Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Environ Toxicol ; 30(2): 129-36, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23765435

RESUMO

Divalent lead ions (Pb(2+) ) are toxic environmental pollutants known to cause serious health problems in humans and animals. Absorption of Pb(2+) from air, water, and food takes place in the respiratory and digestive tracts. The ways in which absorbed Pb(2+) affects cell physiology are just beginning to be understood at the molecular level. Here, we used reverse transcription PCR and Western blotting to analyze cultures of human gastric carcinoma cells exposed to 10 µM lead nitrate. We found that Pb(2+) induces gastrin hormone gene transcription and translation in a time-dependent manner. Promoter deletion analysis revealed that activator protein 1 (AP1) was necessary for gastrin gene transcription in cells exposed to Pb(2+) . MitogIen-activated protein kinase (MAPK)/ERK kinase inhibitor PD98059 suppressed the Pb(2+) -induced increase in messenger RNA. Epidermal growth factor receptor (EGFR) inhibitors AG1478 and PD153035 reduced both transcription and phosphorylation by extracellular signal-regulated kinase (ERK1/2). Cells exposed to Pb(2+) also increased production of c-Jun protein, a component of AP1, and over-expression of c-Jun enhanced activation of the gastrin promoter. In sum, the findings suggest the EGFR-ERK1/2-AP1 pathway mediates the effects of Pb(2+) on gastrin gene activity in cell culture.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Gastrinas/biossíntese , Gastrinas/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Chumbo/toxicidade , Fator de Transcrição AP-1/efeitos dos fármacos , Linhagem Celular Tumoral , Repressão Epigenética/efeitos dos fármacos , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/efeitos dos fármacos , Humanos , Sistema de Sinalização das MAP Quinases , Fosforilação , Proteínas Proto-Oncogênicas c-jun/farmacologia
2.
PLoS One ; 7(5): e36941, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22649503

RESUMO

Our previous study shows that inhibiting activator protein one (AP1) transcription factor function in murine epidermis, using dominant-negative c-jun (TAM67), increases cell proliferation and delays differentiation. To understand the mechanism of action, we compare TAM67 impact in mouse epidermis and in cultured normal human keratinocytes. We show that TAM67 localizes in the nucleus where it forms TAM67 homodimers that competitively interact with AP1 transcription factor DNA binding sites to reduce endogenous jun and fos factor binding. Involucrin is a marker of keratinocyte differentiation that is expressed in the suprabasal epidermis and this expression requires AP1 factor interaction at the AP1-5 site in the promoter. TAM67 interacts competitively at this site to reduce involucrin expression. TAM67 also reduces endogenous c-jun, junB and junD mRNA and protein level. Studies with c-jun promoter suggest that this is due to reduced transcription of the c-jun gene. We propose that TAM67 suppresses keratinocyte differentiation by interfering with endogenous AP1 factor binding to regulator elements in differentiation-associated target genes, and by reducing endogenous c-jun factor expression.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Queratinócitos/fisiologia , Fragmentos de Peptídeos/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Fator de Transcrição AP-1/metabolismo , Animais , Ligação Competitiva , Diferenciação Celular/fisiologia , Células Cultivadas , Imunoprecipitação da Cromatina , Primers do DNA/genética , Ensaio de Desvio de Mobilidade Eletroforética , Imunofluorescência , Humanos , Immunoblotting , Imunoprecipitação , Queratinócitos/metabolismo , Camundongos , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/farmacologia , Precursores de Proteínas , Proteínas Proto-Oncogênicas c-jun/genética , Proteínas Proto-Oncogênicas c-jun/farmacologia , Reação em Cadeia da Polimerase em Tempo Real , Elementos Reguladores de Transcrição/efeitos dos fármacos , Elementos Reguladores de Transcrição/genética
3.
J Immunol ; 186(5): 3173-9, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21257959

RESUMO

Osteopontin (OPN) is expressed by various immune cells and modulates both innate and adaptive immune responses. However, the molecular mechanisms that control opn gene expression, especially at the chromatin level, remain largely unknown. We have previously demonstrated many specific cis- and trans-regulatory elements that determine the extent of endotoxin (LPS)-mediated induction of OPN synthesis in murine macrophages. In the present study, we confirm that NF-κB also plays an important role in the setting of LPS-stimulated OPN expression through binding to a distal regulatory element. Importantly, we demonstrate that LPS stimulates chromosomal loops in the OPN promoter between NF-κB binding site and AP-1 binding site using chromosome conformation capture technology. The crucial role of NF-κB and AP-1 in LPS-stimulated DNA looping was confirmed, as small interfering RNA knock-down of NF-κB p65 and AP-1 c-Jun exhibited decreased levels of DNA looping. Furthermore, we demonstrate that p300 can form a complex with NF-κB and AP-1 and is involved in DNA looping and LPS-induced OPN expression. Therefore, we have identified an essential mechanism to remodel the local chromatin structures and spatial conformations to regulate LPS-induced OPN expression.


Assuntos
DNA/química , Lipopolissacarídeos/fisiologia , Ativação de Macrófagos/genética , NF-kappa B/fisiologia , Conformação de Ácido Nucleico , Osteopontina/genética , Fator de Transcrição AP-1/fisiologia , Transcrição Gênica/imunologia , Animais , Linhagem Celular , DNA/antagonistas & inibidores , DNA/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/biossíntese , Regulação da Expressão Gênica/imunologia , Quinase I-kappa B/biossíntese , Ativação de Macrófagos/imunologia , Camundongos , NF-kappa B/antagonistas & inibidores , NF-kappa B/genética , Osteopontina/biossíntese , Osteopontina/metabolismo , Ligação Proteica/genética , Ligação Proteica/imunologia , Proteínas Proto-Oncogênicas c-jun/genética , Proteínas Proto-Oncogênicas c-jun/farmacologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Fator de Transcrição AP-1/química , Fator de Transcrição AP-1/genética , Fator de Transcrição RelA/antagonistas & inibidores , Fatores de Transcrição de p300-CBP/metabolismo
4.
Biomacromolecules ; 12(1): 19-27, 2011 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-21141810

RESUMO

Polymer therapeutics, including polymeric drugs and polymer-protein conjugates, are clinically established as first-generation nanomedicines. Knowing that the coiled-coil peptide motif is fundamentally important in the regulation of many cellular and pathological processes, the aim of these studies was to examine the feasibility of designing polymer conjugates containing the coiled-coil motif as a putative therapeutic "molecular switch". To establish proof of concept, we prepared a mPEG-FosW(C) conjugate by reacting mPEG-maleimide (M(w) 5522 g mol(-1), M(w)/M(n) 1.1) with a FosW peptide synthesized to contain a terminal cysteine residue (FosW(C)). Its ability to form a stable coil-coil heterodimer with the target c-Jun sequence of the oncogenic AP-1 transcription factor was investigated using 2D (15)N-HSQC NMR together with a recombinantly prepared (15)N-labeled c-Jun peptide ([(15)N]r-c-Jun). Observation that heterodimerization was achieved and that the polymer did not sterically disadvantage hybridization suggests an important future for this new family of polymer therapeutics.


Assuntos
Nanomedicina/métodos , Polietilenoglicóis , Proteínas Proto-Oncogênicas c-fos , Proteínas Proto-Oncogênicas c-jun , Linhagem Celular Tumoral , Humanos , Ressonância Magnética Nuclear Biomolecular , Polietilenoglicóis/química , Polietilenoglicóis/farmacologia , Estrutura Secundária de Proteína , Proteínas Proto-Oncogênicas c-fos/química , Proteínas Proto-Oncogênicas c-fos/farmacologia , Proteínas Proto-Oncogênicas c-jun/química , Proteínas Proto-Oncogênicas c-jun/farmacologia , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia
5.
Mol Carcinog ; 46(11): 901-11, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17477349

RESUMO

Although activating protein-1 (AP-1) transcription factors play an important role in mediating metastasis for nasopharyngeal carcinoma (NPC), the biological and physiological functions of AP-1, in relation to the oncogenic phenotype of NPC, are not fully understood. Our previous study showed that the latent membrane protein 1 (LMP1) mediated a primary dimer form of c-jun and jun B. In this study, we used a NPC cell line that express a specific inhibitor of AP-1, a dominant-negative c-jun mutant (TAM67), to investigate the role of AP-1 in regulating the NPC oncogenic phenotype. First, we observed that TAM67 inhibited cell growth in vitro and in vivo. Next, with Western blotting, we discovered that TAM67 impaired the cyclin D1/cdk4 complex but had little effect on the cyclin E/cdk2 complex, concomitantly with inhibiting Rb phosphorylation. RT-PCR and luciferase assay results demonstrated that the levels of cyclin D1 mRNA and the promoter activity in TAM67 transfectants were reduced as compared with control cells. Thereby, we show that blockade of AP-1 transcriptional activity has a negative impact on cyclin D1 transcription. We obtained the first evidence that TAM67 prevented NPC growth both in vitro and in vivo. AP-1 appears to be a novel target for treating or preventing LMP1-positive NPC effectively.


Assuntos
Neoplasias Nasofaríngeas/metabolismo , Fator de Transcrição AP-1/metabolismo , Proteínas da Matriz Viral/metabolismo , Animais , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Ciclina D1/metabolismo , Quinases Ciclina-Dependentes , Humanos , Camundongos , Camundongos Nus , Neoplasias Nasofaríngeas/patologia , Transplante de Neoplasias , Fragmentos de Peptídeos/farmacologia , Proteínas Proto-Oncogênicas c-jun/farmacologia , Células Tumorais Cultivadas
6.
Oral Microbiol Immunol ; 22(1): 24-9, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17241167

RESUMO

This study aimed to elucidate the modulation by nitric oxide (NO) of the apoptosis of macrophages and osteoblasts, the essential cellular components in the development of periapical lesions. Lipopolysaccharide (LPS) induced prominent nitrite synthesis in J774 mouse macrophage cell lines. Exposure to LPS induced obvious apoptosis in J774 cells, whereas transient transfection with murine inducible nitric oxide synthase (iNOS), small interfering RNA (siRNA) diminished this effect. Tumor necrosis factor-alpha (TNF-alpha) and S-nitroso-N-acetyl-DL-penicillamine (SNAP) (a NO donor) triggered apoptosis in UMR-106 rat osteoblastic cell lines and a synergistic effect was noted when TNF-alpha and SNAP were added to the medium together. Administration of siRNAs for c-Fos and c-Jun: components of activator protein-1 (AP-1) and transforming growth factor-beta1 attenuated the combined effect markedly. Terminal deoxynucleotidyl transferase-mediated nick end-labeling (TUNEL) stain in a rat model of induced periapical lesion showed positive apoptotic signals in macrophages and osteoblasts. Administration of N(G)-monomethyl-l-arginine markedly diminished the extent of bone loss and the amounts of apoptotic macrophages and osteoblasts. In conclusion, NO mediates LPS-stimulated apoptosis of macrophages. It also induces osteoblast apoptosis and augments the pro-apoptotic effect of cytokines. Inhibition of NO synthesis in vivo attenuates apoptosis and the size of periapical lesions. Taken together, these results suggest that NO may promote the progression of periapical lesion by inducing the apoptosis of macrophages and osteoblasts.


Assuntos
Apoptose/efeitos dos fármacos , Sequestradores de Radicais Livres/farmacologia , Macrófagos/efeitos dos fármacos , Óxido Nítrico/farmacologia , Osteoblastos/efeitos dos fármacos , Doenças Periapicais/fisiopatologia , Animais , Linhagem Celular , Células Cultivadas , Modelos Animais de Doenças , Progressão da Doença , Sinergismo Farmacológico , Inibidores Enzimáticos/farmacologia , Lipopolissacarídeos/farmacologia , Camundongos , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico Sintase Tipo II/farmacologia , Nitritos/metabolismo , Proteínas Proto-Oncogênicas c-fos/farmacologia , Proteínas Proto-Oncogênicas c-jun/farmacologia , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Wistar , S-Nitroso-N-Acetilpenicilamina/farmacologia , ômega-N-Metilarginina/farmacologia
7.
Carcinogenesis ; 28(5): 977-87, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17116726

RESUMO

Silibinin is a natural flavonoid antioxidant with anti-hepatotoxic properties and pleiotropic anticancer capabilities. We tested the hypothesis that silibinin inhibits cellular invasiveness by down-regulating the focal adhesion kinase (FAK) and extracellular signal-regulated protein kinase (ERK)-dependent c-Jun/activator protein-1 (AP-1) induction, which leads to inhibition of urokinase-type plasminogen activator (u-PA) and matrix metalloproteinase-2 (MMP-2) expressions in human osteosarcoma MG-63 cells. We found that silibinin decreased cell adhesion and invasiveness, as well as inhibited u-PA and MMP-2 expressions. Silibinin reduced ERK 1/2 phosphorylation, but had no effects on the phosphorylation of c-Jun N-terminal kinases (JNKs) 1/2, p38 and Akt. Silibinin suppressed AP-1-binding activity and c-Jun levels and its phosphorylation without changes of c-Fos and Ets-1 levels. Silibinin also inhibited interleukin-6-induced ERK 1/2 and c-Jun phosphorylation, and cell invasiveness. Thus, silibinin may possess an anti-metastatic activity in MG-63 cells.


Assuntos
Neoplasias Ósseas/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/farmacologia , Metaloproteinase 2 da Matriz/metabolismo , Osteossarcoma/metabolismo , Proteínas Proto-Oncogênicas c-jun/farmacologia , Fator de Transcrição AP-1/farmacologia , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Neoplasias Ósseas/patologia , Adesão Celular , Linhagem Celular Tumoral , Forma Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Progressão da Doença , Quinase 1 de Adesão Focal/metabolismo , Humanos , Masculino , Invasividade Neoplásica , Osteossarcoma/patologia , Transdução de Sinais/efeitos dos fármacos , Silibina , Silimarina/farmacologia
8.
Oncogene ; 24(6): 1001-10, 2005 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-15592508

RESUMO

Disruption of transcriptional control of cellular genes by human T-cell leukemia virus type-1 (HTLV-1) is thought to be associated, at least in part, with the development of adult T-cell leukemia. It has been reported that activating protein-1 (AP-1) is dysregulated by HTLV-1 infection. HTLV-1-encoded Tax elevates AP-1 activity through the induction of AP-1 family member gene expression, including c-Jun, JunD, c-Fos, and Fra-1. However, the precise mechanism by which HTLV-1 regulates AP-1 activity remains to be addressed. Recently, a novel viral protein named HTLV-1 basic leucine-zipper factor, HBZ, has been shown to interact with c-Jun and repress c-Jun-mediated transcription by abrogating its DNA-binding activity. In the course of investigating HBZ function, we found that HBZ reduced the steady-state levels of c-Jun, and the levels were restored by treatment with a proteasome inhibitor. Together, this indicates that HBZ promotes c-Jun degradation through a proteasome-dependent pathway. Furthermore, HBZ deletion mutants revealed that both the N-terminal and leucine-zipper region of HBZ were required for the elimination of c-Jun. These results suggest dual effects of HBZ on the suppression of AP-1 activity by inhibiting c-Jun function, which may contribute to the dysregulation of cell proliferation.


Assuntos
DNA/metabolismo , DNA/farmacologia , Vírus Linfotrópico T Tipo 1 Humano/genética , Vírus Linfotrópico T Tipo 1 Humano/patogenicidade , Proteínas Proto-Oncogênicas c-jun/biossíntese , Fatores de Transcrição/farmacologia , Proteínas Virais/farmacologia , Fatores de Transcrição de Zíper de Leucina Básica , DNA/genética , Regulação da Expressão Gênica , Genes jun , Células HeLa , Humanos , Zíper de Leucina , Leucemia-Linfoma de Células T do Adulto/genética , Leucemia-Linfoma de Células T do Adulto/virologia , Proteínas Proto-Oncogênicas c-jun/farmacologia , Proteínas dos Retroviridae , Fator de Transcrição AP-1 , Transcrição Gênica , Células Tumorais Cultivadas
9.
Int J Cancer ; 114(6): 863-9, 2005 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-15609298

RESUMO

Tumor cells frequently synthesize an N-terminally extended the FGF-2 isoform of 24 kDa devoid of signal peptide but that contains a functional nuclear localization sequence (NLS). Although the signaling pathways elicited by secreted FGF-2 are well described, the molecular mechanisms involved in the growth promoting action of nuclearized 24 kDa FGF-2 remain unknown. The cancer cell line AR4-2J was engineered to stably express only the 24 kDa FGF-2 isoform and cDNA microarrays were used to identify targets implicated in the intracrine-induced cell proliferation. Levels of 27 transcripts were found either upregulated or downregulated compared to control cells. Among the 18 upregulated genes was c-jun, which is often involved in cell proliferation. Real-time PCR and Western blot analyses confirmed c-jun induction at both mRNA and protein levels. The c-jun antisense oligonucleotide strategy pointed out the involvement of c-Jun in the 24 kDa FGF-2-induced cell proliferation. The mitogenic effect was found to depend on ERK pathway and not on phosphoinositide 3-kinase, p38 MAPK, c-Jun NH2-terminal kinase signal transducers. In addition, the MEK inhibitor PD98059 reduced the 24 kDa FGF-2-dependent c-Jun level. These data show that intracrine FGF-2-mediated regulation of cell growth involves ERK activation and consequent c-Jun expression. Thus, despite its incapacity to be secreted, the intracellular-localized 24 kDa FGF-2 can activate a growth-related signaling pathway normally elicited by cell surface receptors.


Assuntos
Proliferação de Células , Fator 2 de Crescimento de Fibroblastos/farmacologia , Proteínas Proto-Oncogênicas c-jun/farmacologia , Animais , Western Blotting , Análise de Sequência com Séries de Oligonucleotídeos , Oligonucleotídeos Antissenso , Neoplasias Pancreáticas/patologia , Reação em Cadeia da Polimerase , Isoformas de Proteínas , Ratos , Retroviridae/genética , Transdução de Sinais , Transfecção , Células Tumorais Cultivadas
10.
Int J Cancer ; 112(4): 570-6, 2004 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-15382037

RESUMO

We investigated the effects of antagonists of growth hormone-releasing hormone (GHRH) alone and in combination with bombesin/gastrin-releasing peptide (BN/GRP) antagonist RC-3940-II on the growth of H-69 human small cell lung carcinoma (SCLC) xenografted into nude mice. Since the activation of the signaling pathways involving protein kinase C (PKC) and the subsequent steps involving mitogen-activated protein kinase (MAPK) and c-fos and c-jun oncogenes are known to be important mechanisms implicated in cellular growth, we investigated how the blockade of tumoral GHRH receptor splice variants and BN/GRP receptors by these antagonists could interfere with these intracellular signaling pathways. Treatment with GHRH antagonists JV-1-65 or MZ-J-7-110 for 4 weeks significantly (p<0.05) decreased the tumor volume by 22.7+/-3.0% and 36.7 +/- 3.6%, respectively, as compared to controls. A larger decrease in tumor volume of 73.0 +/- 9.5% (p<0.01) was produced by BN/GRP antagonist RC-3940-II and its combination with JV-I-65 caused the greatest tumor reduction of 91.0 +/- 9.8% (p<0.01) vs. controls. H-69 SCLC tumors expressed alpha-, betaII-, delta- and eta-PKC isoforms. Antagonists of GHRH and BN/GRP decreased significantly (p<0.05) the expression of betaII- and delta-, but not of alpha- and eta-PKC isoforms. They also inhibited MAPK levels, the effects being significant (p<0.05) in the groups that received BN/GRP antagonist. In addition, expression of c-fos and c-jun mRNA was reduced after combined treatment with JV-1-65 and RC-3940-II. The proliferation of H-69 SCLC cells "in vitro" was also significantly inhibited after incubation of cells with GHRH antagonist, PKC inhibitors or MAPK inhibitor. These findings suggest that the anti-proliferative effects of antagonists of GHRH and BN/GRP on H69-SCLC involve an inhibition of the signaling pathways of specific PKC isoforms, MAPK and c-fos and c-jun oncogenes.


Assuntos
Bombesina/farmacologia , Carcinoma de Células Pequenas/fisiopatologia , Hormônio Liberador de Hormônio do Crescimento/farmacologia , Neoplasias Pulmonares/fisiopatologia , Quinases de Proteína Quinase Ativadas por Mitógeno/farmacologia , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/farmacologia , Animais , Divisão Celular , Hormônio Liberador de Hormônio do Crescimento/antagonistas & inibidores , Humanos , Camundongos , Camundongos Nus , Proteínas Proto-Oncogênicas c-fos/farmacologia , Proteínas Proto-Oncogênicas c-jun/farmacologia , Transdução de Sinais , Transplante Heterólogo
11.
Endocr Res ; 30(4): 807-12, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15666830

RESUMO

We have previously reported that the protein kinase C ligand 12-O-tetradecanoyphorbol-13-acetate (TPA) inhibited the angiotensin II (AII) stimulated CYP11B2 gene expression in the adrenocortical H295R cell line. Here we report that TPA increased the level of phospho-p44/42 MAPK but AII did not. The MEK1 inhibitor PD98059 was found to increase the level of aldosterone synthase mRNA and the activity of a human CYP11B2(-2023 bp)-promoter construct. The cotransfection of H295R with ERK 1 and the hCYP11B2 promoter resulted in the inhibition of the promoter activity. TPA but not AII increased the level of the transcription factor JunB in nuclear extracts and the increase was partially abolished by the MEK1 inhibitor PD98059. The cotransfection of H295R with JunB and the hCYP11B2 promoter abolished the AII stimulating effect. Taken together these results suggest that TPA inhibits the AII-dependent activation of CYP11B2 via the p44/42 MAPK signaling pathway leading to an increase of the level of nuclear JunB.


Assuntos
Citocromo P-450 CYP11B2/genética , Citocromo P-450 CYP11B2/metabolismo , Regulação da Expressão Gênica , Angiotensina II/farmacologia , Linhagem Celular Tumoral , Flavonoides/farmacologia , Humanos , MAP Quinase Quinase 1/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-jun/metabolismo , Proteínas Proto-Oncogênicas c-jun/farmacologia , RNA Mensageiro/metabolismo , Transdução de Sinais/fisiologia , Acetato de Tetradecanoilforbol/farmacologia , Transfecção
12.
J Steroid Biochem Mol Biol ; 87(2-3): 123-31, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14672732

RESUMO

The uterine endometrium and cancers derived from it are classic models of hormone action: estrogen promotes growth and progesterone inhibits proliferation and results in differentiation. We have now identified a major pathway through which progesterone causes these growth-limiting effects. Ligand-bound progesterone receptors modulate the composition and transcriptional activity of members of the activating protein-1 (AP-1) family, and in particular, c-Jun. First, a dominant negative form of c-Jun inhibits the constitutive growth of Hec50co cells in a manner similar to the effects of progesterone through progesterone B receptors. Second, progesterone inhibits the transcriptional activity of the AP-1 complex in reporter gene assays. Third, the DNA binding of AP-1 and the composition of the individual AP-1 factors on DNA is regulated by progesterone on electrophoretic mobility shift assays. Fourth, progesterone strongly inhibits total AP-1 as well as c-Jun recruitment to the cyclin D1 promoter, but enhances AP-1 occupancy on the p53 and p21 promoters, as shown by chromatin immunoprecipitation assays. The effects of progesterone on AP-1 DNA binding are confirmed to result in altered transcription of these AP-1 target genes by RT-PCR. These studies establish that modulation of AP-1 activity is a potential pathway of progesterone-induced growth inhibition in endometrial cancer cells.


Assuntos
Neoplasias do Endométrio/patologia , Progesterona/farmacologia , Fator de Transcrição AP-1/fisiologia , Transcrição Gênica/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Ciclina D1/biossíntese , Ciclina D1/genética , DNA/metabolismo , Neoplasias do Endométrio/tratamento farmacológico , Feminino , Humanos , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-jun/metabolismo , Proteínas Proto-Oncogênicas c-jun/farmacologia , RNA Mensageiro/biossíntese , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Fator de Transcrição AP-1/antagonistas & inibidores , Proteína Supressora de Tumor p53/biossíntese , Proteína Supressora de Tumor p53/genética , Proteínas rho de Ligação ao GTP/biossíntese , Proteínas rho de Ligação ao GTP/genética
14.
J Pharmacol Sci ; 91(2): 145-8, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12686758

RESUMO

The mitogen-activated protein (MAP) kinase pathways has been shown to be necessary for mitogen-stimulated proliferation, but its role in cell migration has not been fully understood. In this study, we investigated the possible contribution of signaling pathways through c-Jun in platelet-derived growth factor (PDGF)-BB directed cell migration in rat aortic vascular smooth muscle cells (VSMCs) infected with a recombinant adenovirus containing the dominant-negative c-Jun (Ad-DN-c-Jun). DN-c-Jun protein was expressed dose-dependently in VSMCs infected with Ad-DN-c-Jun. Expression of DN-c-Jun significantly inhibited VSMC migration induced by PDGF-BB. Our results provide the first evidence that signaling pathways through c-Jun participates in cell migration induced by PDGF-BB in addition to other MAP kinase pathways in VSMCs.


Assuntos
Movimento Celular/efeitos dos fármacos , Proteínas Quinases JNK Ativadas por Mitógeno , Músculo Liso Vascular/citologia , Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteínas Proto-Oncogênicas c-jun/farmacologia , Animais , Aorta Torácica/citologia , Aorta Torácica/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Genes Dominantes , Imidazóis/farmacologia , Técnicas In Vitro , MAP Quinase Quinase 4 , Masculino , Quinases de Proteína Quinase Ativadas por Mitógeno/fisiologia , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Músculo Liso Vascular/efeitos dos fármacos , Piridinas/farmacologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
15.
Am J Physiol Gastrointest Liver Physiol ; 281(5): G1279-89, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11668037

RESUMO

Transforming growth factor-beta (TGF-beta)-activated kinase 1 (TAK1), a serine/threonine kinase, is reported to function in the signaling pathways of TGF-beta, interleukin 1, and ceramide. However, the physiological role of TAK1 in vivo is largely unknown. To assess the function of TAK1 in vivo, dominant-negative TAK1 (dnTAK1) was expressed in the rat liver by adenoviral gene transfer. dnTAK1 expression abrogated c-Jun NH(2)-terminal kinase and c-Jun but not nuclear factor (NF)-kappaB or SMAD activation after partial hepatectomy (PH). Expression of dnTAK1 or TAM-67, a dominant-negative c-Jun, induced G(0) exit in quiescent liver and accelerated cell cycle progression after PH. Finally, dnTAK1 and TAM-67 induced c-myc expression in the liver before and after PH, suggesting that G(0) exit induced by dnTAK1 and TAM-67 is mediated by c-myc induction.


Assuntos
Regulação da Expressão Gênica , Genes Dominantes , Genes myc/fisiologia , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/fisiologia , Fase de Repouso do Ciclo Celular/fisiologia , Animais , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/metabolismo , Proteínas de Ligação a DNA/fisiologia , Ativação Enzimática/efeitos dos fármacos , Genes jun/efeitos dos fármacos , Genes jun/fisiologia , Genes myc/efeitos dos fármacos , Hepatectomia/métodos , Proteínas Quinases JNK Ativadas por Mitógeno , MAP Quinase Quinase Quinases/farmacologia , Masculino , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Fragmentos de Peptídeos/farmacologia , Proteínas Proto-Oncogênicas c-jun/farmacologia , Ratos , Ratos Sprague-Dawley , Fase de Repouso do Ciclo Celular/efeitos dos fármacos , Proteína Smad3 , Proteína Smad4 , Transativadores/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno
16.
Circulation ; 104(13): 1557-63, 2001 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-11571252

RESUMO

BACKGROUND: Activity of voltage-gated K(+) (K(v)) channels controls membrane potential (E(m)) that regulates cytosolic free Ca(2+) concentration ([Ca(2+)](cyt)) by regulating voltage-dependent Ca(2+) channel function. A rise in [Ca(2+)](cyt) in pulmonary artery smooth muscle cells (PASMCs) triggers vasoconstriction and stimulates PASMC proliferation. Whether c-Jun, a transcription factor that stimulates cell proliferation, affects K(v) channel activity in PASMCs was investigated. METHODS AND RESULTS: Infection of primary cultured PASMCs with an adenoviral vector expressing c-jun increased the protein level of c-Jun and reduced K(v) currents (I(K(V))) compared with control cells (infected with an empty adenovirus). Using single-cell reverse transcription-polymerase chain reaction, we observed that the mRNA level of Kv1.5 and the current density of I(K(V)) were both attenuated in c-jun-infected PASMCs compared with control cells and cells infected with antisense c-jun. Overexpression of c-Jun also upregulated protein expression of Kvbeta(2) and accelerated I(K(V)) inactivation. Furthermore, E(m) was more depolarized and [(3)H]thymidine incorporation was greater in PASMCs infected with c-jun than in control cells and cells infected with antisense c-jun. CONCLUSIONS: These results suggest that c-Jun-mediated PASMC proliferation is associated with a decrease in I(K(V)). The resultant membrane depolarization increases [Ca(2+)](cyt) and enhances PASMC growth.


Assuntos
Músculo Liso/efeitos dos fármacos , Canais de Potássio/metabolismo , Proteínas Proto-Oncogênicas c-jun/farmacologia , Adenoviridae/genética , Animais , Divisão Celular/efeitos dos fármacos , Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos , Músculo Liso/citologia , Músculo Liso/metabolismo , Canais de Potássio/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-jun/genética , Artéria Pulmonar/citologia , Artéria Pulmonar/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
17.
J Cell Biochem ; 82(1): 163-70, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11400173

RESUMO

The bradykinin B1 receptor (BKB1R) gene is expressed in selected tissues such as lung and kidney. In these tissues it is expressed at a very low level until induced by inflammatory mediators. Our aim has been to understand the mechanism of this regulatory process. A human BKB1R minigene was constructed. It contained a 1.8 kb promoter, the entire exon I, 1.5 kb of intron I, the entire exon II and intron II, and the luciferase gene as a reporter. Transient transfection of the minigene into SV40-transformed IMR90 cells (IMRSV) resulted in a promoter activity which was activated by the mediators, lipopolysaccharide and (LPS) desArg(10)-kallidin. In contrast, these mediators did not induce the activity of the 1.8 kb promoter construct alone. Thus, motifs exclusive of the promoter such as 5'-UTR and/or intron regions are required for mediator-induced expression of this gene. Promoter activities of both the minigene and the 1.8 kb promoter construct were enhanced in a dose-dependent manner upon cotransfection with c-Jun. Furthermore, cotransfecting c-Jun with the minigene achieved the maximal promoter activity with no further increase in response to mediators. Conversely, the induction of the minigene promoter activity by mediators was abolished upon cotransfection with a dominant negative mutant of c-Jun. Other experiments suggest that multiple AP-1 sites are interactive with the c-Jun upregulation of this gene. Taken together, these results point to c-Jun as a key intermediary in the activation of the expression of this gene by mediators. However, participation of motifs outside of the promoter are necessary to obtain this inducible expression.


Assuntos
Calidina/análogos & derivados , Calidina/metabolismo , Lipopolissacarídeos/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Receptores da Bradicinina/genética , Éxons/genética , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/genética , Humanos , Íntrons/genética , Calidina/farmacologia , Lipopolissacarídeos/farmacologia , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas c-jun/genética , Proteínas Proto-Oncogênicas c-jun/farmacologia , Receptor B1 da Bradicinina , Receptores da Bradicinina/efeitos dos fármacos , TATA Box/genética , Fator de Transcrição AP-1/química , Fator de Transcrição AP-1/genética , Ativação Transcricional , Regulação para Cima
18.
Biochem Biophys Res Commun ; 284(5): 1134-9, 2001 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-11414701

RESUMO

The c-jun gene is a major regulator of proliferative and stress responses of both normal and transformed cells. In general, during immortalization/transformation c-jun cooperates with oncogenic signals rather than acting as an oncogene itself. Here we report a novel example of this cooperation, the requirement for c-jun to sustain expression of the matrix metalloproteinase-2 (MMP-2) gene in cells immortalized by SV40 large T-antigen (TAg). MMP-2 encodes a type IV collagenase that is secreted by cells within normal and tumor microenvironments. We used wild-type and c-jun null primary and TAg-immortalized mouse embryonic fibroblasts (mEFs) to investigate the importance of c-jun for the regulation of this activity, and observed that c-jun is essential for MMP-2 expression in immortalized but not primary mEFs. This finding directly demonstrates a cooperative interaction of c-jun with an oncogene, and suggests that TAg dependent immortalization/transformation may require other c-Jun/AP-1-dependent genes.


Assuntos
Antígenos Transformantes de Poliomavirus/farmacologia , Metaloproteinase 2 da Matriz/biossíntese , Proteínas Proto-Oncogênicas c-jun/farmacologia , Animais , Linhagem Celular Transformada , Transformação Celular Viral , Colagenases/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Fibroblastos/metabolismo , Expressão Gênica/efeitos dos fármacos , Camundongos , Peso Molecular , Retroviridae/genética , Transcrição Gênica/efeitos dos fármacos
19.
Mol Cell Endocrinol ; 175(1-2): 67-79, 2001 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-11325517

RESUMO

The regulation of glucocorticoid receptor gene expression by members of the AP-1 family was examined in glucocorticoid-free NIH3T3 cells transfected with the human glucocorticoid receptor gene promoter driving expression of a CAT reporter gene. c-Jun inhibited the promoter activity by 80% and JunB by 30%, whereas c-Fos and JunD had no inhibitory effect. Electrophoretic mobility shift assays showed that c-Jun is unable to efficiently interact with the AP-1-like site present in the human glucocorticoid receptor promoter. Moreover, c-Jun was still able to repress promoter mutants in which the region containing the AP-1-like site was deleted. NIH3T3 cell clones overexpressing c-Jun exhibited lower glucocorticoid receptor mRNA levels, which suggests that the murine glucocorticoid receptor gene can also be regulated by AP-1. These results provide a new mechanism for cross-talk between the glucocorticoid receptor and the AP-1 family of transcription factors in the absence of glucocorticoid ligands.


Assuntos
Proteínas Proto-Oncogênicas c-jun/farmacologia , Receptores de Glucocorticoides/genética , Transcrição Gênica/efeitos dos fármacos , Células 3T3 , Animais , Regulação para Baixo/efeitos dos fármacos , Humanos , Camundongos , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas c-jun/genética , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Transdução de Sinais , Fator de Transcrição AP-1/metabolismo , Fator de Transcrição AP-1/farmacologia , Transfecção
20.
Mol Cell Endocrinol ; 175(1-2): 205-10, 2001 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-11325530

RESUMO

The CYP11A1 gene encodes cytochrome P450scc, the enzyme catalyzing the first step of steroid biosynthesis in the adrenal and gonad. We generated transgenic mice containing 2.3 kb of the 5'-flanking region of CYP11A1 driving LacZ reporter gene expression, in order to study hormonal control of CYP11A1 gene expression in different tissues. This 2.3 kb fragment contains information for hormonal control; by ACTH and hCG which increased reporter gene expression, in the adrenal and testis of transgenic mice respectively, while dexamethasone administration decreased reporter activity in the adrenal. The 5'-fragment of CYP11A1 has appreciable promoter activities in mouse adrenal Y1 cells but not in non-steroidogenic COS-1 cells, showing cell-type specificity. Transcription factor SF-1 activates the 2.3 kb promoter, which can be potentiated by cotransfection with c-Jun in steroidogenic JEG3 cells but not in COS-1 cells. We conclude that the 2.3 kb region of CYP11A1 contains elements controlling hormonal-dependent, cell-type-specific expression. In addition, c-Jun and SF-1 could act synergistically to activate CYP11A1 gene expression.


Assuntos
Enzima de Clivagem da Cadeia Lateral do Colesterol/genética , Hormônios/farmacologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Glândulas Suprarrenais/citologia , Hormônio Adrenocorticotrópico/farmacologia , Animais , Sequência de Bases/efeitos dos fármacos , Sequência de Bases/genética , Gonadotropina Coriônica/farmacologia , Proteínas de Ligação a DNA/farmacologia , Dexametasona/farmacologia , Sinergismo Farmacológico , Feminino , Fatores de Transcrição Fushi Tarazu , Proteínas de Homeodomínio , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Especificidade de Órgãos , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas c-jun/farmacologia , Receptores Citoplasmáticos e Nucleares , Fator Esteroidogênico 1 , Testículo/citologia , Fatores de Transcrição/farmacologia , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...