Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
Pestic Biochem Physiol ; 187: 105177, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36127041

RESUMO

Insect nicotinic acetylcholine receptors (nAChRs) require cofactors for functional heterologous expression. A previous study revealed that TMX3 was crucial for the functional expression of Drosophila melanogaster Dα1/Dß1 nAChRs in Xenopus laevis oocytes, while UNC-50 and RIC-3 enhanced the acetylcholine (ACh)-induced responses of the nAChRs. However, it is unclear whether the coexpression of UNC-50 and RIC-3 with TMX3 and the subunit stoichiometry affect pharmacology of Dα1/Dß1 nAChRs when expressed in X. laevis oocytes. We have investigated the effects of coexpressing UNC-50 and RIC-3 with TMX3 as well as changing the subunit stoichiometry on the agonist activity of ACh and imidacloprid on the Dα1/Dß1 nAChRs. UNC-50 and RIC-3 hardly affected the agonist affinity of ACh and imidacloprid for the Dα1/Dß1 nAChRs formed by injecting into X. laevis oocytes with an equal amount mixture of the subunit cRNAs, but enhanced current amplitude of the ACh-induced response. Imidacloprid showed higher affinity for the Dß1 subunit-excess Dα1/Dß1 (Dα1/Dß1 = 1/5) nAChRs than the Dα1 subunit-excess Dα1/Dß1 (Dα1/Dß1 = 5/1) nAChRs, suggesting that imidacloprid prefers the Dα1-Dß1 orthosteric site over the Dα1-Dα1 orthosteric site.


Assuntos
Receptores Nicotínicos , Acetilcolina/farmacologia , Animais , Drosophila melanogaster/metabolismo , Neonicotinoides , Nitrocompostos , Oócitos/metabolismo , Receptores Nicotínicos/genética , Receptores Nicotínicos/metabolismo , Xenopus laevis/metabolismo , Proteínas ras/metabolismo , Proteínas ras/farmacologia
3.
Soft Matter ; 13(45): 8483-8491, 2017 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-29091102

RESUMO

The organization of the actin cytoskeleton plays a key role in regulating cell mechanics. It is fundamentally altered during transformation, affecting how cells interact with their environment. We investigated mechanical properties of cells expressing constitutively active, oncogenic Ras (RasV12) in adherent and suspended states. To do this, we utilized atomic force microscopy and a microfluidic optical stretcher. We found that adherent cells stiffen and suspended cells soften with the expression of constitutively active Ras. The effect on adherent cells was reversed when contractility was inhibited with the ROCK inhibitor Y-27632, resulting in softer RasV12 cells. Our findings suggest that increased ROCK activity as a result of Ras has opposite effects on suspended and adhered cells. Our results also establish the importance of the activation of ROCK by Ras and its effect on cell mechanics.


Assuntos
Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Fenômenos Mecânicos/efeitos dos fármacos , Proteínas ras/farmacologia , Animais , Fenômenos Biomecânicos/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Cães , Células Madin Darby de Rim Canino
4.
Mol Carcinog ; 55(5): 563-74, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-25753147

RESUMO

p38δ expression and/or activity are increased in human cutaneous malignancies, including invasive squamous cell carcinoma (SCC) and head and neck SCC, but the role of p38δ in cutaneous carcinogenesis has not been well-defined. We have reported that mice with germline loss of p38δ exhibited a reduced susceptibility to skin tumor development compared with wild-type mice in the two-stage 7,12-dimethylbenz(a)anthracene (DMBA)/12-O-tetradecanoylphorbol-13-acetate (TPA) chemical skin carcinogenesis model. Here, we report that p38δ gene ablation inhibited the growth of tumors generated from v-ras(Ha) -transformed keratinocytes in skin orthografts to nude mice, indicating that keratinocyte-intrinsic p38δ is required for Ras-induced tumorigenesis. Gene expression profiling of v-ras(Ha) -transformed p38δ-null keratinocytes revealed transcriptional changes associated with cellular responses linked to tumor suppression, such as reduced proliferation and increased differentiation, cell adhesion, and cell communications. Notably, a short-term DMBA/TPA challenge, modeling the initial stages of chemical skin carcinogenesis treatment, elicited an enhanced inflammation in p38δ-null skin compared with skin of wild-type mice, as assessed by measuring the expression of pro-inflammatory cytokines, including IL-1ß, IL-6, IL-17, and TNFα. Additionally, p38δ-null skin and p38δ-null keratinocytes exhibited increased p38α activation and signaling in response to acute inflammatory challenges, suggesting a role for p38α in stimulating the elevated inflammatory response in p38δ-null skin during the initial phases of the DMBA/TPA treatment compared with similarly treated p38δ(+/+) skin. Altogether, our results indicate that p38δ signaling regulates skin carcinogenesis not only by keratinocyte cell-autonomous mechanisms, but also by influencing the interaction between between the epithelial compartment of the developing skin tumor and its stromal microenvironment.


Assuntos
Proteína Quinase 13 Ativada por Mitógeno/genética , Neoplasias Cutâneas/genética , Pele/patologia , Proteínas ras/genética , Animais , Benzo(a)Antracenos/toxicidade , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/metabolismo , Camundongos , Camundongos Nus , Proteína Quinase 13 Ativada por Mitógeno/metabolismo , Pele/efeitos dos fármacos , Pele/metabolismo , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Acetato de Tetradecanoilforbol/toxicidade , Proteínas ras/farmacologia
5.
Mol Cancer ; 13: 259, 2014 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-25466244

RESUMO

BACKGROUND: Lung cancer is the major cause of cancer-related deaths and many cases of Non Small Cell Lung Cancer (NSCLC), a common type of lung cancer, have frequent genetic/oncogenic activation of EGFR, KRAS, PIK3CA, BRAF, and others that drive tumor growth. Some patients though initially respond, but later develop resistance to erlotinib/gefitinib with no option except for cytotoxic therapy. Therefore, development of novel targeted therapeutics is imperative to provide improved survival benefit for NSCLC patients. The mTOR cell survival pathway is activated in naïve, or in response to targeted therapies in NSCLC. METHODS: We have discovered P7170, a small molecule inhibitor of mTORC1/mTORC2/ALK1 and investigated its antitumor efficacy using various in vitro and in vivo models of human NSCLC. RESULTS: P7170 inhibited the phosphorylation of AKT, S6 and 4EBP1 (substrates for mTORC2 and mTORC1) levels by 80-100% and growth of NSCLC cells. P7170 inhibited anchorage-independent colony formation of NSCLC patient tumor-derived cells subsistent of disease sub-types. The compound also induced apoptosis in NSCLC cell lines. P7170 at a well-tolerated daily dose of 20 mg/kg significantly inhibited the growth of NSCLC xenografts independent of different mutations (EGFR, KRAS, or PIK3CA) or sensitivity to erlotinib. Pharmacokinetic-pharmacodynamic (PK-PD) analysis showed sub-micro molar tumor concentrations along with mTORC1/C2 inhibition. CONCLUSIONS: Our results provide evidence of antitumor activity of P7170 in the erlotinib -sensitive and -insensitive models of NSCLC.


Assuntos
Receptores de Activinas Tipo II/antagonistas & inibidores , Antineoplásicos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Proliferação de Células/efeitos dos fármacos , Imidazóis/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Complexos Multiproteicos/antagonistas & inibidores , Quinolinas/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular Tumoral , Classe I de Fosfatidilinositol 3-Quinases , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Receptores ErbB/farmacologia , Cloridrato de Erlotinib , Células HeLa , Humanos , Neoplasias Pulmonares/metabolismo , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Alvo Mecanístico do Complexo 2 de Rapamicina , Camundongos , Camundongos Nus , Fosfatidilinositol 3-Quinases/farmacologia , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas/farmacologia , Proteínas Proto-Oncogênicas p21(ras) , Quinazolinas/farmacologia , Proteínas ras/farmacologia
6.
Cell Mol Neurobiol ; 34(3): 409-17, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24414340

RESUMO

Alzheimer's disease (AD) is characterized by deposition of beta-amyloid peptides (Aß) and progressive loss of neurons. Neural stem/progenitor cells (NSPCs) can proliferate and produce immature neurons even in the brain of AD patients. However, Aß42 significantly decreased the expression of RhoC in NSPCs during the co-incubation (P < 0.01). Treating with RhoC siRNA prevented membrane from protrusion and led to a significant reduction in cell migration in responses to SDF-1. Compared with wild-type mice, the numbers of RhoC-immunoreactive cells in hippocampus and cortex were significantly down-regulated in APP/PS1 mice aged 9 months. The results suggest that Aß42 down-regulates the expression of RhoC in NSPCs in vitro and in vivo; down-regulated RhoC expression results in decreased migration of NSPCs.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Movimento Celular/fisiologia , Células-Tronco Neurais/fisiologia , Fragmentos de Peptídeos/toxicidade , Proteínas ras/biossíntese , Animais , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células-Tronco Neurais/efeitos dos fármacos , Gravidez , Ratos , Ratos Sprague-Dawley , Proteínas ras/farmacologia , Proteína de Ligação a GTP rhoC
7.
Nan Fang Yi Ke Da Xue Xue Bao ; 31(8): 1392-5, 2011 Aug.
Artigo em Chinês | MEDLINE | ID: mdl-21868306

RESUMO

OBJECTIVE: To study the effect of binding activities of the NH(2) terminus of E1A to the proteins regulating cell growth on ras-induced cell senescence and explore the mechanism of E1A-mediated escape from ras-induced senescence by E1A in human fibroblast. METHODS: In primary human fibroblasts, the proteins regulating cell growth in association with E1A NH(2) terminus, including the Rb family proteins, p300/CBP, and p400, were inactivated or interfered. The effect of alterations in the binding activities of these proteins on cell senescence bypass mediated by E1A was evaluated by cell growth curve. RESULTS: The Inactivation of Rb family proteins alone was not sufficient to rescue ras-induced cell senescence, whereas inactivation of both the Rb proteins and p300/CBP blocked ras-induced senescence of human fibroblasts. CONCLUSION: Rb and p300/CBP binding activities are both required for E1A to bypass ras-induced senescence in human fibroblasts.


Assuntos
Proteínas E1A de Adenovirus/farmacologia , Senescência Celular/efeitos dos fármacos , Fibroblastos/citologia , Proteínas ras/antagonistas & inibidores , Humanos , Cultura Primária de Células , Proteína do Retinoblastoma/metabolismo , Pele/citologia , Fatores de Transcrição de p300-CBP/metabolismo , Proteínas ras/farmacologia
8.
Biotechnol Prog ; 27(2): 577-80, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21438179

RESUMO

Constitutively active Ras (CA-Ras) is known to enhance cell growth through the induction of various signaling cascades including the phosphoinositide 3-kinase (PI3K)/Akt and mitogen-activated protein kinase (MAPK)/ERK signaling pathways, although the cellular response is highly dependent on the cell type. To evaluate the effect of CA-Ras overexpression on cell growth in recombinant Chinese hamster ovary (rCHO) cells, an erythropoietin (EPO)-producing rCHO cell line with regulated CA-Ras overexpression (EPO-off-CA-Ras) was established using the Tet-off system. The CA-Ras expression level in EPO-off-CA-Ras cells was tightly regulated by doxycycline addition. Although CA-Ras overexpression slightly increased the viable cell concentration during the late exponential phase, it did not increase the maximum viable cell concentration or specific growth rate to a significant degree. Unexpectedly, CA-Ras overexpression in rCHO cells led only to the enhancement in the activation of the MAPK/ERK signaling pathway and not the PI3K/Akt signaling pathway. Taken together, CA-Ras overexpression in rCHO cells did not significantly affect cell growth; it also had no critical impact on viable cell concentration or EPO production, possibly due to a failure to activate the PI3K/Akt signaling pathway.


Assuntos
Proliferação de Células/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas ras/genética , Proteínas ras/farmacologia , Animais , Células CHO , Sobrevivência Celular/efeitos dos fármacos , Cricetinae , Cricetulus , Doxiciclina/farmacologia , Eritropoetina , Transdução de Sinais/efeitos dos fármacos
9.
Oncogene ; 29(45): 6064-70, 2010 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-20818422

RESUMO

RAS mutations are the most common gain-of-function change in human cancer and promise to be a critical therapy target. As a new approach, we have used a surrogate to drug the 'undruggable' (that is, RAS-effector protein-protein interactions inside cancer cells) in pre-clinical mouse models of RAS-dependent cancers. Using this novel reagent, we have specifically targeted RAS signalling in a transgenic mouse model of lung cancer by directly blockading RAS-effector interactions with an antibody fragment that binds to activated RAS, and show that the interaction of RAS and effectors, such as phosphoinositide 3-kinase and RAF, is necessary for tumour initiation. Further, interference with oncogenic RAS-effector interactions result in control of tumour growth in human cancer cells but, crucially, does not necessarily cause tumour regression. These findings support the concept that ablating RAS-dependent signalling in cancer will have chemo-preventive effects that confer a chronic state in cancer and suggest that mutant RAS-targeted therapies may require conjoint targeting of other molecules and/or current cancer therapeutic strategies (for example, radiotherapy and chemotherapy) to be curative. In this context, our findings suggest that the oncogene addiction model is not universally correct in its central thesis that cancer cell death is inevitable after loss of oncogenic protein function.


Assuntos
Neoplasias/metabolismo , Neoplasias/patologia , Proteínas ras/metabolismo , Células 3T3 , Animais , Linhagem Celular Tumoral , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Transgênicos , Mutação , Transdução de Sinais , Proteínas ras/farmacologia
10.
Oncogene ; 29(45): 6004-15, 2010 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-20818427

RESUMO

Overexpression of Ras(V12) in MCF10A cells, an immortalized mammary epithelial cell line, leads to transformation of these cells. We demonstrate that this is accompanied by degradation of C/EBPbeta1. C/EBPbeta is a transcription factor in which three protein isoforms exist because of alternative translation at three in-frame methionines. When C/EBPbeta1 is expressed in MCF10A-Ras(V12) cells, immunoblot analysis reveals that C/EBPbeta1 is degraded in these cells. Treatment of MCF10A-Ras(V12)-C/EBPbeta1 cells with the cdk inhibitor roscovitine leads to stabilization of C/EBPbeta1. It has been previously shown that cdk2 phosphorylates C/EBPbeta on Thr235. We demonstrate that mutation of Thr235 to alanine in C/EBPbeta1 is sufficient to restore the stability of C/EBPbeta1 expression in MCF10A-Ras(V12) cells. Overexpression of Ras(V12) in primary cells induces senescence rather than transformation, thus suppressing tumorigenesis. C/EBPbeta is required for Ras(V12)-induced senescence in primary mouse embryonic fibroblasts. Upregulation of interleukin-6 (IL6) by C/EBPbeta has been shown to be necessary for oncogene-induced senescence, but the specific isoform of C/EBPbeta has not been investigated. We show that the C/EBPbeta1 isoform upregulates IL6 when introduced into normal fibroblasts. In addition, we show that C/EBPbeta1 induces senescence. Taken together, degradation of C/EBPbeta1 by Ras activation may represent a mechanism to bypass OIS.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/genética , Senescência Celular/efeitos dos fármacos , Proteína Oncogênica p21(ras)/farmacologia , Proteínas ras/farmacologia , Mama , Proteína beta Intensificadora de Ligação a CCAAT , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Linhagem Celular , Quinases Ciclina-Dependentes/antagonistas & inibidores , Células Epiteliais , Fibroblastos , Regulação da Expressão Gênica , Humanos , Lactente , Interleucina-6/metabolismo , Mutação , Inibidores de Proteínas Quinases/farmacologia , Purinas/farmacologia , Roscovitina , Regulação para Cima
11.
Clin Immunol ; 131(2): 343-53, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19138568

RESUMO

In experimental membranous nephropathy, complement C5b-9-induced glomerular epithelial cell (GEC) injury leads to loss of glomerular permselectivity and proteinuria. Incubation of cultured GEC with antibody and serially-increasing concentrations of complement induced cytotoxicity in a dose-dependent manner. Stable expression of constitutively-active Ras (V(12)Ras) in GEC attenuated injury significantly. In the V(12)Ras-expressing GEC, disruption of the F-actin cytoskeleton with latrunculin B or swinholide A, or stabilization of F-actin with jasplakinolide reversed the cytoprotective effect of V(12)Ras. GEC displayed cortical F-actin; V(12)Ras-expressing GEC showed smaller and more rounded morphology, and decreased activity of the Rho GTPase, Rac1, compared with control GEC. Thus, the protective effect of V(12)Ras is dependent on remodeling of the actin cytoskeleton, and may be associated with a reduction in Rac activity, thereby altering the equilibrium in the activities of Rho GTPases. Activation of Ras signaling is a novel pathway to consider in developing strategies for cytoprotection in complement-mediated injury.


Assuntos
Complexo de Ataque à Membrana do Sistema Complemento/toxicidade , Glomerulonefrite Membranosa/fisiopatologia , Fatores Imunológicos/toxicidade , Glomérulos Renais/efeitos dos fármacos , Proteínas ras/metabolismo , Animais , Células Cultivadas , Immunoblotting , Glomérulos Renais/lesões , Microscopia de Fluorescência , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Ratos , Transdução de Sinais , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas ras/farmacologia
12.
Methods Mol Biol ; 462: 379-89, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19160682

RESUMO

Phosphatidylinositol-specific phospholipase C (PLC) enzymes catalyze the hydrolysis of phophatidylinositol 4,5-bisphosphate [PtdIns(4,5)P2] to diacylglycerol (DAG) and inositol 1,4,5-triphosphate [Ins(1,4,5)P3]. PLCepsilon is a recently discovered isoform that has been shown to be activated by members of the Ras and Rho families of guanosine trisphosphatases (GTPases) as well as subunits of heterotrimeric G-proteins. We describe a method for expressing a truncated PLCepsilon variant as an MBP fusion protein in E. coli. Subsequently, we describe the methodology necessary to reconstitute this protein with K-Ras-4B and RhoA GTPases and measure its activation.


Assuntos
Baculoviridae/enzimologia , Fosfoinositídeo Fosfolipase C/isolamento & purificação , Fosfoinositídeo Fosfolipase C/metabolismo , Proteínas ras/isolamento & purificação , Proteínas ras/farmacologia , Proteínas rho de Ligação ao GTP/isolamento & purificação , Proteínas rho de Ligação ao GTP/farmacologia , Animais , Linhagem Celular , Membrana Celular/química , Sistema Livre de Células , Cromatografia de Afinidade , Cromatografia em Gel , Ativação Enzimática/efeitos dos fármacos , Escherichia coli/citologia , Isoenzimas/genética , Isoenzimas/isolamento & purificação , Isoenzimas/metabolismo , Fosfoinositídeo Fosfolipase C/genética , Ratos , Deleção de Sequência , Solubilidade
13.
Carcinogenesis ; 28(5): 1021-31, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17148506

RESUMO

Several food polyphenols act as chemopreventers by reducing the incidence of many types of cancer, especially in colon epithelia. In this study, we have investigated whether the flavonoid quercetin can modulate cell proliferation and survival by targeting key molecules and/or biological processes responsible for tumor cell properties. The effect of quercetin on the expression of Ras oncoproteins was specifically studied using systems of either constitutive or conditional expression of oncogenic RAS in human epithelial cells. Our findings suggest that quercetin inhibits cell viability as well as cancer cell properties like anchorage-independent growth. These findings were further supported at the molecular level, since quercetin treatment resulted in a preferential reduction of Ras protein levels in cell lines expressing oncogenic Ras proteins. Notably, in cells that only express wild-type Ras or in those where the oncogenic Ras allele was knocked out, quercetin had no evident effects upon Ras levels. We have shown that quercetin drastically reduces half-life of oncogenic Ras but has no effect when the cells are treated with a proteasome inhibitor. Moreover, in Ha-RAS-transformed cells, quercetin induces autophagic processes. Since quercetin downregulates the levels of oncogenic Ras in cancer cells, we propose that this flavonoid could act as a chemopreventive agent for cancers with frequent mutations of RAS genes.


Assuntos
Autofagia/efeitos dos fármacos , Neoplasias do Colo/genética , Genes ras/efeitos dos fármacos , Quercetina/farmacologia , Proliferação de Células/efeitos dos fármacos , Transformação Celular Neoplásica , Colo , Humanos , Leupeptinas/antagonistas & inibidores , Proteína Oncogênica p21(ras) , Células Tumorais Cultivadas , Proteínas ras/farmacologia
14.
J Neurochem ; 97 Suppl 1: 97-103, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16635257

RESUMO

The dopaminergic transcriptional programme is highly regulated during development and in the adult, in response to activation of membrane receptor signalling cascades. Gene expression of tyrosine hydroxylase (TH), the rate-limiting enzyme in dopamine synthesis, is known to be regulated by receptors that act through protein kinase C (PKC) or Ras signalling. To investigate possible interactions between these two pathways before they converge on Raf activation, we evaluated whether phorbol ester (12-O-tetradecanoyl-phorbol-13-acetate, TPA)-dependent PKC activation required Ras for regulation of TH expression in IMR-32 cells. We found that long-term treatment with TPA, which induces down-regulation of PKC-alpha, led to induction of both protein and message levels of TH by autocrine factors. This was dependent on endogenous Ras, but independent of the transcription factor Nurr1. Moreover, this mechanism of action mimicked the effects of overexpression of the Ras-GAP domain of neurofibromin, GAP-related domain (GRD) I, which is part of the upstream mechanism for regulation of Ras activation and a PKC-alpha substrate. Overexpression of Ras also led to transcriptional and translational up-regulation of TH, independent of Nurr1 induction, as well as distinct phenotypic changes consistent with cell hypertrophy and increased secretory activity shown by induction of expression of vesicular monoamine transporter 2 and synaptosomal-associated protein-25. Most interestingly, overexpression of GRDI and down-regulation of the endogenous GRDII neurofibromin led to significant increases in Nurr1 message, possibly reflecting a transcriptional hierarchy during development. Taken together, these studies suggest that PKC-alpha, neurofibromin and Ras are essential in regulation of TH gene expression in IMR-32 cells.


Assuntos
Dopamina/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Neurofibromina 1/farmacologia , Acetato de Tetradecanoilforbol/farmacologia , Tirosina 3-Mono-Oxigenase/genética , Proteínas ras/farmacologia , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/farmacologia , Ativação Enzimática/efeitos dos fármacos , Genes ras/genética , Humanos , Neuroblastoma , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares , Proteína Quinase C/metabolismo , Proteína Quinase C-alfa/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Fatores de Transcrição/farmacologia
15.
Hum Mol Genet ; 15(7): 1087-98, 2006 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-16513807

RESUMO

Neurofibromatosis type I (NFI) is a common genetic disorder that causes nervous system tumors, and learning and memory defects in humans, and animal models. We identify a novel growth factor stimulated adenylyl cyclase (AC) pathway in the Drosophila brain, which is disrupted by mutations in the epidermal growth factor receptor (EGFR), neurofibromin (NF1) and Ras, but not Galpha(s). This is the first demonstration in a metazoan that a receptor tyrosine kinase (RTK) pathway, acting independently of the heterotrimeric G-protein subunit Galpha(s), can activate AC. We also show that Galpha(s) is the major Galpha isoform in fly brains, and define a second AC pathway stimulated by serotonin and histamine requiring NF1 and Galpha(s), as well as a third, classical Galpha(s)-dependent AC pathway, which is stimulated by Phe-Met-Arg-Phe-amide (FMRFamide) and dopamine. Using mutations and deletions of the human NF1 protein (hNF1) expressed in Nf1 mutant flies, we show that Ras activation by hNF1 is essential for growth factor stimulation of AC activity. Further, we demonstrate that sequences in the C-terminal region of hNF1 are sufficient for NF1/Galpha(s)-dependent neurotransmitter stimulated AC activity, and for rescue of body size defects in Nf1 mutant flies.


Assuntos
Adenilil Ciclases/metabolismo , Regulação da Expressão Gênica , Mutação , Neurofibromina 1/genética , Transdução de Sinais , Proteínas ras/metabolismo , Animais , Animais Geneticamente Modificados , Tamanho Corporal/genética , Drosophila , Ativação Enzimática , Proteínas de Ligação ao GTP/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Deleção de Genes , Substâncias de Crescimento/metabolismo , Substâncias de Crescimento/farmacologia , Humanos , Modelos Biológicos , Neurofibromina 1/metabolismo , Neurofibromina 1/farmacologia , Neurotransmissores , Estrutura Terciária de Proteína , Transdução de Sinais/genética , Proteínas ras/farmacologia
16.
Oncogene ; 25(19): 2708-16, 2006 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-16407839

RESUMO

The proto-oncogene Ras GTPase stimulates transcription of p21Waf1/Cip1 (p21), which is repressed by the RhoA GTPase. We previously showed that Ras also elevates p21 protein levels by reducing its proteasome-mediated degradation. Therefore, we investigated whether RhoA also influenced p21 protein degradation. Pulse-chase analysis of p21 protein stability revealed that inhibitors of Rho function, which disrupt filamentous actin (F-actin), drastically slowed p21 degradation. Direct F-actin disruption mimicked Rho inhibition to stabilize p21. We found that Rho inhibition, or F-actin disruption, activated the JNK stress-activated protein kinase, and that interfering with JNK signalling, but not p38, abrogated p21 stabilization by Rho inhibition or F-actin-disrupting drugs. In addition, Ras-transformation led to increased constitutive JNK activity that contributed to the elevated p21 protein levels. These data suggest that p21 stability is influenced by a mechanism that monitors F-actin downstream of Rho, and which acts through a pathway involving activation of JNK. These results may have significant implications for therapies that target Rho-signalling pathways to induce p21-mediated cell-cycle arrest.


Assuntos
Actinas/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Citoesqueleto/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Northern Blotting , Western Blotting , Transformação Celular Neoplásica , Inibidor de Quinase Dependente de Ciclina p21/genética , Estabilidade Enzimática , Fibroblastos/citologia , Fibroblastos/metabolismo , MAP Quinase Quinase 4/metabolismo , Camundongos , Células NIH 3T3 , Proto-Oncogene Mas , Transdução de Sinais , Células Swiss 3T3 , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas ras/farmacologia , Proteína rhoA de Ligação ao GTP/genética
17.
Biochem Biophys Res Commun ; 332(3): 913-20, 2005 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-15913563

RESUMO

Dexras1/AGS1/RasD1 is a member of the Ras superfamily of monomeric G proteins and has been suggested to disrupt receptor-G protein signaling. We examined the ability of Dexras1 to modulate dopamine D(2L) receptor regulation of adenylyl cyclase (AC) type 1 in HEK293 cells. Acute D(2L) receptor-mediated inhibition of A23187-stimulated AC1 activity (IC50, 4.0+/-1.4 nM; 50+/-3% inhibition) was not altered in the presence of Dexras1 (IC50, 2.4+/-1.3 nM, 50+/-1% inhibition); however, Dexras1 blocked acute D(2L) receptor-mediated activation of ERK 1/2 by approximately 50%. Heterologous sensitization of AC1 induced by persistent activation of D(2L) receptors was completely blocked by Dexras1 under basal and A23187-stimulated conditions. The block of sensitization was concentration-dependent and was not observed with a nucleotide binding-deficient Dexras1G31V mutant. Sensitization of AC1 was Gbetagamma-dependent as demonstrated using the C-terminus of beta-adrenergic receptor kinase (betaARK-ct). These data suggest that Dexras1 selectively regulates receptor-mediated Gbetagamma signaling pathways.


Assuntos
Adenilil Ciclases/metabolismo , Proteínas de Ligação ao GTP/farmacologia , Proteínas ras/farmacologia , Adenilil Ciclases/genética , Sítios de Ligação/genética , Calcimicina/farmacologia , Linhagem Celular , AMP Cíclico/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/metabolismo , Humanos , Cinética , Mutagênese Sítio-Dirigida , Quimpirol/farmacologia , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Transdução de Sinais , Transfecção , Proteínas ras/genética , Proteínas ras/metabolismo
18.
Clin Cancer Res ; 11(5): 2026-37, 2005 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-15756029

RESUMO

It is known that the potential clinical use of farnesyltransferase inhibitors (FTI) could be expanded to include cancers harboring activated receptor tyrosine kinases. Approximately 70% of malignant pleural mesotheliomas (MPM) overexpress epidermal growth factor receptors (EGFR) and a subset express both EGFR and transforming growth factor alpha (TGF-alpha), suggesting an autocrine role for EGFR in MPM. We checked on MPM cells (10 human cell lines, 11 primary cultures obtained by human biopsies, and 7 short-term normal mesothelial cell cultures) concerning the following: (a) the relative overexpression of EGFR (Western blotting, flow cytometry, immunohistochemistry), (b) the relative expression of EGFR ligands (EGF, amphiregulin, TGF-alpha, ELISA), (c) the relative increase of the activated form of Ras (Ras-bound GTP) after EGF stimulation (Ras activation assay), (d) the efficacy of five different FTIs (HDJ2 prenylation, cell cytotoxicity, and apoptosis using ApopTag and gel ladder). EGFR was overexpressed in MPM cells compared with normal pleural mesothelial cells in equivalent levels as in non-small cell lung cancer cells A459. MPM cells constitutively expressed EGFR ligands; however, Ras activation was attenuated at high EGF concentrations (100 ng/mL). Growth of MPM cells was substantially not affected by treatment with different FTIs (SCH66336, BMS-214662, R115777, RPR-115135, and Manumycin). Among these, BMS-214662 was the only one moderately active. BMS-214662 triggered apoptosis in a small fraction of cells (not higher than 30%) that was paralleled by a slight decrease in the levels of TGF-alpha secreted by treated MPM cells. Our data highlighted the concept that the same signaling pathway can be regulated in different ways and these regulations can differ between different cells of different origin.


Assuntos
Alquil e Aril Transferases/farmacologia , Inibidores Enzimáticos/farmacologia , Receptores ErbB/biossíntese , Regulação da Expressão Gênica/efeitos dos fármacos , Mesotelioma/genética , Mesotelioma/fisiopatologia , Neoplasias Pleurais/genética , Neoplasias Pleurais/fisiopatologia , Alquil e Aril Transferases/antagonistas & inibidores , Western Blotting , Proliferação de Células , Farnesiltranstransferase , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Ligantes , Transdução de Sinais , Células Tumorais Cultivadas , Proteínas ras/biossíntese , Proteínas ras/farmacologia
19.
Oncogene ; 24(21): 3492-502, 2005 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-15735677

RESUMO

The Ras-mitogen activated protein kinase (Ras-MAPK) pathway plays an integral role in the formation of human malignancies. Stimulation of this pathway results in phosphorylation of histone H3 at serines 10 and 28 and expression of immediate-early genes. Phosphorylated (serine 10) H3, which is also acetylated on lysine 14, is associated with immediate-early genes. In this report, we investigated the relationship between these two H3 phosphorylation events in parental and ras-transformed fibroblasts. Immunoblot analyses of two-dimensional gel patterns demonstrated that all three H3 variants were phosphorylated after stimulation of the Ras-MAPK pathway and during mitosis. Following stimulation of the Ras-MAPK pathway, H3 phosphorylated on serines 10 and 28 was excluded from regions of highly condensed chromatin and was present in increased levels in ras-transformed cells. Although H3 phosphorylated at serine 10 or 28 was dynamically acetylated, H3 phosphorylated at serine 28 had a higher steady state of acetylation than that of H3 phosphorylated at serine 10. When visualized with indirect immunofluorescence, most foci of phosphorylated serine 28 H3 did not co-localize with foci of H3 phosphorylated on serine 10 or phosphoacetylated on serine 10 and lysine 14, suggesting that these two phosphorylation events act separately to promote gene expression.


Assuntos
Histonas/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/farmacologia , Proteínas ras/farmacologia , Animais , Transformação Celular Neoplásica , Fibroblastos , Técnica Indireta de Fluorescência para Anticorpo , Regulação da Expressão Gênica , Immunoblotting , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/biossíntese , Fosforilação , Serina/metabolismo , Proteínas ras/biossíntese
20.
Oncogene ; 24(6): 1122-7, 2005 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-15592510

RESUMO

Aurora kinases are known to play a key role in maintaining mitotic fidelity, and overexpression of aurora kinases has been noted in various tumors. Overexpression of aurora kinase activity is thought to promote cancer development through a loss of centrosome or chromosome number integrity. Here we observed augmentation of G12V-mutated HRAS-induced neoplastic transformation in BALB/c 3T3 A31-1-1 cells transfected with Aurora-A. Aurora-A-short hairpin RNA (shRNA) experiments showed that the expression level of Aurora-A determines susceptibility to transformation. Aurora-A gene amplification was noted in human patients with tongue or gingival squamous carcinoma (4/11). Amplification was observed even in pathologically normal epithelial tissue taken at sites distant from the tumors in two patients with tongue cancer. However, overexpression of Aurora-A mRNA was observed only within the tumors of all patients examined (11/11). Our data indicate that Aurora-A gene amplification and overexpression play a role in human carcinogenesis, largely due to the effect of Aurora-A on oncogenic cell growth, rather than a loss of maintenance of centrosomal or chromosomal integrity.


Assuntos
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/fisiopatologia , Transformação Celular Neoplásica/genética , Amplificação de Genes , Regulação Neoplásica da Expressão Gênica , Neoplasias Gengivais/genética , Neoplasias Gengivais/fisiopatologia , Proteínas Quinases/biossíntese , Neoplasias da Língua/genética , Neoplasias da Língua/fisiopatologia , Aurora Quinases , Sequência de Bases , Proteínas de Ciclo Celular , Perfilação da Expressão Gênica , Predisposição Genética para Doença , Humanos , Dados de Sequência Molecular , Polimorfismo Genético , Proteínas Serina-Treonina Quinases , RNA Mensageiro/análise , RNA Mensageiro/biossíntese , Proteínas de Xenopus , Proteínas ras/genética , Proteínas ras/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...