Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Semin Cancer Biol ; 50: 90-100, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29175105

RESUMO

FOXO transcription factors are negatively regulated by the PI3K-PKB/AKT signaling pathway and have been mainly considered to be tumor suppressors due to their inhibitory effect on cancer cell growth and survival. However, FOXOs can also support tumor development and progression by maintaining cellular homeostasis, facilitating metastasis and inducing therapy resistance. In agreement with these opposing views on the role of FOXOs in cancer, studies using FOXO levels or activity as prognostic markers for cancer patient disease progression and survival came to contradicting results. While it is clear that FOXOs are involved in various aspects of cancer, it is debatable whether FOXOs function as tumor suppressors or supporters, or may be both depending on the context. In this review, we describe the role of FOXOs in signaling pathways and processes relevant to cancer and evaluate recent advances in understanding the role of FOXOs in cancer. Based on recent insights it becomes clear that FOXOs may not be classical tumor suppressors and that targeting FOXO activity might hold promise in cancer therapy.


Assuntos
Carcinogênese/genética , Fatores de Transcrição Forkhead/genética , Neoplasias/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias/patologia , Proteína Oncogênica v-akt/genética , Fosfatidilinositol 3-Quinases/genética , Transdução de Sinais/genética
2.
Trends Biochem Sci ; 20(1): 18-22, 1995 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-7878738

RESUMO

A powerful combination of genetics and biochemistry has provided details of how Ras-directed signalling interacts with and is regulated by other cellular signalling pathways. This might ultimately lead to the control of deregulated signalling by oncogenic Ras. Recently, progress has been made in understanding the regulation of Ras-mediated activation of the Raf-1-ERK2 kinase cascade through crosstalk with protein kinase C and cyclic-AMP-dependent protein kinase.


Assuntos
Genes ras , Proteínas Serina-Treonina Quinases/fisiologia , Transdução de Sinais/genética , Ativação Enzimática , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-raf
3.
Mol Cell Biol ; 19(6): 4525-34, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10330191

RESUMO

Recent studies indicate that phosphatidylinositide-3OH kinase (PI3K)-induced S6 kinase (S6K1) activation is mediated by protein kinase B (PKB). Support for this hypothesis has largely relied on results obtained with highly active, constitutively membrane-localized alleles of wild-type PKB, whose activity is independent of PI3K. Here we set out to examine the importance of PKB signaling in S6K1 activation. In parallel, glycogen synthase kinase 3beta (GSK-3beta) inactivation and eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1) phosphorylation were monitored as markers of the rapamycin-insensitive and -sensitive branches of the PI3K signaling pathway, respectively. The results demonstrate that two activated PKBalpha mutants, whose basal activity is equivalent to that of insulin-induced wild-type PKB, inhibit GSK-3beta to the same extent as a highly active, constitutively membrane-targeted wild-type PKB allele. However, of these two mutants, only the constitutively membrane-targeted allele of PKB induces S6K1 activation. Furthermore, an interfering mutant of PKB, which blocks insulin-induced PKB activation and GSK-3beta inactivation, has no effect on S6K1 activation. Surprisingly, all the activated PKB mutants, regardless of constitutive membrane localization, induce 4E-BP1 phosphorylation and the interfering PKB mutant blocks insulin-induced 4E-BP1 phosphorylation. The results demonstrate that PKB mediates S6K1 activation only as a function of constitutive membrane localization, whereas the activation of PKB appears both necessary and sufficient to induce 4E-BP1 phosphorylation independently of its intracellular location.


Assuntos
Proteínas de Transporte , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Quinases S6 Ribossômicas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Androstadienos/farmacologia , Western Blotting , Proteínas de Ciclo Celular , Linhagem Celular , Membrana Celular/metabolismo , Inibidores Enzimáticos/farmacologia , Imunofluorescência , Regulação da Expressão Gênica , Humanos , Insulina/farmacologia , Rim/metabolismo , Modelos Biológicos , Modelos Genéticos , Mutagênese , Fosforilação , Testes de Precipitina , Proteínas Proto-Oncogênicas c-akt , Proteínas Recombinantes de Fusão , Transdução de Sinais , Sirolimo/farmacologia , Wortmanina
4.
Mol Cell Biol ; 21(10): 3534-46, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11313479

RESUMO

AFX belongs to a subfamily of Forkhead transcription factors that are phosphorylated by protein kinase B (PKB), also known as Akt. Phosphorylation inhibits the transcriptional activity of AFX and changes the steady-state localization of the protein from the nucleus to the cytoplasm. Our goal was threefold: to identify the cellular compartment in which PKB phosphorylates AFX, to determine whether the nuclear localization of AFX plays a role in regulating its transcriptional activity, and to elucidate the mechanism by which phosphorylation alters the localization of AFX. We show that phosphorylation of AFX by PKB occurs in the nucleus. In addition, nuclear export mediated by the export receptor, Crm1, is required for the inhibition of AFX transcriptional activity. Both phosphorylated and unphosphorylated AFX, however, bind Crm1 and can be exported from the nucleus. These results suggest that export is unregulated and that phosphorylation by PKB is not required for the nuclear export of AFX. We show that AFX enters the nucleus by an active, Ran-dependent mechanism. Amino acids 180 to 221 of AFX comprise a nonclassical nuclear localization signal (NLS). S193, contained within this atypical NLS, is a PKB-dependent phosphorylation site on AFX. Addition of a negative charge at S193 by mutating the residue to glutamate reduces nuclear accumulation. PKB-mediated phosphorylation of AFX, therefore, attenuates the import of the transcription factor, which shifts the localization of the protein from the nucleus to the cytoplasm and results in the inhibition of AFX transcriptional activity.


Assuntos
Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/fisiologia , Fatores de Transcrição/fisiologia , Células 3T3 , Animais , Transporte Biológico/fisiologia , Proteínas de Ciclo Celular , Núcleo Celular/fisiologia , Fatores de Transcrição Forkhead , Camundongos , Fosforilação , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais , Transcrição Gênica
5.
Mol Cell Biol ; 21(23): 8225-35, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11689711

RESUMO

AFX is a Forkhead transcription factor that induces a G(1) cell cycle arrest via upregulation of the cell cycle inhibitor p27(Kip1). Previously we have shown that protein kinase B (PKB) phosphorylates AFX causing inhibition of AFX by nuclear exclusion. In addition, Ras, through the activation of the RalGEF-Ral pathway, induces phosphorylation of AFX. Here we show that the Ras-Ral pathway provokes phosphorylation of threonines 447 and 451 in the C terminus of AFX. A mutant protein in which both threonines are substituted for alanines (T447A/T451A) still responds to PKB-regulated nuclear-cytoplasmic shuttling, but transcriptional activity and consequent G(1) cell cycle arrest are greatly impaired. Furthermore, inhibition of the Ral signaling pathway abolishes both AFX-mediated transcription and regulation of p27(Kip1), while activation of Ral augments AFX activity. From these results we conclude that Ral-mediated phosphorylation of threonines 447 and 451 is required for proper activity of AFX-WT. Interestingly, the T447A/T451A mutation did not affect the induction of transcription and G(1) cell cycle arrest by the PKB-insensitive AFX-A3 mutant, suggesting that Ral-mediated phosphorylation plays a role in the regulation of AFX by PKB.


Assuntos
Regulação da Expressão Gênica/fisiologia , Proteínas Serina-Treonina Quinases , Fatores de Transcrição/metabolismo , Proteínas ral de Ligação ao GTP/metabolismo , Células 3T3 , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Sítios de Ligação/fisiologia , Proteínas de Ciclo Celular , Núcleo Celular/metabolismo , Ativação Enzimática/fisiologia , Fatores de Transcrição Forkhead , Genes Reporter , Humanos , Camundongos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fosforilação , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Relação Estrutura-Atividade , Treonina/metabolismo , Fatores de Transcrição/genética , Transfecção , Células Tumorais Cultivadas , Proteínas ras/metabolismo
6.
Mol Cell Biol ; 18(4): 1802-11, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9528752

RESUMO

Regulation of phosphoinositide 3-kinase (PI 3-kinase) can occur by binding of the regulatory p85 subunit to tyrosine-phosphorylated proteins and by binding of the p110 catalytic subunit to activated Ras. However, the way in which these regulatory mechanisms act to regulate PI 3-kinase in vivo is unclear. Here we show that several growth factors (basic fibroblast growth factor [bFGF], platelet-derived growth factor [PDGF], and epidermal growth factor [EGF; to activate an EGF receptor-Ret chimeric receptor]) all activate PI 3-kinase in vivo in the neuroectoderm-derived cell line SKF5. However, these growth factors differ in their ability to activate PI 3-kinase-dependent signaling. PDGF and EGF(Ret) treatment induced PI 3-kinase-dependent lamellipodium formation and protein kinase B (PKB) activation. In contrast, bFGF did not induce lamellipodium formation but activated PKB, albeit to a small extent. PDGF and EGF(Ret) stimulation resulted in binding of p85 to tyrosine-phosphorylated proteins and strong Ras activation. bFGF, however, induced only strong activation of Ras. In addition, while RasAsn17 abolished bFGF activation of PKB, PDGF- and EGF(Ret)-induced PKB activation was only partially inhibited and lamellipodium formation was unaffected. Interestingly, in contrast to activation of only endogenous Ras (bFGF), ectopic expression of activated Ras did result in lamellipodium formation. From this we conclude that, in vivo, p85 and Ras synergize to activate PI 3-kinase and that strong activation of only endogenous Ras exerts a small effect on PI 3-kinase activity, sufficient for PKB activation but not lamellipodium formation. This differential sensitivity to PI 3-kinase activation could be explained by our finding that PKB activation and lamellipodium formation are independent PI 3-kinase-induced events.


Assuntos
Movimento Celular , Proteínas de Drosophila , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/metabolismo , Proteínas ras/metabolismo , Androstadienos/farmacologia , Cromonas/farmacologia , Citoplasma/metabolismo , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/genética , Receptores ErbB/metabolismo , Fator 2 de Crescimento de Fibroblastos/farmacologia , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Humanos , Morfolinas/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-akt , Proteínas Proto-Oncogênicas c-ret , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais , Células Tumorais Cultivadas , Tirosina/metabolismo , Wortmanina
7.
Mol Cell Biol ; 9(10): 4312-22, 1989 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-2555688

RESUMO

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.


Assuntos
Substâncias de Crescimento/fisiologia , Proteína Oncogênica p21(ras)/fisiologia , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Animais , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Transformada , Transformação Celular Neoplásica/metabolismo , AMP Cíclico/fisiologia , Replicação do DNA/efeitos dos fármacos , Expressão Gênica , Insulina/fisiologia , Fator de Crescimento Insulin-Like I/fisiologia , Fosforilação , Receptor de Insulina/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores de Somatomedina , Transdução de Sinais/fisiologia
8.
Mol Cell Biol ; 20(15): 5469-78, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10891487

RESUMO

The serine/threonine kinase Akt (also known as protein kinase B) (Akt/PKB) is activated upon T-cell antigen receptor (TCR) engagement or upon expression of an active form of phosphatidylinositide (PI) 3-kinase in T lymphocytes. Here we report that the small GTPase Rac1 is implicated in this pathway, connecting the receptor with the lipid kinase. We show that in Jurkat cells, activated forms of Rac1 or Cdc42, but not Rho, stimulate an increase in Akt/PKB activity. TCR-induced Akt/PKB activation is inhibited either by PI 3-kinase inhibitors (LY294002 and wortmannin) or by overexpression of a dominant negative mutant of Rac1 but not Cdc42. Accordingly, triggering of the TCR rapidly stimulates a transient increase in GTP-Rac content in these cells. Similar to TCR stimulation, L61Rac-induced Akt/PKB kinase activity is also LY294002 and wortmannin sensitive. However, induction of Akt/PKB activity by constitutive active PI 3-kinase is unaffected when dominant negative Rac1 is coexpressed, placing Rac1 upstream of PI 3-kinase in the signaling pathway. When analyzing the signaling hierarchy in the pathway leading to cytoskeleton rearrangements, we found that Rac1 acts downstream of PI 3-kinase, a finding that is in accordance with numerous studies in fibroblasts. Our results reveal a previously unrecognized role of the GTPase Rac1, acting upstream of PI 3-kinase in linking the TCR to Akt/PKB. This is the first report of a membrane receptor employing Rac1 as a downstream transducer for Akt/PKB activation.


Assuntos
Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Cromonas/farmacologia , Citoesqueleto/metabolismo , Citoesqueleto/ultraestrutura , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Humanos , Morfolinas/farmacologia , Mutação , Fosfatidilinositol 3-Quinases/genética , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Proteínas Proto-Oncogênicas c-akt , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/metabolismo , Treonina/metabolismo , Células Tumorais Cultivadas , Proteínas rac1 de Ligação ao GTP/genética
9.
Mol Cell Biol ; 20(22): 8480-8, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11046144

RESUMO

The transcription factor c-Jun is critically involved in the regulation of proliferation and differentiation as well as cellular transformation induced by oncogenic Ras. The signal transduction pathways that couple Ras activation to c-Jun phosphorylation are still partially elusive. Here we show that an activated version of the Ras effector Rlf, a guanine nucleotide exchange factor (GEF) of the small GTPase Ral, can induce the phosphorylation of serines 63 and 73 of c-Jun. In addition, we show that growth factor-induced, Ras-mediated phosphorylation of c-Jun is abolished by inhibitory mutants of the RalGEF-Ral pathway. These results suggest that the RalGEF-Ral pathway plays a major role in Ras-dependent c-Jun phosphorylation. Ral-dependent regulation of c-Jun phosphorylation includes JNK, a still elusive JNKK, and possibly Src.


Assuntos
Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fator ral de Troca do Nucleotídeo Guanina/metabolismo , Proteínas ras/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular/efeitos dos fármacos , Ativação Enzimática , Fatores de Troca do Nucleotídeo Guanina , Humanos , Insulina/metabolismo , Insulina/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno , MAP Quinase Quinase 4 , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Mutação , Fosforilação , Pirazóis/farmacologia , Pirimidinas/farmacologia , Serina , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas ral de Ligação ao GTP/metabolismo , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo
10.
Mol Cell Biol ; 9(7): 3114-6, 1989 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-2674680

RESUMO

In 7 of 37 patients with cutaneous melanoma, mutations in the N-ras gene were found. The primary tumors of these seven patients were exclusively localized on body sites continuously exposed to sunlight. Moreover, the ras mutations were all at or near dipyrimidine sites known to be targets of UV damage. Two primary tumors were biclonal with respect to ras mutation. An active role for UV irradiation in induction of the mutations is suggested.


Assuntos
Genes ras , Melanoma/genética , Mutação , Neoplasias Cutâneas/genética , Luz Solar/efeitos adversos , Raios Ultravioleta/efeitos adversos , DNA de Neoplasias/genética , Feminino , Regulação da Expressão Gênica , Humanos , Masculino , Melanoma/patologia , Melanoma/secundário , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/secundário , Células Tumorais Cultivadas
11.
Mol Cell Biol ; 13(12): 7248-56, 1993 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8246947

RESUMO

Expression of p21rasAsn-17, a dominant negative mutant of p21ras that blocks p21ras activation by growth factors, inhibits activation of extracellular signal-regulated kinase 2 (ERK2) by insulin and platelet-derived growth factor in rat-1 cells [A. M. M. de Vries-Smits, B. M. T. Burgering, S. J. Leevers, C. J. Marshall, and J. L. Bos, Nature (London) 357:602-604, 1992]. Here we report that expression of p21rasAsn-17 does not abolish epidermal growth factor (EGF)-induced phosphorylation of ERK2 in fibroblasts. Since EGF activates p21ras in these cells, this indicates that EGF induces a p21ras-independent pathway for the phosphorylation of ERK2 as well. We investigated whether activation of protein kinase C (PKC) or increase in intracellular calcium could be involved in p21ras-independent signaling. In rat-1 cells, inhibition of either PKC, by prolonged 12-O-tetradecanoylphorbol-13-acetate (TPA) pretreatment, or calcium influx, by ethylene glycol-bis(beta-aminoethyl ether)-N,N,N',N'-tetraacetic acid (EGTA) pretreatment, did not abolish EGF-induced ERK2 phosphorylation. However, a combined inhibition of both p21ras and calcium influx, but not PKC, resulted in a complete inhibition of EGF-induced ERK2 phosphorylation. In contrast, in Swiss 3T3 cells, inhibition of both p21ras activation and TPA-sensitive PKC, but not calcium influx, inhibited EGF-induced ERK2 phosphorylation. These results demonstrate that in fibroblasts, EGF induces alternative pathways of ERK2 phosphorylation in a cell-type-specific manner.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Animais , Cálcio/metabolismo , Linhagem Celular , Ácido Egtázico/farmacologia , Camundongos , Proteína Quinase 1 Ativada por Mitógeno , Fosforilação , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Ratos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Acetato de Tetradecanoilforbol/farmacologia
12.
Mol Cell Biol ; 20(24): 9138-48, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11094066

RESUMO

Interleukin-3 (IL-3), IL-5, and granulocyte-macrophage colony-stimulating factor regulate the survival, proliferation, and differentiation of hematopoietic lineages. Phosphatidylinositol 3-kinase (PI3K) has been implicated in the regulation of these processes. Here we investigate the molecular mechanism by which PI3K regulates cytokine-mediated proliferation and survival in the murine pre-B-cell line Ba/F3. IL-3 was found to repress the expression of the cyclin-dependent kinase inhibitor p27(KIP1) through activation of PI3K, and this occurs at the level of transcription. This transcriptional regulation occurs through modulation of the forkhead transcription factor FKHR-L1, and IL-3 inhibited FKHR-L1 activity in a PI3K-dependent manner. We have generated Ba/F3 cell lines expressing a tamoxifen-inducible active FKHR-L1 mutant [FKHR-L1(A3):ER*]. Tamoxifen-mediated activation of FKHR-L1(A3):ER* resulted in a striking increase in p27(KIP1) promoter activity and mRNA and protein levels as well as induction of the apoptotic program. The level of p27(KIP1) appears to be critical in the regulation of cell survival since mere ectopic expression of p27(KIP1) was sufficient to induce Ba/F3 apoptosis. Moreover, cell survival was increased in cytokine-starved bone marrow-derived stem cells from p27(KIP1) null-mutant mice compared to that in cells from wild-type mice. Taken together, these observations indicate that inhibition of p27(KIP1) transcription through PI3K-induced FKHR-L1 phosphorylation provides a novel mechanism of regulating cytokine-mediated survival and proliferation.


Assuntos
Proteínas de Ciclo Celular , Citocinas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Inibidores Enzimáticos/metabolismo , Interleucina-3/metabolismo , Proteínas Associadas aos Microtúbulos/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica/genética , Proteínas Supressoras de Tumor , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Linfócitos B/citologia , Linfócitos B/metabolismo , Linhagem Celular , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p27 , Quinases Ciclina-Dependentes/antagonistas & inibidores , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Inibidores Enzimáticos/farmacologia , Eosinófilos/efeitos dos fármacos , Eosinófilos/metabolismo , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead , Regulação da Expressão Gênica , Genes Reporter/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Hidroxitestosteronas/farmacologia , Interleucina-3/genética , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Mutação , PTEN Fosfo-Hidrolase , Monoéster Fosfórico Hidrolases/genética , Monoéster Fosfórico Hidrolases/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , RNA/metabolismo , Transdução de Sinais , Sirolimo/farmacologia , Tamoxifeno/farmacologia , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética
13.
Cell Death Differ ; 23(9): 1483-92, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27035620

RESUMO

Loss of cellular adhesion leads to the progression of breast cancer through acquisition of anchorage independence, also known as resistance to anoikis. Although inactivation of E-cadherin is essential for acquisition of anoikis resistance, it has remained unclear how metastatic breast cancer cells counterbalance the induction of apoptosis without E-cadherin-dependent cellular adhesion. We report here that E-cadherin inactivation in breast cancer cells induces PI3K/AKT-dependent FOXO3 inhibition and identify FOXO3 as a novel and direct transcriptional activator of the pro-apoptotic protein BMF. As a result, E-cadherin-negative breast fail to upregulate BMF upon transfer to anchorage independence, leading to anoikis resistance. Conversely, expression of BMF in E-cadherin-negative metastatic breast cancer cells is sufficient to inhibit tumour growth and dissemination in mice. In conclusion, we have identified repression of BMF as a major cue that underpins anoikis resistance and tumour dissemination in E-cadherin-deficient metastatic breast cancer.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Neoplasias da Mama/patologia , Caderinas/metabolismo , Proteína Forkhead Box O3/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Anoikis/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Proteína 11 Semelhante a Bcl-2/antagonistas & inibidores , Proteína 11 Semelhante a Bcl-2/genética , Proteína 11 Semelhante a Bcl-2/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/mortalidade , Caderinas/genética , Linhagem Celular Tumoral , Doxiciclina/farmacologia , Doxiciclina/uso terapêutico , Feminino , Humanos , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Células MCF-7 , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , Transdução de Sinais , Ativação Transcricional
14.
Oncogene ; 35(17): 2166-77, 2016 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-26279295

RESUMO

Melanoma is the most lethal form of skin cancer and successful treatment of metastatic melanoma remains challenging. BRAF/MEK inhibitors only show a temporary benefit due to rapid occurrence of resistance, whereas immunotherapy is mainly effective in selected subsets of patients. Thus, there is a need to identify new targets to improve treatment of metastatic melanoma. To this extent, we searched for markers that are elevated in melanoma and are under regulation of potentially druggable enzymes. Here, we show that the pro-proliferative transcription factor FOXM1 is elevated and activated in malignant melanoma. FOXM1 activity correlated with expression of the enzyme Pin1, which we found to be indicative of a poor prognosis. In functional experiments, Pin1 proved to be a main regulator of FOXM1 activity through MEK-dependent physical regulation during the cell cycle. The Pin1-FOXM1 interaction was enhanced by BRAF(V600E), the driver oncogene in the majority of melanomas, and in extrapolation of the correlation data, interference with\ Pin1 in BRAF(V600E)-driven metastatic melanoma cells impaired both FOXM1 activity and cell survival. Importantly, cell-permeable Pin1-FOXM1-blocking peptides repressed the proliferation of melanoma cells in freshly isolated human metastatic melanoma ex vivo and in three-dimensional-cultured patient-derived melanoids. When combined with the BRAF(V600E)-inhibitor PLX4032 a robust repression in melanoid viability was obtained, establishing preclinical value of patient-derived melanoids for prognostic use of drug sensitivity and further underscoring the beneficial effect of Pin1-FOXM1 inhibitory peptides as anti-melanoma drugs. These proof-of-concept results provide a starting point for development of therapeutic Pin1-FOXM1 inhibitors to target metastatic melanoma.


Assuntos
Proteína Forkhead Box M1/genética , Melanoma/tratamento farmacológico , Peptidilprolil Isomerase de Interação com NIMA/genética , Proteínas Proto-Oncogênicas B-raf/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Indóis/administração & dosagem , Melanoma/genética , Melanoma/patologia , Terapia de Alvo Molecular , Mutação , Metástase Neoplásica , Inibidores de Proteínas Quinases/administração & dosagem , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Transdução de Sinais , Sulfonamidas/administração & dosagem , Vemurafenib
15.
Oncogene ; 10(5): 919-25, 1995 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-7898933

RESUMO

Association of the p21ras guanine nucleotide exchange factor mSOS with tyrosine-phosphorylated Shc has been implicated in the activation of p21ras. In addition, after growth factor stimulation mSOS becomes phosphorylated as indicated by the appearance of a form of mSOS with reduced electrophoretic mobility. This phosphorylation is delayed with respect to Shc-Grb2-mSOS complex formation and activation of p21ras. To investigate the role of mSOS phosphorylation in further detail we have investigated the effect of phosphorylation on mSOS complex formation and p21ras activation. We found that Shc is associated with the unphosphorylated, faster migrating form of mSOS. Furthermore, although there is a correlation between the amount of complexes formed and the activation of p21ras, there is no such a correlation between mSOS phosphorylation and p21ras activation. In addition, inhibition of mSOS phosphorylation did not affect complex formation of mSOS with tyrosine phosphorylated Shc. Also, induction of mSOS phosphorylation prior to complex formation did not affect EGF-induced association of mSOS with Shc significantly, and Shc still associated predominantly with the faster migrating form of mSOS. From these results we conclude that the unphosphorylated form of mSOS is associated with Shc and that perhaps a phosphorylation-dephosphorylation step is part of the mSOS activation-inactivation cycle.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas/metabolismo , Células 3T3 , Animais , Receptores ErbB/metabolismo , Proteína Adaptadora GRB2 , Fatores de Troca do Nucleotídeo Guanina , Camundongos , Fosforilação , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Tirosina/metabolismo , Fatores ras de Troca de Nucleotídeo Guanina
16.
Oncogene ; 11(3): 561-9, 1995 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-7543196

RESUMO

The exposure of mammalian cells to ultraviolet radiation (UV) may lead to DNA damage resulting in mutation and thus possibly cancer, while irradiation can further act as a potent tumor promoter. In addition UV induces p21ras-mediated signalling leading to activation of transcription factors such as AP-1 and NF-kappa B, as well as activation of the Src tyrosine kinase. This 'UV-response' has been well studied in mammalian cells and furthermore is conserved in yeast, however the most upstream components of this signal transduction pathway have remained elusive. Here we show that UV rapidly activates both the EGF receptor and insulin receptor, as shown by tyrosine phosphorylation of these receptors. We demonstrate that this activation is due to autophosphorylation as it only occurs in cells containing receptors with a functional kinase domain. We have further analysed the propagation of the UV-induced signal to downstream events such as, IRS-1 and Shc tyrosine phosphorylation, phosphatidylinositol 3-kinase activation, leukotriene synthesis, MAP kinase activation and gene induction all of which are activated by UV irradiation. Importantly, we demonstrate that in cells expressing a 'kinase-dead' receptor mutant the UV-response is inhibited, blocking leukotriene synthesis, MAP kinase activation and transcriptional induction. Furthermore, prior-stimulation of cells with UV appears to reduce further responsiveness to addition of growth factor suggesting a common signaling pathway. These data demonstrate a critical role for receptor-mediated events in regulating the response mammalian cells to UV exposure.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas Adaptadoras de Transporte Vesicular , Receptores ErbB/efeitos da radiação , Receptor de Insulina/efeitos da radiação , Células 3T3 , Animais , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/metabolismo , Insulina/farmacologia , Proteínas Substratos do Receptor de Insulina , Camundongos , Proteína Quinase 1 Ativada por Mitógeno , Fosfoproteínas/metabolismo , Fosforilação , Fosfotirosina , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-raf , Receptor de Insulina/metabolismo , Proteínas Adaptadoras da Sinalização Shc , Transdução de Sinais/efeitos da radiação , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src , Tirosina/análogos & derivados , Tirosina/metabolismo , Raios Ultravioleta
17.
Oncogene ; 11(11): 2207-14, 1995 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-8570170

RESUMO

Ret is a receptor tyrosine kinase predominantly expressed in tissue derived from the neuroectoderm and is involved in multiple endocrine neoplasia type 2A and 2B, familiar medullary thyroid carcinoma, and Hirschsprung's disease. The ligand for the receptor is still unknown. Previously, using a human epidermal growth factor receptor - Ret chimaeric receptor (HERRet) stably transfected into fibroblasts, it was shown that Ret activation induces the activation of p21ras, but, surprisingly, activation of extracellular signal-regulated kinase 2 (ERK2) was not observed (Santoro et al. (1994) Mol. Cell. Biol., 14, 663). In this report we describe early signaling events induced by the activated HERRet fusion receptor in a cell line derived from neuroectodermal tissue, SK-N-MC. In these cells, activated HERRet induces tyrosine phosphorylation of Shc, complex formation of Shc with Grb2 and Sos and activation of p21ras. Importantly, also ERK2 is activated. This activation was strong and sustained for at least 2 h. Activation was abolished by the dominant negative p21rasasn17 mutant, showing that activation of ERK2 is mediated by p21ras. These results suggest that Ret can induce ERK2 activation in a p21ras dependent manner in cells derived from tissue where Ret is endogenously expressed.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Proteínas de Drosophila , Proteínas Proto-Oncogênicas/fisiologia , Receptores Proteína Tirosina Quinases/fisiologia , AMP Cíclico/farmacologia , Ativação Enzimática , Proteína Adaptadora GRB2 , Humanos , Proteínas de Membrana/metabolismo , Proteína Quinase 1 Ativada por Mitógeno , Fosforilação , Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-ret , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Transdução de Sinais , Proteínas Son Of Sevenless , Células Tumorais Cultivadas , Tirosina/metabolismo
18.
Oncogene ; 17(22): 2811-8, 1998 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-9879987

RESUMO

Expression of the proto-oncogene c-myc stimulates cell proliferation in the presence of the appropriate survival factors and triggers apoptosis in their absence; this dual capacity ensures that cell growth is restricted to the correct paracrine environment and is thereby strictly controlled. Recently our laboratory demonstrated that c-Myc-induced apoptosis requires the CD95 death receptor pathway and that insulin-like growth factor (IGF-1) signalling suppresses this killing. To investigate further the links between c-Myc and IGF-1 pathways in CD95-induced apoptosis, we examined the effects of c-Myc and a downstream IGF-1 survival kinase, Akt, on killing mediated by CD95 and its recruited effector proteins (FADD and caspase-8). Here, we show that c-Myc activation does not exacerbate killing induced by FADD or pro-caspase-8, which narrows the point at which c-Myc exerts its action downstream of the interaction of CD95 with its ligand and upstream of FADD. We show further that activated Akt suppresses CD95-induced apoptosis and that Akt exerts its activity at a point downstream of FADD but upstream of caspase-8. These results restrict the possible mechanisms by which CD95-induced apoptosis is modulated by death signals and survival factors.


Assuntos
Apoptose , Proteínas de Arabidopsis , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Proto-Oncogênicas , Transdução de Sinais , Receptor fas/metabolismo , Células 3T3 , Animais , Anticorpos/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Caspase 8 , Caspase 9 , Caspases/genética , Caspases/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Meios de Cultura Livres de Soro , Ácidos Graxos Dessaturases/antagonistas & inibidores , Ácidos Graxos Dessaturases/genética , Ácidos Graxos Dessaturases/metabolismo , Camundongos , Proteínas de Plantas/antagonistas & inibidores , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Precursores de Proteínas/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas c-akt , Ratos , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacologia , Transfecção , Receptor fas/imunologia
19.
Oncogene ; 15(21): 2529-39, 1997 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-9399641

RESUMO

The binding of insulin to its receptor initiates multiple signal transduction pathways regulating such diverse processes as proliferation, differentiation, glucose transport, and glycogen metabolism. The STAT-family of transcription factors has been demonstrated to play a critical role in gene induction by a variety of hemopoietic cytokines and hormones. Furthermore, constitutive activation of STATs is observed in transformed cells. Here we describe activation of a transcriptional complex binding to a consensus STAT-transcriptional element in response to insulin challenge. This complex is induced rapidly after tyrosine autophosphorylation of the insulin receptor, and is sustained for several hours. Supershift analysis of the insulin-induced complex reveals that it specifically contains the transcription factor Stat3. DAN binding of this complex is inhibited by pre-incubation with tyrosine, but not serine/threonine protein kinase inhibitors, whereas transcriptional activation is inhibited by both. Utilising a dominant negative mutant of p21ras we demonstrate that both insulin-induced Stat3 DNA-binding and also transactivation do not require p21ras. Furthermore, although previous studies have suggested a role for MAP kinases (ERKs) and PI-3K in STAT activation, utilising the specific MEK inhibitor PD098059 and the PI-3K inhibitor wortmannin, we demonstrate that activation of ERKs or PI-3K are not required for insulin induced Stat3 phosphorylation or transactivation.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/fisiologia , Proteínas de Ligação a DNA/metabolismo , Insulina/farmacologia , Proteínas Quinases Ativadas por Mitógeno , Fosfatidilinositol 3-Quinases/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transativadores/metabolismo , Células 3T3 , Animais , Células Cultivadas , DNA/metabolismo , Camundongos , Proteína Quinase 1 Ativada por Mitógeno , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Proto-Oncogênicas p21(ras)/fisiologia , Fator de Transcrição STAT3 , Ativação Transcricional
20.
Oncogene ; 13(2): 353-62, 1996 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-8710374

RESUMO

The small GTPase Rap 1A is a close relative of Ras that, when overexpressed, is able to revert oncogenic transformation induced by active Ras. We screened a mouse embryonic cDNA library using the yeast two-hybrid system and isolated the cDNA of a novel Rap 1A-interacting protein. The open reading frame encodes for an 84 kDa protein with a Cdc25-homology domain which shares approximately 30% identity with Ral guanine nucleotide dissociation stimulator (RalGDS) and RalGDS-like (Rg1). The C-terminal region reveals a striking conservation of sequences with the Ras-binding domain of RalGDS. We designated this protein Rlf, for RalGDS-like factor. In the yeast system, Rlf interacts with Rap 1A, H-Ras and R-Ras, but not with Rac and Rho. In addition, we found that Rlf interacts with Rap 1Aval12 but not with Rap 1AAsn17. In vitro binding studies revealed that a C-terminally located 91 amino acid region of Rlf is sufficient for direct association with the GTP-bound form of Ras and Rap 1A. The observed dissociation constants are 0.6 microM and 0.4 microM, respectively. No significant association with Ras-GDP or Rap 1A-GDP could be detected. These binding characteristics indicate that Rlf is a putative effector for Ras and Rap 1A.


Assuntos
DNA Complementar/genética , DNA Complementar/isolamento & purificação , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/isolamento & purificação , Fatores de Troca do Nucleotídeo Guanina , Fatores de Transcrição/genética , Fatores de Transcrição/isolamento & purificação , Proteínas ras/genética , Proteínas ras/isolamento & purificação , ras-GRF1 , Sequência de Aminoácidos , Animais , Sequência de Bases , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Clonagem Molecular , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Guanosina Trifosfato/metabolismo , Camundongos , Dados de Sequência Molecular , Ligação Proteica , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae , Homologia de Sequência de Aminoácidos , Fatores de Transcrição/metabolismo , Fator ral de Troca do Nucleotídeo Guanina , Proteínas rap de Ligação ao GTP , Proteínas ras/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA