Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell Death Differ ; 13(9): 1475-84, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16322753

RESUMO

Although cyclin G1 has been implicated in certain p53-related biological phenomena, other aspects of its function remain unclear. Here we report hitherto unknown mechanism by which cyclin G1 increases radiation sensitivity by regulating the level of cyclin B1. Overexpression of cyclin G1 was observable in lung carcinoma tissues. Irradiation of human lung cells with cyclin G1 overexpression resulted in increased cell death and gamma-H2AX foci suggesting that cyclin G1 rendered the cells more susceptible to DNA damage. Enhanced radiosensitivity by cyclin G1 was correlated with increased cyclin B1, CDC2/cyclin B1 complex, and MPM2. Cell cycle synchronization clearly showed coexpression of cyclin G1 and cyclin B1 in G2/M phase. Depletion of cyclin G1 by interference RNA revealed that cyclin G1 regulated transcription of cyclin B1 in a p53-independent manner, and confirmed that the increased mitotic cells and cell death by cyclin G1 were dependent upon cyclin B1. Therefore, our data suggest that cyclin G1 enhanced radiation sensitivity by overriding radiation-induced G2 arrest through transcriptional upregulation of cyclin B1.


Assuntos
Ciclina B/biossíntese , Ciclinas/fisiologia , Dano ao DNA , Fase G2/efeitos da radiação , Ativação Transcricional , Adenocarcinoma Papilar/metabolismo , Adenocarcinoma Papilar/patologia , Proteína Quinase CDC2/metabolismo , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Morte Celular/efeitos da radiação , Linhagem Celular , Linhagem Celular Tumoral , Ciclina B1 , Ciclina G , Ciclina G1 , Ciclinas/biossíntese , Fibroblastos/citologia , Fibroblastos/efeitos da radiação , Histonas/metabolismo , Humanos , Pulmão/citologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Análise Serial de Tecidos , Proteína Supressora de Tumor p53/metabolismo
2.
Mol Cell Biol ; 19(2): 1313-24, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9891065

RESUMO

Protein kinase C (PKC) is a multigene family of enzymes consisting of at least 11 isoforms. It has been implicated in the induction of c-fos and other immediate response genes by various mitogens. The serum response element (SRE) in the c-fos promoter is necessary and sufficient for induction of transcription of c-fos by serum, growth factors, and the phorbol ester 12-O-tetradecanoylphorbol-13-acetate (TPA). It forms a complex with the ternary complex factor (TCF) and with a dimer of the serum response factor (SRF). TCF is the target of several signal transduction pathways and SRF is the target of the rhoA pathway. In this study we generated dominant-negative and constitutively active mutants of PKC-alpha, PKC-delta, PKC-epsilon, and PKC-zeta to determine the roles of individual isoforms of PKC in activation of the SRE. Transient-transfection assays with NIH 3T3 cells, using an SRE-driven luciferase reporter plasmid, indicated that PKC-alpha and PKC-epsilon, but not PKC-delta or PKC-zeta, mediate SRE activation. TPA-induced activation of the SRE was partially inhibited by dominant negative c-Raf, ERK1, or ERK2, and constitutively active mutants of PKC-alpha and PKC-epsilon activated the transactivation domain of Elk-1. TPA-induced activation of the SRE was also partially inhibited by a dominant-negative MEKK1. Furthermore, TPA treatment of serum-starved NIH 3T3 cells led to phosphorylation of SEK1, and constitutively active mutants of PKC-alpha and PKC-epsilon activated the transactivation domain of c-Jun, a major substrate of JNK. Constitutively active mutants of PKC-alpha and PKC-epsilon could also induce a mutant c-fos promoter which lacks the TCF binding site, and they also induce transactivation activity of the SRF. Furthermore, rhoA-mediated SRE activation was blocked by dominant negative mutants of PKC-alpha or PKC-epsilon. Taken together, these findings indicate that PKC-alpha and PKC-epsilon can enhance the activities of at least three signaling pathways that converge on the SRE: c-Raf-MEK1-ERK-TCF, MEKK1-SEK1-JNK-TCF, and rhoA-SRF. Thus, specific isoforms of PKC may play a role in integrating networks of signal transduction pathways that control gene expression.


Assuntos
Genes fos , Isoenzimas/genética , Isoenzimas/metabolismo , MAP Quinase Quinase 4 , MAP Quinase Quinase Quinase 1 , Quinases de Proteína Quinase Ativadas por Mitógeno , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Células 3T3 , Animais , Sequência de Bases , Sítios de Ligação/genética , Células COS , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , MAP Quinase Quinase 1 , Camundongos , Mutação , Plasmídeos/genética , Regiões Promotoras Genéticas , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-raf/genética , Proteínas Proto-Oncogênicas c-raf/metabolismo , Transdução de Sinais , Acetato de Tetradecanoilforbol/farmacologia
3.
Mol Cell Biol ; 20(15): 5392-403, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10891480

RESUMO

Although mitogenic and differentiating factors often activate a number of common signaling pathways, the mechanisms leading to their distinct cellular outcomes have not been elucidated. In a previous report, we demonstrated that mitogen-activated protein (MAP) kinase (ERK) activation by the neurogenic agents fibroblast growth factor (FGF) and nerve growth factor is dependent on protein kinase Cdelta (PKCdelta), whereas MAP kinase activation in response to the mitogen epidermal growth factor (EGF) is independent of PKCdelta in rat hippocampal (H19-7) and pheochromocytoma (PC12) cells. We now show that EGF activates MAP kinase through a PKCzeta-dependent pathway involving phosphatidylinositol 3-kinase and PDK1 in H19-7 cells. PKCzeta, like PKCdelta, acts upstream of MEK, and PKCzeta can potentiate Raf-1 activation by EGF. Inhibition of PKCzeta also blocks EGF-induced DNA synthesis as monitored by bromodeoxyuridine incorporation in H19-7 cells. Finally, in embryonic rat brain hippocampal cell cultures, inhibitors of PKCzeta or PKCdelta suppress MAP kinase activation by EGF or FGF, respectively, indicating that these factors activate distinct signaling pathways in primary as well as immortalized neural cells. Taken together, these results implicate different PKC isoforms as determinants of growth factor signaling specificity within the same cell. Furthermore, these data provide a mechanism whereby different growth factors can differentially activate a common signaling intermediate and thereby generate biological diversity.


Assuntos
Fator de Crescimento Epidérmico/metabolismo , Isoenzimas/metabolismo , Neurônios/metabolismo , Proteína Quinase C/metabolismo , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Animais , Células Cultivadas , Fator de Crescimento Epidérmico/farmacologia , Hipocampo/citologia , Hipocampo/embriologia , Isoenzimas/genética , MAP Quinase Quinase 1 , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Oligonucleotídeos Antissenso/farmacologia , Células PC12/metabolismo , Fosfatidilinositol 3-Quinases/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Proteína Quinase C/genética , Proteína Quinase C-delta , Proteína Quinase C-theta , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-raf/metabolismo , Ratos , Ratos Sprague-Dawley
4.
Cancer Res ; 61(2): 725-31, 2001 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-11212275

RESUMO

Epidemiological studies link increased garlic (Allium sativum) consumption with a reduced incidence of colon cancer in various human populations. Experimental carcinogenesis studies in animal models and in cell culture systems indicate that several allium-derived compounds exhibit inhibitory effects and that the underlying mechanisms may involve both the initiation and promotion phases of carcinogenesis. To provide a better understanding of the effects of allium derivatives on the prevention of colon cancer, we examined two water-soluble derivatives of garlic, S-allylcysteine (SAC) and S-allylmercaptocysteine (SAMC), for their effects on proliferation and cell cycle progression in two human colon cancer cell lines, SW-480 and HT-29. For comparison, we included the compound sulindac sulfide (SS), because sulindac compounds are well-established colon cancer chemopreventive agents. We found that SAMC, but not SAC, inhibited the growth of both cell lines at doses similar to that of SS. SAMC also induced apoptosis, and this was associated with an increase in caspase3-like activity. These affects of SAMC were accompanied by induction of jun kinase activity and a marked increase in endogenous levels of reduced glutathione. Although SS caused inhibition of cell cycle progression from G1 to S, SAMC inhibited progression at G2-M, and a fraction of the SW-480 and HT-29 cells were specifically arrested in mitosis. Coadministration of SS with SAMC enhanced the growth inhibitory and apoptotic effects of SS. These findings suggest that SAMC may be useful in colon cancer prevention when used alone or in combination with SS or other chemopreventive agents.


Assuntos
Antineoplásicos/farmacologia , Divisão Celular/efeitos dos fármacos , Cisteína/análogos & derivados , Cisteína/farmacologia , Sulindaco/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Caspase 3 , Caspases/efeitos dos fármacos , Caspases/metabolismo , Ciclo Celular/efeitos dos fármacos , Cisteína/química , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Fase G2/efeitos dos fármacos , Alho/química , Glutationa/efeitos dos fármacos , Glutationa/metabolismo , Células HT29 , Humanos , Hibridização in Situ Fluorescente , Isoenzimas/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno , Cinética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mitose/efeitos dos fármacos , Plantas Medicinais , Proteínas Proto-Oncogênicas c-bcl-2/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Sulindaco/análogos & derivados , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo
5.
Cancer Res ; 58(15): 3448-54, 1998 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-9699679

RESUMO

We previously reported increased expression of p27Kip1 in a series of human breast cancer cell lines, as compared to cell lines established from normal mammary epithelial cells. These data were surprising because this protein exerts a growth-inhibitory function in normal cells, and p27Kip1 has been proposed as a candidate tumor suppressor gene. A possible explanation for the paradoxical increase in p27Kip1 in the breast cancer cell lines is that they had become refractory to the inhibitory effects of this protein. To address this question, here, we transfected the MCF7 breast cancer cell line and the MCF10F nontumorigenic mammary epithelial cell line with a vector containing the p27Kip1 cDNA to obtain derivatives that express increased levels of p27Kip1. The increased expression of p27Kip1 in both of these cell lines was associated with lengthening of the G1 phase, an increase in the doubling time, a decreased saturation density, and a decreased plating efficiency. In the MCF7 cells, anchorage-independent growth and in vivo tumorigenicity were also suppressed. These effects were associated with decreased cyclin E-associated in vitro kinase activity in both cell lines. The increased expression of p27Kip1 was associated with a decreased level of expression of cyclin D1 in the MCF10F cells but an increased level of the cyclin D1 protein in the MCF7 cell line. Both derivatives expressed slightly increased levels of the cyclin E protein. Thus, breast cancer cells are still responsive to p27Kip1-mediated inhibition of cell growth despite the high basal level of this protein. These results suggest that therapeutic strategies that further increase the level of expression of p27Kip1 or mimic its activity might be useful in cancer therapy.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Mama/citologia , Mama/metabolismo , Proteínas de Ciclo Celular , Transformação Celular Neoplásica , Proteínas Associadas aos Microtúbulos/biossíntese , Proteínas Associadas aos Microtúbulos/fisiologia , Proteínas Supressoras de Tumor , Adesão Celular/fisiologia , Ciclo Celular/fisiologia , Divisão Celular/fisiologia , Linhagem Celular , Inibidor de Quinase Dependente de Ciclina p27 , DNA Complementar/genética , DNA Complementar/metabolismo , Células Epiteliais/patologia , Humanos , Proteínas Associadas aos Microtúbulos/genética , Transfecção , Células Tumorais Cultivadas
6.
Oncogene ; 18(1): 103-15, 1999 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-9926925

RESUMO

Recent studies have shown that decreased expression of p27Kip1 is associated with high grade tumors and an unfavorable prognosis in several types of human cancer. To clarify the role of p27Kip1 in colon cancer, we have overexpressed this protein in the HT29 colon cancer cell line. The derivatives displayed an increase in the p27Kip1 protein in cyclin E/CDK2 immunoprecipitates and a decrease in cyclin E-associated kinase activity when compared to vector control clones, providing evidence that the overexpressed protein was functional. Clones with a high level of p27Kip1 displayed partial growth inhibition in monolayer culture and a decrease in plating efficiency, even though they expressed increased levels of the cyclin D1 protein. Using alkaline phosphatase expression as a marker, we found that the p27Kip1 overexpressor clones displayed a 2-3-fold increase in sensitivity to induction of differentiation by 2 mM sodium butyrate. In contrast to these results, derivatives of HT29 cells that stably overexpressed p21Cip1/Waf1 displayed decreased sensitivity to the induction of differentiation. These findings may explain why decreased levels of p27Kip1 in certain human cancers is associated with high grade (poorly differentiated) tumors, and suggest that strategies that increase the level of p27Kip1 may be useful in cancer therapy.


Assuntos
Neoplasias do Colo/patologia , Ciclinas/fisiologia , Proteínas Associadas aos Microtúbulos/fisiologia , Proteínas Supressoras de Tumor , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Neoplasias do Colo/metabolismo , Inibidor de Quinase Dependente de Ciclina p21 , Inibidor de Quinase Dependente de Ciclina p27 , Ciclinas/biossíntese , Ciclinas/genética , Expressão Gênica , Células HT29 , Humanos , Proteínas Associadas aos Microtúbulos/biossíntese , Proteínas Associadas aos Microtúbulos/genética , Fatores de Tempo
7.
Clin Cancer Res ; 6(10): 4136-41, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11051267

RESUMO

Sulindac sulfone (Exisulind) induces apoptosis and exhibits cancer chemopreventive activity, but in contrast to sulindac, it does not inhibit cyclooxygenases 1 or 2. We found that sulindac sulfone and two potent derivatives, CP248 and CP461, inhibited the cyclic GMP (cGMP) phosphodiesterases (PDE) 2 and 5 in human colon cells, and these compounds caused rapid and sustained activation of the c-Jun NH2-terminal kinase 1 (JNK1). Rapid activation of stress-activated protein/ERK kinase 1 (SEK1) and mitogen-activated protein kinase kinase kinase (MEKK1), which are upstream of JNK1, was also observed. Other compounds that increase cellular levels of cGMP also activated JNK1, and an inhibitor of protein kinase G (PKG), Rp-8-pCPT-cGMPS, inhibited JNK1 activation by the sulindac sulfone derivatives. Expression of a dominant-negative JNK1 protein inhibited CP248-induced cleavage of poly(ADP-ribose) polymerase, a marker of apoptosis. Thus, it appears that sulindac sulfone and related compounds induce apoptosis, at least in part, through activation of PKG, which then activates the MEKK1-SEK1-JNK1 cascade. These studies also indicate a role for cGMP and PKG in the JNK pathway.


Assuntos
Apoptose , GMP Cíclico/metabolismo , MAP Quinase Quinase 4 , MAP Quinase Quinase Quinase 1 , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Sulindaco/farmacologia , Neoplasias do Colo/tratamento farmacológico , Relação Dose-Resposta a Droga , Ativação Enzimática , Genes Dominantes , Humanos , Proteína Quinase 8 Ativada por Mitógeno , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Modelos Biológicos , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Sulindaco/análogos & derivados , Fatores de Tempo , Células Tumorais Cultivadas
8.
Clin Cancer Res ; 5(7): 1805-15, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10430085

RESUMO

The retinoblastoma (Rb) gene is inactivated in a variety of human cancers, but in colorectal carcinomas there is frequently increased expression of this gene. This is paradoxical in view of the known role of Rb as a tumor suppressor gene. In the present study, we compared the levels of expression of the Rb protein (pRb) in normal human colorectal mucosa, adenomatous polyps, and carcinomas by immunohistochemistry. In vitro studies were also done to examine the phenotypic effects of an antisense oligodeoxynucleotide (AS-Rb) targeted to Rb mRNA in the HCT116 colon carcinoma cell line that expresses a relatively high level of pRb. The incidence of pRb-positive cells was increased during multistage colorectal carcinogenesis. In vitro treatment of HCT116 cells with AS-Rb decreased the level of pRb by about 70% and also decreased the levels of the cyclin D1 protein and cyclin D1-associated kinase activity. AS-Rb inhibited growth of HCT116 cells and induced apoptosis. Reporter assays indicated about a 17-fold increase in E2F activity. These findings suggest that the increased expression of pRb in colorectal carcinoma cells may provide a homeostatic mechanism that protects them from growth inhibition and apoptosis, perhaps by counterbalancing potentially toxic effects of excessive E2F activity.


Assuntos
Apoptose , Proteínas de Transporte , Neoplasias Colorretais/metabolismo , Proteínas de Ligação a DNA , Proteína do Retinoblastoma/metabolismo , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Divisão Celular/genética , Tamanho Celular , Células Cultivadas , Neoplasias Colorretais/patologia , Fatores de Transcrição E2F , Humanos , Imuno-Histoquímica , Oligonucleotídeos Antissenso/genética , Proteína do Retinoblastoma/genética , Proteína 1 de Ligação ao Retinoblastoma , Fator de Transcrição DP1 , Fatores de Transcrição/metabolismo , Células Tumorais Cultivadas
9.
Anticancer Res ; 19(6B): 4935-45, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10697493

RESUMO

The process of terminal differentiation is associated with exit from the cell cycle and loss of the proliferative potential of cells. The cyclin-dependent kinase inhibitors (CDIs) play critical roles in check-point functions during the cell cycle and as inhibitors of cell proliferation. Loss of their activities can impair development and differentiation and contribute to the uncontrolled proliferation characteristic of cancer cells. When the promyelocytic leukemia cell line HL60 is induced to differentiate in vitro, by a variety of agents, cellular levels of the CD1 proteins p21Cip1 and p27Kip1 are increased. To further address the roles of these two proteins in differentiation, we have overexpressed either a human p21Cip1 or p27Kip1 construct in HL60 cells. The overexpression of p21Cip1 accelerated both the monocytic and granulocytic differentiation of HL60 cells triggered by TPA or DMSO, respectively. The accelerated and more dramatic induction of differentiation seen in the p21Cip1 overexpressors was associated with a more rapid reduction of CDK2 kinase-associated activity, increased levels and more rapid dephosphorylation of the Rb protein, and increased levels of the cyclin D3 protein. Stable overexpression of p27Kip1 also enhanced TPA-induced differentiation of HL60 cells. These studies provide direct evidence that the increased expression of p21Cip1 and p27Kip1 play a causal role in the process of terminal differentiation of HL60 cells. Therefore, agents that enhance the expression of one or both of these proteins might be useful in therapy by enhancing the terminal differentiation of leukemia cells.


Assuntos
Quinases relacionadas a CDC2 e CDC28 , Proteínas de Ciclo Celular , Diferenciação Celular , Ciclinas/genética , Leucemia Promielocítica Aguda/genética , Proteínas Associadas aos Microtúbulos/genética , Proteínas Supressoras de Tumor , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula , Ciclina D3 , Quinase 2 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p21 , Inibidor de Quinase Dependente de Ciclina p27 , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/metabolismo , Dimetil Sulfóxido/farmacologia , Granulócitos/citologia , Granulócitos/efeitos dos fármacos , Células HL-60 , Humanos , Leucemia Promielocítica Aguda/enzimologia , Leucemia Promielocítica Aguda/patologia , Monócitos/citologia , Monócitos/efeitos dos fármacos , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Proteína do Retinoblastoma/metabolismo , Acetato de Tetradecanoilforbol/farmacologia
10.
Anticancer Res ; 21(3B): 1665-71, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11497245

RESUMO

Propolis has numerous biologic activities including antibiotic, antifungal, antiviral and anti-inflammatory properties. Several components isolated from propolis have been shown to have anticancer activity. This study demonstrates that the compound PM-3 (3-[2-dimethyl-8-(3-methyl-2-butenyl)benzopyran]-6-propenoic acid) isolated from Brazilian propolis markedly inhibits the growth of MCF-7 human breast cancer cells. This effect was associated with inhibition of cell cycle progression and induction of apoptosis. Treatment of MCF-7 cells with PM-3 arrested cells in the G1 phase and resulted in a decrease in the protein levels of cyclin D1 and cyclin E. PM-3 also inhibited the expression of cyclin D1 at the transcriptional level when examined in cyclin D1 promoter luciferase assays. Induction of apoptosis by PM-3 occurred within 48 hours after treatment of MCF-7 cells. The MCF-7 treated cells also displayed a decrease in the level of the estrogen receptor (ER) protein and inhibition of estrogen response element (ERE) promoter activity. Therefore, PM-3 merits further investigation with respect to breast cancer chemoprevention or therapy.


Assuntos
Antineoplásicos/farmacologia , Benzopiranos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Metacrilatos/farmacologia , Extratos Vegetais/farmacologia , Própole/metabolismo , Anexina A5/metabolismo , Apoptose , Benzopiranos/química , Northern Blotting , Western Blotting , Ciclo Celular/efeitos dos fármacos , Divisão Celular , Corantes/farmacologia , Ciclina D1/biossíntese , Ciclina E/biossíntese , Relação Dose-Resposta a Droga , Regulação para Baixo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Genes Reporter , Humanos , Metacrilatos/química , Modelos Químicos , Regiões Promotoras Genéticas , Receptores de Estrogênio/biossíntese , Receptores de Estrogênio/efeitos dos fármacos , Elementos de Resposta , Sais de Tetrazólio/farmacologia , Tiazóis/farmacologia , Fatores de Tempo , Transcrição Gênica , Transfecção , Células Tumorais Cultivadas
11.
Cell Prolif ; 45(5): 438-44, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22925503

RESUMO

OBJECTIVES: This study compared rate of cell proliferation, viability, cell size, expression patterns of genes related to pluripotency and epigenetic modification between canine foetal fibroblasts (cFF) and canine adipose tissue-derived mesenchymal stem cells (cAd-MSC). MATERIALS AND METHODS: Proliferation pattern, cell viability as well as cell size at each passage of cFF and cAd-MSC were measured when cultures reached confluence. In addition, real-time PCR was performed to investigate expression of Dnmt1, HDAC1, OCT4, SOX2, BAX, BCL2 genes with reference to ß-actin gene expression as an endogenous control in both cell lines. RESULTS: cFF and cAd-MSC differed in number of generations, but not in doubling times, at all passages. Mean cell size of cAd-MSC was significantly smaller than that of cFF. Cell viability was significantly lower in cFFs and apoptotic level was significantly lower in cAd-MSC compared to passage-matched cFF. In the expression of genes related to pluripotency and epigenetic modification, level of HDAC1 in cAd-MSC was significantly higher than in cFF, but expression of Dnmt1 did not differ between the two groups. OCT4 and SOX2 were significantly more highly expressed in cAd-MSC compared to cFF. CONCLUSIONS: cAd-MSC have higher stem-cell potential than cFF in terms of proliferation patterns, epigenetic modification and pluripotency, thus cAd-MSC could be more appropriate than cFF as donors of nuclei in somatic cell nuclear transfer for transgenesis.


Assuntos
Tecido Adiposo/citologia , Proliferação de Células , Epigênese Genética/fisiologia , Fibroblastos/citologia , Células-Tronco Mesenquimais/citologia , Animais , Técnicas de Cultura de Células/métodos , Sobrevivência Celular/fisiologia , Cães , Feminino , Feto/citologia , Fibroblastos/fisiologia , Células-Tronco Mesenquimais/fisiologia , Gravidez
12.
J Biol Chem ; 276(45): 41879-88, 2001 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-11551972

RESUMO

We have studied the role of phosphorylation in the activation of metal-regulatory transcription factor-1 (MTF-1) and metallothionein (MT) gene expression. We showed that MTF-1 is phosphorylated in vivo and that zinc stimulates MTF-1 phosphorylation 2-4-fold. Several kinase inhibitors were used to examine the possible involvement of kinase cascades in the activation of MTF-1. Metal-induced MT gene expression was abrogated by protein kinase C (PKC), c-Jun N-terminal kinase (JNK), phosphoinositide 3-kinase, and tyrosine-specific protein kinases inhibitors, as assayed by Northern analysis and by cotransfection experiments using a metal regulatory element-luciferase reporter plasmid. The extracellular signal-activated protein kinase and the p38 kinase cascades did not appear to be essential for the activation of MT gene transcription by metals. By using dominant-negative mutants of PKC, JNK, mitogen-activated kinase kinase 4 (MKK4), and MKK7, we provide further evidence supporting a role for PKC and JNK in the activation of MTF-1 in response to metals. Notably, increased MTF-1 DNA binding in response to zinc and MTF-1 nuclear localization was not inhibited in cells preincubated with the different kinase inhibitors despite strong inhibition of MTF-1-mediated gene expression. This suggests that phosphorylation is essential for MTF-1 transactivation function. We hypothesize that metal-induced phosphorylation of MTF-1 is one of the primary events leading to increased MTF-1 activity. Thus, metal ions such as cadmium could activate MTF-1 and induce MT gene expression by stimulating one or several kinases in the MTF-1 signal transduction pathway.


Assuntos
Metais/farmacologia , Transdução de Sinais , Fatores de Transcrição/metabolismo , Animais , Cádmio/farmacologia , DNA/metabolismo , Proteínas de Ligação a DNA , Proteínas Quinases JNK Ativadas por Mitógeno , Camundongos , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Fosforilação , Proteína Quinase C/fisiologia , Zinco/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno , Fator MTF-1 de Transcrição
13.
J Biol Chem ; 276(19): 16406-10, 2001 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-11278263

RESUMO

We recently obtained evidence that treatment of human colon cancer cells with exisulind (sulindac sulfone) and related compounds induces apoptosis by activation of protein kinase G (PKG) and c-Jun kinase (JNK1). The present study further explores this mechanism. We demonstrate that in NIH3T3 cells a constitutively active mutant of PKG causes a dose-dependent activation of JNK1 and thereby transactivates c-Jun and stimulates transcription from the AP-1 enhancer element. The activation of JNK1 and the transactivation of c-Jun by this mutant of PKG were inhibited by a dominant negative MEKK1. In vitro assays showed that a purified PKG directly phosphorylated the N-terminal domain of MEKK1. PKG also directly phosphorylated a full-length MEKK1, and this was associated with enhanced MEKK1 phosphorylation. Thus, it appears that PKG activates JNK1 through a novel PKG-MEKK1-SEK1-JNK1 pathway, by directly phosphorylating and activating MEKK1.


Assuntos
Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Genes jun , MAP Quinase Quinase 4 , MAP Quinase Quinase Quinase 1 , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Células 3T3 , Animais , Ativação Enzimática , Regulação da Expressão Gênica , Camundongos , Proteína Quinase 8 Ativada por Mitógeno , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-jun/metabolismo , Ativação Transcricional , Transfecção
14.
J Biol Chem ; 276(27): 24506-10, 2001 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-11333276

RESUMO

Expression of the COOH-terminal residues 179-330 of the LSP1 protein in the LSP1(+) B-cell line W10 increases anti-IgM- or ionomycin-induced apoptosis, suggesting that expression of this LSP1 truncate (B-LSP1) interferes with a Ca(2+)-dependent step in anti-IgM signaling. Here we show that inhibition of Ca(2+)-dependent conventional protein kinase C (cPKC) isoforms with Gö6976 increases anti-IgM-induced apoptosis of W10 cells and that expression of B-LSP1 inhibits translocation of PKCbetaI but not of PKCbetaII or PKCalpha to the plasma membrane. The increased anti-IgM-induced apoptosis is partially reversed by overexpression of PKCbetaI. This shows that the B-LSP1-mediated inhibition of PKCbetaI leads to increased anti-IgM-induced apoptosis. Expression of constitutively active PKCbetaI protein in W10 cells activates the mitogen-activated protein kinase ERK2, whereas expression of B-LSP1 inhibits anti-IgM-induced activation of ERK2, suggesting that anti-IgM-activated PKCbetaI is involved in the activation of ERK2 and that inhibition of ERK2 activation contributes to the increased anti-IgM-induced apoptosis. Pull-down assays show that LSP1 interacts with PKCbetaI but not with PKCbetaII or PKCalpha in W10 cell lysates, while in vitro LSP1 and B-LSP1 bind directly to PKCbetaI. Thus, B-LSP1 is a unique reagent that binds PKCbetaI and inhibits anti-IgM-induced PKCbetaI translocation, leading to inhibition of ERK2 activation and increased apoptosis.


Assuntos
Anticorpos Anti-Idiotípicos/metabolismo , Apoptose , Proteínas de Ligação ao Cálcio/metabolismo , Inibidores Enzimáticos/metabolismo , Isoenzimas/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase C/metabolismo , Ativação Enzimática , Humanos , Proteínas dos Microfilamentos , Proteína Quinase C beta , Células Tumorais Cultivadas
15.
Proc Natl Acad Sci U S A ; 98(2): 609-14, 2001 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-11149941

RESUMO

Pre-T cell receptor (preTCR)-derived signals mediate the transition of thymocytes from the CD4(-) CD8(-) double-negative (DN) to CD4(+) CD8(+) double-positive stage of T lymphocyte development. This progression, termed beta-selection, is limited to thymocytes that have generated a functional TCR-beta chain able to associate with pTalpha to form the preTCR complex. Formation of the preTCR complex not only induces differentiation, survival, and proliferation of DN thymocytes; it also inhibits further TCR-beta gene rearrangement through an ill-defined process known as allelic exclusion. The signaling pathways controlling this critical developmental checkpoint have not been characterized. Here we demonstrate that formation of the preTCR complex leads to the activation of protein kinase C (PKC), and that activation of PKC is necessary for the differentiation and expansion of DN thymocytes. Importantly, we also show that allelic exclusion at the TCR-beta gene loci is enforced by PKC-mediated signals. These results define PKC as a central mediator of both differentiation and allelic exclusion during thymocyte development.


Assuntos
Alelos , Deleção Clonal/fisiologia , Hematopoese/fisiologia , Isoenzimas/fisiologia , Proteína Quinase C/fisiologia , Subpopulações de Linfócitos T/citologia , Timo/citologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Biolística , Diferenciação Celular , Proteínas de Ligação a DNA , Ativação Enzimática , Rearranjo Gênico da Cadeia beta dos Receptores de Antígenos dos Linfócitos T , Genes Dominantes , Genes Reporter , Isoenzimas/genética , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/fisiologia , Camundongos , Camundongos Knockout , Técnicas de Cultura de Órgãos , Fosfatidilinositol Diacilglicerol-Liase , Fosfoproteínas/fisiologia , Proteína Quinase C/genética , Proteína Quinase C-alfa , Proteínas Tirosina Quinases/fisiologia , Proteínas Proto-Oncogênicas c-raf/fisiologia , Proteínas Proto-Oncogênicas p21(ras)/fisiologia , Proteínas Recombinantes de Fusão/fisiologia , Transdução de Sinais , Transfecção , Fosfolipases Tipo C/fisiologia , Proteína-Tirosina Quinase ZAP-70
16.
J Biol Chem ; 276(1): 522-8, 2001 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-11016926

RESUMO

Multiple studies have shown that intracellular signal transduction by the protein kinase C (PKC) family participates in the initiation of megakaryocyte differentiation. In this study, multiple approaches addressed the functional contributions by specific PKC isozymes to megakaryocytic lineage commitment of two independent cell lines, K562 and human erythroleukemia (HEL). Pharmacologic profiles of induction and inhibition of megakaryocytic differentiation in both cell lines suggested a role for the calcium-independent novel PKCs, in particular PKC-epsilon. In transfection studies, the isolated variable domain of PKC-epsilon selectively blocked exogenous activation of the megakaryocyte-specific alpha IIb promoter. Constitutively active mutants of PKC-epsilon, but not of other PKC isozymes, cooperated with the transcription factor GATA-1 in the activation of the alpha IIb promoter. The functional cooperation between GATA-1 and PKC-epsilon displayed dependence on cellular milieu, as well as on the promoter context of GATA binding sites. In aggregate, the data suggest that PKC-epsilon specifically participates in megakaryocytic lineage commitment through functional cooperation with GATA-1 in the activation of megakaryocytic promoters.


Assuntos
Diferenciação Celular , Isoenzimas/metabolismo , Megacariócitos/citologia , Megacariócitos/enzimologia , Proteína Quinase C/metabolismo , Carbazóis/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Proteínas de Ligação a DNA/metabolismo , Diterpenos/farmacologia , Fatores de Ligação de DNA Eritroide Específicos , Imunofluorescência , Fator de Transcrição GATA1 , Regulação da Expressão Gênica/efeitos dos fármacos , Genes Reporter , Humanos , Indóis/farmacologia , Isoenzimas/antagonistas & inibidores , Isoenzimas/química , Isoenzimas/genética , Células K562 , Maleimidas/farmacologia , Megacariócitos/efeitos dos fármacos , Megacariócitos/metabolismo , Mutação , Regiões Promotoras Genéticas/genética , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/química , Proteína Quinase C/genética , Proteína Quinase C-épsilon , Transporte Proteico/efeitos dos fármacos , Elementos de Resposta/genética , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Transfecção , Células Tumorais Cultivadas
17.
Cell Growth Differ ; 7(6): 699-710, 1996 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8780883

RESUMO

Cyclin D1 is frequently amplified and/or overexpressed in human breast cancer and several other types of cancer. To examine the role of cyclin D1 in normal mammary epithelial cells, in the present study we have overexpressed human cyclin D1 in the mouse mammary epithelial cell line HC11, using retrovirus-mediated transduction. We found that the cyclin D1 overexpresser clones displayed a decrease in saturation density, a decrease in anchorage-independent growth, an increased fraction of cells in the G(zero)-G1 phase, and increased expression of beta-casein, when compared to the control cells. The latter finding suggested that they were more differentiated. Furthermore, the cyclin D1 overexpressers displayed a marked increase in susceptibility to induction of apoptosis by serum withdrawal or by treatment with hydroxyurea or the protein kinase C inhibitors CGP 41251 and Ro31-8220. Thus, in some mammary epithelial cells, increased expression of cyclin D1 can inhibit growth, induce differentiation, and enhance apoptosis. These effects might be due, at least in part, to the fact that these derivatives displayed increased expression of the p27kip1 inhibitory protein.


Assuntos
Apoptose/fisiologia , Quinases relacionadas a CDC2 e CDC28 , Proteínas de Ciclo Celular , Ciclinas/biossíntese , Genes Supressores de Tumor , Inibidores do Crescimento/biossíntese , Glândulas Mamárias Animais/metabolismo , Proteínas Associadas aos Microtúbulos/biossíntese , Proteínas Oncogênicas/biossíntese , Proteínas Supressoras de Tumor , Animais , Ciclo Celular/genética , Linhagem Celular , Ciclina D1 , Quinase 2 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p27 , Quinases Ciclina-Dependentes/metabolismo , Células Epiteliais , Epitélio/metabolismo , Feminino , Regulação da Expressão Gênica/fisiologia , Técnicas de Transferência de Genes , Vetores Genéticos , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Serina-Treonina Quinases/metabolismo , Retroviridae/genética
18.
Gastroenterology ; 118(3): 507-14, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10702201

RESUMO

BACKGROUND & AIMS: We have previously reported that nonsteroidal anti-inflammatory drugs (NSAIDs) could induce apoptosis of gastric epithelial cells both in vivo and in vitro. This study investigated the role of protein kinase C (PKC) isoforms in the regulation of NSAID-induced apoptosis. METHODS: Protein levels of 12 PKC isoforms in AGS cells, in the presence or absence of indomethacin, were determined by Western blot. The effect of PKC-beta1 overexpression by transfection with its complementary DNA (cDNA) on indomethacin-induced apoptosis and apoptosis-related genes, including p53, p21(waf1/cip1), and c-myc, was further investigated. RESULTS: Treatment with indomethacin decreased the abundance of PKC-beta1 and increased that of PKC-beta2, eta, and epsilon, but did not alter the expression of PKC alpha, gamma, zeta, delta, iota, and micro. Overexpression of PKC-beta1 attenuated the apoptotic response of AGS cells to indomethacin, associated with overexpression of p21(waf1/cip1) in both messenger RNA and protein levels. Inhibition of PKC-beta1-mediated overexpression of p21(waf1/cip1) by its antisense cDNA partially reduced the antiapoptotic effect of PKC-beta1. CONCLUSIONS: Indomethacin-induced apoptosis in gastric cancer cells is partly mediated by differential regulation of PKC isoform expression. Enhanced expression of exogenous PKC-beta1 protects against indomethacin-induced apoptosis through up-regulation of p21(waf1/cip1).


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Apoptose/efeitos dos fármacos , Mucosa Gástrica/enzimologia , Mucosa Gástrica/fisiopatologia , Indometacina/farmacologia , Isoenzimas/metabolismo , Proteína Quinase C/metabolismo , Elementos Antissenso (Genética)/farmacologia , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/genética , Ciclinas/metabolismo , DNA Complementar/farmacologia , Mucosa Gástrica/patologia , Proteína Quinase C beta , Proteínas Proto-Oncogênicas c-myc/metabolismo , Transfecção , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA