Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 13(1): 2835, 2022 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-35595767

RESUMO

Cyclin-dependent kinase 2 (CDK2) complex is significantly over-activated in many cancers. While it makes CDK2 an attractive target for cancer therapy, most inhibitors against CDK2 are ATP competitors that are either nonspecific or highly toxic, and typically fail clinical trials. One alternative approach is to develop non-ATP competitive inhibitors; they disrupt interactions between CDK2 and either its partners or substrates, resulting in specific inhibition of CDK2 activities. In this report, we identify two potential druggable pockets located in the protein-protein interaction interface (PPI) between CDK2 and Cyclin A. To target the potential druggable pockets, we perform a LIVS in silico screening of a library containing 1925 FDA approved drugs. Using this approach, homoharringtonine (HHT) shows high affinity to the PPI and strongly disrupts the interaction between CDK2 and cyclins. Further, we demonstrate that HHT induces autophagic degradation of the CDK2 protein via tripartite motif 21 (Trim21) in cancer cells, which is confirmed in a leukemia mouse model and in human primary leukemia cells. These results thus identify an autophagic degradation mechanism of CDK2 protein and provide a potential avenue towards treating CDK2-dependent cancers.


Assuntos
Autofagia , Quinases relacionadas a CDC2 e CDC28 , Ciclina A , Quinase 2 Dependente de Ciclina , Leucemia , Animais , Linhagem Celular Tumoral/metabolismo , Ciclina A/antagonistas & inibidores , Ciclina A/metabolismo , Quinase 2 Dependente de Ciclina/antagonistas & inibidores , Quinase 2 Dependente de Ciclina/metabolismo , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/metabolismo , Mepesuccinato de Omacetaxina/metabolismo , Mepesuccinato de Omacetaxina/farmacologia , Humanos , Camundongos , Ribonucleoproteínas
2.
Chem Biol Interact ; 272: 72-79, 2017 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-28506552

RESUMO

Ovarian cancer is the most lethal gynecological malignancy worldwide. Thiophenes such as terthiophene have been shown to have anti-tumor effects on several cancer cell lines, including ovarian cancer cells. However, the underlying mechanisms behind the anti-proliferative effect of thiophenes are poorly understood. In this study, we investigated the molecular mechanisms underlying the anti-proliferative effect of α-terthienylmethanol, a terthiophene isolated from Eclipta prostrata (False Daisy), on human ovarian cancer cells. We found that α-terthienylmethanol is a more potent inhibitor of cell growth than is cisplatin in human ovarian cancer cells. α-Terthienylmethanol induces cell cycle arrest in ovarian cancer cells, as shown by the accumulation of cells in S phase. In addition, α-terthienylmethanol induced a change in S phase-related proteins cyclin A, cyclin-dependent kinase 2, and cyclin D2. Knockdown of cyclin A using specific siRNAs significantly compromised α-terthienylmethanol-induced S phase arrest. We further demonstrated that α-terthienylmethanol induced an increase in intracellular ROS, and the antioxidant N-acetyl-l-cysteine significantly reversed the S phase arrest induced by α-terthienylmethanol. Moreover, α-terthienylmethanol significantly increased the levels of p-H2AX, a DNA damage marker. These results suggest that α-terthienylmethanol inhibits the growth of human ovarian cancer cells by S phase cell cycle arrest via induction of ROS stress and DNA damage.


Assuntos
Espécies Reativas de Oxigênio/metabolismo , Pontos de Checagem da Fase S do Ciclo Celular/efeitos dos fármacos , Tiofenos/toxicidade , Acetilcisteína/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Ciclina A/antagonistas & inibidores , Ciclina A/genética , Ciclina A/metabolismo , Quinase 2 Dependente de Ciclina/metabolismo , Dano ao DNA/efeitos dos fármacos , Eclipta/química , Eclipta/metabolismo , Feminino , Histonas/metabolismo , Humanos , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Fosforilação/efeitos dos fármacos , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Tiofenos/química
3.
Mol Biol Cell ; 27(13): 2051-63, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27170181

RESUMO

Regulation of cell cycle arrest in premeiotic G2 phase coordinates germ cell maturation and meiotic cell division with hormonal and developmental signals by mechanisms that control Cyclin B synthesis and inhibitory phosphorylation of the M-phase kinase, Cdk1. In this study, we investigated how inhibitory phosphorylation of Cdk1 by Myt1 kinase regulates premeiotic G2 phase of Drosophila male meiosis. Immature spermatocytes lacking Myt1 activity exhibit two distinct defects: disrupted intercellular bridges (fusomes) and premature centriole disengagement. As a result, the myt1 mutant spermatocytes enter meiosis with multipolar spindles. These myt1 defects can be suppressed by depletion of Cyclin A activity or ectopic expression of Wee1 (a partially redundant Cdk1 inhibitory kinase) and phenocopied by expression of a Cdk1F mutant defective for inhibitory phosphorylation. We therefore conclude that Myt1 inhibition of Cyclin A/Cdk1 is essential for normal fusome behavior and centriole engagement during premeiotic G2 arrest of Drosophila male meiosis. The novel meiotic functions we discovered for Myt1 kinase are spatially and temporally distinct from previously described functions of Myt1 as an inhibitor of Cyclin B/Cdk1 to regulate G2/MI timing.


Assuntos
Proteína Quinase CDC2/metabolismo , Ciclina A/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Animais , Proteína Quinase CDC2/antagonistas & inibidores , Proteínas de Ciclo Celular/metabolismo , Centríolos/metabolismo , Ciclina A/antagonistas & inibidores , Ciclina B/metabolismo , Drosophila/metabolismo , Fase G2 , Masculino , Meiose , Mitose , Proteínas Nucleares/metabolismo , Fosforilação
4.
Biochem Pharmacol ; 96(4): 315-22, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26074263

RESUMO

REV-ERBα and REV-ERBß are nuclear receptors that are ligand-dependent transcriptional repressors. Heme is the natural ligand for these receptors, but several synthetic agonists and antagonists have been designed recently. The gene that encodes REV-ERBα, NR1D1, is closely associated with ERBB2, the gene that encodes the HER2 oncogene, which is amplified in HER2(+) breast cancers. We examined the effect of a synthetic REV-ERB agonist, SR9011, on a range of estrogen receptor positive (ER(+)), ER(-), HER2(+), HER2(-) and triple negative breast cancer cell lines. We found that SR9011 suppressed proliferation of the breast cancer cell lines regardless of their ER or HER2 status. SR9011 had no effect on MCF10A cell proliferation. SR9011 appears to pause the cell cycle of the breast cancer cells prior to M phase. Cyclin A (CCNA2) was identified as a direct target gene of REV-ERB suggesting that suppression of expression of this cyclin by SR9011 may mediate the cell cycle arrest. These data indicate that synthetic REV-ERB ligands may hold utility in treatment of diseases associated with uncontrolled cellular proliferation such as cancer.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/patologia , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/agonistas , Pirrolidinas/farmacologia , Tiofenos/farmacologia , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ciclina A/antagonistas & inibidores , Ciclina A/genética , Ciclina A/metabolismo , Feminino , Humanos , Receptor ErbB-2/metabolismo , Receptores de Estrogênio/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia
5.
PLoS One ; 10(4): e0123736, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25860957

RESUMO

Inhibitor of growth (ING) proteins have multiple functions in the control of cell proliferation, mainly by regulating processes associated with chromatin regulation and gene expression. ING5 has been described to regulate aspects of gene transcription and replication. Moreover deregulation of ING5 is observed in different tumors, potentially functioning as a tumor suppressor. Gene transcription in late G1 and in S phase and replication is regulated by cyclin-dependent kinase 2 (CDK2) in complex with cyclin E or cyclin A. CDK2 complexes phosphorylate and regulate several substrate proteins relevant for overcoming the restriction point and promoting S phase. We have identified ING5 as a novel CDK2 substrate. ING5 is phosphorylated at a single site, threonine 152, by cyclin E/CDK2 and cyclin A/CDK2 in vitro. This site is also phosphorylated in cells in a cell cycle dependent manner, consistent with it being a CDK2 substrate. Furthermore overexpression of cyclin E/CDK2 stimulates while the CDK2 inhibitor p27KIP1 represses phosphorylation at threonine 152. This site is located in a bipartite nuclear localization sequence but its phosphorylation was not sufficient to deregulate the subcellular localization of ING5. Although ING5 interacts with the tumor suppressor p53, we could not establish p53-dependent regulation of cell proliferation by ING5 and by phospho-site mutants. Instead we observed that the knockdown of ING5 resulted in a strong reduction of proliferation in different tumor cell lines, irrespective of the p53 status. This inhibition of proliferation was at least in part due to the induction of apoptosis. In summary we identified a phosphorylation site at threonine 152 of ING5 that is cell cycle regulated and we observed that ING5 is necessary for tumor cell proliferation, without any apparent dependency on the tumor suppressor p53.


Assuntos
Quinase 2 Dependente de Ciclina/metabolismo , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Substituição de Aminoácidos , Sítios de Ligação , Linhagem Celular , Proliferação de Células/fisiologia , Ciclina A/antagonistas & inibidores , Ciclina A/genética , Ciclina A/metabolismo , Ciclina E/antagonistas & inibidores , Ciclina E/genética , Ciclina E/metabolismo , Técnicas de Silenciamento de Genes , Células HEK293 , Células HeLa , Humanos , Mutagênese Sítio-Dirigida , Fosforilação , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Treonina/química , Fatores de Transcrição/química , Fatores de Transcrição/genética , Transfecção , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/genética
6.
Curr Biol ; 25(6): 811-816, 2015 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-25754637

RESUMO

The timing mechanism for mitotic progression is still poorly understood. The spindle assembly checkpoint (SAC), whose reversal upon chromosome alignment is thought to time anaphase [1-3], is functional during the rapid mitotic cycles of the Drosophila embryo; but its genetic inactivation had no consequence on the timing of the early mitoses. Mitotic cyclins-Cyclin A, Cyclin B, and Cyclin B3-influence mitotic progression and are degraded in a stereotyped sequence [4-11]. RNAi knockdown of Cyclins A and B resulted in a Cyclin B3-only mitosis in which anaphase initiated prior to chromosome alignment. Furthermore, in such a Cyclin B3-only mitosis, colchicine-induced SAC activation failed to block Cyclin B3 destruction, chromosome decondensation, or nuclear membrane re-assembly. Injection of Cyclin B proteins restored the ability of SAC to prevent Cyclin B3 destruction. Thus, SAC function depends on particular cyclin types. Changing Cyclin B3 levels showed that it accelerated progress to anaphase, even in the absence of SAC function. The impact of Cyclin B3 on anaphase initiation appeared to decline with developmental progress. Our results show that different cyclin types affect anaphase timing differently in the early embryonic divisions. The early-destroyed cyclins-Cyclins A and B-restrain anaphase-promoting complex/cyclosome (APC/C) function, whereas the late-destroyed cyclin, Cyclin B3, stimulates function. We propose that the destruction schedule of cyclin types guides mitotic exit by affecting both Cdk1 and APC/C, whose activities change as each cyclin type is lost.


Assuntos
Ciclina B/metabolismo , Proteínas de Drosophila/metabolismo , Mitose/fisiologia , Anáfase/genética , Anáfase/fisiologia , Animais , Animais Geneticamente Modificados , Ciclina A/antagonistas & inibidores , Ciclina A/genética , Ciclina A/metabolismo , Ciclina B/antagonistas & inibidores , Ciclina B/genética , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/genética , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Pontos de Checagem da Fase M do Ciclo Celular/genética , Pontos de Checagem da Fase M do Ciclo Celular/fisiologia , Masculino , Metáfase/genética , Metáfase/fisiologia , Mitose/genética , Modelos Biológicos
7.
Chembiochem ; 16(3): 432-9, 2015 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-25619419

RESUMO

Cyclin-dependent kinases (CDKs) control many cellular processes and are considered important therapeutic targets. Large collections of inhibitors targeting CDK active sites have been discovered, but their use in chemical biology or drug development has been often hampered by their general lack of specificity. An alternative approach to develop more specific inhibitors is targeting protein interactions involving CDKs. CKS proteins interact with some CDKs and play important roles in cell division. We discovered two small-molecule inhibitors of CDK-CKS interactions. They bind to CDK2, do not inhibit its enzymatic activity, inhibit the proliferation of tumor cell lines, induce an increase in G1 and/or S-phase cell populations, and cause a decrease in CDK2, cyclin A, and p27(Kip1) levels. These molecules should help decipher the complex contributions of CDK-CKS complexes in the regulation of cell division, and they might present an interesting therapeutic potential.


Assuntos
Quinases relacionadas a CDC2 e CDC28/metabolismo , Quinase 2 Dependente de Ciclina/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/farmacologia , Quinases relacionadas a CDC2 e CDC28/antagonistas & inibidores , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/metabolismo , Divisão Celular/efeitos dos fármacos , Ciclina A/antagonistas & inibidores , Ciclina A/metabolismo , Quinase 2 Dependente de Ciclina/metabolismo , Relação Dose-Resposta a Droga , Ensaios de Triagem em Larga Escala , Humanos , Células MCF-7/efeitos dos fármacos , Simulação de Acoplamento Molecular , Estrutura Molecular , Terapia de Alvo Molecular , Mapas de Interação de Proteínas/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/metabolismo
8.
Environ Toxicol ; 30(10): 1205-15, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24706497

RESUMO

Latex of Euphorbia antiquorum (EA) has demonstrated great chemotherapeutic potential for cancer. However, the mechanisms of anti-proliferation of EA on cancer cell remain to be further investigated. The purpose of this study was to explore the influence of EA in human cervical cancer cells. Here, the cell cycle distribution by flow cytometry was examined and the protein expression by the western blotting methods was analyzed. From the cytometric results it was shown that EA-induced S-phase arrest in a concentration manner both in human cervical cancer HeLa and CaSki cells. According the western blot results it was illustrated that EA could downregulate early cyclin E1-Cdk2; and cyclin A-Cdc2 provides a significant additional quantity of S-phase promotion, that in turn promoted the expression of p21(waf1/cip1) and p27(kip1) which were the inhibitors in the complex of cyclin A and Cdc2 that led to cell cycle arrest. Moreover, EA promoted the activation of ataxia telangiectasia mutated (ATM) and check-point kinase-2 (Chk2); however, it negatively regulated the expression of Topoisomerases I and II, Cdc25A, and Cdc25C signaling. Caffeine, an ATM/ATR inhibitor significantly reversed EA downregulation in the levels of Cdc25A. Furthermore, JNK inhibitor SP600125 and p38 MAPK inhibitor SB203580 both could reverse the EA upregulation of the protein of Chk2 level, significantly. This study, therefore, revealed that EA could downregulate topoisomerase, and activate ATM kinase, which then induce parallel Chk 1/2 and MAPK signaling pathways to promote the degradation of Cdc25A to induced S-phase arrest in human cervical cancer HeLa cells.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Euphorbia/química , Látex/toxicidade , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Pontos de Checagem da Fase S do Ciclo Celular/efeitos dos fármacos , Proteínas Mutadas de Ataxia Telangiectasia/antagonistas & inibidores , Proteína Quinase CDC2/antagonistas & inibidores , Proteína Quinase CDC2/metabolismo , Linhagem Celular Tumoral , Quinase 1 do Ponto de Checagem , Ciclina A/antagonistas & inibidores , Ciclina A/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , DNA Topoisomerases Tipo I/metabolismo , Euphorbia/metabolismo , Feminino , Células HeLa , Humanos , Látex/química , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/química , Proteínas Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Neoplasias do Colo do Útero/metabolismo , Neoplasias do Colo do Útero/patologia , Fosfatases cdc25/metabolismo
9.
Rapid Commun Mass Spectrom ; 28(24): 2681-9, 2014 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-25380489

RESUMO

RATIONALE: Matrix-assisted laser desorption/ionization (MALDI) mass spectrometry combined with isotope labeling methods are effective for protein and peptide quantification, but limited in their multiplexing capacity, cost-effectiveness and dynamic range. This study investigates MALDI-MS-based quantification of peptide phosphorylation without labeling, and aims to overcome the shot-to-shot variability of MALDI using a mathematical transformation and extended data acquisition times. METHODS: A linear relationship between the reciprocal of phosphopeptide mole fraction and the reciprocal of phosphorylated-to-unphosphorylated signal ratio is derived, and evaluated experimentally using three separate phosphopeptide systems containing phosphorylated serine, threonine and tyrosine residues: mixtures of phosphopeptide and its des-phospho-analog with known stoichiometry measured by vacuum MALDI-linear ion trap mass spectrometry and fit to the linear model. The model is validated for quantifying in vitro phosphorylation assays with inhibition studies on Cdk2/cyclinA. RESULTS: Dynamic range of picomoles to femtomoles, good accuracy (deviations of 1.5-3.0% from expected values) and reproducibility (relative standard deviation (RSD) = 4.3-6.3%) are achieved. Inhibition of cyclin-dependent kinase phosphorylation by the classical inhibitors olomoucine and r-roscovitine was evaluated and IC50 values found to be in agreement with reported literature values. These results, achieved with single-point calibration, without isotope or chromatography, compare favorably to those arrived at using isotope dilution (p > 0.5 for accuracy). CONCLUSIONS: The mathematical relationship derived here can be applied to a method that we term Double Reciprocal Isotope-free Phosphopeptide Quantification (DRIP-Q), as a strategy for quantification of in vitro phosphorylation assays, the first MALDI-based, isotope- and calibration curve-free method of its type. These results also pave the way for further systematic studies investigating the effect of peptide composition and experimental conditions on quantitative, label-free MALDI.


Assuntos
Cromatografia Líquida de Alta Pressão/métodos , Fosfopeptídeos/análise , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Sequência de Aminoácidos , Calibragem , Ciclina A/antagonistas & inibidores , Ciclina A/metabolismo , Quinase 2 Dependente de Ciclina/antagonistas & inibidores , Quinase 2 Dependente de Ciclina/metabolismo , Modelos Lineares , Dados de Sequência Molecular , Fosfopeptídeos/química , Fosfopeptídeos/metabolismo , Fosforilação , Reprodutibilidade dos Testes
10.
Int J Oncol ; 45(6): 2393-402, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25340978

RESUMO

Cantharidin (CTD), a component of natural mylabris (Mylabris phalerata Pallas) was reported to have high cytotoxicity in many human cancer cell lines. However, it was not reported to affect human melanoma A375.S2 cells. In the present study, we found that CTD induced cell morphological changes and decreased the percentage of viable cells and induced G2/M phase arrest and induction of apoptosis in A375.S2 cells. Results also showed that CTD induced the generation of reactive oxygen species (ROS) and Ca2+ and decreased mitochondria membrane potential and lead to the release of cytochrome c, AIF and Endo G. Further investigation revealed that CTD induced A375.S2 cells with an increase of caspase activation and caspase-dependent apoptotic proteins to trigger correlated pathway mechanisms according to western blotting results. Western blotting was used for examining the changes of G2/M phase arrest and apoptosis-associated protein expression and confocal laser microscopy was used to examine the translocation apoptosis-associated protein. Results showed that CTD increased the protein expression of caspase-3, -8 and -9, cytochrome c, Bax, Bid, Endo G and AIF but inhibited the levels of Bcl-2 and Bcl-x. CTD induced ER stress-associated protein expression such as GRP78, IRE1ß, ATF6α and caspase-12. Based on those observations, we suggest that CTD may have potential as a novel anti-cancer agent for the treatment of skin cancer.


Assuntos
Cantaridina/administração & dosagem , Ciclina A/biossíntese , Melanoma/tratamento farmacológico , Fosfatases cdc25/biossíntese , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Ciclina A/antagonistas & inibidores , Chaperona BiP do Retículo Endoplasmático , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Melanoma/genética , Melanoma/patologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteínas de Neoplasias/biossíntese , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Fosfatases cdc25/antagonistas & inibidores
11.
PLoS One ; 9(10): e109170, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25271736

RESUMO

The majority of prostate cancer (PCa) patient receiving androgen ablation therapy eventually develop castration-resistant prostate cancer (CRPC). We previously reported that androgen treatment suppresses Skp2 and c-Myc through androgen receptor (AR) and induced G1 cell cycle arrest in androgen-independent LNCaP 104-R2 cells, a late stage CRPC cell line model. However, the mechanism of androgenic regulation of Skp2 in CRPC cells was not fully understood. In this study, we investigated the androgenic regulation of Skp2 in two AR-positive CRPC cell line models, the LNCaP 104-R1 and PC-3AR Cells. The former one is an early stage androgen-independent LNCaP cells, while the later one is PC-3 cells re-expressing either wild type AR or mutant LNCaP AR. Proliferation of LNCaP 104-R1 and PC-3AR cells is not dependent on but is suppressed by androgen. We observed in this study that androgen treatment reduced protein expression of Cdk2, Cdk7, Cyclin A, cyclin H, Skp2, c-Myc, and E2F-1; lessened phosphorylation of Thr14, Tyr15, and Thr160 on Cdk2; decreased activity of Cdk2; induced protein level of p27(Kip1); and caused G1 cell cycle arrest in LNCaP 104-R1 cells and PC-3AR cells. Overexpression of Skp2 protein in LNCaP 104-R1 or PC-3AR cells partially blocked accumulation of p27(Kip1) and increased Cdk2 activity under androgen treatment, which partially blocked the androgenic suppressive effects on proliferation and cell cycle. Analyzing on-line gene array data of 214 normal and PCa samples indicated that gene expression of Skp2, Cdk2, and cyclin A positively correlates to each other, while Cdk7 negatively correlates to these genes. These observations suggested that androgen suppresses the proliferation of CRPC cells partially through inhibition of Cyclin A, Cdk2, and Skp2.


Assuntos
Androgênios/fisiologia , Proliferação de Células/fisiologia , Ciclina A/antagonistas & inibidores , Quinase 2 Dependente de Ciclina/antagonistas & inibidores , Orquiectomia , Neoplasias da Próstata/patologia , Proteínas Quinases Associadas a Fase S/antagonistas & inibidores , Linhagem Celular Tumoral , Humanos , Masculino
12.
Bioorg Med Chem Lett ; 24(1): 199-203, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24332088

RESUMO

A new class of quinoline-based kinase inhibitors has been discovered that both disrupt cyclin dependent 2 (CDK2) interaction with its cyclin A subunit and act as ATP competitive inhibitors. The key strategy for discovering this class of protein-protein disrupter compounds was to screen the monomer CDK2 in an affinity-selection/mass spectrometry-based technique and to perform secondary assays that identified compounds that bound only to the inactive CDK2 monomer and not the active CDK2/cyclin A heterodimer. Through a series of chemical modifications the affinity (Kd) of the original hit improved from 1 to 0.005µM.


Assuntos
Ciclina A/antagonistas & inibidores , Quinase 2 Dependente de Ciclina/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Quinolinas/farmacologia , Cristalografia por Raios X , Ciclina A/química , Ciclina A/metabolismo , Quinase 2 Dependente de Ciclina/química , Quinase 2 Dependente de Ciclina/metabolismo , Relação Dose-Resposta a Droga , Humanos , Modelos Moleculares , Estrutura Molecular , Inibidores de Proteínas Quinases/química , Quinolinas/química , Relação Estrutura-Atividade
13.
Cancer Lett ; 333(1): 103-12, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23354589

RESUMO

p27(Kip1) (p27) binds and inhibits the cyclin E- or cyclin A-associated cyclin-dependent kinases (CDKs)2 and other CDKs, and negatively regulates G1-G2 cell cycle progression. To develop specific CDK inhibitors, we have modeled the interaction between p27 and cyclin A-CDK2, and designed a novel compound that mimics p27 binding to cyclin A-CDK2. The chemically synthesized inhibitor exhibited high potency and selective inhibition towards cyclin E/cyclin A-CDK2 kinase in vitro but not other kinases. To facilitate permeability of the inhibitor, a cell penetrating peptide (CPP) was conjugated to the inhibitor to examine its effect in several cancer cell lines. The CPP-conjugated inhibitor significantly inhibited the proliferation of cancer cells. The treatment of the inhibitor resulted in the increased accumulation of p27 and p21(Cip1/Waf1) (p21) and hypo-phosphorylation of retinoblastoma protein (Rb). The degradation of p27, mediated through the phosphorylation of threonine-187 in p27, was also inhibited. Consequently, exposure of cells to the inhibitor caused cell cycle arrest and apoptosis. We conclude that specific cyclinE/cyclin A-CDK2 inhibitors can be developed based on the interaction between p27 and cyclin/CDK to block cell cycle progression to prevent tumor growth and survival.


Assuntos
Antineoplásicos/farmacologia , Ciclina A/antagonistas & inibidores , Ciclina E/antagonistas & inibidores , Quinase 2 Dependente de Ciclina/antagonistas & inibidores , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Sequência de Aminoácidos , Ciclo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Peptídeos Penetradores de Células/farmacologia , Humanos , Dados de Sequência Molecular
14.
J Mol Model ; 19(1): 371-82, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22926267

RESUMO

We employ ensemble docking simulations to characterize the interactions of two enantiomeric forms of a Ru-complex compound (1-R and 1-S) with three protein kinases, namely PIM1, GSK-3ß, and CDK2/cyclin A. We show that our ensemble docking computational protocol adequately models the structural features of these interactions and discriminates between competing conformational clusters of ligand-bound protein structures. Using the determined X-ray crystal structure of PIM1 complexed to the compound 1-R as a control, we discuss the importance of including the protein flexibility inherent in the ensemble docking protocol, for the accuracy of the structure prediction of the bound state. A comparison of our ensemble docking results suggests that PIM1 and GSK-3ß bind the two enantiomers in similar fashion, through two primary binding modes: conformation I, which is very similar to the conformation presented in the existing PIM1/compound 1-R crystal structure; conformation II, which represents a 180° flip about an axis through the NH group of the pyridocarbazole moiety, relative to conformation I. In contrast, the binding of the enantiomers to CDK2 is found to have a different structural profile including a suggested bound conformation, which lacks the conserved hydrogen bond between the kinase and the ligand (i.e., ATP, staurosporine, Ru-complex compound). The top scoring conformation of the inhibitor bound to CDK2 is not present among the top-scoring conformations of the inhibitor bound to either PIM1 or GSK-3ß and vice-versa. Collectively, our results help provide atomic-level insights into inhibitor selectivity among the three kinases.


Assuntos
Ciclina A/metabolismo , Quinase 2 Dependente de Ciclina/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Simulação de Acoplamento Molecular , Proteínas Proto-Oncogênicas c-pim-1/metabolismo , Rutênio/metabolismo , Trifosfato de Adenosina/metabolismo , Sítios de Ligação , Ciclina A/antagonistas & inibidores , Ciclina A/química , Quinase 2 Dependente de Ciclina/antagonistas & inibidores , Quinase 2 Dependente de Ciclina/química , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/química , Glicogênio Sintase Quinase 3 beta , Ligação de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Ligantes , Simulação de Dinâmica Molecular , Conformação Proteica , Proteínas Proto-Oncogênicas c-pim-1/química , Rutênio/química , Estereoisomerismo , Termodinâmica
15.
Cell Death Dis ; 3: e415, 2012 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-23096116

