Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 200
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Curr Opin Cell Biol ; 2(2): 285-95, 1990 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-2114126

RESUMEN

The study of oncogenes offers insights into many steps in signal transduction. Rapid progress is possible because of the combination of biochemistry and genetics--unique in vertebrate cell biology--the availability of specific clones and antibodies, sequence information, dominant mutants, and biochemical assays of function. The wealth of detail on oncogenes and proto-oncogenes continues to increase dramatically. Hopefully, in the next year or two some of the gaps will be filled in and all the steps along at least one pathway from the cell membrane to the nucleus will be understood.


Asunto(s)
Transformación Celular Neoplásica/genética , Oncogenes/fisiología , Animales , Núcleo Celular/metabolismo , Proteínas de Unión al GTP/fisiología , Proteínas Activadoras de GTPasa , Humanos , Meiosis/fisiología , Proteínas de Neoplasias/fisiología , Proteína Oncogénica p21(ras)/fisiología , Proteínas Tirosina Quinasas/fisiología , Proteínas/fisiología , Transducción de Señal/fisiología , Proteínas Activadoras de ras GTPasa
2.
Nat Cell Biol ; 6(9): 872-83, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15322553

RESUMEN

Vascular proliferative disorders, such as atherosclerosis and restenosis, are the most common causes of severe cardiovascular diseases, but a common molecular mechanism remains elusive. Here, we identify and characterize a novel hyperplasia suppressor gene, named HSG (later re-named rat mitofusin-2). HSG expression was markedly reduced in hyper-proliferative vascular smooth muscle cells (VSMCs) from spontaneously hypertensive rat arteries, balloon-injured Wistar Kyoto rat arteries, or ApoE-knockout mouse atherosclerotic arteries. Overexpression of HSG overtly suppressed serum-evoked VSMC proliferation in culture, and blocked balloon injury induced neointimal VSMC proliferation and restenosis in rat carotid arteries. The HSG anti-proliferative effect was mediated by inhibition of ERK/MAPK signalling and subsequent cell-cycle arrest. Deletion of the p21(ras) signature motif, but not the mitochondrial targeting domain, abolished HSG-induced growth arrest, indicating that rHSG-induced anti-proliferation was independent of mitochondrial fusion. Thus, rHSG functions as a cell proliferation suppressor, whereas dysregulation of rHSG results in proliferative disorders.


Asunto(s)
Enfermedades Cardiovasculares/etiología , Proteínas de la Membrana/fisiología , Proteínas Mitocondriales/fisiología , Músculo Liso Vascular/patología , Animales , Arteriosclerosis/etiología , Enfermedades Cardiovasculares/patología , División Celular , Reestenosis Coronaria/etiología , GTP Fosfohidrolasas , Regulación de la Expresión Génica , Humanos , Sistema de Señalización de MAP Quinasas , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Proteínas Mitocondriales/biosíntesis , Proteínas Mitocondriales/metabolismo , Datos de Secuencia Molecular , Miocitos del Músculo Liso/patología , Proteína Oncogénica p21(ras)/metabolismo , Proteína Oncogénica p21(ras)/fisiología , Ratas
3.
J Proteome Res ; 9(2): 1007-19, 2010 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-19954229

RESUMEN

Epithelial-mesenchymal transition (EMT) describes a process whereby immotile epithelial cells escape structural constraints imposed by cellular architecture and acquire a phenotype characteristic of migratory mesenchymal cells. Implicated in carcinoma progression and metastasis, EMT has been the focus of several recent proteomics-based studies aimed at identifying new molecular players. To gain insights into extracellular mediators associated with EMT, we conducted an extensive proteomic analysis of the secretome from MDCK cells following oncogenic Ras-induced EMT (21D1 cells). Using Orbitrap technology and a label-free quantitative approach, differential expression of several secreted modulators were revealed. Proteomic findings were further substantiated by mRNA transcript expression analysis with 71% concordance. MDCK cells undergoing Ras-induced EMT remodel the extracellular matrix (ECM) via diminished expression of basement membrane constituents (collagen type IV and laminin 5), up-regulation of extracellular proteases (MMP-1, kallikreins -6 and -7), and increased production and secretion of ECM constituents (SPARC, collagen type I, fibulins -1 and -3, biglycan, and decorin). Collectively, these findings suggest that hierarchical regulation of a subset of extracellular effectors may coordinate a biological response during EMT that enhances cell motility. Transient silencing of MMP-1 in 21D1 cells via siRNA-mediated knockdown attenuated cell migration. Many of the secretome proteins identified broaden our understanding of the EMT process.


Asunto(s)
Movimiento Celular , Células Epiteliales/citología , Matriz Extracelular/metabolismo , Mesodermo/citología , Proteína Oncogénica p21(ras)/fisiología , Animales , Transformación Celular Neoplásica , Células Cultivadas , Perros , Perfilación de la Expresión Génica , Microscopía Confocal , Microscopía Fluorescente , Proteína Oncogénica p21(ras)/genética , ARN Mensajero/genética
4.
Hum Cell ; 33(4): 1186-1196, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32700262

RESUMEN

Pancreatic cancer is the fourth most common lethal malignancy with an overall 5-year survival rate of less than 5%. ERas, a novel Ras family member, was first identified in murine embryonic stem cells and is upregulated in various cancers. However, the expression and potential role of ERas in pancreatic cancer have not been investigated. In this study, we found that ERas mRNA and protein were upregulated in pancreatic cancer tissues and cells compared with controls. Knockdown of ERas in pancreatic cancer cells by siRNA significantly decreased cell proliferation, colony formation, migration, and invasion and promoted cell apoptosis in vitro. Epithelial-mesenchymal transition (EMT) is closely related to tumor progression. We observed a significant decrease in N-cadherin expression in pancreatic cancer cells in response to ERas gene silencing by immunofluorescence assay and western blot. Furthermore, tumor growth and EMT were inhibited in xenografts derived from pancreatic cancer cells with ERas downregulation. We further investigated the regulatory mechanisms of ERas in pancreatic cancer and found that ERas may activate the Erk/Akt signaling pathway. Moreover, Erk inhibitor decreased pancreatic cancer cells proliferation and colony formation activities. Our data suggest that targeting ERas and its relevant signaling pathways might represent a novel therapeutic approach for the treatment of pancreatic cancer.


Asunto(s)
Proliferación Celular/genética , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica/genética , Sistema de Señalización de MAP Quinasas/genética , Proteína Oncogénica p21(ras)/genética , Proteína Oncogénica p21(ras)/fisiología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/genética , Animales , Línea Celular Tumoral , Movimiento Celular/genética , Progresión de la Enfermedad , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones Desnudos , Terapia Molecular Dirigida , Invasividad Neoplásica/genética , Proteína Oncogénica p21(ras)/metabolismo , Neoplasias Pancreáticas/terapia , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/uso terapéutico , Transducción de Señal/fisiología , Regulación hacia Arriba/genética
5.
J Cell Biol ; 121(3): 665-72, 1993 May.
Artículo en Inglés | MEDLINE | ID: mdl-8486743

RESUMEN

Little is known about the signal transduction mechanisms involved in the response to neurotrophins and other neurotrophic factors in neurons, beyond the activation of the tyrosine kinase activity of the neurotrophin receptors belonging to the trk family. We have previously shown that the introduction of the oncogene product ras p21 into the cytoplasm of chick embryonic neurons can reproduce the survival and neurite-outgrowth promoting effects of the neurotrophins nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF), and of ciliary neurotrophic factor (CNTF). To assess the potential signal-transducing role of endogenous ras p21, we introduced function-blocking anti-ras antibodies or their Fab fragments into cultured chick embryonic neurons. The BDNF-induced neurite outgrowth in E12 nodose ganglion neurons was reduced to below control levels, and the NGF-induced survival of E9 dorsal root ganglion (DRG) neurons was inhibited in a specific and dose-dependent fashion. Both effects could be reversed by saturating the epitope-binding sites with biologically inactive ras p21 before microinjection. Surprisingly, ras p21 did not promote the survival of NGF-dependent E12 chick sympathetic neurons, and the NGF-induced survival in these cells was not inhibited by the Fab-fragments. The survival effect of CNTF on ras-responsive ciliary neurons could not be blocked by anti-ras Fab fragments. These results indicate an involvement of ras p21 in the signal transduction of neurotrophic factors in sensory, but not sympathetic or ciliary neurons, pointing to the existence of different signaling pathways not only in CNTF-responsive, but also in neurotrophin-responsive neuronal populations.


Asunto(s)
Ganglios Simpáticos/fisiología , Factores de Crecimiento Nervioso/fisiología , Neuronas Aferentes/fisiología , Ganglio Nudoso/fisiología , Proteína Oncogénica p21(ras)/fisiología , Animales , Anticuerpos Monoclonales/farmacología , Factor Neurotrófico Derivado del Encéfalo , Células Cultivadas/efectos de los fármacos , Embrión de Pollo , Factor Neurotrófico Ciliar , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Ratas , Transducción de Señal/efectos de los fármacos
6.
Science ; 249(4969): 635-40, 1990 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-2116664

RESUMEN

A substantial number of novel guanine nucleotide binding regulatory proteins have been identified over the last few years but the function of many of them is largely unknown. This article will discuss a particular family of these proteins, structurally related to the Ras oncoprotein. Approximately 30 Ras-related small guanosine triphosphate (GTP)-binding proteins are known, and from yeast to man they appear to be involved in controlling a diverse set of essential cellular functions including growth, differentiation, cytoskeletal organization, and intracellular vesicle transport and secretion.


Asunto(s)
Proteínas de Unión al GTP/fisiología , Genes ras , Proteína Oncogénica p21(ras)/fisiología , Proteínas ras , Animales , Proteínas Fúngicas/metabolismo , Regulación de la Expresión Génica , Modelos Biológicos
7.
Mol Cell Biol ; 26(22): 8252-66, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16966377

RESUMEN

Oncogene-mediated premature senescence has emerged as a potential tumor-suppressive mechanism in early cancer transitions. Previous work shows that RAS and p38 MAPK participate in premature senescence, but transcriptional effectors have not been identified. Here, we demonstrate that the HBP1 transcriptional repressor participates in RAS- and p38 MAPK-induced premature senescence. In cell lines, we had previously isolated HBP1 as a retinoblastoma (RB) target but have determined that it functions as a proliferation regulator by inhibiting oncogenic pathways as a transcriptional repressor. In primary cells, the results indicate that HBP1 is a necessary component of premature senescence by RAS and p38 MAPK. Similarly, a knockdown of WIP1 (a p38 MAPK phosphatase) induced premature senescence that also required HBP1. Furthermore, HBP1 requires regulation by RB, in which few transcriptional regulators for premature senescence have been shown. Together, the data suggest a model in which RAS and p38 MAPK signaling engage HBP1 and RB to trigger premature senescence. As an initial step toward clinical relevance, a bioinformatics approach shows that the relative expression levels of HBP1 and WIP1 correlated with decreased relapse-free survival in breast cancer patients. Together, these studies highlight p38 MAPK, HBP1, and RB as important components for a premature-senescence pathway with possible clinical relevance to breast cancer.


Asunto(s)
Neoplasias de la Mama/diagnóstico , Senescencia Celular , Proteínas del Grupo de Alta Movilidad/fisiología , Proteína Oncogénica p21(ras)/fisiología , Proteínas Represoras/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Sitios de Unión , Línea Celular , Replicación del ADN , Bases de Datos Genéticas , Humanos , Modelos Biológicos , Fosfoproteínas Fosfatasas/metabolismo , Proteína Fosfatasa 2C , Proteína de Retinoblastoma/metabolismo , Proteína de Retinoblastoma/fisiología , Transducción de Señal , Transfección
8.
Oncogene ; 26(28): 4124-34, 2007 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-17213803

RESUMEN

Reovirus shows considerable potential as an oncolytic agent for Ras-activated tumors and is currently in clinical trials. Here we ask whether such tumor cell lines can acquire resistance to reoviral oncolysis. We challenged human HT1080 fibrosarcoma cells that carry a Ras mutation by prolonged exposure to reovirus, thereby yielding highly virus-resistant HTR1 cells. These cells are persistently infected with reovirus, exhibit high Ras activity and retain the original Ras gene mutation, showing that resistance to reovirus can be displayed in cells with active Ras. The HTR1 cells also exhibit reduced cellular cathepsin B activity, which normally contributes to viral entry and activation. Persistently infected HTR1 cells were not tumorigenic in vivo, whereas immunologically cured virus-free HTR1 cells were highly tumorigenic. Thus, acquisition of resistance to reovirus may constrain therapeutic strategies. To determine whether reoviral resistance is associated with a general reduction in apoptotic potential, we challenged the HTR1 cells with apoptotic inducers and E1B-defective adenovirus, resulting in significant apoptosis and cell death following both approaches. Therefore, even if resistance to reoviral oncolysis should arise in tumor cells in vivo, other therapeutic strategies may nevertheless remain effective.


Asunto(s)
Fibrosarcoma/patología , Proteína Oncogénica p21(ras)/fisiología , Reoviridae/fisiología , Secuencia de Bases , Catepsina B/metabolismo , Línea Celular , Línea Celular Tumoral , Cartilla de ADN , Fibrosarcoma/virología , Humanos , Mutación , Proteína Oncogénica p21(ras)/genética
9.
Neuron ; 2(1): 1087-96, 1989 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-2696501

RESUMEN

Although evidence obtained with the PC12 cell line has suggested a role for the ras oncogene proteins in the signal transduction of nerve growth factor-mediated fiber outgrowth, little is known about the signal transduction mechanisms involved in the neuronal response to neurotrophic factors in nontransformed cells. We report here that the oncogene protein T24-ras, when introduced into the cytoplasm of freshly dissociated chick embryonic neurons, promotes the in vitro survival and neurite outgrowth of nerve growth factor-responsive dorsal root ganglion neurons, brain-derived neurotrophic factor-responsive nodose ganglion neurons, and ciliary neuronotrophic factor-responsive ciliary ganglion neurons. The proto-oncogene product c-Ha-ras also promotes neuronal survival, albeit less strongly. No effect could be observed with truncated counterparts of T24-ras and c-Ha-ras lacking the 23 C-terminal amino acids including the membrane-anchoring, palmityl-accepting cysteine. These results suggest a generalized involvement of ras or ras-like proteins in the intracellular signal transduction pathway for neurotrophic factors.


Asunto(s)
Axones/ultraestructura , Ganglios Parasimpáticos/citología , Ganglios Espinales/citología , Neuronas/citología , Ganglio Nudoso/citología , Proteína Oncogénica p21(ras)/fisiología , Nervio Vago/citología , Neoplasias de las Glándulas Suprarrenales , Animales , Axones/fisiología , Línea Celular , Supervivencia Celular , Células Cultivadas , Embrión de Pollo , Genes ras , Neuronas/fisiología , Proteína Oncogénica p21(ras)/genética , Feocromocitoma
10.
FASEB J ; 21(9): 2086-100, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17351128

RESUMEN

Prostate cancer is one of the most common cancers in men, with more than 500,000 new worldwide cases reported annually, resulting in 200,000 deaths of mainly older men in developed countries. Existing treatments have not proved very effective in managing prostate cancer, and continuing efforts therefore are ongoing to explore novel targets and strategies for future therapies. LAPSER1 has been identified as a candidate tumor suppressor gene in prostate cancer, but its true functions remain unknown. We report here that LAPSER1 colocalizes to the centrosomes and midbodies in mitotic cells with gamma-tubulin, MKLP1, and p80 katanin, and is involved in cytokinesis. Moreover, RNAi-mediated disruption of LAPSER1, which is accompanied by the mislocalization of p80 katanin, results in malformation of the central spindle. Significantly, the enhanced expression of LAPSER1 induces binucleation and renders the cells resistant to oncogenic transformation. In cells transformed by the v-Fps oncogene, overexpressed LAPSER1 induces abortive cytokinesis, followed by mitotic catastrophe in a p80 katanin-dependent manner. Cells that are rescued from this apoptotic pathway with Z-VAD-fmk display karyokinesis. These results suggest that LAPSER1 participates in cytokinesis by interacting with p80 katanin, the disruption of which may potentially cause genetic instability and cancer.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Centrosoma/química , Citocinesis/fisiología , Genes Supresores de Tumor , Proteínas de la Membrana/fisiología , Proteínas Supresoras de Tumor/fisiología , Adenocarcinoma/patología , Adenosina Trifosfatasas/química , Clorometilcetonas de Aminoácidos/farmacología , Animales , Apoptosis , Neoplasias Óseas/patología , Células CHO , Línea Celular , Línea Celular Transformada , Línea Celular Tumoral/química , Línea Celular Tumoral/ultraestructura , Transformación Celular Viral , Centrosoma/ultraestructura , Cricetinae , Cricetulus , Proteínas de Fusión gag-onc/fisiología , Humanos , Katanina , Leucina Zippers , Masculino , Proteínas de la Membrana/análisis , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas Asociadas a Microtúbulos/análisis , Proteína Oncogénica p21(ras)/fisiología , Proteínas Oncogénicas v-abl/fisiología , Osteosarcoma/patología , Poliploidía , Neoplasias de la Próstata/patología , Subunidades de Proteína , Proteínas Tirosina Quinasas/fisiología , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Ratas , Proteínas Recombinantes de Fusión/fisiología , Huso Acromático/ultraestructura , Fracciones Subcelulares/química , Tubulina (Proteína)/análisis , Proteínas Supresoras de Tumor/análisis , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/genética
11.
Cancer Chemother Pharmacol ; 62(3): 491-8, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18066549

RESUMEN

PURPOSE: We investigated the effects of two peptides from the ras-p21 protein, corresponding to residues 35-47 (PNC-7) and 96-110 (PNC-2), on two ras-transformed human cancer cell lines, HT1080 fibrosarcoma and MIAPaCa-2 pancreatic cancer cell lines. In prior studies, we found that both peptides block oncogenic, but not insulin-activated wild-type, ras-p21-induced oocyte maturation. When linked to a transporter penetratin peptide, these peptides induce reversion of ras-transformed rat pancreatic cancer cells (TUC-3) to the untransformed phenotype. METHODS: These peptides and a control peptide, linked to a penetratin peptide, were incubated with each cell lines. Cell counts were obtained over several weeks. The cause of cell death was determined by measuring caspase as an indicator of apoptosis and lactate dehydrogenase (LDH) as marker of necrosis. Since both peptides block the phosphorylation of jun-N-terminal kinase (JNK) in oocytes, we blotted cell lysates of the two cancer cell lines for the levels of phosphorylated JNK to determine if the peptides reduced these levels. RESULTS: We find that both peptides, but not control peptides linked to the penetratin sequence, induce phenotypic reversion of the HT-1080 cell line but cause tumor cell necrosis of the MIA-PaCa-2 cell line. On the other hand, neither peptide has any effect on the viability of an untransformed pancreatic acinar cell line, BMRPA1. We find that, while total JNK levels remain constant during peptide treatment, phosphorylated JNK levels decrease dramatically, consistent with the mechanisms of action of these peptides. CONCLUSION: We conclude that these peptides block tumor but not normal cell growth likely by blocking oncogenic ras-p21-induced phosphorylation of JNK, an essential step on the oncogenic ras-p21-protein pathway. These peptides are therefore promising as possible anti-tumor agents.


Asunto(s)
Antineoplásicos/farmacocinética , Transformación Celular Neoplásica/efectos de los fármacos , Proteína Oncogénica p21(ras)/fisiología , Fragmentos de Péptidos/farmacología , Antineoplásicos/química , Caspasas/biosíntesis , Técnicas de Cultivo de Célula , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/biosíntesis , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Necrosis , Proteína Oncogénica p21(ras)/química , Fragmentos de Péptidos/química , Fosforilación
12.
Tohoku J Exp Med ; 216(1): 25-34, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18719335

RESUMEN

Survivin, a member of the inhibitor of apoptosis protein (IAP) family, has been widely studied because of its aberrant expression in human cancer. Survivin has multiple functions, including cell-cycle regulation at mitosis, inhibition of apoptosis and caspase-independent cytoprotection. Clinical studies have shown that survivin is associated with resistance to treatment and its expression is linked to poor prognosis. Recent studies indicated that Ras pathways up-regulate survivin expression in hematopoietic cells. Here we analyzed downstream pathways of Ras in interleukin-3 (IL-3)-dependent Baf-3 murine-derived pro-B lymphocytic cells that express constitutively active Ras mutants, using signaling pathway-specific inhibitors. Both mitogen-activated protein kinase (MAPK) and phosphatidylinositol-3 kinase (PI3-K) pathways are involved in the induction of survivin. Downstream of PI3-K, the signaling pathway is composed of two kinases, Akt and mammalian target of rapamycin (mTOR) pathways. In the downstream targets of PI3-K, mTOR but not Akt is responsible for survivin expression. Using a counterflow centrifugal elutriator, we observed G2/M phase-dominant survivin expression in Baf-3 cells. Interestingly, constitutively active Ras mutants also induced survivin in a cell cycle-dependent manner. Reporter assays of the survivin gene promoter revealed a transcriptional regulatory cis-acting region that is responsible for Ras signaling, indicating that Ras increases the transcription of the survivin gene through specific enhancer elements. These data illustrate the pathways regulating survivin expression by Ras. Ras activates the MAPK, PI3-K and mTOR pathways, and these signals enhance survivin transcription. Our data will provide the new information about mechanisms of survivin expression by Ras-signalling pathways.


Asunto(s)
Linfocitos B/metabolismo , Regulación de la Expresión Génica , Genes ras , Proteínas Asociadas a Microtúbulos/biosíntesis , Proteína Oncogénica p21(ras)/fisiología , Transducción de Señal/fisiología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Linfocitos B/efectos de los fármacos , Proteínas Portadoras/fisiología , Ciclo Celular/fisiología , Línea Celular/efectos de los fármacos , Línea Celular/metabolismo , Cromonas/farmacología , Elementos de Facilitación Genéticos , Flavonoides/farmacología , Humanos , Proteínas Inhibidoras de la Apoptosis , Interleucina-3/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Proteínas Asociadas a Microtúbulos/genética , Morfolinas/farmacología , Proteína Oncogénica p21(ras)/genética , Fosfatidilinositol 3-Quinasas/fisiología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Mutación Puntual , Proteínas Proto-Oncogénicas c-akt/fisiología , Proteínas Recombinantes de Fusión/fisiología , Proteínas Represoras , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Survivin , Serina-Treonina Quinasas TOR , Transfección
13.
Oncogene ; 37(12): 1594-1609, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29326437

RESUMEN

Personalized medicine for cancer patients requires a deep understanding of the underlying genetics that drive cancer and the subsequent identification of predictive biomarkers. To discover new genes and pathways contributing to oncogenesis and therapy resistance in HER2+ breast cancer, we performed Mouse Mammary Tumor Virus (MMTV)-induced insertional mutagenesis screens in ErbB2/cNeu-transgenic mouse models. The screens revealed 34 common integration sites (CIS) in mammary tumors of MMTV-infected mice, highlighting loci with multiple independent MMTV integrations in which potential oncogenes are activated, most of which had never been reported as MMTV CIS. The CIS most strongly associated with the ErbB2-transgenic genotype was the locus containing Eras (ES cell-expressed Ras), a constitutively active RAS-family GTPase. We show that upon expression, Eras acts as a potent oncogenic driver through hyperactivation of the PI3K/AKT pathway, in contrast to other RAS proteins that signal primarily via the MAPK/ERK pathway and require upstream activation or activating mutations to induce signaling. We additionally show that ERAS synergistically enhances HER2-induced tumorigenesis and, in this role, can functionally replace ERBB3/HER3 by acting as a more powerful activator of PI3K/AKT signaling. Although previously reported as pseudogene in humans, we observed ERAS RNA and protein expression in a substantial subset of human primary breast carcinomas. Importantly, we show that ERAS induces primary resistance to the widely used HER2-targeting drugs Trastuzumab (Herceptin) and Lapatinib (Tykerb/Tyverb) in vivo, and is involved in acquired resistance via selective upregulation during treatment in vitro, indicating that ERAS may serve as a novel clinical biomarker for PI3K/AKT pathway hyperactivation and HER2-targeted therapy resistance.


Asunto(s)
Neoplasias de la Mama/patología , Transformación Celular Neoplásica/genética , Resistencia a Antineoplásicos/genética , Neoplasias Mamarias Experimentales/patología , Mutagénesis Insercional/fisiología , Proteína Oncogénica p21(ras)/fisiología , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Células Cultivadas , Femenino , Humanos , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ratones Transgénicos , Proteína Oncogénica p21(ras)/genética , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo
14.
Oncogene ; 25(10): 1485-95, 2006 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-16278678

RESUMEN

Heat shock proteins (Hsps) are overexpressed in many tumors, but are downregulated in some tumors. To check for a direct effect of Ha-Ras(val12) on HSP70 transcription, we transiently expressed the oncoprotein in Rat1 fibroblasts and monitored its effect on HSP70b promoter-driven reporter gene. We show that expression of Ha-Ras(val12) induced this promoter. Promoter analysis via systematic deletions and point mutations revealed that Ha-Ras(val12) induces HSP70b transcription via heat shock elements (HSEs). Also, Ha-Ras(val12) induction of HSE-mediated transcription was dramatically reduced in HSF1-/- cells. Yet, residual effect of Ha-Ras(val12) that was still measured in HSF1-/- cells suggests that some of the Ha-Ras(val12) effect is Hsf1-independent. When HSF1-/- cells, stably expressing Ha-Ras(val12), were grown on soft agar only small colonies were formed suggesting a role for heat shock factor 1 (Hsf1) in Ha-Ras(val12)-mediated transformation. Although Ha-ras(Val12) seems to be an inducer of HSP70's expression, we found that in Ha-ras(Val12-)transformed fibroblasts expression of this gene is suppressed. This suppression is correlated with higher sensitivity of Ha-ras(val12)-transformed cells to heat shock. We suggest that Ha-ras(Val12) is involved in Hsf1 activation, thereby inducing the cellular protective response. Cells that repress this response are perhaps those that acquire the capability to further proliferate and become transformed clones.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Regulación de la Expresión Génica/fisiología , Proteínas HSP70 de Choque Térmico/biosíntesis , Proteínas HSP70 de Choque Térmico/genética , Proteína Oncogénica p21(ras)/fisiología , Factores de Transcripción/fisiología , Transcripción Genética , Transporte Activo de Núcleo Celular , Animales , Línea Celular Transformada , Genes Reporteros , Proteínas HSP70 de Choque Térmico/antagonistas & inhibidores , Células HeLa , Factores de Transcripción del Choque Térmico , Humanos , Ratones , Ratones Desnudos , Células 3T3 NIH , Oxidación-Reducción , Fosforilación , Ratas
15.
Endocrinology ; 148(6): 2806-14, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17332061

RESUMEN

GnRH neurons migrate into the hypothalamus during development. Although migratory defects may result in disordered activation of the reproductive axis and lead to delayed or absent sexual maturation, specific factors regulating GnRH neuronal migration remain largely unknown. The receptor tyrosine kinase, adhesion-related kinase (Ark) (also known as Axl, UFO, and Tyro7), has been implicated in the migration of GnRH neuronal cells. Binding of its ligand, growth arrest-specific gene 6 (Gas6), promotes cytoskeletal remodeling and migration of NLT GnRH neuronal cells via Rac and p38 MAPK. Here, we examined the Axl effectors proximal to Rac in the signaling pathway. Gas6/Axl-induced lamellipodia formation and migration were blocked after phosphatidylinositol-3-kinase (PI3K) inhibition in GnRH neuronal cells. The p85 subunit of PI3K coimmunoprecipitated with Axl and was phosphorylated in a Gas6-sensitive manner. In addition, PI3K inhibition in GnRH neuronal cells diminished Gas6-induced Rac activation. Exogenous expression of a dominant-negative form of Ras also decreased GnRH neuronal lamellipodia formation, migration, and Rac activation. PI3K inhibition blocked Ras in addition to Rac activation and migration. In contrast, pharmacological blockade of the phospholipase C gamma effectors, protein kinase C or calcium/calmodulin protein kinase II, had no effect on Gas6/Axl signaling to promote Rac activation or stimulate cytoskeletal reorganization and migration. Together, these data show that the PI3K-Ras pathway is a major mediator of Axl actions upstream of Rac to induce GnRH neuronal cell migration.


Asunto(s)
Movimiento Celular/fisiología , Hormona Liberadora de Gonadotropina/metabolismo , Neuronas/metabolismo , Proteína Oncogénica p21(ras)/fisiología , Proteínas Oncogénicas/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Tirosina Quinasas Receptoras/fisiología , Animales , Línea Celular Transformada , Citoesqueleto/metabolismo , Péptidos y Proteínas de Señalización Intercelular/fisiología , Ratones , Modelos Biológicos , Proteínas Oncogénicas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas , Proteínas Tirosina Quinasas Receptoras/metabolismo , Tirosina Quinasa del Receptor Axl
16.
Mol Cell Biol ; 10(1): 310-5, 1990 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-2152963

RESUMEN

Treatment with insulin or progesterone or microinjection of the transforming protein product of Ha-rasVal-12,Thr-59 (p21) is known to induce germinal vesicle breakdown in Xenopus oocytes. We have investigated the effect of p21 on S6 kinase and the H1 histone kinase of maturation-promoting factor in the presence and absence of antisense oligonucleotides against the c-mosxe proto-oncogene. Injection of p21 led to a rapid increase in S6 phosphorylation, with kinetics similar to those previously observed with insulin. Microinjection of c-mosxe antisense oligonucleotides inhibited germinal vesicle breakdown induced by p21 and totally abolished S6 kinase activation by insulin or progesterone but only partially inhibited activation by p21. However, the activation of p34cdc2 protein kinase by all three stimuli was blocked by antisense oligonucleotides. The results suggest that in oocyte maturation c-mosxe functions downstream of p21 but upstream of p34cdc2 and S6 kinase activation, although not all p21-induced events require c-mosxe.


Asunto(s)
Sustancias de Crecimiento/metabolismo , Proteína Oncogénica p21(ras)/fisiología , Oocitos/fisiología , Protamina Quinasa/metabolismo , Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas/fisiología , Proteínas Ribosómicas/metabolismo , Animales , Activación Enzimática , Insulina/farmacología , Factor Promotor de Maduración , Oligonucleótidos , Oligonucleótidos Antisentido , Progesterona/farmacología , Proteínas Proto-Oncogénicas c-mos , Proteína S6 Ribosómica , Proteínas Quinasas S6 Ribosómicas , Xenopus laevis
17.
Mol Cell Biol ; 10(10): 5314-23, 1990 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-2118993

RESUMEN

We used a dominant inhibitory mutation of c-Ha-ras which changes Ser-17 to Asn-17 in the gene product p21 [p21(Asn-17)Ha-ras] to investigate ras function in mitogenic signal transduction. An NIH 3T3 cell line [NIH(M17)] was isolated that displayed inducible expression of the mutant Ha-ras gene (Ha-ras Asn-17) via the mouse mammary tumor virus long terminal repeat and was growth inhibited by dexamethasone. The effect of dexamethasone induction on response of quiescent NIH(M17) cells to mitogens was then analyzed. Stimulation of DNA synthesis by epidermal growth factor (EGF) and 12-O-tetradecanoylphorbol-13-acetate (TPA) was completely blocked by p21(Asn-17) expression, and stimulation by serum, fibroblast growth factor, and platelet-derived growth factor was partially inhibited. However, the induction of fos, jun, and myc by EGF and TPA was not significantly inhibited in this cell line. An effect of p21(Asn-17) on fos induction was, however, demonstrated in transient expression assays in which quiescent NIH 3T3 cells were cotransfected with a fos-cat receptor plasmid plus a Ha-ras Asn-17 expression vector. In this assay, p21(Asn-17) inhibited chloramphenicol acetyltransferase expression induced by EGF and other growth factors. In contrast to its effect on DNA synthesis, however, Ha-ras Asn-17 expression did not inhibit fos-cat expression induced by TPA. Conversely, downregulation of protein kinase C did not inhibit fos-cat induction by activated ras or other oncogenes. These results suggest that ras proteins are involved in at least two parallel mitogenic signal transduction pathways, one of which is independent of protein kinase C. Although either pathway alone appears to be sufficient to induce fos, both appear to be necessary to induce the full mitogenic response.


Asunto(s)
Mitógenos/farmacología , Proteína Oncogénica p21(ras)/fisiología , Animales , Línea Celular , ADN/biosíntesis , Proteínas de Unión al ADN/genética , Genes Dominantes , Sustancias de Crecimiento/farmacología , Ratones , Mutación , Proteínas Nucleares/genética , Oncogenes , Proteína Quinasa C/fisiología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-fos , Proteínas Proto-Oncogénicas c-jun , Proteínas Proto-Oncogénicas c-myc , Transducción de Señal , Acetato de Tetradecanoilforbol/farmacología , Factores de Transcripción/genética , Transfección
18.
Mol Cell Biol ; 15(10): 5531-41, 1995 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-7565705

RESUMEN

BCR-ABL is a deregulated tyrosine kinase expressed in Philadelphia chromosome-positive human leukemias. Prolongation of hematopoietic cell survival by inhibition of apoptosis has been proposed to be an integral component of BCR-ABL-induced chronic myelogenous leukemia. BCR-ABL elicits transformation of both fibroblast and hematopoietic cells and blocks apoptosis following cytokine deprivation in various factor-dependent cells. To elucidate the mechanisms whereby BCR-ABL induces transformation and blocks apoptosis in hematopoietic cells, we examined the biological effects of expression of a series of BCR-ABL mutants. Single amino acid substitutions in the GRB2 binding site (Y177F), Src homology 2 domain (R552L), or an autophosphorylation site in the tyrosine kinase domain (Y793F) do not diminish the antiapoptotic and transforming properties of BCR-ABL in hematopoietic cells, although these mutations were previously shown to drastically reduce the transforming activity of BCR-ABL in fibroblasts. A BCR-ABL molecule containing all three mutations (Y177F/R552L/Y793F) exhibits a severe decrease in transforming and antiapoptotic activities compared with the wild-type BCR-ABL protein in 32D myeloid progenitor cells. Ras is activated, the SHC adapter protein is tyrosine phosphorylated and binds GRB2, and myc mRNA levels are increased following expression of all kinase active BCR-ABL proteins with the exception of the Y177F/R552L/Y793F BCR-ABL mutant in 32D cells. We propose that BCR-ABL uses multiple pathways to activate Ras in hematopoietic cells and that this activation is necessary for the transforming and antiapoptotic activities of BCR-ABL. However, Ras activation is not sufficient for BCR-ABL-mediated transformation. A BCR-ABL deletion mutant (delta 176-427) that activates Ras and blocks apoptosis but has severely impaired transforming ability in 32D cells has been identified. These data suggest that BCR-ABL requires additional signaling components to elicit tumorigenic growth which are distinct from those required to block apoptosis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Apoptosis/fisiología , Transformación Celular Neoplásica , Proteínas de Fusión bcr-abl/fisiología , Proteína Oncogénica p21(ras)/fisiología , Proteínas Tirosina Quinasas/fisiología , Transducción de Señal/fisiología , Animales , Linfocitos B , Secuencia de Bases , Línea Celular , Células Precursoras Eritroides , Proteínas de Fusión bcr-abl/genética , Proteína Adaptadora GRB2 , Guanosina Trifosfato/metabolismo , Humanos , Ratones , Ratones Desnudos , Datos de Secuencia Molecular , Mutación , Fosforilación , Proteínas Tirosina Quinasas/genética , Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , ARN Mensajero/biosíntesis
19.
Mol Cell Biol ; 9(10): 4312-22, 1989 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-2555688

RESUMEN

Expression of a mutant H-ras gene confers a transformed phenotype to rat-1 fibroblasts which is basically independent of exogenous growth factors (GFs). Rat-1 cells induced to express high levels of the normal H-ras gene were also found to display a transformed phenotype. In contrast to cells expressing mutant H-ras, these cells were dependent on GFs. We used this difference in GF dependence to analyze a possible involvement of exogenous GFs in H-ras function. Compared with untransformed rat-1 cells, cells overexpressing normal H-ras displayed an elevated response toward insulinlike growth factor 1 (IGF-1), insulin, and bombesin and an increased sensitivity toward phosphatidic acids. It was found that 8-bromo-cyclic AMP inhibited the responses to all GFs in rat-1 cells but had no effect on mutant-H-ras-transformed cells. In cells overexpressing normal H-ras, 8-bromo-cyclic AMP inhibited the responses to all GFs except those to insulin and IGF-1. This implies that overexpression of normal H-ras in the presence of insulin/IGF-1 is functionally similar to the expression of mutant H-ras, since mutant H-ras can circumvent this block by itself. These and other results strongly suggest a functional linkage between insulin/IGF-1 and normal p21 H-ras.


Asunto(s)
Sustancias de Crecimiento/fisiología , Proteína Oncogénica p21(ras)/fisiología , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Animales , División Celular/efectos de los fármacos , Línea Celular , Línea Celular Transformada , Transformación Celular Neoplásica/metabolismo , AMP Cíclico/fisiología , Replicación del ADN/efectos de los fármacos , Expresión Génica , Insulina/fisiología , Factor I del Crecimiento Similar a la Insulina/fisiología , Fosforilación , Receptor de Insulina/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores de Somatomedina , Transducción de Señal/fisiología
20.
Mol Cell Biol ; 17(1): 506-18, 1997 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-8972231

RESUMEN

Constitutive activation of mitogen-activated protein kinase (MAPK) is a property common to many oncoproteins, including Mos, Ras, and Raf, and is essential for their transforming activities. We have shown that high levels of expression of the Mos/MAPK pathway in Swiss 3T3 fibroblast cause cells in S phase to undergo apoptosis, while cells in G1 irreversibly growth arrest. Interestingly, cells in G2 and M phases also arrest at a G1-like checkpoint after proceeding through mitosis. These cells fail to undergo cytokinesis and are binucleated. Thus, constitutive overexpression of Mos and MAPK cannot be tolerated, and fibroblasts transformed by Mos express only low levels of the mos oncogene product. Here, we show that p53 plays a key role in preventing oncogene-mediated activation of MAPK. In the absence of p53 (p53-/-), the growth arrest normally observed in wild-type p53 (p53+/+) mouse embryo fibroblasts (MEFs) is markedly reduced. The mos transformation efficiency in p53-/- MEFs is two to three orders of magnitude higher than that in p53+/+ cells, and p53-/- cells tolerate > 10-fold higher levels of both Mos and activated MAPK. Moreover, we show that, like Mos, both v-ras and v-raf oncogene products induce apoptosis in p53+/+ MEFs. These oncogenes also display a high transforming activity in p53-/- MEFs, as does a gain-of-function MAPK kinase mutant (MEK*). Thus, the p53-dependent checkpoint pathway is responsive to oncogene-mediated MAPK activation in inducing irreversible G1 growth arrest and apoptosis. Moreover, we show that the chromosome instability induced by the loss of p53 is greatly enhanced by the constitutive activation of the Mos/MAPK pathway.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Transformación Celular Neoplásica , Cromosomas/genética , Quinasa 1 de Quinasa de Quinasa MAP , Proteínas Proto-Oncogénicas c-mos/fisiología , Transducción de Señal/fisiología , Proteína p53 Supresora de Tumor/fisiología , Células 3T3 , Animales , Apoptosis , Ciclo Celular/fisiología , División Celular , Células Cultivadas , Activación Enzimática , Fibroblastos , Expresión Génica , Ratones , Virus de la Leucemia Murina de Moloney/genética , Virus del Sarcoma Murino de Moloney/genética , Proteína Oncogénica p21(ras)/genética , Proteína Oncogénica p21(ras)/fisiología , Proteínas Oncogénicas v-raf , Oncogenes , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Oncogénicas de Retroviridae/genética , Proteínas Oncogénicas de Retroviridae/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA