Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 200
Filtrar
1.
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
2.
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
3.
Exp Biol Med (Maywood) ; 240(12): 1606-13, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26041389

RESUMEN

Oxidative stress is a main risk factor of vascular aging, which may lead to age-associated diseases. Related transcriptional enhancer factor-1 (RTEF-1) has been suggested to regulate many genes expression which are involved in the endothelial angiogenesis and vasodilation. However, whether RTEF-1 has a direct role in anti-oxidation and what specific genes are involved in RTEF-1-driven anti-oxidation have not been elucidated. In this study, we found that overexpressing RTEF-1 in H2O2-treated human umbilical vein endothelial cells decreased senescence-associated-ß-galactosidase (SA-ß-gal)-positive cells and G0/G1 cells population. The expressions of p53 and p21 were decreased in H2O2-treated RTEF-1 o/e human umbilical vein endothelial cells. However, specific small interfering RNA of RTEF-1 totally reversed the anti-oxidation effect of RTEF-1 and inhibited RTEF-1-induced decreased p53 and p21 expressions. It demonstrated that RTEF-1 could protect cells from H2O2-induced oxidative damage. In addition, we demonstrated that RTEF-1 could up-regulate Klotho gene expression and activate its promoter. Furthermore, Klotho small interfering RNA significantly blocked RTEF-1-driven endothelial cell protection from H2O2-induced oxidative damage and increased p53 and p21 expressions. These results reveal that RTEF-1 is a potential anti-oxidation gene and can prevent H2O2-induced endothelial cell oxidative damage by activating Klotho.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Glucuronidasa/fisiología , Peróxido de Hidrógeno/toxicidad , Proteínas Musculares/fisiología , Estrés Oxidativo/efectos de los fármacos , Factores de Transcripción/fisiología , Venas Umbilicales/citología , Western Blotting , Senescencia Celular/fisiología , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiología , Humanos , Proteínas Klotho , Proteína Oncogénica p21(ras)/fisiología , Estrés Oxidativo/fisiología , ARN Interferente Pequeño/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Transcripción de Dominio TEA , Proteína p53 Supresora de Tumor/fisiología , Venas Umbilicales/efectos de los fármacos , Venas Umbilicales/fisiología
4.
J Clin Invest ; 125(4): 1393-5, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25798614

RESUMEN

RAS-driven cancers exhibit variable dependency on autophagy for survival; however, it is not fully understood how. In this issue of the JCI, Cheong and colleagues demonstrate that RAS-dependent elevation of casein kinase 1α (CK1α) negatively regulates autophagy at the level of autophagy gene transcription. Moreover, combined inhibition of both CK1α and autophagy reduced proliferation of RAS-driven tumors. The results of this study provide insight into the connection between mutant RAS and autophagy, and suggest targeting CK1α as a potential therapeutic strategy to modulate autophagy in RAS-driven cancers.


Asunto(s)
Autofagia/fisiología , Quinasa de la Caseína I/fisiología , Proteínas de Neoplasias/fisiología , Neoplasias/enzimología , Proteína Oncogénica p21(ras)/fisiología , Animales , Humanos
5.
J Clin Invest ; 125(4): 1401-18, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25798617

RESUMEN

Activating mutations in the RAS oncogene are common in cancer but are difficult to therapeutically target. RAS activation promotes autophagy, a highly regulated catabolic process that metabolically buffers cells in response to diverse stresses. Here we report that casein kinase 1α (CK1α), a ubiquitously expressed serine/threonine kinase, is a key negative regulator of oncogenic RAS-induced autophagy. Depletion or pharmacologic inhibition of CK1α enhanced autophagic flux in oncogenic RAS-driven human fibroblasts and multiple cancer cell lines. FOXO3A, a master longevity mediator that transcriptionally regulates diverse autophagy genes, was a critical target of CK1α, as depletion of CK1α reduced levels of phosphorylated FOXO3A and increased expression of FOXO3A-responsive genes. Oncogenic RAS increased CK1α protein abundance via activation of the PI3K/AKT/mTOR pathway. In turn, elevated levels of CK1α increased phosphorylation of nuclear FOXO3A, thereby inhibiting transactivation of genes critical for RAS-induced autophagy. In both RAS-driven cancer cells and murine xenograft models, pharmacologic CK1α inactivation synergized with lysosomotropic agents to inhibit growth and promote tumor cell death. Together, our results identify a kinase feedback loop that influences RAS-dependent autophagy and suggest that targeting CK1α-regulated autophagy offers a potential therapeutic opportunity to treat oncogenic RAS-driven cancers.


Asunto(s)
Autofagia/fisiología , Quinasa de la Caseína I/fisiología , Proteínas de Neoplasias/fisiología , Neoplasias/enzimología , Proteína Oncogénica p21(ras)/fisiología , Secuencia de Aminoácidos , Animales , Quinasa de la Caseína I/antagonistas & inhibidores , Retroalimentación Fisiológica , Fibroblastos/enzimología , Fibroblastos/fisiología , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Datos de Secuencia Molecular , Trasplante de Neoplasias , Fosfatidilinositol 3-Quinasas/fisiología , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-akt/fisiología , ARN Interferente Pequeño/farmacología , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Serina-Treonina Quinasas TOR/fisiología , Activación Transcripcional
7.
Cancer Res ; 71(21): 6708-17, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21937682

RESUMEN

The p19ARF tumor suppressor limits ribosome biogenesis and responds to hyperproliferative signals to activate the p53 checkpoint response. Although its activation of p53 has been well characterized, the role of ARF in restraining nucleolar ribosome production is poorly understood. Here we report the use of a mass spectroscopic analysis to identify protein changes within the nucleoli of Arf-deficient mouse cells. Through this approach, we discovered that ARF limited the nucleolar localization of the RNA helicase DDX5, which promotes the synthesis and maturation of rRNA, ultimately increasing ribosome output and proliferation. ARF inhibited the interaction between DDX5 and nucleophosmin (NPM), preventing association of DDX5 with the rDNA promoter and nuclear pre-ribosomes. In addition, Arf-deficient cells transformed by oncogenic RasV12 were addicted to DDX5, because reduction of DDX5 was sufficient to impair RasV12-driven colony formation in soft agar and tumor growth in mice. Taken together, our findings indicate that DDX5 is a key p53-independent target of the ARF tumor suppressor and is a novel non-oncogene participant in ribosome biogenesis.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/fisiología , ARN Helicasas DEAD-box/fisiología , Ribosomas/metabolismo , Animales , División Celular , Nucléolo Celular/metabolismo , Transformación Celular Neoplásica/genética , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina/deficiencia , ADN Ribosómico/genética , ADN Ribosómico/metabolismo , Fibroblastos , Ratones , Ratones Noqueados , Ratones Desnudos , Proteínas Nucleares/metabolismo , Nucleofosmina , Proteína Oncogénica p21(ras)/fisiología , Regiones Promotoras Genéticas , Transporte de Proteínas , Procesamiento Postranscripcional del ARN , ARN Ribosómico/metabolismo , Ensayo de Tumor de Célula Madre , Proteína p53 Supresora de Tumor/fisiología
8.
BMB Rep ; 43(6): 432-7, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20587334

RESUMEN

LBH is a transcription factor as a candidate gene for CHD associated with partial trisomy 2p syndrome. To identify potential LBH-interacting partners, a yeast two-hybrid screen using LBH as a bait was performed with a human heart cDNA library. One of the clones identified encodes alphaB-crystallin. Co-immunoprecipitation and GST pull-down assays showed that LBH interacts with alphaB-crystallin, which is further confirmed by mammalian two-hybrid assays. Co-localization analysis showed that in COS-7 cells, alphaB-crystallin that is cytoplasmic alone, accumulates partialy in the nucleus when co-transfected with LBH. Transient transfection assays indicated that overexpression of LBH or alphaB-crystallin reduced the transcriptional activities of p53 and p21, respectively, Overexpression of both alphaB-crystallin and LBH together resulted in a stronger repression of the transcriptional activities of p21 and p53. These results showed that the interaction of LBH and alphaB-crystallin may inhibit synergistically the transcriptional regulation of p53 and p21.


Asunto(s)
Cristalinas/fisiología , Proteína Oncogénica p21(ras)/fisiología , Transactivadores/fisiología , Transcripción Genética , Proteína p53 Supresora de Tumor/fisiología , Animales , Células COS , Chlorocebus aethiops , Humanos , Inmunoprecipitación , Fracciones Subcelulares/metabolismo , Factores de Transcripción , Técnicas del Sistema de Dos Híbridos
9.
Oncogene ; 29(36): 5083-94, 2010 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-20581871

RESUMEN

Oncogene-mediated premature senescence has emerged as a potential tumor-suppressive mechanism in early cancer transitions. Many studies showed that Ras and p38 mitogen-activated protein kinase (MAPK) participate in premature senescence. Our previous work indicated that the HMG box-containing protein 1 (HBP1) transcription factor is involved in Ras- and p38 MAPK-induced premature senescence, but the mechanism of which has not yet been identified. Here, we showed that the p16(INK4A) cyclin-dependent kinase inhibitor is a novel target of HBP1 participating in Ras-induced premature senescence. The promoter of the p16(INK4A) gene contains an HBP1-binding site at position -426 to -433 bp from the transcriptional start site. HBP1 regulates the expression of the endogenous p16(INK4A) gene through direct sequence-specific binding. With HBP1 expression and the subsequent increase of p16(INK4A) gene expression, Ras induces premature senescence in primary cells. The data suggest a model in which Ras and p38 MAPK signaling engage HBP1 and p16(INK4A) to trigger premature senescence. In addition, we report that HBP1 knockdown is also required for Ras-induced transformation. All the data indicate that the mechanism of HBP1-mediated transcriptional regulation is important for not only premature senescence but also tumorigenesis.


Asunto(s)
Senescencia Celular/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Proteínas del Grupo de Alta Movilidad/fisiología , Proteína Oncogénica p21(ras)/fisiología , Proteínas Represoras/fisiología , Animales , Transformación Celular Neoplásica/genética , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Regulación de la Expresión Génica , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Modelos Biológicos , Proteína Oncogénica p21(ras)/genética , Proteína Oncogénica p21(ras)/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factores de Tiempo , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología , Trasplante Heterólogo , Regulación hacia Arriba
10.
Endocr Relat Cancer ; 17(1): 113-23, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19903742

RESUMEN

RET/papillary thyroid carcinoma (PTC), TRK-T, or activating mutations of Ras and BRaf are frequent genetic alterations in PTC, all leading to the activation of the extracellular-regulated kinase (Erk) cascade. The aim of this study was to investigate the role of calmodulin-dependent kinase II (CaMKII) in the signal transduction leading to Erk activation in PTC cells. In normal thyroid cells, CaMKII and Erk were in the inactive form in the absence of stimulation. In primary PTC cultures and in PTC cell lines harboring the oncogenes RET/PTC-1 or BRaf(V600E), CaMKII was active also in the absence of any stimulation. Inhibition of calmodulin or phospholipase C (PLC) attenuated the level of CaMKII activation. Expression of recombinant RET/PTC-3, BRaf(V600E), or Ras(V12) induced CaMKII activation. Inhibition of CaMKII attenuated Erk activation and DNA synthesis in thyroid papillary carcinoma (TPC-1), a cell line harboring RET/PTC-1, suggesting that CaMKII is a component of the Erk signal cascade in this cell line. In conclusion, PTCs contain an active PLC/Ca(2+)/calmodulin-dependent signal inducing constitutive activation of CaMKII. This kinase is activated by BRaf(V600E), oncogenic Ras, and by RET/PTC. CaMKII participates to the activation of the Erk pathway by oncogenic Ras and RET/PTC and contributes to their signal output, thus modulating tumor cell proliferation.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Carcinoma Papilar/enzimología , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Proteínas de Neoplasias/fisiología , Neoplasias de la Tiroides/enzimología , Secuencia de Aminoácidos , Animales , Señalización del Calcio/fisiología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Calmodulina/fisiología , Carcinoma Papilar/genética , Carcinoma Papilar/patología , División Celular , Activación Enzimática/efectos de los fármacos , Estrenos/farmacología , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Datos de Secuencia Molecular , Mutación , Proteínas de Neoplasias/antagonistas & inhibidores , Proteína Oncogénica p21(ras)/fisiología , Piperidinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/fisiología , Proteínas Proto-Oncogénicas c-ret/fisiología , Pirrolidinonas/farmacología , Quinazolinas/farmacología , Ratas , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología , Fosfolipasas de Tipo C/fisiología
11.
Cancer Biol Ther ; 9(2): 122-33, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19923925

RESUMEN

A common metabolic change in cancer is the acquisition of glycolytic phenotypes. Increased expression of glycolytic enzymes is considered as one contributing factor. The role of mitochondrial defects in acquisition of glycolytic phenotypes has been postulated but remains controversial. Here we show that functional defects in mitochondrial respiration could be induced by oncogenic H-Ras(Q61L) transformation, even though the mitochondrial contents or mass was not reduced in the transformed cells. First, mitochondrial respiration, as measured by mitochondrial oxygen consumption, was suppressed in NIH-3T3 cells transformed with H-Ras(Q61L). Second, oligomycin or rotenone did not reduce the cellular ATP levels in the H-Ras(Q61L) transformed cells, suggesting a diminished role of mitochondrial respiration in the cellular energy metabolism. Third, inhibition of glycolysis with iodoacetic acid reduced ATP levels at a much faster rate in H-Ras(Q61L) transformed cells than in the vector control cells. The reduction of cellular ATP levels was reversed by exogenously added pyruvate in the vector control cells but not in H-Ras(Q61L) transformed cells. Finally when compared to the HRas(Q61L) transformed cells, the vector control cells had increased resistance toward glucose deprivation. The increased resistance was dependent on mitochondrial oxidative phosphorylation since rotenone or oligomycin abolished the increased survival of the vector control cells under glucose deprivation. The results also suggest an inability of the H-Ras(Q61L) transformed cells to reactivate mitochondrial respiration under glucose deprivation. Taken together, the data suggest that mitochondrial respiration can be impaired during transformation of NIH-3T3 cells by oncogeneic H-Ras(Q61L).


Asunto(s)
Transformación Celular Neoplásica , Fibroblastos/metabolismo , Genes ras , Mitocondrias/metabolismo , Proteína Oncogénica p21(ras)/fisiología , Adenosina Trifosfato/metabolismo , Animales , Antimicina A/farmacología , Transporte de Electrón/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Glucosa/metabolismo , Glucólisis/efectos de los fármacos , Ácido Yodoacético/farmacología , Ratones , Mitocondrias/efectos de los fármacos , Mutación Missense , Células 3T3 NIH/metabolismo , Oligomicinas/farmacología , Proteína Oncogénica p21(ras)/genética , Fosforilación Oxidativa/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Mutación Puntual , Ácido Pirúvico/farmacología , Rotenona/farmacología
12.
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
13.
Cell Signal ; 21(4): 502-8, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19146951

RESUMEN

Protein kinase C delta (PKCdelta) modulates cell survival and apoptosis in diverse cellular systems. We recently reported that PKCdelta functions as a critical anti-apoptotic signal transducer in cells containing activated p21(Ras) and results in the activation of AKT, thereby promoting cell survival. How PKCdelta is regulated by p21(Ras), however, remains incompletely understood. In this study, we show that PKCdelta, as a transducer of anti-apoptotic signals, is activated by phosphotidylinositol 3' kinase/phosphoinositide-dependent kinase 1 (PI(3)K-PDK1) to deliver the survival signal to Akt in the environment of activated p21(Ras). PDK1 is upregulated in cells containing an activated p21Ras. Knock-down of PDK1, PKCdelta, or AKT forces cells containing activated p21(Ras) to undergo apoptosis. PDK1 regulates PKCdelta activity, and constitutive expression of PDK1 increases PKCdelta activity in different cell types. Conversely, expression of a kinase-dead (dominant-negative) PDK1 significantly suppresses PKCdelta activity. p21(Ras)-mediated survival signaling is therefore regulated by via a PI(3)K-AKT pathway, which is dependent upon both PDK1 and PKCdelta, and PDK1 activates and regulates PKCdelta to determine the fate of cells containing a mutated, activated p21(Ras).


Asunto(s)
Proteína Oncogénica p21(ras)/fisiología , Proteína Quinasa C-delta/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Proteínas Quinasas Dependientes de 3-Fosfoinosítido , Animales , Apoptosis , Células 3T3 BALB/metabolismo , División Celular , Línea Celular Tumoral/metabolismo , Supervivencia Celular , Activación Enzimática , Técnicas de Silenciamiento del Gen , Genes ras , Humanos , Ratones , Células 3T3 NIH/metabolismo , Proteína Oncogénica p21(ras)/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Proteína Quinasa C-delta/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/fisiología , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/fisiología , ARN Interferente Pequeño/farmacología , Proteínas Recombinantes de Fusión/fisiología
14.
Cancer Lett ; 275(2): 221-6, 2009 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-19022560

RESUMEN

The NM23 family proteins are involved in a variety of biological processes including tumor metastasis, development, and differentiation; however, their functions in the regulation of cellular proliferation are poorly understood. We have investigated the role of one NM23 family protein, NM23H2, in the regulation of cellular proliferation directed by the extracellular signal regulated kinase (ERK) pathway. The activity of ERKs was enhanced by knockdown of endogenous NM23H2 and blocked by overexpression of NM23H2 in both NIH3T3 and HEK293 cells. Additionally, the epidermal growth factor (EGF)- and oncogenic Ras(G12R)-induced proliferation of both HEK293 and NIH3T3 cells was reduced by NM23H2 overexpression. Furthermore, activation of Raf-1, MEK and the ERKs by either EGF or Ras(G12R) was inhibited by NM23H2 overexpression. Together, our data indicate that NM23H2 is a negative regulator of cellular proliferation stimulated by EGF- and Ras-mediated activation of the ERK pathway.


Asunto(s)
Proliferación Celular , Factor de Crecimiento Epidérmico/fisiología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Nucleósido Difosfato Quinasas NM23/fisiología , Proteína Oncogénica p21(ras)/fisiología , Animales , Secuencia de Bases , Western Blotting , Línea Celular , Cartilla de ADN , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Citometría de Flujo , Técnicas de Silenciamiento del Gen , Humanos , Ratones
15.
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
16.
Cancer Res ; 68(15): 6215-23, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18676845

RESUMEN

Fibroblast growth factor 2 (FGF2) is considered to be a bona fide oncogenic factor, although results from our group and others call this into question. Here, we report that exogenous recombinant FGF2 irreversibly inhibits proliferation by inducing senescence in Ras-dependent malignant mouse cells, but not in immortalized nontumorigenic cell lines. We report the following findings in K-Ras-dependent malignant Y1 adrenocortical cells and H-Ras V12-transformed BALB-3T3 fibroblasts: (a) FGF2 inhibits clonal growth and tumor onset in nude and immunocompetent BALB/c mice, (b) FGF2 irreversibly blocks the cell cycle, and (c) FGF2 induces the senescence-associated beta-galactosidase with no accompanying signs of apoptosis or necrosis. The tyrosine kinase inhibitor PD173074 completely protected malignant cells from FGF2. In Y1 adrenal cells, reducing the constitutively high levels of K-Ras-GTP using the dominant-negative RasN17 mutant made cells resistant to FGF2 cytotoxicity. In addition, transfection of the dominant-negative RhoA-N19 into either Y1 or 3T3-B61 malignant cell lines yielded stable clonal transfectants that were unable to activate RhoA and were resistant to the FGF2 stress response. We conclude that in Ras-dependent malignant cells, FGF2 interacts with its cognate receptors to trigger a senescence-like process involving RhoA-GTP. Surprisingly, attempts to select FGF2-resistant cells from the Y1 and 3T3-B61 cell lines yielded only rare clones that (a) had lost the overexpressed ras oncogene, (b) were dependent on FGF2 for proliferation, and (c) were poorly tumorigenic. Thus, FGF2 exerted a strong negative selection that Ras-dependent malignant cells could rarely overcome.


Asunto(s)
Proliferación Celular , Senescencia Celular/fisiología , Factor 2 de Crecimiento de Fibroblastos/fisiología , Proteína Oncogénica p21(ras)/fisiología , Proteína de Unión al GTP rhoA/fisiología , Células 3T3 , Animales , Línea Celular , Electroforesis en Gel de Poliacrilamida , Citometría de Flujo , Ratones , Ratones Endogámicos BALB C
17.
Cancer Res ; 68(15): 6224-31, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18676846

RESUMEN

Transforming growth factor-beta (TGF-beta) cooperates with oncogenic members of the Ras superfamily to promote cellular transformation and tumor progression. Apart from the classic (H-, K-, and N-) Ras GTPases, only the R-Ras subfamily (R-Ras, R-Ras2/TC21, and R-Ras3/M-Ras) has significant oncogenic potential. In this study, we show that oncogenic R-Ras transformation of EpH4 cells requires TGF-beta signaling. When murine EpH4 cells were stably transfected with a constitutively active R-Ras(G38V) mutant, they were no longer sensitive to TGF-beta-mediated growth inhibition and showed increased proliferation and transformation in response to exogenous TGF-beta. R-Ras/EpH4 cells require TGF-beta signaling for transformation to occur and they produce significantly elevated levels of endogenous TGF-beta, which signals in an autocrine fashion. The effects of TGF-beta are independent of Smad2/3 activity and require activation of TGF-beta-associated kinase 1 (TAK1) and its downstream effectors c-Jun NH(2)-terminal kinase and p38 mitogen-activated protein kinase as well as the phosphoinositide 3-kinase/Akt and mammalian target of rapamycin pathways. Thus, TAK1 is a novel link between TGF-beta signaling and oncogenic R-Ras in the promotion of tumorigenesis.


Asunto(s)
Transformación Celular Neoplásica , Quinasas Quinasa Quinasa PAM/fisiología , Glándulas Mamarias Humanas/citología , Proteína Oncogénica p21(ras)/fisiología , Factor de Crecimiento Transformador beta/fisiología , Western Blotting , Línea Celular , Proliferación Celular , Humanos , Glándulas Mamarias Humanas/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
18.
Oncogene ; 27(36): 4921-32, 2008 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-18454176

RESUMEN

Ras oncogene upregulates the expression of nicotinamide adenine dinucleotide phosphate oxidase (Nox) 1 via the Raf/MEK/ERK pathway, leading to the elevated production of reactive oxygen species that is essential for maintenance of Ras-transformation phenotypes. However, the precise transcriptional control mechanism underlying Ras-induced Nox1 expression remains to be elucidated. Here we demonstrated that via the MEK/ERK pathway, Ras signaling enhances the activity of the functional Nox1 promoter (nt -321 to -1) in colon cancer CaCo-2 cells and thereby induces the formation of the specific protein-DNA complexes in the two GATA-binding site-containing regions (nt -161 to -136 and -125 to -100). Supershift assays with GATA antibodies, protein analyses and chromatin immunoprecipitation revealed that GATA-6 is a component of the specific protein-DNA complexes at the Nox1 promoter. GATA-6 was able to trans-activate the Nox1 promoter but not a promoter in which the GATA-binding sites are mutated. Moreover, GATA-6 was phosphorylated at serine residues by MEK-activated ERK, which increased GATA-6 DNA binding, correlating with suppression of the Nox1 promoter activity by an MEK inhibitor PD98059. Finally, the site-directed mutation of the consensus ERK phosphorylation site (PYS(120)P to PYA(120)P) of GATA-6 abolished its trans-activation activity, suppressing of the growth of CaCo-2 cells. On the basis of these results, we propose that oncogenic Ras signaling upregulates the transcription of Nox1 through MEK-ERK-dependent phosphorylation of GATA-6.


Asunto(s)
Factor de Transcripción GATA6/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , NADH NADPH Oxidorreductasas/genética , Proteína Oncogénica p21(ras)/fisiología , Regulación hacia Arriba/fisiología , Secuencia de Bases , Células CACO-2 , Cartilla de ADN , Ensayo de Cambio de Movilidad Electroforética , Genes Reporteros , Humanos , NADPH Oxidasa 1 , Fosforilación , Regiones Promotoras Genéticas , ARN Mensajero/genética , Transducción de Señal
19.
Sci Signal ; 1(13): pe14, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18385037

RESUMEN

Oncogenic transformation is associated with genetic changes and epigenetic alterations. A study now shows that oncogenic Ras uses a complex and elaborate epigenetic silencing program to specifically repress the expression of multiple unrelated cancer-suppressing genes through a common pathway. These results suggest that cancer-related epigenetic modifications may arise through a specific and instructive mechanism and that genetic changes and epigenetic alterations are intimately connected and contribute to tumorigenesis cooperatively.


Asunto(s)
Epigénesis Genética/fisiología , Silenciador del Gen , Proteína Oncogénica p21(ras)/fisiología , Islas de CpG , Metilación de ADN
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...