RESUMO

Inhibitors of the tyrosine kinase activity of epidermal growth factor receptor, as erlotinib, have an established role in treating several cancer types. However, resistance to erlotinib, particularly in breast cancer cell lines, and erlotinib treatment-associated disorders have also been described. Also, methods and combination therapies that could reverse resistance and ameliorate non-desirable effects represent a clinical challenge. Here, we show that the ATP non-competitive CDK2/cyclin A inhibitor NBI1 sensitizes erlotinib-resistant tumor cells to the combination treatment (co-treatment) for apoptosis-mediated cell death. Furthermore, in erlotinib-sensitive cells, the effective dose of erlotinib was lower in the presence of NBI1. The analysis in the breast cancer MDA-MB-468 erlotinib-resistant and in lung cancer A549 cell lines of the molecular mechanism underlying the apoptosis induced by co-treatment highlighted that the accumulation of DNA defects and depletion of cIAP and XIAP activates the ripoptosome that ultimately activates caspases-8 and -10 and apoptosis. This finding could have significant implications for future treatment strategies in clinical settings.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Caspase 8/metabolismo , Ciclina A/antagonistas & inibidores , Quinase 2 Dependente de Ciclina/antagonistas & inibidores , Resistencia a Medicamentos Antineoplásicos , Quinazolinas/farmacologia , Apoptose/efeitos dos fármacos , Caspase 8/genética , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Quimioterapia Combinada , Cloridrato de Erlotinib , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Neoplasias/genética , Neoplasias/fisiopatologia
16.
Mol Carcinog ; 51(6): 475-90, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21678498

RESUMO

Nimbolide, a plant-derived limonoid has been shown to exert its antiproliferative effects in various cell lines. We demonstrate that nimbolide effectively inhibited proliferation of WiDr colon cancer cells through inhibition of cyclin A leading to S phase arrest. It also caused activation of caspase-mediated apoptosis through the inhibition of ERK1/2 and activation of p38 and JNK1/2. Further nimbolide effectively retarded tumor cell migration and invasion through inhibition of metalloproteinase-2/9 (MMP-2/9) expression, both at the mRNA and protein level. It was also a strong inhibitor of VEGF expression, promoter activity, and in vitro angiogenesis. Finally, nimbolide suppressed the nuclear translocation of p65/p50 and DNA binding of NF-κB, which is an important transcription factor for controlling MMP-2/9 and VEGF gene expression.


Assuntos
Antineoplásicos/farmacologia , Neoplasias do Colo/genética , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Limoninas/farmacologia , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 9 da Matriz/genética , NF-kappa B/metabolismo , Caspases/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Neoplasias do Colo/enzimologia , Neoplasias do Colo/metabolismo , Ciclina A/antagonistas & inibidores , Ciclina D1/antagonistas & inibidores , Regulação para Baixo/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Limoninas/química , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Invasividade Neoplásica , Neovascularização Patológica , Fosforilação/efeitos dos fármacos , Regiões Promotoras Genéticas/efeitos dos fármacos , RNA Mensageiro/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
17.
Eur J Med Chem ; 46(12): 5825-32, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22000924

RESUMO

The design and synthesis of a small library of 4-aminopyrido[2,3-d]pyrimidine derivatives is reported. The potential activity of these compounds as CDK2/Cyclin A, CDK4/Cyclin D, EGFR and anti-tumor was evaluated by cytotoxicity studies in A431a, SNU638b, HCT116 and inhibition of CDK2-Cyclin A, CDK4/Cyclin D and EGFR enzyme activity in vitro. The anti-proliferative and CDK2-Cyclin A inhibitory activity of compounds 4c and 11a was significantly more active than roscovotine with IC(50) 0.3 and 0.09 µM respectively. Molecular modeling study, including fitting to a 3D-pharmacophore model, docking into cyclin dependant kinase2 (CDK2) active site and binding energy calculations were carried out and these studies suggested the same binding orientation inside the CDK2 binding pocket for these analogs compared to ATP.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Quinase 2 Dependente de Ciclina/antagonistas & inibidores , Pirimidinas/química , Pirimidinas/farmacologia , Antineoplásicos/síntese química , Sítios de Ligação , Linhagem Celular Tumoral , Ciclina A/antagonistas & inibidores , Ciclina A/metabolismo , Ciclina D/antagonistas & inibidores , Ciclina D/metabolismo , Quinase 2 Dependente de Ciclina/química , Quinase 2 Dependente de Ciclina/metabolismo , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Quinase 4 Dependente de Ciclina/metabolismo , Desenho de Fármacos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Humanos , Modelos Moleculares , Neoplasias/tratamento farmacológico , Pirimidinas/síntese química
18.
Org Biomol Chem ; 9(22): 7629-32, 2011 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-21863156
19.
Eur J Cell Biol ; 90(8): 657-64, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21514001

RESUMO

Fenofibrate, a peroxisome proliferator-activated receptor (PPAR) α agonist, is a hypolipidemic drug. Although several studies have explored the fenofibrate-induced antiproliferative effect in cultured human cells, it is not clear which role PPARα plays in this antiproliferative effect. Therefore, we investigated the antiproliferative mechanism of fenofibrate in Huh7 (human hepatoma cell line). Cell viability was measured by the WST-8 assay and cell proliferation was assessed using the BrdU incorporation assay. The cell cycle was analyzed by flow cytometry. The cyclins, tumor suppressor proteins and regulators of the AKT signaling pathway were analyzed by immunoblotting. Using flow cytometry, we showed that fenofibrate blocks entry into the S phase of the cell cycle. We certified that this G1 arrest is caused by the reduction of cyclin A and E2F1 and the accumulation of the cyclin-dependent kinase inhibitor p27. Interestingly, the antiproliferative effect of fenofibrate was not affected by the PPARα antagonist (GW6471) or by PPARα-specific siRNA. These results suggest that fenofibrate suppresses Huh7 cell growth through a PPARα independent mechanism. Furthermore, we showed that treatment of Huh7 cells with fenofibrate leads to suppression of AKT phosphorylation. We also found for the first time that fenofibrate increased the C-terminal modulator protein (CTMP), which inhibits AKT phosphorylation. Our data suggest that fenofibrate inhibits the proliferation of Huh7 cells by blocking Akt activation, and that CTMP is one of the key players for this antiproliferative property of fenofibrate in Huh7 cells.


Assuntos
Carcinoma Hepatocelular/tratamento farmacológico , Ciclina A/antagonistas & inibidores , Fenofibrato/farmacologia , Neoplasias Hepáticas/tratamento farmacológico , PPAR alfa/metabolismo , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Western Blotting , Carcinoma Hepatocelular/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células , Ciclina A/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/biossíntese , DNA Polimerase Dirigida por DNA/metabolismo , Fator de Transcrição E2F1/metabolismo , Citometria de Fluxo , Humanos , Immunoblotting , Neoplasias Hepáticas/metabolismo , Oxazóis/farmacologia , PPAR alfa/antagonistas & inibidores , PPAR alfa/genética , Fosforilação/efeitos dos fármacos , Interferência de RNA , RNA Interferente Pequeno , Transdução de Sinais/efeitos dos fármacos , Proteínas Supressoras de Tumor/análise , Tirosina/análogos & derivados , Tirosina/farmacologia
20.
Am J Physiol Renal Physiol ; 300(2): F393-402, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21123490

RESUMO

Mesangial cell (MC) proliferation is a key feature in the pathogenesis of a number of renal diseases. Peroxisome proliferator-activated receptor-γ (PPARγ) has attracted considerable attention for its effects on stimulating cell differentiation and on inducing cell cycle arrest. We previously showed that aldosterone (Aldo) stimulates MC proliferation via the phosphoinositide 3-kinase (PI3K)/Akt signaling pathway, which was dependent on reactive oxygen species (ROS)-mediated epithelial growth factor receptor (EGFR) transactivation (Huang S, Zhang A, Ding G, and Chen R. Am J Physiol Renal Physiol 296: F1323-F1333, 2009). In this study, we examined whether the PPARγ agonist rosiglitazone inhibited Aldo-induced MC proliferation by modulating ROS-dependent EGFR intracellular signaling. Rosiglitazone at 1-10 µM dose dependently inhibited Aldo-induced MC proliferation of cultured mouse MCs. The inhibitory effect was blocked by the PPARγ antagonist PD-68235, indicating that the rosiglitazone effect acted through PPARγ activation. Rosiglitazone also arrested Aldo-induced cell cycle progression and suppressed expression of cyclins D1 and A. Moreover, rosiglitazone dose dependently blocked Aldo-induced ROS production, EGFR phosphorylation, and PI3K/Akt activation. These results suggest that the PPARγ agonist rosiglitazone may inhibit Aldo-induced MC proliferation directly, by affecting ROS/EGFR/PI3K/Akt signaling pathways and cell cycle-regulatory proteins. PPARγ might be a novel therapeutic target against glomerular diseases.


Assuntos
Aldosterona/metabolismo , Proliferação de Células/efeitos dos fármacos , Receptores ErbB/metabolismo , Células Mesangiais/efeitos dos fármacos , PPAR gama/agonistas , Espécies Reativas de Oxigênio/metabolismo , Tiazolidinedionas/farmacologia , Animais , Células Cultivadas , Ciclina A/antagonistas & inibidores , Ciclina D1/antagonistas & inibidores , Células Mesangiais/citologia , Células Mesangiais/metabolismo , Camundongos , Antagonistas de Receptores de Mineralocorticoides/farmacologia , Nitrocompostos/farmacologia , PPAR gama/antagonistas & inibidores , PPAR gama/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Rosiglitazona , Transdução de Sinais/efeitos dos fármacos , Tiazóis/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...