Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Oncogene ; 37(4): 427-438, 2018 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-28967905

RESUMO

Deregulated AKT kinase activity due to PTEN deficiency in cancer cells contributes to oncogenesis by incompletely understood mechanisms. Here, we show that PTEN deletion in HCT116 and DLD1 colon carcinoma cells leads to suppression of CHK1 and CHK2 activation in response to irradiation, impaired G2 checkpoint proficiency and radiosensitization. These defects are associated with reduced expression of MRE11, RAD50 and NBS1, components of the apical MRE11/RAD50/NBS1 (MRN) DNA damage response complex. Consistent with reduced MRN complex function, PTEN-deficient cells fail to resect DNA double-strand breaks efficiently after irradiation and show greatly diminished proficiency for DNA repair via the error-free homologous recombination (HR) repair pathway. MRE11 is highly unstable in PTEN-deficient cells but stability can be significantly restored by inhibiting mTORC1 or p70S6 kinase (p70S6K), downstream kinases whose activities are stimulated by AKT, or by mutating a residue in MRE11 that we show is phosphorylated by p70S6K in vitro. In primary human fibroblasts, activated AKT suppresses MRN complex expression to escalate RAS-induced DNA damage and thereby reinforce oncogene-induced senescence. Taken together, our data demonstrate that deregulation of the PI3K-AKT/ mTORC1/ p70S6K pathways, an event frequently observed in cancer, exert profound effects on genome stability via MRE11 with potential implications for tumour initiation and therapy.


Assuntos
Instabilidade Genômica/genética , Proteína Homóloga a MRE11/genética , Neoplasias/genética , PTEN Fosfo-Hidrolase/deficiência , Reparo de DNA por Recombinação/genética , Dano ao DNA/efeitos da radiação , Regulação para Baixo , Fibroblastos , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Instabilidade Genômica/efeitos da radiação , Células HCT116 , Humanos , Proteína Homóloga a MRE11/antagonistas & inibidores , Proteína Homóloga a MRE11/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Neoplasias/radioterapia , PTEN Fosfo-Hidrolase/genética , Fosforilação , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Pirimidinonas/farmacologia , RNA Interferente Pequeno/metabolismo , Tolerância a Radiação/genética , Reparo de DNA por Recombinação/efeitos da radiação , Proteínas Quinases S6 Ribossômicas 70-kDa/antagonistas & inibidores , Proteínas Quinases S6 Ribossômicas 70-kDa/genética , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais/genética , Tionas/farmacologia , Raios X/efeitos adversos
2.
Oncogene ; 25(39): 5359-69, 2006 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-16619043

RESUMO

Chk1 plays a crucial role in the DNA damage and replication checkpoints in vertebrates and may therefore be an important determinant of tumour cell responses to genotoxic anticancer drugs. To evaluate this concept we compared the effects of the nucleoside analogue 5-fluorouracil (5FU) on cell cycle progression and clonogenic survival in DT40 B-lymphoma cells with an isogenic mutant derivative in which Chk1 function was ablated by gene targeting. We show that 5FU activates Chk1 in wild-type DT40 cells and that 5FU-treated cells accumulate in the S phase of the cell cycle due to slowing of the overall rate of DNA replication. In marked contrast, Chk1-deficient DT40 cells fail to slow DNA replication upon initial exposure to 5FU, despite equivalent inhibition of the target enzyme thymidylate synthase, and instead accumulate progressively in the G1 phase of the following cell cycle. This G1 accumulation cannot be reversed rapidly by exogenous thymidine or removal of 5FU, and is associated with increased incorporation of 5FU into genomic DNA and severely diminished clonogenic survival. Taken together, these results demonstrate that a Chk1-dependent replication checkpoint which slows S phase progression can protect tumour cells against the cytotoxic effects of 5FU.


Assuntos
Fluoruracila/toxicidade , Proteínas Quinases/metabolismo , Fase S/fisiologia , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Quinase 1 do Ponto de Checagem , Dano ao DNA , Replicação do DNA , Ativação Enzimática , Humanos , Linfoma de Células B , Fase S/efeitos dos fármacos
3.
Curr Biol ; 10(18): 1119-22, 2000 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-10996792

RESUMO

Re-entry into the cell cycle from quiescence requires the activation of mitogen-activated protein (MAP) kinases of the extracellular-signal-regulated kinase (ERK) family [1,2]. The relationship between ERK and cell-cycle control is, however, complex, as ERK activation can also lead to terminal differentiation [3] or a senescence-like growth arrest [4]. Here, we report that reversible cell-cycle exit induced by serum withdrawal in primary avian fibroblasts is associated with rapid deactivation of ERK, but ERK activity is subsequently regenerated and sustained at high levels in fully quiescent (G0) cells. As in proliferating cells, ERK activation during G0 required the MAPkinase kinase MEK and was partially dependent on cell adhesion. Active, phosphorylated ERK was concentrated in the nucleus in cycling cells, but was largely confined to the cytoplasm during G0. This was unexpected, as activatory phosphorylation mediated by MEK is thought to play an important role in promoting nuclear translocation [5,6]. These results indicate that transient deactivation of ERK signalling can be sufficient for stable cell-cycle exit, and that MEK-mediated phosphorylation is not sufficient for nuclear translocation of active ERK in G0. Cytoplasmic sequestration may prevent active ERK from accessing critical nuclear cell-cycle targets, thus allowing quiescent or post-mitotic cells to retain ERK activity for other physiological functions.


Assuntos
Ciclo Celular , MAP Quinase Quinase Quinase 1 , Sistema de Sinalização das MAP Quinases , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Fase de Repouso do Ciclo Celular , Células 3T3 , Animais , Adesão Celular , Núcleo Celular/enzimologia , Células Cultivadas , Embrião de Galinha , Meios de Cultura , Citoplasma/enzimologia , Fibroblastos , Citometria de Fluxo , Substâncias de Crescimento/metabolismo , Camundongos , Microscopia Confocal , Proteínas Quinases Ativadas por Mitógeno/imunologia , Ratos
4.
Curr Biol ; 8(2): 117-20, 1998 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-9427647

RESUMO

Stimulation of c-Jun transcriptional activity via phosphorylation mediated by the stress-activated or c-Jun amino-terminal (SAPK/JNK) subgroup of mitogen-activated protein kinases (MAP kinases) is thought to depend on a kinase-docking site (the delta region) within the amino-terminal activation domain, which is deleted from the oncogenic derivative, v-Jun [1] [2] [3]. This mutation markedly enhances v-Jun oncogenicity [4] [5]; however, its transcriptional consequences have not been resolved. In part, this reflects uncertainty as to whether binding of SAPK/JNK inhibits c-Jun function directly [6] [7] or, alternatively, serves to facilitate and maintain the specificity of positive regulatory phosphorylation [8]. Using a two-hybrid approach, we show that SAPK/JNK stimulates c-Jun transactivation in yeast and that this depends on both catalytic activity and physical interaction between the kinase and its substrate. Furthermore, c-Jun is active when tethered to DNA via SAPK/JNK, demonstrating that kinase binding does not preclude transactivation. Taken together, these results suggest that SAPK/JNK acts primarily as a positive regulator of c-Jun transactivation in situ, and that loss of the docking site physically uncouples v-Jun from this control. This loss-of-function model accounts for the deficit of v-Jun regulatory phosphorylation and repression of TPA response element (TRE)-dependent transcription observed in v-Jun-transformed cells and predicts that an important property of the oncoprotein is to antagonise SAPK/JNK-dependent gene expression.


Assuntos
Regulação da Expressão Gênica , Proteínas Quinases Ativadas por Mitógeno , Proteína Oncogênica p65(gag-jun)/metabolismo , Proteínas Quinases/metabolismo , Transdução de Sinais , Animais , Humanos , Proteína Quinase 12 Ativada por Mitógeno , Mutação , Proteína Oncogênica p65(gag-jun)/genética , Oncogenes , Proteínas Quinases/genética , Ratos , Proteínas Recombinantes de Fusão
5.
Mol Cell Biol ; 9(2): 865-8, 1989 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-2651907

RESUMO

A single nuclear protein (Myc-associated protein) can be specifically cross-linked to avian Myc proteins by treatment of nuclei or cells with the reversible cross-linker dimethyl 3,3'-dithiobis-propionimidate. Myc-associated protein has a molecular weight of approximately 500,000, is not detectably phosphorylated and, in contrast to Myc, has a long apparent half-life of greater than 3 h.


Assuntos
Proteínas Proto-Oncogênicas/isolamento & purificação , Animais , Linhagem Celular Transformada , Reagentes de Ligações Cruzadas , Meia-Vida , Imidoésteres , Peso Molecular , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-myc
6.
Mol Cell Biol ; 20(7): 2529-42, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10713176

RESUMO

v-Jun accelerates G(1) progression and shares the capacity of the Myc, E2F, and E1A oncoproteins to sustain S-phase entry in the absence of mitogens; however, how it does so is unknown. To gain insight into the mechanism, we investigated how v-Jun affects mitogen-dependent processes which control the G(1)/S transition. We show that v-Jun enables cells to express cyclin A and cyclin A-cdk2 kinase activity in the absence of growth factors and that deregulation of cdk2 is required for S-phase entry. Cyclin A expression is repressed in quiescent cells by E2F acting in conjunction with its pocket protein partners Rb, p107, and p130; however, v-Jun overrides this control, causing phosphorylated Rb and proliferation-specific E2F-p107 complexes to persist after mitogen withdrawal. Dephosphorylation of Rb and destruction of cyclin A nevertheless occur normally at mitosis, indicating that v-Jun enables cells to rephosphorylate Rb and reaccumulate cyclin A without exogenous mitogenic stimulation each time the mitotic "clock" is reset. D-cyclin-cdk activity is required for Rb phosphorylation in v-Jun-transformed cells, since ectopic expression of the cdk4- and cdk6-specific inhibitor p16(INK4A) inhibits both DNA synthesis and cell proliferation. Despite this, v-Jun does not stimulate D-cyclin-cdk activity but does induce a marked deregulation of cyclin E-cdk2. In particular, hormonal activation of a conditional v-Jun-estrogen receptor fusion protein in quiescent, growth factor-deprived cells stimulates cyclin E-cdk2 activity and triggers Rb phosphorylation and DNA synthesis. Thus, v-Jun overrides the mitogen dependence of S-phase entry by deregulating Rb phosphorylation, E2F-pocket protein interactions, and ultimately cyclin A-cdk2 activity. This is the first report, however, that cyclin E-cdk2, rather than D-cyclin-cdk, is likely to be the critical Rb kinase target of v-Jun.


Assuntos
Quinases relacionadas a CDC2 e CDC28 , Ciclo Celular/genética , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/metabolismo , Mitógenos/farmacologia , Proteína Oncogênica p65(gag-jun)/metabolismo , Proteínas Oncogênicas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteína do Retinoblastoma/metabolismo , Animais , Proteínas de Transporte/genética , Divisão Celular/genética , Embrião de Galinha , Quinase 2 Dependente de Ciclina , Inibidor p16 de Quinase Dependente de Ciclina , Fibroblastos , Fase G1 , Microinjeções , Fosforilação , Plasmídeos , Fase S , Transformação Genética
7.
Nucleic Acids Res ; 33(16): 5235-42, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16166655

RESUMO

Interaction with Max via the helix-loop-helix/leucine zipper (HLH-LZ) domain is essential for Myc to function as a transcription factor. Myc is commonly upregulated in tumours, however, its activity can also be potentiated by virally derived mutations. vMyc, derived from the virus, MC29 gag-Myc, differs from its cellular counterpart by five amino acids. The N-terminal mutation stabilizes the protein, however, the significance of the other mutations is not known. We now show that vMyc can sustain longer deletions in the LZ domain than cMyc before complete loss in transforming activity, implicating the viral mutations in contributing to Myc:Max complex formation. We confirmed this both in vitro and in vivo, with loss of Max binding correlating with a loss in the biological activity of Myc. A specific viral mutation, isoleucine383>leucine (I383>L) in helix 2 of the HLH domain, extends the LZ domain from four to five heptad repeats. Significantly, introduction of I383>L into a Myc mutant that is defective for Max binding substantially restored its ability to complex with Max in vitro and in vivo. We therefore propose that this virally derived mutation is functional by significantly contributing to establishing a more hydrophobic interface between the LZs of Myc and Max.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteína Oncogênica p55(v-myc)/genética , Proteína Oncogênica p55(v-myc)/metabolismo , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Animais , Fatores de Transcrição de Zíper de Leucina Básica , Sítios de Ligação , Embrião de Galinha , Dimerização , Sequências Hélice-Alça-Hélice , Zíper de Leucina , Dados de Sequência Molecular , Mutação , Proteína Oncogênica p55(v-myc)/química , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-myc/química , Proteínas Proto-Oncogênicas c-myc/metabolismo , Deleção de Sequência
8.
Oncogene ; 36(5): 678-686, 2017 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-27375025

RESUMO

Stresses such as hypoxia, nutrient deprivation and acidification disturb protein folding in the endoplasmic reticulum (ER) and activate the Unfolded Protein Response (UPR) to trigger adaptive responses through the effectors, PERK, IRE1 and ATF6. Most of these responses relate to ER homoeostasis; however, here we show that the PERK branch of the UPR also controls DNA replication. Treatment of cells with the non-genotoxic UPR agonist thapsigargin led to a rapid inhibition of DNA synthesis that was attributable to a combination of DNA replication fork slowing and reduced replication origin firing. DNA synthesis inhibition was dependent on the UPR effector PERK and was associated with phosphorylation of the checkpoint adaptor protein Claspin and activation of the Chk1 effector kinase, both of which occurred in the absence of detectable DNA damage. Remarkably, thapsigargin did not inhibit bulk DNA synthesis or activate Chk1 in cells depleted of Claspin, or when Chk1 was depleted or subject to chemical inhibition. In each case thapsigargin-resistant DNA synthesis was due to an increase in replication origin firing that compensated for reduced fork progression. Taken together, our results unveil a new aspect of PERK function and previously unknown roles for Claspin and Chk1 as negative regulators of DNA replication in the absence of genotoxic stress. Because tumour cells proliferate in suboptimal environments, and frequently show evidence of UPR activation, this pathway could modulate the response to DNA replication-targeted chemotherapies.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Quinase 1 do Ponto de Checagem/metabolismo , Replicação do DNA/fisiologia , Resposta a Proteínas não Dobradas/fisiologia , eIF-2 Quinase/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Linhagem Celular Tumoral , Quinase 1 do Ponto de Checagem/genética , Humanos , Transfecção , eIF-2 Quinase/genética
9.
Oncogene ; 5(5): 683-9, 1990 May.
Artigo em Inglês | MEDLINE | ID: mdl-2189105

RESUMO

Small deletions of 7 to 48 amino acids have been generated in the leucine zipper domain of the avian cMyc protein and the mutant cMyc proteins expressed using an avian retroviral vector. Retrovirally encoded cMyc protein transforms primary chick embryo fibroblasts and leads to abnormal regulation of the endogenous c-myc gene. Deletion of the most C-terminal leucine of the zipper motif confers a partial phenotype affecting some but not all parameters of transformation. Complete loss of transforming activity results from deletion of further leucine residues, including one which is not part of the heptad repeat. In cMyc transformed cells endogenous c-myc mRNA is expressed at a low level and is abnormally refractory to induction by serum stimulation. In contrast, a non-transforming cMyc protein which lacks the zipper does not affect normal c-myc expression. These results demonstrate that the leucine zipper domain of avian cMyc is required for both transformation and autoregulation, and suggests that essential leucine residues within the motif may be spaced differently from those in the zippers of Fos and Jun.


Assuntos
Homeostase , Leucina/análise , Proteínas Proto-Oncogênicas/genética , Transformação Genética/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Sequência de Bases , Embrião de Galinha , DNA/análise , DNA/genética , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Dados de Sequência Molecular , Fenótipo , Proteínas Proto-Oncogênicas/análise , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-myc , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
10.
Oncogene ; 19(51): 5906-18, 2000 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-11127822

RESUMO

v-Jun shares the ability of the Myc, E1A, and E2F oncogenes to both sustain cell cycle progression and promote apoptosis in the absence of mitogenic stimulation. To gain an insight into the mechanism of apoptosis sensitization, we examined the possible involvement of key regulatory proteins previously implicated in oncogene-induced cell death during v-Jun-induced apoptosis triggered by serum withdrawal. We observed that ectopic expression of the anti-apoptotic Bcl-2 protein, or of two downstream effectors of growth factor signalling, v-PI 3-Kinase and v-Src, partially or completely suppressed apoptosis. Apoptosis was also observed in the presence of serum growth factors when endogenous PI3K activity was blocked using the synthetic inhibitor LY294002, further suggesting an important role for PI3-K in cell survival. Cytochrome C was released into the cytosol of apoptotic v-Jun expressing cells, and this release was inhibited by Bcl-2, suggesting an important role for mitochondrial dysfunction in v-Jun induced apoptosis. In contrast, inhibition of Fas signalling using dominant negative FADD did not inhibit apoptosis, nor was there any evidence for accumulation or activation of p53 in v-Jun transformed cells. Consistent with this latter observation, inhibition of p53 function by HPV16 E6 protein had no effect on v-Jun induced cell death. Taken together, these results suggest that mitochondrial dysfunction is an important component of the mechanism through which v-Jun sensitizes cells to apoptosis, but that the apoptotic signals elicited by v-Jun upstream of the mitochondria do not depend on increased levels of p53 activity or Fas signalling.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Apoptose/fisiologia , Grupo dos Citocromos c/metabolismo , Genes jun/fisiologia , Mitocôndrias/metabolismo , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Proteínas de Transporte/fisiologia , Inibidores de Caspase , Células Cultivadas , Embrião de Galinha , Inibidores de Cisteína Proteinase/farmacologia , Proteína de Domínio de Morte Associada a Fas , Fibroblastos/citologia , Substâncias de Crescimento/sangue , Proteína Oncogênica p65(gag-jun)/antagonistas & inibidores , Proteína Oncogênica p65(gag-jun)/biossíntese , Proteína Oncogênica p65(gag-jun)/fisiologia , Proteína Oncogênica pp60(v-src)/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/fisiologia
11.
Oncogene ; 6(11): 1949-58, 1991 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-1658706

RESUMO

Chicken c-Jun proteins synthesized in vitro in reticulocyte extract consist of several electrophoretic isoforms resulting from phosphorylation which can be specifically reversed by purified protein phosphatase 2A (PP2A). Using the phosphatase inhibitors okadaic acid and microcystin-LR, we conclude that the isoforms seen in vitro represent a balance between the action of an unidentified kinase(s) which phosphorylates c-Jun and dephosphorylation by an endogenous PP2A-like phosphatase. c-Jun proteins are also subject to phosphorylation in vivo in chick embryo fibroblasts (CEF), which can be reversed by PP2A. In contrast, the viral Jun oncoprotein encoded by ASV17 is not subject to PP2A-sensitive phosphorylation in vitro and is hypophosphorylated in comparison with c-Jun in ASV17-transformed CEF. Hybrids between c-Jun and v-Jun demonstrate that differential phosphorylation in vitro is a consequence of deletion of 27 amino acids in the N-terminal third of v-Jun. The deletion is important for oncogenic activation and lies in a domain, termed delta, which regulates c-Jun transactivation function. PP2A-sensitive phosphorylation in vitro correlates with the differential responsiveness of c-Jun and v-Jun to a recently identified cell type-specific inhibitor of transactivation function.


Assuntos
Genes jun , Fosfoproteínas Fosfatases/farmacologia , Sequência de Aminoácidos , Animais , Galinhas , Mapeamento Cromossômico , Clonagem Molecular , Eletroforese em Gel de Poliacrilamida , Éteres Cíclicos/farmacologia , Isotipos de Imunoglobulinas/química , Toxinas Marinhas , Camundongos , Microcistinas , Dados de Sequência Molecular , Ácido Okadáico , Peptídeos Cíclicos/farmacologia , Fosfoproteínas Fosfatases/antagonistas & inibidores , Fosfoproteínas Fosfatases/fisiologia , Fosforilação/efeitos dos fármacos , Testes de Precipitina , Biossíntese de Proteínas , Proteína Fosfatase 2 , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-jun/análise , Processamento Pós-Transcricional do RNA , Transcrição Gênica
12.
Oncogene ; 9(12): 3499-508, 1994 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-7970710

RESUMO

The c-Myc oncoprotein is a basic-helix-loop-helix-leucine zipper (b-HLH-LZ) transcription factor involved in regulating cell proliferation and differentiation. We have used retrovirus-mediated gene transfer to investigate the effect of ectopic c-Myc expression on the spontaneous differentiation of primary quail myoblasts in vitro. Unlike normal myoblasts, c-Myc-expressing myoblasts are unable to form myotubes or express muscle-specific genes, such as myosin, and show severely reduced expression of the myogenic regulatory factors myoD, myogenin, and myf5. The c-Myc leucine zipper (LZ) motif is essential for the differentiation block since myoblasts expressing a mutant with a partial deletion of this region, c-Myc delta 7, differentiate and express myoD family regulators and muscle-specific genes normally. Remarkably, c-Myc delta 7, like wild-type c-Myc, retains the capacity to transform the growth phenotype of myoblasts, and associates with the b-HLH-LZ Myc partner protein Max in transformed cells. We conclude that the block to myogenic differentiation induced by c-Myc can be dissociated from cell transformation per se, and that this attribute correlates more closely with down-regulation of myoD family gene expression. These findings are discussed in the light of current models of Myc function.


Assuntos
Músculos/citologia , Proteína MyoD/genética , Proteínas Proto-Oncogênicas c-myc/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem Celular Transformada , Coturnix , Regulação da Expressão Gênica , Dados de Sequência Molecular , Proteína MyoD/biossíntese , Oligodesoxirribonucleotídeos
13.
Oncogene ; 12(11): 2409-18, 1996 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-8649782

RESUMO

We have investigated the expression of Jun family proteins and composition of AP-1 in chicken embryo fibroblasts before and after transformation by the v-Jun oncoprotein of ASV17. We show that p39 c-Jun is the predominant Jun family protein expressed in normal fibroblasts, and that heterodimers of c-Jun and Fos-related partners (Fra's) account for the majority of the AP-1 DNA binding activity. Unexpectedly, because ASV17-transformed fibroblasts do not express p39 c-Jun, v-Jun replaces c-Jun as the predominant AP-1 constituent in association with similar or identical Fra's. This substitution has little effect on the overall level of TRE-specific DNA binding activity, however it results in a profound reduction in TRE-dependent transcriptional activity and a striking defect in signal-regulated phosphorylation of the Jun component of AP-1; whilst agonists of SAPK/JNK kinases trigger transient N-terminal phosphorylation of c-Jun in normal fibroblasts, no corresponding modification of v-Jun occurs in ASV17-transformed cells. Because SAPK/JNK-mediated phosphorylation is thought to regulate c-Jun transcriptional activity and thereby cellular gene expression in response to extracellular signals, we propose that subversion of this signal transduction process by v-Jun is likely to contribute to oncogenesis by ASV17.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/fisiologia , Proteínas Quinases Ativadas por Mitógeno , Proteína Oncogênica p65(gag-jun)/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Transdução de Sinais , Fator de Transcrição AP-1/metabolismo , Células 3T3 , Sequência de Aminoácidos , Animais , Transformação Celular Neoplásica/metabolismo , Embrião de Galinha , Cloranfenicol O-Acetiltransferase/metabolismo , DNA/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno , Camundongos , Dados de Sequência Molecular , Fosforilação , Proteínas Proto-Oncogênicas c-jun/análise , Fator de Transcrição AP-1/química
14.
Oncogene ; 9(8): 2363-8, 1994 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8036019

RESUMO

Growth factors, phorbol esters, and oncogenes such as ras, src, and sis are believed to stimulate c-Jun transcriptional activation by inducing increased phosphorylation at two serine residues (S63 and S73) within the N-terminal transactivation domain. Although S63 and S73 are conserved in the mutant v-Jun oncoprotein, they are not phosphorylated by two enzymes which target the corresponding residues in c-Jun in vitro; namely a partially purified c-Jun kinase from TPA-stimulated U937 cells and purified p54 mitogen activated protein (MAP) kinase. In addition, v-Jun activates transcription more strongly than c-Jun when fused to the Gal4 DNA-binding domain, and transcriptional activation by Gal4-v-Jun is unaffected when S63, S73, or both, are replaced with non-phosphorylatable alanine residues, amino acid substitutions which severely impair transcriptional activation by Gal4-c-Jun. The novel biochemical and transcriptional properties of v-Jun result from deletion of a 27 amino acid segment, termed delta, which is important for transforming activity. On the basis of these results we propose that unlike c-Jun, v-Jun transcriptional activation is independent of positive regulatory phosphorylation and that this may contribute to oncogenesis by v-Jun.


Assuntos
Proteína Oncogênica p65(gag-jun)/fisiologia , Ativação Transcricional , Animais , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Embrião de Galinha , Humanos , Proteína Oncogênica p65(gag-jun)/química , Fosforilação , Proteínas Proto-Oncogênicas c-jun/fisiologia , Relação Estrutura-Atividade
15.
Oncogene ; 22(16): 2383-95, 2003 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-12717415

RESUMO

Previous studies have shown that the viral Jun (v-Jun) oncoprotein induces marked alterations in cell cycle control, which are associated with, and may be caused by, increased cdk2 kinase activity. Since p21 CIP1 is an important regulator of cdk2, we investigated whether aberrant expression of this cyclin-dependent kinase inhibitor might contribute to cell cycle deregulation by v-Jun. We find that the basal levels of p21 CIP1 mRNA and protein expression are greatly reduced in chick embryo fibroblasts (CEF) transformed by v-Jun, and that v-Jun blocks the increases in p21 CIP1 expression that normally accompany growth inhibition induced by serum deprivation or confluency in untransformed CEF. Importantly, ectopic expression of p21 CIP1 in v-Jun-transformed CEF inhibits both cdk2 kinase activity and cell cycle progression, indicating that these alterations in p21 CIP1 expression are likely to be functionally significant for growth deregulation. We also investigated the mechanism through which v-Jun disturbs p21 CIP1 expression and the possible involvement of a known p21 CIP1 regulator, p53, as an intermediate in this process. This analysis revealed that repression is mediated primarily at the level of p21 CIP1 gene transcription, however the mechanism is complex; both p53-dependent and -independent mechanisms contribute as judged by analysis of p21 CIP1 promoter mutants and other assays of p53 transcriptional activity.


Assuntos
Quinases relacionadas a CDC2 e CDC28 , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/metabolismo , Fase G1/fisiologia , Proteína Oncogênica p65(gag-jun)/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Embrião de Galinha , Quinase 2 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/genética , Ciclinas/isolamento & purificação , Regulação para Baixo , Regulação da Expressão Gênica/fisiologia , Dados de Sequência Molecular , Proteína Oncogênica p65(gag-jun)/genética , Fase S/fisiologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
16.
Oncogene ; 8(7): 1849-55, 1993 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8510929

RESUMO

Max is a basic helix-loop-helix/leucine zipper (bHLH/LZ) protein that forms sequence-specific DNA-binding complexes with the c-Myc oncoprotein (Myc). Using Saccharomyces cerevisiae, we have shown that the Max bHLH/LZ domain enables Myc to activate transcription through CACGTG and CACATG sequences in vivo, and that the number and context of such sites determines the level of activation. In addition, we have used yeast to investigate the role of the Myc helix-loop-helix (HLH) and leucine zipper (LZ) motifs in mediating Max-dependent DNA-binding and transcriptional activation in vivo using HLH/LZ mutants generated by site-directed mutagenesis. The results show that, while both motifs are essential for Myc to activate transcription, helix 2 of the HLH together with the contiguous LZ suffice to mediate complex formation with Max, whilst helix 1 is essential for sequence-specific DNA binding of Myc-Max complexes. Furthermore, the ability of Myc HLH/LZ mutants to bind DNA and activate transcription in collaboration with Max correlates closely with their neoplastic transforming activity in higher eukaryotic cells.


Assuntos
Transformação Celular Neoplásica , Proteínas de Ligação a DNA/metabolismo , DNA/metabolismo , Zíper de Leucina/fisiologia , Proteínas Proto-Oncogênicas c-myc/fisiologia , Saccharomyces cerevisiae/genética , Fatores de Transcrição , Ativação Transcricional , Sequência de Bases , Fatores de Transcrição de Zíper de Leucina Básica , Dados de Sequência Molecular , Mutação , Estrutura Secundária de Proteína , Proteínas Proto-Oncogênicas c-myc/química
17.
Oncogene ; 6(2): 205-9, 1991 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-1900354

RESUMO

The level of AP-1 DNA-binding activity exhibited in vitro by unfractionated extracts of Hela nuclei can be stimulated by a low molecular weight fraction from rabbit reticulocyte lysate. Stimulation also requires a heat labile component of the nuclear extract, probably a protein. Stimulated and unstimulated extracts with high and low AP-1 DNA-binding activities contain the same levels of proteins reactive with antisera against Jun and Fos, proteins which are shown to be involved in the AP-1/DNA complexes detected in vitro. The low molecular weight fraction from reticulocyte lysate can be substituted by the reducing agent dithiothreitol (DTT) in the stimulation reaction and conversely oxidised glutathione greatly reduces formation of AP-1/DNA complexes. The binding activities of transcription factors SP-1, NF-1 and CBP are unaffected by DTT or oxidised glutathione. These observations, taken together, suggest that the efficiency with which pre-existing Fos and Jun proteins can bind an AP-1 target sequence in vitro can be controlled by a nuclear activity which is sensitive to oxidation/reduction and that this control mechanism is specific for AP-1.


Assuntos
Proteínas de Ligação a DNA/metabolismo , DNA/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Transcrição/metabolismo , Animais , Ditiotreitol/farmacologia , Células HeLa , Humanos , Proteínas Proto-Oncogênicas c-fos , Proteínas Proto-Oncogênicas c-jun , Coelhos
18.
Oncogene ; 10(5): 849-55, 1995 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-7898927

RESUMO

The transactivating function of the c-Jun proto-oncogene component of the AP-1 transcription factor is acutely regulated by a wide variety of cellular signals via modulation of phosphorylation of two serines (63 and 73). The viral oncoprotein, v-Jun, while containing homologous serines, is not phosphorylated in cells. A novel family of stress-activated protein kinases (SAPKs), also termed Jun N-terminal domain kinases (JNKs), are responsible for mediating S63/73 phosphorylation in response to a variety of cellular stimuli including tumor necrosis factor-alpha, heat stress and u.v. light. The p54 alpha 1, alpha 2, p54 beta and p46 beta SAPKs are shown to bind directly to c-Jun but not to v-Jun, with an absolute requirement for c-Jun amino acids 31-47, a region deleted in v-Jun. Inactive SAPKs tightly bind c-Jun in resting cells and may be a manifestation of the 'delta' inhibitor, a previously described repressor of c-Jun function.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/fisiologia , Proteínas Quinases Ativadas por Mitógeno , Proteínas Proto-Oncogênicas c-jun/fisiologia , Proteínas Repressoras/fisiologia , Linhagem Celular , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , Fosforilação , Proto-Oncogene Mas
19.
Oncogene ; 31(11): 1366-75, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-21804609

RESUMO

Chk1 is a key regulator of DNA damage checkpoint responses and genome stability in eukaryotes. To better understand how checkpoint proficiency relates to cancer development, we investigated the effects of genetic ablation of Chk1 in the mouse skin on tumors induced by chemical carcinogens. We found that homozygous deletion of Chk1 immediately before carcinogen exposure strongly suppressed benign tumor (papilloma) formation, and that the few, small lesions that formed in the ablated skin always retained Chk1 expression. Remarkably, Chk1 deletion rapidly triggered spontaneous cell proliferation, γ-H2AX staining and apoptosis within the hair follicle, a principal site of origin for carcinogen-induced tumors. At later times, the ablated skin was progressively repopulated by non-recombined Chk1-expressing cells and ultimately normal sensitivity to tumor induction was restored when carcinogen treatment was delayed. In marked contrast, papillomas formed normally in Chk1 hemizygous skin but showed an increased propensity to progress to carcinoma. Thus, complete loss of Chk1 is incompatible with epithelial tumorigenesis, whereas partial loss of function (haploinsufficiency) fosters benign malignant tumor progression.


Assuntos
Carcinoma de Células Escamosas/genética , Transformação Celular Neoplásica/genética , Papiloma/induzido quimicamente , Proteínas Quinases/genética , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/genética , Animais , Apoptose , Carcinógenos , Quinase 1 do Ponto de Checagem , Dano ao DNA , Progressão da Doença , Deleção de Genes , Instabilidade Genômica , Camundongos , Camundongos Knockout
20.
Oncogene ; 31(9): 1086-94, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21765472

RESUMO

Here, we show that activation of the checkpoint effector kinase Chk1 in response to irradiation-induced DNA damage is minimal in G1, maximal during S-phase and diminishes as cells enter G2. In addition, formation of irradiation-induced replication protein A (RPA)-coated single-stranded DNA (RPA-ssDNA), a structure required for ATM and Rad3-related (ATR)-Chk1 activation, occurs in a broadly similar pattern. Cyclin-dependent kinase (Cdk) activity is thought to promote RPA-ssDNA formation by stimulating DNA strand resection at double-strand breaks (DSBs), providing one possible mechanism of imposing cell cycle dependence on DNA damage signaling. However, it has recently been shown that Chk1 itself is also subject to Cdk-mediated phosphorylation at serines 286 and 301 (S286 and 301). We show that Chk1 S301 phosphorylation increases as cells progress through S and G2 and that both Cdk1 and Cdk2 are likely to contribute to this modification in vivo. We also find that substitution of S286 and S301 with non-phosphorylatable alanine residues strongly attenuates DNA damage-induced Chk1 activation and G2 checkpoint proficiency, but does not eliminate the underlying cell cycle dependence of Chk1 regulation. Taken together, these data indicate that Cdk activity regulates multiple steps in the DNA damage response pathway including full activation of Chk1 and checkpoint proficiency.


Assuntos
Pontos de Checagem do Ciclo Celular , Quinases Ciclina-Dependentes/metabolismo , Dano ao DNA/efeitos da radiação , Proteínas Quinases/metabolismo , Proteína Quinase CDC2/metabolismo , Pontos de Checagem do Ciclo Celular/genética , Pontos de Checagem do Ciclo Celular/efeitos da radiação , Linhagem Celular Tumoral , Quinase 1 do Ponto de Checagem , Quinase 2 Dependente de Ciclina/metabolismo , Ativação Enzimática , Humanos , Modelos Biológicos , Fosforilação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA