Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Taiwan J Obstet Gynecol ; 62(6): 874-883, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-38008508

RESUMO

OBJECTIVE: The data on the association between phthalates and breast cancer risk remains inconsistent. This study aimed to explore the possible mechanism of low-dose exposures of phthalates, including Butyl benzyl phthalate (BBP), di(n-butyl) phthalate (DBP), and di(20ethylhexyl) phthalate (DEHP), on breast tumorigenesis. METHODS AND METHODS: MCF-10A normal breast cells were treated with phthalates (10 and 100 nM) and 17ß-estradiol (E2, 10 nM), which were co-cultured with fibroblasts from normal mammary tissue. Cell viability, cycle, and apoptosis were detected by MTT assay, flow cytometry, and TUNEL assay respectively. The expression levels of related proteins were determined by Western blot. RESULTS: Like E2, both 10 nM and 100 nM phthalates exerted significantly higher cell viability, lower apoptosis, and increased cell numbers in the S and G2/M phases with up-regulation of cyclin D/CDK4, cyclin E/CDK2, cyclin A/CDK2, cyclin A/CDK1, and cyclin B/CDK1, compared with the control group. Significant increase in PDK1, P13K, p-AKT, p-mTOR, and BCL-2 expression and a decrease in Bax protein, cytochrome C, caspase 8, and caspase 3 levels were noted in cells treated with 10 nM and 100 nM phthalates and E2, compared with the control group and MCF-10A cells co-cultured with fibroblasts. The effects of the three phthalates were noted to be dose-dependent. CONCLUSIONS: The results indicate that phthalates at a level below its no-observed-adverse-effect concentration, as defined by the current standards, still induce cell cycle progression and proliferation as well as inhibit apoptosis of normal breast cells. Thus, the possibility of breast tumorigenesis through chronic phthalate exposure should be considered.


Assuntos
Ácidos Ftálicos , Humanos , Nível de Efeito Adverso não Observado , Proliferação de Células , Ácidos Ftálicos/toxicidade , Divisão Celular , Dibutilftalato/farmacologia , Ciclina A/farmacologia , Carcinogênese
2.
Recent Pat Anticancer Drug Discov ; 18(2): 187-199, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35319392

RESUMO

BACKGROUND: Hepatocellular carcinoma (HCC) is one of the most common cancers, associated with a high rate of mortality. A disturbance between cell proliferation and cell death is one of the cancer hallmarks including HCC. Cell proliferation is mainly controlled by the cell cycle. The arrest of the cell cycle is one of the important targets of anticancer agents. OBJECTIVES: The present study tries to clarify the exact role of some natural products such as daidzein (DAZ) and alcoholic chicory leaf extract (CE), as possible regulators of cell cycle and apoptosis. METHODS: HCC in rats was induced using diethylnitrosamine (DENA). Ninety rats were allocated and divided equally into nine groups, treated with CE, DAZ, a combination of both, and sorafenib with non-treated control groups. RESULTS: Treatment with CE, DAZ, and their combination significantly downregulated hepatic tissue expression of cyclin D1/CDK4 axis as well as cyclin A/CDK2 axis. The suggested therapeutic protocol inhibited the proliferation and dampened Bcl-2 expression. Furthermore, the efficiency of combining CE and DAZ demonstrated a potency comparable to sorafenib in terms of cyclin D/CDK4 axis expression, as well as; this combination protocol was more potent in revealing a potentiated inhibitory effect on cyclin A and Ki-67 expression. CONCLUSION: Treatment with DAZ or CE alone, or in combination, could possess an inhibitory effect on hepatocarcinogenesis via cell cycle arrest, inhibition of proliferation through suppression of Ki-67 expression, and apoptosis induction, mediated by downregulation of Bcl-2.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Ratos , Animais , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/genética , Sorafenibe/farmacologia , Sorafenibe/uso terapêutico , Ciclina A/farmacologia , Antígeno Ki-67 , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Quinase 4 Dependente de Ciclina/genética , Quinase 4 Dependente de Ciclina/metabolismo , Ciclo Celular , Pontos de Checagem do Ciclo Celular , Proliferação de Células , Proteínas Proto-Oncogênicas c-bcl-2 , Expressão Gênica , Quinase 2 Dependente de Ciclina
3.
Nutr Cancer ; 74(5): 1734-1744, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34353198

RESUMO

Diabetes mellitus (DM) is associated with an increased risk and progression of cholangiocarcinoma (CCA). High glucose underlying the association between DM and CCA by modulating the intracellular signaling has been demonstrated. However, the effects of DM and hyperglycemia on cell cycle machineries and progression of CCA remain elucidated. CCA cells, KKU-213A and KKU-213B were cultured in normal (NG, 5.6 mM) or high glucose (HG, 25 mM) resembling euglycemia and hyperglycemia. Western blotting was used to determine expressions of cell cycle machineries in CCA cells. The expression of cyclin A in CCA tissues from patients with or without hyperglycemia was determined by immunohistochemistry. Pan-cyclin dependent kinases (CDKs) inhibitor and silencing of cyclin A expression were investigated as a possible modality targeting CCA treatment in patients with DM. High glucose induced expression of cell cycle machinery proteins in both CCA cells. Among these, cyclin A was consistently and significantly upregulated. Nuclear cyclin A was significantly increased in tumor tissues from CCA patients with hyperglycemia and was significantly associated with post-operative survival of shorter than 5 mo. Silencing cyclin A expression sensitized CCA cells to pan-CDKs inhibitor, suggesting the combined treatment as an alternative approach for treatment of CCA patients with DM.


Assuntos
Neoplasias dos Ductos Biliares , Colangiocarcinoma , Diabetes Mellitus , Hiperglicemia , Neoplasias dos Ductos Biliares/tratamento farmacológico , Neoplasias dos Ductos Biliares/metabolismo , Ductos Biliares Intra-Hepáticos/metabolismo , Ductos Biliares Intra-Hepáticos/patologia , Linhagem Celular Tumoral , Proliferação de Células , Colangiocarcinoma/tratamento farmacológico , Colangiocarcinoma/metabolismo , Ciclina A/metabolismo , Ciclina A/farmacologia , Ciclinas/metabolismo , Glucose/farmacologia , Humanos , Inibidores de Proteínas Quinases/farmacologia , Regulação para Cima
4.
J Appl Biomed ; 19(2): 113-124, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34754259

RESUMO

Oplopanax elatus (Nakai) Nakai has a long history of use as an ethnomedicine by the people living in eastern Asia. However, its bioactive constituents and cancer chemopreventive mechanisms are largely unknown. The aim of this study was to prepare O. elatus extracts, fractions, and single compounds and to investigate the herb's antiproliferative effects on colon cancer cells and the involved mechanisms of action. Two polyyne compounds were isolated from O. elatus, falcarindiol and oplopandiol. Based on our HPLC analysis, falcarindiol and oplopandiol are major constituents in the dichloromethane (CH2Cl2) fraction. For the HCT-116 cell line, the dichloromethane fraction showed significant effects. Furthermore, the IC50 for falcarindiol and oplopandiol was 1.7 µM and 15.5 µM, respectively. In the mechanistic study, after treatment with 5 µg/ml for 48 h, dichloromethane fraction induced cancer cell apoptosis by 36.5% (p < 0.01% vs. control of 3.9%). Under the same treatment condition, dichloromethane fraction caused cell cycle arrest at the G2/M phase by 32.6% (p < 0.01% vs. control of 23.4%), supported by upregulation of key cell cycle regulator cyclin A to 21.6% (p < 0.01% vs. control of 8.6%). Similar trends were observed by using cell line HT-29. Data from this study filled the gap between phytochemical components and the cancer chemoprevention of O. elatus. The dichloromethane fraction is a bioactive fraction, and falcarindiol is identified as an active constituent. The mechanisms involved in cancer chemoprevention by O. elatus were apoptosis induction and G2/M cell cycle arrest mediated by a key cell cycle regulator cyclin A.


Assuntos
Neoplasias do Colo , Oplopanax , Apoptose , Pontos de Checagem do Ciclo Celular , Quimioprevenção , Ciclina A/farmacologia , Ciclinas/farmacologia , Di-Inos , Álcoois Graxos , Humanos , Cloreto de Metileno/farmacologia , Oplopanax/química , Regulação para Cima
5.
J Biol Chem ; 283(39): 26577-90, 2008 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-18667424

RESUMO

Platelet-derived growth factor BB (PDGF-BB) induced cyclin A expression and CDK2 activity in vascular smooth muscle cells (VSMC). Inhibition of nuclear factors of activated T cell (NFAT) activation by cyclosporin A (CsA) and VIVIT suppressed PDGF-BB-induced cyclin A expression and CDK2 activity, resulting in blockade of VSMC in the G(1) phase. In addition, CsA- and VIVIT-mediated inhibition of NFATs and small interfering RNA-targeted down-regulation of cyclin A levels suppressed PDGF-BB-induced VSMC DNA synthesis. PDGF-BB also induced cyclin A mRNA levels in VSMC in an NFAT-dependent manner. Cloning and bioinformatic analysis of rat cyclin A promoter revealed the presence of NFAT-binding elements, and PDGF-BB induced the binding of NFATs to these regulatory sequences in a CsA- and VIVIT-sensitive manner. Chromatin immunoprecipitation analysis showed that NFATc1 binds to the cyclin A promoter in response to PDGF-BB in a VIVIT-sensitive manner. Furthermore, PDGF-BB induced cyclin A promoter-luciferase reporter gene activity in VSMC, and it was inhibited by both CsA and VIVIT. Balloon injury induced cyclin A expression and CDK2 activity in rat carotid arteries, and these responses were also blocked by VIVIT. In addition, VIVIT attenuated balloon injury-induced SMC proliferation, resulting in reduced restenosis. Down-regulation of NFATc1 by its small interfering RNA inhibited PDGF-BB-induced cyclin A expression and DNA synthesis both in rat and human VSMC. Together, these findings demonstrate that the cyclin A-CDK2 complex may be a potential effector of NFATs, specifically NFATc1, in mediating SMC multiplication leading to neointima formation. Therefore, NFATs may be used as target molecules for the development of therapeutic agents against vascular diseases such as restenosis.


Assuntos
Estenose das Carótidas/metabolismo , Fase G1 , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Fatores de Transcrição NFATC/metabolismo , Animais , Becaplermina , Estenose das Carótidas/patologia , Células Cultivadas , Ciclina A/farmacologia , Quinase 2 Dependente de Ciclina/metabolismo , Ciclosporina/farmacologia , Replicação do DNA/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Fase G1/efeitos dos fármacos , Humanos , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Fatores de Transcrição NFATC/antagonistas & inibidores , Oligopeptídeos/farmacologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , Ligação Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-sis , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/farmacologia , Ratos , Elementos de Resposta
6.
EMBO Rep ; 8(1): 91-8, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17159919

RESUMO

The anaphase-promoting complex (APC) early mitotic inhibitor 1 (Emi1) is required to induce S- and M-phase entries by stimulating the accumulation of cyclin A and cyclin B through APC(Cdh1/cdc20) inhibition. In this report, we show that Emi1 proteolysis can be induced by cyclin A/cdk (cdk for cyclin-dependent kinase). Paradoxically, Emi1 is stable during G2 phase, when cyclin A/cdk, Plx1 and SCF(betatrcp) (SCF for Skp1-Cul1-Fbox protein)--which play a role in its degradation--are active. Here, we identify Pin1 as a new regulator of Emi1 that induces Emi1 stabilization by preventing its association with SCF(betatrcp). We show that Pin1 binds to Emi1 and prevents its association with betatrcp in an isomerization-dependent pathway. We also show that Emi1-Pin1 binding is present in vivo in XL2 cells during G2 phase and that this association protects Emi1 from being degraded during this phase of the cell cycle. We propose that S- and M-phase entries are mediated by the accumulation of cyclin A and cyclin B through a Pin1-dependent stabilization of Emi1 during G2.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Peptidilprolil Isomerase/metabolismo , Proteínas de Xenopus/metabolismo , Proteínas Contendo Repetições de beta-Transducina/metabolismo , Animais , Proteínas de Ciclo Celular/análise , Proteínas de Ciclo Celular/genética , Extratos Celulares/química , Ciclina A/metabolismo , Ciclina A/farmacologia , Ciclina B/metabolismo , Ciclina B/farmacologia , Quinases Ciclina-Dependentes/farmacologia , Fase G2 , Humanos , Imunoprecipitação , Mitose , Peptidilprolil Isomerase de Interação com NIMA , Xenopus , Proteínas de Xenopus/análise , Proteínas de Xenopus/genética
7.
Bioorg Chem ; 33(4): 264-73, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16023486

RESUMO

Cdc25s, dual-specificity phosphatases that dephosphorylate and activate cyclin-dependent kinases, are important regulators of the eukaryotic cell cycle. Herein, we probe the protonation state of the phosphate on the protein substrate of Cdc25 by pH-dependent studies and thiosubstitution. We have extended the useable range of pH for this enzyme substrate pair by using high concentrations of glycerol under acidic conditions. Using the protein substrate, we find a slope of 2 for the acidic side of the bell-shaped pH-rate profile, as found with other protein tyrosine phosphatases. Using thiophosphorylated protein substrate, we find no change in the basic side of the pH-rate profile, despite a large reduction in activity as measured by kcat/Km (0.18%) or kcat (0. 11%). In contrast, the acidic side of the profile changes shows a slope of 1, consistent with the 1.5 pH unit shift associated with thiosubstitution. Thus, Cdc25, like other protein phosphatases, uses a dianionic phosphorylated substrate.


Assuntos
Fosfoproteínas/metabolismo , Compostos de Sulfidrila/metabolismo , Fosfatases cdc25/metabolismo , Ciclina A/farmacologia , Estabilidade Enzimática , Concentração de Íons de Hidrogênio , Cinética , Estrutura Molecular , Fosforilação , Ligação Proteica , Especificidade por Substrato , Compostos de Sulfidrila/química , Fosfatases cdc25/química
8.
J Cell Sci ; 117(Pt 25): 6019-30, 2004 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-15536124

RESUMO

In most eukaryotes, replication origins fire asynchronously throughout S-phase according to a precise timing programme. When replication fork progression is inhibited, an intra-S-phase checkpoint is activated that blocks further origin firing and stabilizes existing replication forks to prevent them undergoing irreversible collapse. We show that chromatin incubated in Xenopus egg extracts displays a replication-timing programme in which firing of new replication origins during S phase depends on the continued activity of S-phase-inducing cyclin-dependent kinases. We also show that low concentrations of the DNA-polymerase inhibitor aphidicolin, which only slightly slows replication-fork progression, strongly suppress further initiation events. This intra-S-phase checkpoint can be overcome by caffeine, an inhibitor of the ATM/ATR checkpoint kinases, or by neutralizing antibodies to ATR. However, depletion or inhibition of Chk1 did not abolish the checkpoint. We could detect no significant effect on fork stability when this intra-S-phase checkpoint was inhibited. Interestingly, although caffeine could prevent the checkpoint from being activated, it could not rescue replication if added after the timing programme would normally have been executed. This suggests that special mechanisms might be necessary to reverse the effects of the intra-S-phase checkpoint once it has acted on particular origins.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas de Xenopus/fisiologia , Animais , Afidicolina/metabolismo , Afidicolina/farmacologia , Proteínas Mutadas de Ataxia Telangiectasia , Cafeína/metabolismo , Cafeína/farmacologia , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/metabolismo , Quinase 1 do Ponto de Checagem , Cromatina/metabolismo , Ciclina A/farmacologia , Replicação do DNA , Eletroforese em Gel de Ágar , Inibidores Enzimáticos/farmacologia , Cinética , Masculino , Inibidores da Síntese de Ácido Nucleico , Antígeno Nuclear de Célula em Proliferação/metabolismo , Purinas/farmacologia , Roscovitina , Fase S , Espermatozoides/metabolismo , Fatores de Tempo , Xenopus , Proteínas de Xenopus/metabolismo
9.
Int J Oncol ; 24(2): 273-8, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14719102

RESUMO

We investigated the role of cyclin A in the cytotoxic effect of 5-fluorouracil (5-FU) on cancer cell lines. Experiments were performed using gastric cancer chemosensitive NUGC3, and chemoresistant NUGC3/5FU/L established by repeated exposure to 5-FU. 5-FU inhibited cell growth of NUGC3 in a dose-dependent manner. Low concentrations of 5-FU did not inhibit cell growth of NUGC3/5FU/L, while high concentrations slightly inhibited cell growth. Examination of the cell cycle pattern of NUGC3 cells showed accumulation at S-phase at 10 micro M and at G1-S-phase at 100 micro M of 5-FU. Cell cycle pattern of NUGC3/5FU/L cells did not change 5-FU concentrations. 5-FU increased cyclin A mRNA level in NUGC3 cells but not NUGC3/5FU/L cells. In the presence of 100 micro M 5-FU, cyclin A protein level increased 2.6-fold in NUGC3 and 1.47-fold in NUGC3/5FU/L. 5-FU dose-dependently increased the percentage of cyclin A-positive NUGC3 cells, but not NUGC3/5FU/L cells. The percentage of cyclin A-positive cells in other 5-FU sensitive esophageal, colon and gastric cancer cell lines (T.Tn, LOVO, DLD-1, MKN-7), increased in the presence of 1 and 10 micro M 5-FU, while cyclin A-positive cells in 5-FU resitant hepatocellular and colon carcinoma cell lines (HCC50 and C-1), did not increase with the same treatment. Our results indicate that the cytotoxic effects of 5-FU in human cancer cell lines correlate with cyclin A and it may be used as a predictive factor for chemotherapy response.


Assuntos
Ciclina A/biossíntese , Fluoruracila/farmacologia , Antimetabólitos Antineoplásicos/farmacologia , Carcinoma Hepatocelular/tratamento farmacológico , Linhagem Celular Tumoral , Colágeno/metabolismo , Neoplasias do Colo/tratamento farmacológico , Corantes/farmacologia , Ciclina A/metabolismo , Ciclina A/farmacologia , Relação Dose-Resposta a Droga , Citometria de Fluxo , Humanos , Immunoblotting , Neoplasias Hepáticas/tratamento farmacológico , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fase S , Sais de Tetrazólio/farmacologia , Tiazóis/farmacologia , Fatores de Tempo
10.
J Biol Chem ; 279(9): 7438-46, 2004 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-14681231

RESUMO

Cyclin A is required for cell cycle S phase entry, and its overexpression contributes to tumorigenesis. Release of pre-existing E2Fs from inactive complexes of E2F and hypophosphorylated retinoblastoma (RB) is the prevailing dogma for E2F transcriptional activation of target genes such as cyclin A. Here we explored the hypothesis that new synthesis of E2F-1 is required for insulin-like growth factor-I (IGF-I) to induce cyclin A accumulation and RB hyperphosphorylation, events that are targeted by tumor necrosis factor alpha (TNFalpha) to arrest cell cycle progression. We first established that IGF-I increases expression of cyclin A, causes hyperphosphorylation of RB, and augments the mass of E2F-1 in a time-dependent manner. As expected, E2F-1 small interfering RNA blocks the ability of IGF-I to increase synthesis of E2F-1. Most important, this E2F-1 small interfering RNA also blocks the ability of IGF-I to increase cyclin A accumulation and to hyperphosphorylate RB. We next established that TNFalpha dose-dependently inhibits IGF-I-induced phosphorylation of both RB and histone H1 by cyclin A-dependent cyclin-dependent kinases. Cyclin-dependent kinase 2 (Cdk2) mediates this suppression because co-immunoprecipitation experiments revealed that TNFalpha reduces the amount of IGF-I-induced cyclin A that binds Cdk2, leading to a reduction in Cdk2 enzymatic activity. TNFalpha antagonizes the ability of IGF-I to increase mass of both E2F-1 and cyclin A but not cyclin E or D1. The cytostatic property of TNFalpha is also shown by its ability to block IGF-I-stimulated luciferase activity of a cyclin A promoter reporter. Deletion of an E2F recognition site from this reporter eliminates the regulatory effects of both IGF-I and TNFalpha on cyclin A transcription, indicating the essential role of E2F-1 in mediating their cross-talk. Collectively, these results establish that TNFalpha targets IGF-I-induced E2F-1 synthesis, leading to inhibition of the subsequent accumulation in cyclin A, formation of cyclin A-Cdk2 complexes, hyperphosphorylation of RB, and cell cycle arrest.


