by TGF-beta is also mediated by the NADPH oxidase (NOX) system. TGF-beta-treated FaO cells induce nox1 expression. However, the treatment with TGF-beta and AG1478 greatly enhanced the expression of another family member: nox4. NOX1 and NOX4 targeted knock-down by siRNA experiments suggest that they play opposite roles, because NOX1 knockdown increases caspase-3 activity and cell death, whilst NOX4 knock-down attenuates the apoptotic process. This attenuation correlates with maintenance of GSH and antioxidant enzymes levels. In summary, EGFR inhibition enhances apoptosis induced by TGF-beta in FaO rat hepatoma cells through an increased oxidative stress coincident with a change in the expression pattern of NOX enzymes.


Asunto(s)
Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/enzimología , Factor de Crecimiento Epidérmico/metabolismo , Neoplasias Hepáticas/enzimología , NADPH Oxidasas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología , Animales , Antioxidantes/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Receptores ErbB/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Isoenzimas/genética , Isoenzimas/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , NADH NADPH Oxidorreductasas/genética , NADH NADPH Oxidorreductasas/metabolismo , NADPH Oxidasa 1 , NADPH Oxidasa 4 , NADPH Oxidasas/genética , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas , ARN Interferente Pequeño/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Tirfostinos/farmacología
8.
Biochem J ; 405(2): 251-9, 2007 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-17407446

RESUMEN

The TGF-beta (transforming growth factor-beta) induces survival signals in foetal rat hepatocytes through transactivation of EGFR (epidermal growth factor receptor). The molecular mechanism is not completely understood, but both activation of the TACE (tumour necrosis factor alpha-converting enzyme)/ADAM17 (a disintegrin and metalloproteinase 17; one of the metalloproteases involved in shedding of the EGFR ligands) and up-regulation of TGF-alpha and HB-EGF (heparin-binding epidermal growth factor-like growth factor) appear to be involved. In the present study, we have analysed the molecular mechanisms that mediate up-regulation of the EGFR ligands by TGF-beta in foetal rat hepatocytes. The potential involvement of ROS (reactive oxygen species), an early signal induced by TGF-beta, and the existence of an amplification loop triggered by initial activation of the EGFR, have been studied. Results indicate that DPI (diphenyleneiodonium) and apocynin, two NOX (NADPH oxidase) inhibitors, and SB431542, an inhibitor of the TbetaR-I (TGF-beta receptor I), block up-regulation of EGFR ligands and Akt activation. Different members of the NOX family of genes are expressed in hepatocytes, included nox1, nox2 and nox4. TGF-beta up-regulates nox4 and increases the levels of Rac1 protein, a known regulator of both Nox1 and Nox2, in a TbetaR-I-dependent manner. TGF-beta mediates activation of the nuclear factor-kappaB pathway, which is inhibited by DPI and is required for up-regulation of TGF-alpha and HB-EGF. In contrast, EGFR activation is not required for TGF-beta-induced up-regulation of those ligands. Considering previous work that has established the role of ROS in apoptosis induced by TGF-beta in hepatocytes, the results of the present study indicate that ROS might mediate both pro- and anti-apoptotic signals in TGF-beta-treated cells.


Asunto(s)
Receptores ErbB/metabolismo , NADPH Oxidasas/metabolismo , FN-kappa B/fisiología , Factor de Crecimiento Transformador beta/fisiología , Acetofenonas/farmacología , Animales , Antracenos/farmacología , Benzamidas/farmacología , Cromonas/farmacología , Dioxoles/farmacología , Activación Enzimática/efectos de los fármacos , Factor de Crecimiento Epidérmico/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Flavonoides/farmacología , Factor de Crecimiento Similar a EGF de Unión a Heparina , Hepatocitos/enzimología , Humanos , Imidazoles/farmacología , Péptidos y Proteínas de Señalización Intercelular , Morfolinas/farmacología , NADH NADPH Oxidorreductasas/biosíntesis , NADPH Oxidasa 1 , NADPH Oxidasa 4 , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/biosíntesis , Compuestos Onio/farmacología , Péptidos/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Quinazolinas , Ratas , Especies Reactivas de Oxígeno/farmacología , Tirfostinos/farmacología , Regulación hacia Arriba , Proteína de Unión al GTP rac1/biosíntesis
9.
Comp Hepatol ; 6: 1, 2007 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-17311678

RESUMEN

BACKGROUND: The activation of hepatic stellate cells (HSCs) plays a pivotal role during liver injury because the resulting myofibroblasts (MFBs) are mainly responsible for connective tissue re-assembly. MFBs represent therefore cellular targets for anti-fibrotic therapy. In this study, we employed activated HSCs, termed M1-4HSCs, whose transdifferentiation to myofibroblastoid cells (named M-HTs) depends on transforming growth factor (TGF)-beta. We analyzed the oxidative stress induced by TGF-beta and examined cellular defense mechanisms upon transdifferentiation of HSCs to M-HTs. RESULTS: We found reactive oxygen species (ROS) significantly upregulated in M1-4HSCs within 72 hours of TGF-beta administration. In contrast, M-HTs harbored lower intracellular ROS content than M1-4HSCs, despite of elevated NADPH oxidase activity. These observations indicated an upregulation of cellular defense mechanisms in order to protect cells from harmful consequences caused by oxidative stress. In line with this hypothesis, superoxide dismutase activation provided the resistance to augmented radical production in M-HTs, and glutathione rather than catalase was responsible for intracellular hydrogen peroxide removal. Finally, the TGF-beta/NADPH oxidase mediated ROS production correlated with the upregulation of AP-1 as well as platelet-derived growth factor receptor subunits, which points to important contributions in establishing antioxidant defense. CONCLUSION: The data provide evidence that TGF-beta induces NADPH oxidase activity which causes radical production upon the transdifferentiation of activated HSCs to M-HTs. Myofibroblastoid cells are equipped with high levels of superoxide dismutase activity as well as glutathione to counterbalance NADPH oxidase dependent oxidative stress and to avoid cellular damage.

10.
Cell Signal ; 19(4): 683-94, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17055226

RESUMEN

The transforming growth factor-beta (TGF-beta) regulates hepatocyte growth, inhibiting proliferation and inducing apoptosis. Indeed, escaping from the TGF-beta suppressor actions might be a prerequisite for liver tumour progression. In this work we show that TGF-beta plays a dual role in regulating apoptosis in FaO rat hepatoma cells, since, in addition to its pro-apoptotic effect, TGF-beta also activates survival signals, such as AKT, the epidermal growth factor receptor (EGFR) being required for its activation. TGF-beta induces the expression of the EGFR ligands transforming growth factor-alpha (TGF-alpha) and heparin-binding EGF-like growth factor (HB-EGF) and induces intracellular re-localization of the EGFR. Cells that overcome the apoptotic effects of TGF-beta undergo morphological changes reminiscent of an epithelial-mesenchymal transition (EMT) process. In contrast, TGF-beta does not activate AKT in adult hepatocytes, which correlates with lack of EGFR transactivation and no response to EGFR inhibitors. Although TGF-beta induces TGF-alpha and HB-EGF in adult hepatocytes, these cells show very low expression of TACE/ADAM 17 (TNF-alpha converting enzyme), which is required for EGFR ligand proteolysis and activation. Furthermore, adult hepatocytes do not undergo EMT processes in response to TGF-beta, which might be due, at least in part, to the fact that F-actin re-organization induced by TGF-beta in FaO cells require the EGFR pathway. Finally, results indicate that EGFR transactivation does not block TGF-beta-induced cell cycle arrest in FaO cells, but must be interfering with the pro-apoptotic signalling. In conclusion, TGF-beta is a suppressor factor for adult quiescent hepatocytes, but not for hepatoma cells, where it plays a dual role, both suppressing and promoting carcinogenesis.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAM17 , Actinas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Masculino , Mesodermo/citología , Mesodermo/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Activación Transcripcional/efectos de los fármacos , Familia-src Quinasas/metabolismo
11.
Exp Cell Res ; 312(15): 2860-71, 2006 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-16828470

RESUMEN

Transforming growth factor-beta (TGF-beta) induces apoptosis in fetal rat hepatocytes. However, a subpopulation of these cells survives, concomitant with changes in phenotype, reminiscent of an epithelial-mesenchymal transition (EMT). We have previously suggested that EMT might confer cell resistance to apoptosis (Valdés et al., Mol. Cancer Res., 1: 68-78, 2002). However, the molecular mechanisms responsible for this resistance are not explored yet. In this work, we have isolated and subcultured the population of hepatocytes that suffered the EMT process and are resistant to apoptosis (TGF-beta-treated fetal hepatocytes: TbetaT-FH). We prove that they secrete mitogenic and survival factors, as analyzed by the proliferative and survival capacity of conditioned medium. Inhibition of the epidermal growth factor receptor (EGFR) sensitizes TbetaT-FH to die after serum withdrawal. TbetaT-FH expresses high levels of transforming growth factor-alpha (TGF-alpha) and heparin-binding EGF-like growth factor (HB-EGF) and shows constitutive activation of the EGFR pathway. A blocking anti-TGF-alpha antibody restores the capacity of cells to die. TGF-beta, which is expressed by TbetaT-FH, mediates up-regulation of TGF-alpha and HB-EGF expression in those cells. In summary, results suggest that an autocrine loop of TGF-beta confers resistance to apoptosis after an EMT process in hepatocytes, through the increase in the expression of EGFR ligands.


Asunto(s)
Apoptosis , Comunicación Autocrina , Receptores ErbB/metabolismo , Hepatocitos/citología , Hepatocitos/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Comunicación Autocrina/efectos de los fármacos , Células Cultivadas , Células Epiteliales/citología , Células Epiteliales/metabolismo , Receptores ErbB/antagonistas & inhibidores , Humanos , Ligandos , Mesodermo/citología , Mesodermo/metabolismo , Modelos Biológicos , Ratas , Ratas Wistar , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1 , Regulación hacia Arriba
12.
Oncogene ; 24(28): 4580-7, 2005 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-15856020

RESUMEN

Transforming growth factor beta1 (TGF-beta1) belongs to a family of polypeptide factors, whose cytostatic and apoptotic functions help restrain the growth of mammalian cells. Although solid data established the role of TGF-beta's as suppressor factors in tumorigenic processes, in the context of an advanced stage of disease, TGF-beta's could also play a pro-oncogenic role. We have previously shown that TGF-beta1 induces both pro- and anti-apoptotic signals in foetal rat hepatocytes. In this work, we have focused on its anti-apoptotic mechanism. We show that TGF-beta1 activates the epidermal growth factor receptor (EGFR) and phosphorylates c-Src. EGFR is required for Akt activation. Blocking EGFR signalling amplifies the apoptotic response to TGF-beta1. TGF-beta1 induced a rapid activation of the tumour necrosis factor-alpha-converting enzyme (TACE/ADAM (a disintegrin and metalloprotease) 17). Inhibitors of TACE considerably attenuated Akt activation, which suggests that TGF-beta1 activates EGF signalling in hepatocytes by promoting shedding of EGF-like ligands. The activation of c-Src by TGF-beta1 is EGFR dependent and is required for full Akt phosphorylation and cell survival. Inhibition of EGFR does not block the epithelial-mesenchymal transition (EMT) induced by TGF-beta1 in hepatocytes, which indicates that activation of EGFR plays an essential role in impairing apoptosis, but it is dispensable for the EMT process.


Asunto(s)
Receptores ErbB/metabolismo , Hepatocitos/citología , Hepatocitos/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteínas ADAM , Proteína ADAM17 , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Caspasa 3 , Caspasas/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Fosfatidilinositol 3-Quinasa Clase Ib , Células Epiteliales/metabolismo , Hepatocitos/efectos de los fármacos , Isoenzimas/metabolismo , Hígado/citología , Hígado/embriología , Mesodermo/metabolismo , Metaloendopeptidasas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas pp60(c-src)/efectos de los fármacos , Ratas , Ratas Wistar , Transducción de Señal , Factor de Crecimiento Transformador beta/farmacología , Factor de Crecimiento Transformador beta1 , Factor de Necrosis Tumoral alfa/metabolismo
13.
Free Radic Biol Med ; 36(1): 16-26, 2004 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-14732287

RESUMEN

Transforming growth factor-beta (TGF-beta) induces an oxidative stress process in hepatocytes that mediates its apoptotic activity. To determine the cellular source of the early reactive oxygen species (ROS) generated by fetal rat hepatocytes in response to TGF-beta, we used inhibitors that block different ROS-producing systems. Diphenyleneiodonium, which inhibits NADPH oxidase and other flavoproteins, completely blocked the increase in ROS induced by TGF-beta, coincidently with an impairment of caspase-3 activation and cell death. Rotenone, an inhibitor of the NADH dehydrogenase in mitochondrial complex I, attenuated, but did not completely inhibit, ROS-production, caspase activation, and cell death mediated by TGF-beta. No significant protection was observed with inhibitors of other ROS-producing systems, such as cytochrome P450 (metyrapone), cyclooxygenase (indomethacin), and xanthine oxidase (allopurinol). Additional experiments have indicated that two different mechanisms could be involved in the early ROS production by TGF-beta. First, an inducible (cycloheximide-inhibited) NADPH oxidase-like system could account for the extramitochondrial production of ROS. Second, TGF-beta could increase ROS by a rapid downregulation of antioxidant genes. In particular, intramitochondrial ROS would increase by depletion of MnSOD. Finally, glutathione depletion is a late event and it would be more the consequence than the cause of the increase in ROS induced by TGF-beta.


Asunto(s)
Apoptosis/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Animales , Caspasa 3 , Caspasas/metabolismo , Catalasa/genética , Células Cultivadas , Glutatión/metabolismo , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , Hígado/citología , Hígado/efectos de los fármacos , Hígado/embriología , Microscopía Confocal , NADPH Oxidasas/metabolismo , Compuestos Onio/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Rotenona/farmacología , Superóxido Dismutasa/genética , Factores de Tiempo , alfa-Tocoferol/farmacología
14.
Exp Cell Res ; 292(1): 209-18, 2004 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-14720520

RESUMEN

Transforming growth factor-beta (TGF-beta) induces apoptosis in fetal rat hepatocytes. However, a subpopulation of these cells survives concomitant with changes in morphology and phenotype, reminiscent of an epithelial mesenchymal transition (EMT) [Exp. Cell Res. 252 (1999) 281-291]. In this work, we have isolated the subpopulation that survives to TGF-beta-induced apoptosis, showing that these cells maintain the response to TGF-beta in terms of Smads activation and growth inhibition. Analyses of the intracellular signals that could impair the apoptotic effects of TGF-beta have indicated that the phosphatidylinositol 3-kinase (PI 3-K)/Akt pathway is activated in these resistant cells. Experiments in fetal rat hepatocytes have shown that TGF-beta is able to transiently activate PI 3-K/Akt by a mechanism independent of protein synthesis but dependent on a tyrosine kinase activity. Pro-apoptotic signals, such as oxidative stress and caspases, contribute to the loss of Akt at later times. Inhibiting PI 3-K sensitizes fetal hepatocytes (FH) to the apoptosis induced by TGF-beta and causes spontaneous death in the resistant cells. In conclusion, TGF-beta, as it is known for other cytokines, could be inducing pro-apoptotic and survival signals in hepatocytes, the balance among them will decide cell fate.


Asunto(s)
Apoptosis/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Proteínas Serina-Treonina Quinasas , Transducción de Señal , Factor de Crecimiento Transformador beta/farmacología , Animales , Caspasa 3 , Caspasas/metabolismo , Supervivencia Celular , Células Cultivadas , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Femenino , Feto , Hepatocitos/citología , Hepatocitos/fisiología , Humanos , Estrés Oxidativo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Embarazo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...