Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
1.
Br J Cancer ; 112(4): 755-64, 2015 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-25633035

RESUMO

BACKGROUND: Transcription factor EGR3 (Early Growth Response 3) is a little-studied member of the EGR family that is highly expressed in human prostate tumours compared with normal tissue. Its function in prostate cancer, however, is unknown. METHODS: Stable shRNA silencing was achieved in naturally overexpressing prostate cancer cells, followed by Affymetrix expression analysis. Fold changes of ⩾2 and ⩽-2 were considered valid and t-tests P-values of ⩽0.01 were considered statistically significant. Potential EGR3 target genes were validated by real-time qPCR, chromatin immunoprecipitation, and gain-of-function experiments. Promoter analysis confirmed the presence of consensus binding sites in the promoters of target genes. RESULTS: Early Growth Response 3 regulates the expression of ∼330 genes, 35% of which are involved in immune responses and inflammatory processes, and 15% crosstalk with the NF-κB signalling pathway. In particular, EGR3 induces the expression of over 50 secreted cytokines, growth factors, and matrix remodelling factors. Two interleukins of great relevance to prostate cancer, IL6 and IL8, were further validated as EGR3 target genes: both promoters contain EGR consensus binding sites and are pulled down in intact cells by EGR3 chromatin immunoprecipitation. Silencing of EGR3 decreased IL6 and IL8 expression, whereas overexpression of EGR3 in nontransformed cells induced IL6 and IL8 expression. CONCLUSIONS: Chronic inflammation plays a critical role in prostate cancer and elevated production of pro-inflammatory cytokines IL8 and IL6, in particular, contributes to disease progression and to the onset of castration resistance. It is shown for the first time that EGR3 is involved in the upregulation of both IL6 and IL8. Together with our previous observation that EGR3 is highly expressed in prostate tumours compared with normal tissue and strongly correlates with IL6 and IL8 expression in clinical samples, the present study suggests that EGR3 promotes excessive production of IL6 and IL8 observed during the progression of prostate cancer.


Assuntos
Proteína 3 de Resposta de Crescimento Precoce/fisiologia , Inflamação/genética , Interleucina-6/genética , Interleucina-8/genética , Neoplasias da Próstata/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Análise em Microsséries , Ativação Transcricional , Transfecção , Células Tumorais Cultivadas
2.
Cell Death Differ ; 14(3): 436-46, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16990849

RESUMO

p73, a transcription factor rarely mutated in cancer, regulates a subset of p53 target genes that cause cells to respond to genotoxic stress by growth arrest and apoptosis. p73 is produced in two main forms; only TAp73 reiterates the roles of p53, while DeltaNp73 can be oncogenic in character. We show that the TAp73 form produced by TP73 P1 promoter has five distinct Egr1-binding sites, each contributing to the transcriptional upregulation of TAp73 by Egr1 in several cell types. In contrast, TP73 P2 promoter transcribes DeltaNp73, is not induced by Egr1, but is induced by TAp73 and p53. Induction of TAp73 by genotoxic stress requires Egr1 in mouse in vivo. Newly discovered non-consensus p53-binding sites in p73, p53 and Egr1 promoters reveal inter-regulating networks and sustained expression by feedback loops in response to stress, resulting in prolonged expression of the p53 family of genes and efficient apoptosis.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteínas Nucleares/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Antineoplásicos Fitogênicos/farmacologia , Apoptose , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Proteína 1 de Resposta de Crescimento Precoce/genética , Etoposídeo/farmacologia , Raios gama , Humanos , Camundongos , Modelos Biológicos , Proteínas Nucleares/genética , Regiões Promotoras Genéticas , Fatores de Tempo , Transcrição Gênica , Transfecção , Proteínas Supressoras de Tumor/genética , Regulação para Cima
3.
Cancer Gene Ther ; 13(2): 115-24, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16138117

RESUMO

Recent studies are reviewed indicating that the transcription factor early growth response-1 (Egr1) is a direct regulator of multiple tumor suppressors including TGFbeta1, PTEN, p53, and fibronectin. The downstream pathways of these factors display multiple nodes of interaction with each other, suggesting the existence of a functional network of suppressor factors that serve to maintain normal growth regulation and resist the emergence of transformed variants. Paradoxically, Egr1 is oncogenic in prostate cancer. In the majority of these cancers, PTEN or p53 is inactive. It is suggested that these defects in the suppressor network allow for the unopposed induction of TGFbeta1 and fibronectin, which favor transformation and survival of prostate tumor epithelial cells, and explain the role of Egr1 in prostate cancer. Egr1 is a novel and logical target for intervention by gene therapy methods, and targeting methods are discussed.


Assuntos
Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Fibronectinas/metabolismo , Regulação Neoplásica da Expressão Gênica , Genes p53/genética , Terapia Genética/métodos , PTEN Fosfo-Hidrolase/metabolismo , Neoplasias da Próstata/terapia , Fator de Crescimento Transformador beta/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/genética , Células Epiteliais/metabolismo , Humanos , Masculino , Modelos Genéticos , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Neoplasias da Próstata/metabolismo , Fator de Crescimento Transformador beta1
4.
Leukemia ; 16(5): 799-812, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11986940

RESUMO

A relationship was proved between constitutive activity of leukemic cell c-jun-N-terminal kinase (JNK) and treatment failure in AML. Specifically, early treatment failure was predicted by the presence of constitutive JNK activity. The mechanistic origins of this association was sought. A multidrug resistant leukemic cell line, HL-60/ADR, characterized by hyperexpression of c-jun and JNK activity, was transfected with a mutant c-jun vector, whose substrate N-terminal c-jun serines were mutated. Down-regulated expression occurred of c-jun/AP-1-dependent genes, catalase and glutathione-S-transferase (GST) pi, which participate in cellular homeostasis to oxidative stress and xenobiotic exposure. MRP-efflux was abrogated in HL-60/ADR cells with dominant-negative c-jun, perhaps because MRP1 protein expression was also lost. Heightened sensitivity to daunorubicin resulted in cells subjected to this change. Biochemical analysis in 67 primary adult AML samples established a statistical correlation between cellular expression of c-jun and JNK activity, JNK activity with hyperleukocytosis at presentation of disease, and with exuberant MRP efflux. These findings reflect the survival role for c-jun/AP-1 and its regulatory kinase previously demonstrated for yeast in homeostatic response to oxidative stress and in operation of ATP-binding cassette efflux pumps, and may support evolutionary conservation of such function. Thus, JNK and c-jun may be salient drug targets in multidrug resistant AML.


Assuntos
Resistencia a Medicamentos Antineoplásicos/fisiologia , Leucemia Mieloide/enzimologia , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Doença Aguda , Células da Medula Óssea/patologia , Divisão Celular , Daunorrubicina , Resistência a Múltiplos Medicamentos/fisiologia , Células HL-60 , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , Leucemia Mieloide/tratamento farmacológico , Leucemia Mieloide/patologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-jun/metabolismo , Transdução de Sinais
5.
Clin Cancer Res ; 7(9): 2788-96, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11555594

RESUMO

PURPOSE: EGR-1 is an immediate early gene with diverse functions that include the suppression of growth. EGR-1 is down-regulated many cancer cell types, suggesting a tumor suppressor role, and may critically involve the p53 pathway. The aim of this work was to measure the expression of EGR-1 and the p16/INK4a/ARF-Mdm2-p53 pathway status in fresh human gliomas. EXPERIMENTAL DESIGN: Thirty-one human gliomas with different grades of malignancy were investigated for Egr-1 mRNA and the protein expression, frequency, and spectrum of p53 gene mutations, mdm2 gene amplification, and p16/INK4a/ARF allele loss. RESULTS: The amplification of Mdm2 and the deletion of the p16/INK4a gene was found in 3 and 5 cases, respectively, whereas mutations of p53, including two novel mutations, were observed in 10 other cases. The three types of changes occurred strictly mutually exclusively, emphasizing that these genes operate in a common pathway critical to glioma progression. EGR-1 mRNA was significantly down-regulated in astrocytomas (14.7 +/- 5.1%) and in glioblastomas (33.6 +/- 10.0%) versus normal brain. Overall, EGR-1 mRNA was strongly suppressed (average, 15.2 +/- 13.9%) in 27 of 31 cases (87%), independent of changes in p16/INK4a/ARF and Mdm2; whereas 4 of 31 cases with residual EGR-1 expression as well as the highest EGR-1 variance segregated with p53 mutations. Immunohistochemical analyses confirmed the suppression of EGR-1 protein. CONCLUSIONS: These results indicate that EGR-1 is commonly suppressed in gliomas independent of p16/INK4a/ARF and Mdm2 and that suppression is less crucial in tumors bearing p53 mutations, and these results implicate an EGR-1 growth regulatory mechanism as a target of inactivation during tumor progression.


Assuntos
Neoplasias Encefálicas/genética , Proteínas de Ligação a DNA/genética , Glioma/genética , Proteínas Imediatamente Precoces , Proteínas Nucleares , Proteínas/fisiologia , Fatores de Transcrição/genética , Northern Blotting , Neoplasias Encefálicas/patologia , Inibidor p16 de Quinase Dependente de Ciclina/genética , Análise Mutacional de DNA , DNA de Neoplasias/química , DNA de Neoplasias/genética , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo , Proteína 1 de Resposta de Crescimento Precoce , Deleção de Genes , Regulação Neoplásica da Expressão Gênica , Glioma/patologia , Humanos , Imuno-Histoquímica , Mutação de Sentido Incorreto , Proteínas/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-mdm2 , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p14ARF , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/fisiologia
7.
J Biol Chem ; 276(30): 28546-53, 2001 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-11352915

RESUMO

c-Jun, a member of the activation protein 1 (AP-1) family of transcription factors, has been implicated in the regulation of many important biological processes including cell cycle progression, transformation, differentiation, and apoptosis. Accordingly, its expression and function are upregulated in response to diverse stimuli including mitogens and a wide range of stresses. Transcriptional activation of the c-Jun protein is dependent on its phosphorylation at Ser-63 and Ser-73, a process mediated by c-Jun N-terminal kinase. Active c-Jun is required for AP-1 transactivation and c-Jun-mediated transformation, but its role during stress remains unclear as both pro-apoptotic and pro-survival effects of c-Jun have been observed. Here we investigated the importance of c-Jun N-terminal phosphorylation in influencing the sensitivity of human T98G glioblastoma cells to a variety of cytotoxic agents. Stable expression of a nonphosphorylatable dominant negative protein c-Jun(S63A,S73A) markedly inhibited the activation of AP-1-driven transcription and greatly increased the cytotoxic effects of DNA-damaging agents associated with enhanced apoptosis. However, the same cells expressing the mutant Jun protein did not differ from parental cells in their sensitivity to several non-DNA-damaging cytotoxic agents. Our results suggest that activated c-Jun has a selective role in protecting human tumor cells from apoptosis induced by DNA damage.


Assuntos
Dano ao DNA , Proteínas Proto-Oncogênicas c-jun/fisiologia , Antineoplásicos/farmacologia , Apoptose , Cloranfenicol O-Acetiltransferase/metabolismo , Cisplatino/farmacologia , Citometria de Fluxo , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mutagênicos , Fosforilação , Serina/química , Estresse Fisiológico , Fatores de Tempo , Fator de Transcrição AP-1/metabolismo , Transcrição Gênica , Ativação Transcricional , Transfecção , Células Tumorais Cultivadas
8.
J Immunol ; 166(4): 2437-43, 2001 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-11160303

RESUMO

Ligation of the T cell coreceptor CD28 or CD2 by its cognate ligands B7-1 or LFA-3, respectively, greatly aids the Ag-induced up-regulation of several genes, including IL-2 and CD40 ligand (CD40L). Using luciferase reporter constructs under the control of the 1.2 kb of 5' noncoding region of the human CD40L gene, we have found that stimulation through CD28 was required for a strong transcriptional activity of the CD40L promoter in response to TCR ligation, while the activity induced by CD2 was slightly lower than CD28. Deletion analysis demonstrated that the transcriptional elements mediating this effect were located within a 300-bp region upstream of the start site. Further dissection of this region and gel shift analyses demonstrated the presence of a CD28 response element in a region located between nucleotides -170 to -164 relative to the start site. Transcriptional studies with a CD40L enhancer-promoter carrying a mutation in this putative CD28 response element revealed that the activity was reduced by 80 and 70% after B7-1 and LFA-3 costimulation, respectively. The transcription factor complex bound to this site contained at least JunD, c-Fos, p50, p65, and c-REL:, but not c-Jun. Mutations introduced into the CD28RE also blocked the binding of this complex. These observations identify an important role for the CD28 signaling pathway in the regulation of CD40L promoter transcriptional activity.