Assuntos
Proteínas de Ciclo Celular , Ciclina A/genética , Proteínas de Ligação a DNA , Fator de Crescimento Insulin-Like I/farmacologia , Proteína do Retinoblastoma/metabolismo , Fatores de Transcrição/biossíntese , Fator de Necrose Tumoral alfa/fisiologia , Adenocarcinoma , Neoplasias da Mama , Quinases relacionadas a CDC2 e CDC28/metabolismo , Ciclo Celular , Ciclina A/metabolismo , Ciclina A/farmacologia , Quinase 2 Dependente de Ciclina , Fatores de Transcrição E2F , Fator de Transcrição E2F1 , Expressão Gênica/efeitos dos fármacos , Humanos , Immunoblotting , Técnicas de Imunoadsorção , Luciferases/genética , Fosforilação , Fosfosserina/metabolismo , Regiões Promotoras Genéticas/genética , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/farmacologia
11.
Mol Cell ; 11(4): 997-1008, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12718885

RESUMO

Eukaryotic cells control the initiation of DNA replication so that origins that have fired once in S phase do not fire a second time within the same cell cycle. Failure to exert this control leads to genetic instability. Here we investigate how rereplication is prevented in normal mammalian cells and how these mechanisms might be overcome during tumor progression. Overexpression of the replication initiation factors Cdt1 and Cdc6 along with cyclin A-cdk2 promotes rereplication in human cancer cells with inactive p53 but not in cells with functional p53. A subset of origins distributed throughout the genome refire within 2-4 hr of the first cycle of replication. Induction of rereplication activates p53 through the ATM/ATR/Chk2 DNA damage checkpoint pathways. p53 inhibits rereplication through the induction of the cdk2 inhibitor p21. Therefore, a p53-dependent checkpoint pathway is activated to suppress rereplication and promote genetic stability.


Assuntos
Quinases relacionadas a CDC2 e CDC28 , Proteínas de Ciclo Celular/genética , Divisão Celular/genética , Transformação Celular Neoplásica/genética , Replicação do DNA/genética , Células Eucarióticas/metabolismo , Genes cdc/fisiologia , Proteína Supressora de Tumor p53/genética , Animais , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Quinase do Ponto de Checagem 2 , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Ciclina A/genética , Ciclina A/metabolismo , Ciclina A/farmacologia , Quinase 2 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p21 , Quinases Ciclina-Dependentes/genética , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/genética , Ciclinas/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Humanos , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo
12.
J Biol Chem ; 277(32): 28592-600, 2002 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-12036957

RESUMO

Fully grown Xenopus oocyte is arrested at prophase I of meiosis. Re-entry into meiosis depends on the activation of MPF (M-phase promoting factor or cyclin B.Cdc2 complex), triggered by progesterone. The prophase-arrested oocyte contains a store of Cdc2. Most of the protein is present as a monomer whereas a minor fraction, called pre-MPF, is found to be associated with cyclin B. Activation of Cdc2 depends on two key events: cyclin binding and an activating phosphorylation on Thr-161 residue located in the T-loop. To get new insights into the regulation of Thr-161 phosphorylation of Cdc2, monomeric Cdc2 was isolated from prophase oocytes. Based on its activation upon cyclin addition and detection by an antibody directed specifically against Cdc2 phosphorylated on Thr-161, we show for the first time that the prophase oocyte contains a significant amount of monomeric Cdc2 phosphorylated on Thr-161. PP2C, a Mg2+-dependent phosphatase, negatively controls Thr-161 phosphorylation of Cdc2. The unexpected presence of a population of free Cdc2 already phosphorylated on Thr-161 could contribute to the generation of the Cdc2 kinase activity threshold required to initiate MPF amplification.


Assuntos
Proteína Quinase CDC2/metabolismo , Quinases relacionadas a CDC2 e CDC28 , Proteínas Serina-Treonina Quinases , Proteínas de Saccharomyces cerevisiae , Treonina/química , Animais , Cromatografia em Gel , Clonagem Molecular , Ciclina A/farmacologia , Quinase 2 Dependente de Ciclina , Quinases Ciclina-Dependentes/metabolismo , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica no Desenvolvimento , Biblioteca Gênica , Glutationa Transferase/metabolismo , Histonas/metabolismo , Immunoblotting , Magnésio/farmacologia , Meiose , Dados de Sequência Molecular , Fosfoproteínas Fosfatases/metabolismo , Fosforilação , Ligação Proteica , Proteína Fosfatase 2 , Proteína Fosfatase 2C , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas de Xenopus , Xenopus laevis , Quinase Ativadora de Quinase Dependente de Ciclina
13.
Exp Cell Res ; 274(2): 334-41, 2002 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11900493

RESUMO

Nuclei from terminally differentiated Xenopus erythrocytes lack essential components of the prereplication complex, including the origin recognition complex (ORC) proteins XORC1 and XORC2. In Xenopus egg extract, these proteins are able to bind erythrocyte chromatin from permeable nuclei, but not from intact nuclei, even though they are able to cross an intact nuclear envelope. In this report we use both permeable and intact erythrocyte nuclei to investigate the role of cyclin-dependent kinase activity in modulating the binding of XORC2 to chromatin. We find that elevating the level of cyclin A-dependent kinase in egg extract prevents the binding of XORC2 to chromatin from permeable nuclei and that kinase inhibition reverses this effect. We also observe a nuclear transport-dependent accumulation of H1 kinase activity within intact nuclei incubated in the extract. However, inhibiting this kinase activity does not facilitate the binding of XORC2 to chromatin, suggesting that other molecules and/or mechanisms exist to prevent association of XORC proteins with replication origins within intact nuclei from terminally differentiated cells.


Assuntos
Quinases relacionadas a CDC2 e CDC28 , Diferenciação Celular/genética , Divisão Celular/fisiologia , Núcleo Celular/genética , Cromatina/metabolismo , Replicação do DNA/fisiologia , Proteínas de Ligação a DNA/metabolismo , Eritroblastos/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Animais , Cromatina/genética , Ciclina A/metabolismo , Ciclina A/farmacologia , Quinase 2 Dependente de Ciclina , Quinases Ciclina-Dependentes/metabolismo , Quinases Ciclina-Dependentes/farmacologia , Proteínas de Ligação a DNA/genética , Feminino , Histonas/metabolismo , Fator Promotor de Maturação/antagonistas & inibidores , Fator Promotor de Maturação/metabolismo , Membrana Nuclear/genética , Membrana Nuclear/metabolismo , Oócitos , Complexo de Reconhecimento de Origem , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/farmacologia , Xenopus , Proteínas de Xenopus
14.
Blood ; 99(3): 939-45, 2002 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-11806997

RESUMO

Previously, it was reported that homocysteine (Hcy) specifically inhibits the growth of endothelial cells (ECs), suppresses Ras/mitogen-activated protein (MAP) signaling, and arrests cell growth at the G(1)/S transition of the cell cycle. The present study investigated the molecular mechanisms underlying this cell-cycle effect. Results showed that clinically relevant concentrations (50 microM) of Hcy significantly inhibited the expression of cyclin A messenger RNA (mRNA) in ECs in a dose- and time-dependent manner. G(1)/S-associated molecules that might account for this block were not changed, because Hcy did not affect mRNA and protein expression of cyclin D1 and cyclin E. Cyclin D1- and E-associated kinase activities were unchanged. In contrast, cyclin A-associated kinase activity and CDK2 kinase activity were markedly suppressed. Nuclear run-on assay demonstrated that Hcy decreased the transcription rate of the cyclin A gene but had no effect on the half-life of cyclin A mRNA. In transient transfection experiments, Hcy significantly inhibited cyclin A promoter activity in endothelial cells, but not in vascular smooth muscle cells. Finally, adenovirus-transduced cyclin A expression restored EC growth inhibition and overcame the S phase block imposed by Hcy. Taken together, these findings indicate that cyclin A is a critical functional target of Hcy-mediated EC growth inhibition.


Assuntos
Ciclina A/antagonistas & inibidores , Regulação para Baixo/efeitos dos fármacos , Endotélio Vascular/citologia , Homocisteína/farmacologia , Animais , Aorta/citologia , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Ciclina A/genética , Ciclina A/farmacologia , Regulação para Baixo/genética , Endotélio Vascular/efeitos dos fármacos , Homocisteína/fisiologia , Humanos , Músculo Liso Vascular/citologia , Regiões Promotoras Genéticas/efeitos dos fármacos , RNA Mensageiro/análise , RNA Mensageiro/efeitos dos fármacos , Ratos , Transcrição Gênica/efeitos dos fármacos , Veias Umbilicais/citologia
15.
Lung Cancer ; 31(2-3): 181-91, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11165397

RESUMO

TGF-beta is a potent inhibitory regulator of cell growth, which is transduced through interaction between type I (RI) and type II (RII) receptors that form heteromeric kinase complexes. Abnormal expression of these receptors has been identified in several human epithelial cancers and has been shown to be highly associated with resistance to TGF-beta. In this study, we investigated the expression of RI and RII in 13 human non-small cell lung cancer cell lines (NSCLCs) and demonstrated decreased or loss of RII expression in five lung cancer cell lines, but not of RI. Of these cell lines, the role of RII in NCI-H358 adenocarcinoma, which lacks RII and is insensitive to TGF-beta, was investigated by transducing this cell line with a recombinant retrovirus expressing full-length TGF-beta RII. Stably transfected cells showed significant increase in RII mRNA and protein expression. These cells responded to exogenous TGF-beta1 with suppressed proliferation in a dose-dependent manner and G1 arrest accompanied by morphological change distinct from control cells. We also investigated whether overexpression of dominant-negative RII (dnRII) in NCI-H441 adenocarcinoma, which is sensitive but expresses low levels of RII, could block signaling through the receptor complex. The overexpression of this kinase-domain-truncated RII by expressing the retroviral dnRII construct led to loss of the ability to respond to TGF-beta1 and an exhibition of uncontrolled growth. These results suggest a close association between the loss of the expression of wild-type TGF-beta RII and carcinogenesis in human lung cancer cells.


Assuntos
Adenocarcinoma/fisiopatologia , Transformação Celular Neoplásica , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/fisiopatologia , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Adenocarcinoma/patologia , Divisão Celular , Ciclina A/farmacologia , Humanos , Neoplasias Pulmonares/patologia , RNA Mensageiro , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Proteína do Retinoblastoma/biossíntese , Transfecção , Fator de Crescimento Transformador beta/biossíntese , Fator de Crescimento Transformador beta/farmacologia , Células Tumorais Cultivadas
16.
J Mol Biol ; 293(5): 1005-15, 1999 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-10547281

RESUMO

Promoters of growth and cell cycle regulated genes frequently carry binding sites for transcription factors of the E2F and Sp1 families. We have demonstrated recently that direct interaction between Sp1 and a subgroup of the E2F factors is essential for the regulation of certain promoters. We show here that the amino acids necessary for this interaction in both cases are located within the DNA binding domain. This is in line with the assumption, that the interaction between E2F and Sp-factors contributes to promoter-specificity. Cyclin A, which binds to E2F-1 in close vicinity to Sp1 does not interfere with this interaction. Moreover we have investigated the ability of other members of the Sp1 family to interact with E2F-1 and to regulate the activity of the E2F and Sp1 dependent murine thymidine kinase promoter. All four factors of the Sp1 family are able to bind E2F-1 in co-immunoprecipitation and GST-pull down experiments. Mobility shift assays with oligonucleotides comprising the Sp1, or both the Sp1 and the E2F binding site suggest that Sp1 and Sp3 supply most if not all activity binding to the GC-box of the thymidine kinase promoter in murine fibroblasts. Reporter gene assays in Drosophila melanogaster SL2 cells and murine fibroblast 3T6 cells demonstrate that the thymidine kinase promoter is activated strongly by Sp1 and Sp3, weakly by Sp4, and not at all by Sp2. Co-expression of E2F-1 results in synergistic activation in 3T6 but not in SL2 cells.


Assuntos
Proteínas de Transporte , Proteínas de Ciclo Celular , Proteínas de Ligação a DNA , Proteínas de Drosophila , Família Multigênica , Regiões Promotoras Genéticas/genética , Fator de Transcrição Sp1/metabolismo , Timidina Quinase/genética , Transativadores , Fatores de Transcrição/metabolismo , Ativação Transcricional , Sequência de Aminoácidos , Animais , Sítios de Ligação , Linhagem Celular , Ciclina A/metabolismo , Ciclina A/farmacologia , DNA/genética , DNA/metabolismo , Drosophila melanogaster , Fatores de Transcrição E2F , Fator de Transcrição E2F1 , Fibroblastos , Camundongos , Dados de Sequência Molecular , Família Multigênica/genética , Família Multigênica/fisiologia , Ligação Proteica/efeitos dos fármacos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Elementos de Resposta/genética , Proteína 1 de Ligação ao Retinoblastoma , Alinhamento de Sequência , Deleção de Sequência , Fator de Transcrição Sp1/química , Fator de Transcrição Sp1/genética , Fator de Transcrição DP1 , Fatores de Transcrição/química , Fatores de Transcrição/genética
17.
J Cell Biochem ; 75(2): 300-9, 1999 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-10502302

RESUMO

Transcriptional activation is important for the elevated expression of human thymidine kinase (hTK) in tumor cells. Here, we used TK(-133/+33)-luciferase reporter gene construct and bandshift assay to study the cis-elements involved in transcriptional activation of the hTK promoter. We found that two CCAAT boxes at -71/-67 and -40/-36 and Sp1 binding site located at -118/-113 were critical for maximal expression of the hTK promoter activity. As Sp1-mediated activation of the hTK promoter was not detectable for the promoter construct with double mutations at two CCAAT boxes, we proposed that NF-Y binding to the hTK promoter sequence is a requisite step for the functional interaction with Sp1. Here, we further showed that the hTK promoter activity was reduced in HeLa cells transfected with p16 or p21, both of which are inhibitors of cyclin-dependent kinases (CDKs). Inhibition of the hTK promoter activity by p16 could be abrogated by overexpression of cyclin A, indicating that the cyclin A activating event is more directly involved in transcriptional activation of the hTK promoter. We thus proposed that NF-Y-mediated activation of the hTK promoter is closely linked to the activation of CDK2/cyclin A pathway.


Assuntos
Quinases Ciclina-Dependentes/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regiões Promotoras Genéticas , Timidina Quinase/genética , Transativadores/genética , Transativadores/metabolismo , Sequência de Bases , Proteínas Estimuladoras de Ligação a CCAAT , Ciclina A/farmacologia , Inibidor p16 de Quinase Dependente de Ciclina/farmacologia , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/farmacologia , Relação Dose-Resposta a Droga , Genes Reporter , Células HL-60 , Células HeLa , Humanos , Immunoblotting , Luciferases/metabolismo , Dados de Sequência Molecular , Mutagênese , Plasmídeos/metabolismo , Fatores de Tempo , Transfecção
18.
Genes Dev ; 12(15): 2278-92, 1998 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-9694794

RESUMO

The retinoblastoma tumor suppressor protein, RB, is a negative regulator of cell proliferation. Growth inhibitory activity of RB is attenuated by phosphorylation. Mutation of a combination of phosphorylation sites leads to a constitutively active RB. In Rat-1 cells, the phosphorylation-site-mutated (PSM)-RB, but not wild-type RB, can inhibit S-phase entry. In PSM-RB-arrested G1 cells, normal levels of cyclin E and cyclin E-associated kinase activity were detected, but the expression of cyclin A was inhibited. The ectopic expression of cyclin E restored cyclin A expression and drove the PSM-RB expressing cells into S phase. Interestingly, Rat-1 cells coexpressing cyclin E and PSM-RB could not complete DNA replication. Microinjection of cells that have passed through the G1 restriction point with plasmids expressing PSM-RB also led to the inhibition of DNA synthesis. The S-phase inhibitory activity of PSM-RB could be attenuated by the coinjection of SV40 T-antigen, adenovirus E1A, or a high level of E2F-1 expression plasmids. However, the S-phase inhibitory activity of PSM-RB could not be overcome by the coinjection of cyclin E or cyclin A expression plasmids. These results reveal a novel role for RB in the inhibition of S-phase progression that is distinct from the inhibition of the G1/S transition, and suggest that continued phosphorylation of RB beyond G1/S is required for the completion of DNA replication.


Assuntos
Proteínas de Transporte , Proteínas de Ciclo Celular , Proteínas de Ligação a DNA , DNA/biossíntese , Fase G1/fisiologia , Proteína do Retinoblastoma/fisiologia , Fase S/fisiologia , Proteínas E1A de Adenovirus/farmacologia , Animais , Antígenos Transformantes de Poliomavirus/genética , Antígenos Transformantes de Poliomavirus/farmacologia , Sequência de Bases , Sítios de Ligação/genética , Linhagem Celular , Ciclina A/genética , Ciclina A/metabolismo , Ciclina A/farmacologia , Ciclina E/genética , Ciclina E/metabolismo , Ciclina E/farmacologia , Primers do DNA/genética , Fatores de Transcrição E2F , Fator de Transcrição E2F1 , Fase G1/genética , Expressão Gênica , Mutagênese Sítio-Dirigida , Mutação , Fosforilação , Reação em Cadeia da Polimerase , Ratos , Proteína do Retinoblastoma/genética , Proteína 1 de Ligação ao Retinoblastoma , Fase S/efeitos dos fármacos , Fase S/genética , Fator de Transcrição DP1 , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
19.
Dev Biol ; 196(2): 160-70, 1998 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-9576829

RESUMO

The germ cells of metazoans follow a program of proliferation that is distinct from proliferation programs of somatic cells. Despite their developmental importance, the cell proliferation program in the metazoan primordial germ cells is not well characterized and the regulatory controls are not understood. In Drosophila melanogaster, germ cell precursors (called pole cells) proliferate early in embryogenesis and then enter a prolonged quiescence. We found that polar nuclear divisions are asynchronous and lag behind somatic nuclear divisions during syncytial cycles 9 and 10. Thus, the polar division program deviates from the somatic division program when pole nuclei and somatic nuclei still share a common cytoplasm, earlier than previously thought. The lag in polar nuclear divisions is independent of grapes, which is required for lengthening somatic cell cycles 10-13. Mapping of the last S phase in pole cells and measurement of their DNA content indicate that pole cells become quiescent in G2 phase of the cell cycle. We were able to drive quiescent pole cells into mitosis by induction of either an activator of Cdc2 (Cdc25string phosphatase) or a mutant form of Cdc2 that cannot be inhibited by phosphorylation. In contrast, induction of wild-type Cdc2 with a mitotic cyclin did not induce mitosis in pole cells. We propose that inhibition of Cdc2 by phosphorylation contributes to G2 arrest in pole cells during embryogenesis. Furthermore, pole cells enter G1 following induced mitoses, indicating that entry into both mitosis and S phase is blocked in quiescent pole cells. These studies represent the first molecular characterization of proliferation in embryonic germ cells of Drosophila.


Assuntos
Proteína Quinase CDC2/fisiologia , Ciclo Celular/fisiologia , Drosophila melanogaster/embriologia , Células Germinativas/citologia , Animais , Proteínas de Ciclo Celular/metabolismo , Ciclina A/análise , Ciclina A/farmacologia , Replicação do DNA , Drosophila melanogaster/citologia , Embrião não Mamífero/química , Ativação Enzimática , Fase G2/fisiologia , Histonas/análise , Mitose , Fosfoproteínas Fosfatases/metabolismo , Fosforilação , Fosfatases cdc25
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...