Assuntos
Antígenos CD28/genética , Antígenos CD28/imunologia , Ligante de CD40/genética , Proteínas Nucleares , Regiões Promotoras Genéticas/imunologia , Elementos de Resposta/imunologia , Animais , Antígeno B7-1/fisiologia , Composição de Bases , Antígenos CD28/metabolismo , Ligante de CD40/metabolismo , Antígenos CD58/fisiologia , Células CHO , Cricetinae , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica/imunologia , Humanos , Células Jurkat , Ativação Linfocitária/genética , Família Multigênica/imunologia , Fatores de Transcrição NFATC , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Proteínas Proto-Oncogênicas c-rel/metabolismo , Fatores de Transcrição/genética , Transcrição Gênica/imunologia
9.
Nat Cell Biol ; 3(12): 1124-8, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11781575

RESUMO

The PTEN tumour suppressor and pro-apoptotic gene is frequently mutated in human cancers. We show that PTEN transcription is upregulated by Egr-1 after irradiation in wild-type, but not egr-1-/-, mice in vivo. We found that Egr-1 specifically binds to the PTEN 5' untranslated region, which contains a functional GCGGCGGCG Egr-1-binding site. Inducing Egr-1 by exposing cells to ultraviolet light upregulates expression of PTEN messenger RNA and protein, and leads to apoptosis. egr-1-/- cells, which cannot upregulate PTEN expression after irradiation, are resistant to ultraviolet-light-induced apoptosis. Therefore, Egr-1 can directly regulate PTEN, triggering the initial step in this apoptotic pathway. Loss of Egr-1 expression, which often occurs in human cancers, could deregulate the PTEN gene and contribute to the radiation resistance of some cancer cells.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas Imediatamente Precoces , Monoéster Fosfórico Hidrolases/genética , Monoéster Fosfórico Hidrolases/metabolismo , Transdução de Sinais/efeitos da radiação , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Animais , Apoptose/fisiologia , Apoptose/efeitos da radiação , Células Cultivadas , Derme/citologia , Proteína 1 de Resposta de Crescimento Precoce , Etoposídeo/farmacologia , Fibroblastos/citologia , Raios gama , Regulação Neoplásica da Expressão Gênica/fisiologia , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Humanos , Glândulas Mamárias Animais/citologia , Camundongos , Camundongos Knockout , Neoplasias/fisiopatologia , Inibidores da Síntese de Ácido Nucleico/farmacologia , PTEN Fosfo-Hidrolase , Regiões Promotoras Genéticas/genética , RNA Mensageiro/análise , Transdução de Sinais/fisiologia , Raios Ultravioleta
10.
Biotechniques ; 29(1): 162-9, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10907091

RESUMO

A methodology is described that allows the in vivo trapping of transcription factors to their target regulatory elements in multiple genes simultaneously. Cross-linking using formaldehyde is the first of several steps to isolate, purify, clone and characterize multiple gene promoter DNA fragments. The example that we use indicates that the TGF beta 1 gene is a direct target induced by Egr-1 in HT1080 cells that express constitutive Egr-1, thus explaining the growth retardation that follows Egr-1 expression. The genes identified using this procedure reflect the specific activities of Egr-1 at that moment in the cell and provide a method for confirmation of genes that are the direct targets of Egr-1 action.


Assuntos
Clonagem Molecular/métodos , Proteínas de Ligação a DNA/genética , Proteínas Imediatamente Precoces , Fatores de Transcrição/genética , Sítios de Ligação , Neoplasias da Mama/metabolismo , Reagentes de Ligações Cruzadas , DNA/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/farmacologia , Proteína 1 de Resposta de Crescimento Precoce , Fibrossarcoma , Formaldeído , Expressão Gênica/efeitos dos fármacos , Humanos , Técnicas de Imunoadsorção , Reação em Cadeia da Polimerase , Sensibilidade e Especificidade , Acetato de Tetradecanoilforbol/farmacologia , Fatores de Transcrição/metabolismo , Fatores de Transcrição/farmacologia , Transcrição Gênica , Ativação Transcricional , Transfecção , Fator de Crescimento Transformador beta/genética , Células Tumorais Cultivadas
11.
J Biol Chem ; 275(32): 24767-75, 2000 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-10825181

RESUMO

The c-Jun N-terminal kinase/stress-activated protein kinase (JNK/SAPK) pathway is activated by numerous cellular stresses. Although it has been implicated in mediating apoptosis and growth factor signaling, its role in regulating cell growth is not yet clear. Here, the influence of JNK on basal (unstimulated) growth of human tumor glioblastoma T98G cells was investigated using highly specific JNK antisense oligonucleotides to inhibit JNK expression. Transient depletion of either JNK1 or JNK2 suppressed cell growth associated with an inhibition of DNA synthesis and cell cycle arrest in S phase. The growth-inhibitory potency of JNK2 antisense ((JNK)2 IC(50) = 0.14 micrometer) was greater than that of JNK1 antisense ((JNK)1 IC(50) = 0.37 micrometer), suggesting that JNK2 plays a dominant role in regulating growth of T98G cells. Indeed, JNK2 antisense-treated populations exhibited greater inhibition of DNA synthesis and accumulation of S-phase cells than did the JNK1 antisense-treated cultures, with a significant proportion of these cells detaching from the tissue culture plate. JNK2 (but not JNK1) antisense-treated cultures exhibited marked elevation in the expression of the cyclin-dependent kinase inhibitor p21(cip1/waf1) accompanied by inhibition of Cdk2/Cdc2 kinase activities. Taken together, these results indicate that JNK is required for growth of T98G cells in nonstress conditions and that p21(cip1/waf1) may contribute to the sustained growth arrest of JNK2-depleted T98G cultures.


Assuntos
Ciclo Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Proteínas Quinases Ativadas por Mitógeno/genética , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Transcrição Gênica/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , DNA de Neoplasias/biossíntese , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , Cinética , Proteína Quinase 8 Ativada por Mitógeno , Proteína Quinase 9 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fase S , Tionucleotídeos , Células Tumorais Cultivadas
13.
J Biol Chem ; 275(27): 20315-23, 2000 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-10783396

RESUMO

EGR-1, a transcription factor with important functions in the regulation of growth and differentiation, is highly expressed in brain. Previous studies have shown that EGR-1 suppresses the transformed phenotype. However, the expression and role of EGR-1 in human glioblastoma cells are not yet determined. In this study, we found that the basal expression of the EGR-1 protein is undetectable, but is inducible in four human glioblastoma cell lines. To determine EGR-1 functions, we re-expressed EGR-1 in human glioblastoma U251 cells and found that the secretion of transforming growth factor-beta1 (TGF-beta1), plasminogen activator inhibitor-1 (PAI-1), and fibronectin (FN) was greatly enhanced. Addition of anti-TGF-beta antibodies completely inhibited the secretion of PAI-1, but had little effect on secretion of FN, indicating that PAI-1 is under the control of EGR-1-induced TGF-beta1. An examination of the promoter of the FN gene revealed two EGR-1-binding sites between positions -75 and -52 and positions -4 and +14 that specifically bound EGR-1 in gel mobility shift experiments. Utilizing wild-type and mutant FN promoter/luciferase reporter genes, we demonstrated that EGR-1 positively regulated the activity of the FN gene. In addition, cell adhesion and migration were greatly increased in the EGR-1-expressing cells, and adhesion was reversed by addition of RGD-containing peptides. These results suggest that EGR-1 may regulate cell interaction with the extracellular matrix by coordinated induction of TGF-beta1, FN, and PAI-1 in human glioblastoma cells.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Fibronectinas/genética , Proteínas Imediatamente Precoces , Fatores de Transcrição/metabolismo , Ativação Transcricional/genética , Anticorpos/farmacologia , Sítios de Ligação , Adesão Celular , Movimento Celular , Proteína 1 de Resposta de Crescimento Precoce , Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Genes Reporter , Glioblastoma , Humanos , Mutação , Oligopeptídeos/farmacologia , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Regiões Promotoras Genéticas , Fator de Crescimento Transformador beta/imunologia , Células Tumorais Cultivadas
14.
Methods Enzymol ; 314: 342-62, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10565024

RESUMO

Methods for the selection and characterization of antisense oligonucleotides for specifically eliminating closely related gene family members are available. High-throughput semiautomated methods using 96-well plate formats and array technology and improved assays are under active development that will streamline many steps and will likely merge. Second-generation 20-mer antisense phosphorothioate oligonucleotides containing 2'-methoxyethyl groups at the first and last 6 nucleotides with improved nuclease resistance and RNA affinity are becoming available.


Assuntos
Proteínas Quinases Ativadas por Mitógeno/genética , Família Multigênica , Oligonucleotídeos Antissenso/farmacologia , Animais , Transformação Celular Neoplásica , Expressão Gênica/efeitos dos fármacos , Meia-Vida , Isoenzimas/genética , Isoenzimas/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno , Lipossomos , Camundongos , Camundongos Knockout , Proteína Quinase 10 Ativada por Mitógeno , Proteína Quinase 9 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fenótipo , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Tionucleotídeos/farmacologia , Transfecção , Transplante Heterólogo
15.
J Biol Chem ; 274(42): 29779-85, 1999 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-10514454

RESUMO

Phorbol ester 12-O-tetradecanoylphorbol-13-acetate (TPA) and all-trans-retinoic acid (trans-RA) are potent regulators of growth of cancer cells. In this study, we investigated the effect of TPA and trans-RA alone or their combination on proliferation of human breast cancer ZR75-1 and T47D and lung cancer H460 and H292 cell lines. trans-RA caused various degrees of growth inhibition of these cell lines. However, TPA showed inhibition of proliferation of H460 and H292 cells and induction of ZR75-1 cell growth. Although trans-RA did not significantly regulate the growth inhibitory effect of TPA, it completely prevented its growth stimulating function. The divergent effects of TPA were associated with specific disruption of cell cycle events, an induction of G(0)/G(1) arrest in H460 and H292 cells and inhibition of G(0)/G(1) arrest with increase of S phase in ZR75-1 cells. Induction of G(0)/G(1) arrest was accompanied by induction of p21(WAF1) and ERK activity, whereas inhibition of G(0)/G(1) arrest was associated with enhanced activity of JNK and AP-1 but not ERK. trans-RA did not affect TPA-induced p21(WAF1) expression. However, it inhibited TPA-induced AP-1 activity in ZR75-1 cells and the constitutive AP-1 activity in H460 and H292 cells. Thus, trans-RA modulates TPA activity through its interaction through TPA-induced JNK/AP-1 pathway but not TPA-induced ERK/p21(WAF1) pathway.


Assuntos
Divisão Celular/efeitos dos fármacos , Acetato de Tetradecanoilforbol/farmacologia , Tretinoína/farmacologia , Neoplasias da Mama/enzimologia , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Ciclo Celular/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/biossíntese , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fator de Transcrição AP-1/antagonistas & inibidores , Fator de Transcrição AP-1/metabolismo , Células Tumorais Cultivadas
16.
J Biol Chem ; 274(44): 31648-54, 1999 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-10531373

RESUMO

We have studied the roles of c-Jun N-terminal protein kinase (JNK) and extracellular signal-regulated protein kinase (ERK) cascade in both the cisplatin-resistant Caov-3 and the cisplatin-sensitive A2780 human ovarian cancer cell lines. Treatment of both cells with cisplatin but not transplatin isomer activates JNK and ERK. Activation of JNK by cisplatin occurred at 30 min, reached a plateau at 3 h, and declined thereafter, whereas activation of ERK by cisplatin showed a biphasic pattern, indicating the different time frame. Activation of JNK by cisplatin was maximal at 1000 microM, whereas activation of ERK was maximal at 100 microM and was less at higher concentrations, indicating the different dose dependence. Cisplatin-induced JNK activation was neither extracellular and intracellular Ca(2+)- nor protein kinase C-dependent, whereas cisplatin-induced ERK activation was extracellular and intracellular Ca(2+)- dependent and protein kinase C-dependent. A mitogen-activated protein kinase/extracellular signal-regulated kinase kinase inhibitor, PD98059, had no effect on the cisplatin-induced JNK activity, suggesting an absence of cross-talk between the ERK and JNK cascades. We further examined the effect of each cascade on the viability following cisplatin treatment. Either exogenous expression of dominant negative c-Jun or the treatment by PD98059 induced sensitivity to cisplatin in both cells. Our findings suggest that cisplatin-induced DNA damage differentially activates JNK and ERK cascades and that inhibition of either of these cascades sensitizes ovarian cancer cells to cisplatin.


Assuntos
Antineoplásicos/farmacologia , Cisplatino/farmacologia , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Neoplasias Ovarianas/metabolismo , Cálcio/metabolismo , Interações Medicamentosas , Ativação Enzimática , Feminino , Flavonoides/farmacologia , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas c-jun/genética , Receptor Cross-Talk , Transdução de Sinais , Estereoisomerismo , Células Tumorais Cultivadas
17.
Cell Growth Differ ; 10(8): 545-54, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10470854

RESUMO

We have previously shown, by expression of a nonphosphorylatable dominant inhibitor mutant of c-Jun [cJun(S63A,S73A)], that activation of the NH2-terminal Jun kinase/stress-activated protein kinase by genotoxic damage is required for DNA repair. Here, we examine the consequences of inhibition of DNA repair on p53-induced apoptosis in T98G cells, which are devoid of endogenous wild-type p53. Relative to parental or wild-type c-Jun-expressing control cells, mutant Jun-expressing T98G clones show similar growth rates and plating efficiencies. However, these cells are unable to repair DNA (PCR-stop assays) and exhibit up to an 80-fold increased methotrexate-induced colony formation due to amplification of the dihydrofolate reductase gene. Moreover, the mutant c-Jun clones exhibit increased apoptosis and elevated bax:bcl2 ratios on expression of wild-type p53. These results indicate that inhibition of DNA repair leads to accumulation of DNA damage in tumor cells with unstable genomes and this, in turn, enhances p53mediated apoptosis.


Assuntos
Apoptose , Reparo do DNA/genética , Proteínas Proto-Oncogênicas c-jun/genética , Proteína Supressora de Tumor p53/fisiologia , Western Blotting , Divisão Celular/efeitos dos fármacos , Divisão Celular/genética , Cisplatino/metabolismo , Células Clonais , Adutos de DNA/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética , Humanos , Metotrexato/farmacologia , Mutação , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Tetra-Hidrofolato Desidrogenase/genética , Células Tumorais Cultivadas , Ensaio Tumoral de Célula-Tronco , Proteína Supressora de Tumor p53/biossíntese , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética , Proteína X Associada a bcl-2
18.
Clin Cancer Res ; 5(9): 2359-65, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10499605

RESUMO

The purpose of this study was to determine the safety, toxicity, and antitumor immune response following S.C. immunizations with a mixture of irradiated, autologous tumor cells and autologous fibroblasts that were genetically modified to express the gene for interleukin 2 (IL-2) in patients with colorectal carcinoma. Ten patients were treated with a fixed dose of tumor cells (10(7)) and escalating doses of fibroblasts secreting IL-2 (per 24 h): 100 units (three patients), 200 units (three patients), 400 units (three patients), and 800 units (one patient). Pre- and posttreatment peripheral blood mononuclear cells were evaluated for evidence of antitumor immune responses. Fatigue and/or flu-like symptoms were experienced by seven patients and delayed-type hypersensitivity-like skin reactions were observed at the sites of the second or subsequent vaccinations in five patients. Low frequencies of tumor cytotoxic T-cell precursors (range, 1/190,000-1/1,320,000 peripheral blood mononuclear cells) were detected prior to therapy in four of seven patients. There was a 5-fold increase following treatment in the frequency of tumor cytotoxic T-cell precursors in two of six evaluable patients. Some patients with colorectal cancer have low frequencies of tumor cytotoxic T-cell precursors that may be increased by this well-tolerated form of IL-2 gene therapy, which warrants continued clinical evaluation.


Assuntos
Vacinas Anticâncer/uso terapêutico , Neoplasias Colorretais/terapia , Fibroblastos/metabolismo , Terapia Genética/métodos , Imunoterapia Adotiva/métodos , Interleucina-2/biossíntese , Interleucina-2/genética , Vacinas Anticâncer/imunologia , Transplante de Células , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Terapia Combinada , Fibroblastos/fisiologia , Fibroblastos/transplante , Engenharia Genética , Terapia Genética/efeitos adversos , Humanos , Hipersensibilidade Tardia/etiologia , Hipersensibilidade Tardia/imunologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/efeitos da radiação , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/efeitos da radiação , Linfócitos T Citotóxicos/transplante
19.
Oncogene ; 18(24): 3633-42, 1999 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-10380885

RESUMO

The human fibrosarcoma cell line, HT1080, clone H4, was used to determine if the transformation suppressive functions of p53 and Egr-1 have the same underlying mechanism. This cell line expresses only mutant p53 and no detectable Egr-1. H4 clones stably expressing Egr-1 are less transformed in proportion to the level of Egr-1 expressed, acting through the induction of the TGFbeta1 gene. Here, H4 cells and the highest Egr-1 expressing clone were transfected with a vector expressing normal human p53 to derive stable clones expressing p53. The expression of p53 in H4 cells inhibited transformed growth and reduced tumorigenicity. The effect of coexpression of both p53 and Egr-1 was additive, producing cell lines with 30% of normal growth rate and sevenfold reduced tumorigenicity compared with control lines. These results indicated that each factor may act independently by different pathways, although each additively increased the level of p21WAF1 cell cycle inhibitor. However, exposure of the H4-derived cells to UV-C irradiation produced contrasting effects. Cell cycle analyses showed that the presence of p53 was associated with loss of the G1 and S cells to apoptosis after irradiation. In contrast, the expression of Egr-1 increased entry into S/G2 phase of the cell cycle with little apoptosis via a mechanism involving elevated FAK and low caspase activities. Apoptosis was observed only in the cell lines that expressed no Egr-1, especially those expressing wt-p53, and was preceded by high caspase activity. In summary, Egr-1 suppressed transformation and counteracted apoptosis by the coordinated activation of TGFbeta1, FN, p21 and FAK, leading to enhanced cell attachment and reduced caspase activity. In the doubly expressing cell line, the survival effect of Egr-1 was dominant over the apoptotic effect of p53.


Assuntos
Apoptose , Transformação Celular Neoplásica , Proteínas de Ligação a DNA/metabolismo , Proteínas Imediatamente Precoces , Proteínas Quinases JNK Ativadas por Mitógeno , Quinases de Proteína Quinase Ativadas por Mitógeno , Proteínas Quinases Ativadas por Mitógeno , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/metabolismo , Apoptose/efeitos da radiação , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Testes de Carcinogenicidade , Caspase 3 , Caspases/metabolismo , Adesão Celular , Moléculas de Adesão Celular/metabolismo , Ciclo Celular/efeitos da radiação , Divisão Celular/efeitos da radiação , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/metabolismo , Fragmentação do DNA/efeitos da radiação , Proteínas de Ligação a DNA/genética , Proteína 1 de Resposta de Crescimento Precoce , Fibrossarcoma , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Humanos , MAP Quinase Quinase 4 , Proteína Quinase 3 Ativada por Mitógeno , Mutação , Proteínas Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Fatores de Transcrição/genética , Transfecção , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética , Raios Ultravioleta
20.
J Biol Chem ; 274(7): 4400-11, 1999 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-9933644

RESUMO

Re-expression of EGR-1 in fibrosarcoma HT1080 suppresses transformation including tumorigenicity (Huang, R.-P., Liu, C., Fan, Y., Mercola, D., and Adamson, E. (1995) Cancer Res. 55, 5054-5062) owing in part to up-regulation of the transforming growth factor (TGF)-beta1 promoter by EGR-1 which suppresses growth by an autocrine mechanism (Liu, C., Adamson, E., and Mercola, D. (1996) Proc. Natl. Acad. Sci. U. S. A. 93, 11831-11836). Here we show that enhanced cell attachment contributes to the suppression via increased secretion of fibronectin (FN) and also of plasminogen activator inhibitor-1 (PAI-1). The secretion of FN and PAI-1 is strongly correlated with EGR-1 expression (RPEARSON = 0.971 and 0. 985, respectively). Addition of authentic TGF-beta1 to parental cells greatly stimulated secretion of PAI-1 but not FN, whereas addition of TGF-beta antibody or lipofection with specific antisense TGF-beta1 oligonucleotides to EGR-1-regulated cells completely inhibits the secretion of PAI-1 but not FN. However, in gel mobility shift assays pure EGR-1 or nuclear extracts of EGR-1-regulated cells specifically bind to two GC-rich elements of the human FN promoter at positions -75/-52 and -4/+18, indicating that the increased secretion of FN is likely due to direct up-regulation by EGR-1. Moreover, adhesion was greatly enhanced in EGR-1-regulated cells and was reversed by treatment with Arg-Gly-Asp (RGD) or PAI-1 antibody indicating that the secreted proteins are functional. We conclude that EGR-1 regulates the coordinated expression of gene products important for cell attachment ("oikis" factor) and normal growth control.


Assuntos
Transformação Celular Neoplásica , Proteínas de Ligação a DNA/farmacologia , Fibronectinas/biossíntese , Fibrossarcoma/metabolismo , Proteínas Imediatamente Precoces/farmacologia , Inibidor 1 de Ativador de Plasminogênio/biossíntese , Fatores de Transcrição/farmacologia , Fator de Crescimento Transformador beta/biossíntese , Dedos de Zinco , Adesão Celular/efeitos dos fármacos , Proteína 1 de Resposta de Crescimento Precoce , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Oligopeptídeos/farmacologia , Regiões Promotoras Genéticas , Transfecção , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA