Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Biochim Biophys Acta Mol Cell Res ; 1866(5): 945-956, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30826331

RESUMO

The maintenance of stem cells often requires the support of feeder cells. Primary mouse embryonic fibroblasts (MEFs) have traditionally been used as feeder cells, and although these MEF-derived feeder cells have exhibited a reasonable performance, they require repeated cell isolation, since MEFs cannot expand indefinitely. To overcome this limitation, immortalized cells, such as STO cells, have been used. However, one major disadvantage is that previously reported immortalized cells can only support stem cell cultures for a relatively short period, typically 4 to 7 days. In this study, we found that our newly established rat-derived fibroblasts immortalized by the expression of mutant cyclin-dependent kinase 4, cyclin D, and telomerase reverse transcriptase, can function as feeder cells for relatively long cell culture periods of approximately 14 days. The rat-derived immortalized cells developed in this study should be a useful source of feeder cells to support stem cell research.


Assuntos
Ciclina D/biossíntese , Quinase 4 Dependente de Ciclina/biossíntese , Células Alimentadoras/metabolismo , Fibroblastos/metabolismo , Mutação , Células-Tronco/metabolismo , Telomerase/biossíntese , Animais , Linhagem Celular Transformada , Técnicas de Cocultura , Ciclina D/genética , Quinase 4 Dependente de Ciclina/genética , Células Alimentadoras/citologia , Fibroblastos/citologia , Camundongos , Ratos , Células-Tronco/citologia , Telomerase/genética
2.
Pharmacology ; 102(1-2): 26-36, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29763912

RESUMO

BACKGROUND: Wnt signaling plays an essential role in tumor cell growth, including the development of malignant mesothelioma (MM). Epigenetic silencing of negative Wnt regulators leading to constitutive Wnt signaling has been observed in various cancers and warrants further attention. We have reported that a succinate ether derivative of α-tocotrienol (T3E) has potent cytotoxic effects in MM cells. Thus, in this study, we investigated whether the anti-MM effect of T3E could be mediated via the epigenetic alteration of the Wnt antagonist gene, Dickkopf-1 (DKK1). METHODS: WST-1 and cell analyzers were employed to analyze the effects of T3E on cell viability and apoptosis of human MM cell lines (H2452, H28). Real-time PCR and Western blot were performed to evaluate the expression at mRNA and protein levels. Methylation status and epigenetic modifications of DKK1's promoter regions after T3E treatment in MM cells were studied using methylation-specific PCR and Chromatin immunoprecipitation. Small interfering RNA-mediated knockdown -(siRNA), and specific inhibitors, were used to validate DKK1 as a target of T3E. RESULTS: T3E markedly impaired MM cell viability, increased the expression of phosphorylated-JNK and DKK1 and suppressed cyclin D, a downstream target gene of Wnt signaling. Knockdown of DKK1 expression by siRNA or a specific JNK inhibitor confirmed the contribution of DKK1 and JNK to T3E-induced cytotoxicity in MM cells. On the other hand, cytoskeleton-associated protein 4 (CKAP4) expression, which promotes cell proliferation as a Wnt-independent DKK1 receptor was inhibited by T3E. Silencing CKAP4 by -siRNA did not appear to directly affect MM cell viability, thereby indicating that expression of both DKK1 and CKAP4 is required. Furthermore, T3E-mediated inhibition of both DNA methyltransferases (DNMT1, 3A, and 3B) and histone deacetylases (HDAC1, 2, 3, and 8) in MM cells leads to increased DKK1 expression, thereby promoting tumor growth inhibition. MM cells treated with Zebularine (a DNMT inhibitor) and sodium butyrate (an HDAC inhibitor) exhibited cytotoxic effects, which may explain the inhibitory action of T3E on MM cells. In addition, an enhanced expression of DKK1 in MM cells following T3E treatment is positively correlated with the methylation status of its promoter; T3E decreased DNA methylation and increased histone acetylation. Moreover, T3E specifically increased histone H3 lysine 4 (H3K4) methylation activity, whereas no effects were observed on histone H3K9 and H3K27. CONCLUSIONS: Targeting the epigenetic induction of DKK1 may lead to effective treatment of MM, and T3E has great potential to induce anti-MM activity.


Assuntos
Epigênese Genética/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Neoplasias Pulmonares/genética , Mesotelioma/genética , Tocotrienóis/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ciclina D/biossíntese , DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , Metilação de DNA/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Proteínas de Membrana/biossíntese , Mesotelioma Maligno , RNA Interferente Pequeno/farmacologia
3.
PLoS One ; 11(8): e0156701, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27580131

RESUMO

Lung cancer is one of the most malignant tumors and the leading cause of cancer-related deaths worldwide. Among lung cancers, 40% are diagnosed as adenocarcinoma. Bromodomain containing 7 (BRD7) is a member of bromodomain-containing protein family. It was proved to be downregulated in various cancers. However, the role of BRD7 in lung adenocarcinoma is still unknown. Western blot and qRT-PCR was performed to measure the BRD7 expression in lung adenocarcinoma tissues and cells. CCK8 and migration assay was done to detect the functional role of BRD7 in lung adenocarcinoma. In this study, we showed that the expression of BRD7 was downregulated in lung adenocarcinoma tissues and cells. The lower of BRD7 levels in patients with lung adenocarcinoma was associated with shortened disease-free survival. Furthermore, overexpression of BRD7 inhibited lung adenocarcinoma cell proliferation and migration. Inhibition of BRD7 expression promoted cell proliferation and migration by activating ERK phosphorylation. Overexpression of BRD7 inhibited cyclin D and myc expression. Our findings are consistent with a tumor suppressor role for BRD7 in lung adenocarcinoma tumorigenesis.


Assuntos
Adenocarcinoma/metabolismo , Movimento Celular , Proteínas Cromossômicas não Histona/biossíntese , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas Supressoras de Tumor/biossíntese , Células A549 , Adenocarcinoma/genética , Adenocarcinoma/patologia , Proteínas Cromossômicas não Histona/genética , Ciclina D/biossíntese , Ciclina D/genética , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Masculino , Proteínas Supressoras de Tumor/genética
4.
J Pharm Pharmacol ; 68(7): 901-11, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27138740

RESUMO

OBJECTIVES: Our study aimed to evaluate the effect of cordycepin on human lung cancer cell lines A549 and NCI-H460. METHODS: Human lung cancer A549 cells and NCI-H460 cells were treated with different concentrations of cordycepin for different times. Cells incubated without cordycepin were defined as a control. The cell proliferation, migration and apoptosis were, respectively, determined by MTT assay, transwell migration assay and flow cytometry. Additionally, the expression levels of related proteins associated with cell cycle, epithelial-mesenchymal transition (EMT) and apoptosis were examined. KEY FINDINGS: The survival rate of A549 cells and NCI-H460 cells treated with cordycepin significantly decreased compared with untreated cells in a concentration-dependent manner, while the apoptosis rate increased. The migration number of cells treated with cordycepin significantly decreased as the increase in concentration. qRT-PCR and Western blot analysis showed that the aberrant expression of related molecules associated with cell cycle, migration and apoptosis was observed in the lung cancer cells, such as cyclin B, cyclin E, MMP-9, caspase-3 and Bcl-2. CONCLUSIONS: Cordycepin may exert inhibitory effects on the development of human lung cancer via inhibiting cell proliferation, suppressing migration and inducing apoptosis, suggesting that cordycepin may have therapeutic potential for the treatment of this disease.


Assuntos
Apoptose/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Desoxiadenosinas/farmacologia , Caderinas/biossíntese , Caspase 3/biossíntese , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ciclina B/biossíntese , Ciclina D/biossíntese , Ciclina E/biossíntese , Relação Dose-Resposta a Droga , Humanos , Metaloproteinase 9 da Matriz/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Fatores de Tempo , Vimentina/biossíntese
5.
BMC Cancer ; 16: 17, 2016 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-26762417

RESUMO

BACKGROUND: Silvestrol is a cyclopenta[b]benzofuran that was isolated from the fruits and twigs of Aglaia foveolata, a plant indigenous to Borneo in Southeast Asia. The purpose of the current study was to determine if inhibition of protein synthesis caused by silvestrol triggers autophagy and apoptosis in cultured human cancer cells derived from solid tumors. METHODS: In vitro cell viability, flow cytometry, fluorescence microscopy, qPCR and immunoblot was used to study the mechanism of action of silvestrol in MDA-MB-435 melanoma cells. RESULTS: By 24 h, a decrease in cyclin B and cyclin D expression was observed in silvestrol-treated cells relative to control. In addition, silvestrol blocked progression through the cell cycle at the G2-phase. In silvestrol-treated cells, DAPI staining of nuclear chromatin displayed nucleosomal fragments. Annexin V staining demonstrated an increase in apoptotic cells after silvestrol treatment. Silvestrol induced caspase-3 activation and apoptotic cell death in a time- and dose-dependent manner. Furthermore, both silvestrol and SAHA enhanced autophagosome formation in MDA-MB-435 cells. MDA-MB-435 cells responded to silvestrol treatment with accumulation of LC3-II and time-dependent p62 degradation. Bafilomycin A, an autophagy inhibitor, resulted in the accumulation of LC3 in cells treated with silvestrol. Silvestrol-mediated cell death was attenuated in ATG7-null mouse embryonic fibroblasts (MEFs) lacking a functional autophagy protein. CONCLUSIONS: Silvestrol potently inhibits cell growth and induces cell death in human melanoma cells through induction of early autophagy and caspase-mediated apoptosis. Silvestrol represents a natural product scaffold that exhibits potent cytotoxic activity and could be used for the further study of autophagy and its relationship to apoptosis in cancer cells.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Caspase 3/biossíntese , Melanoma/tratamento farmacológico , Triterpenos/administração & dosagem , Animais , Antineoplásicos Fitogênicos/administração & dosagem , Caspase 3/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ciclina B/biossíntese , Ciclina D/biossíntese , Regulação Neoplásica da Expressão Gênica , Humanos , Macrolídeos/administração & dosagem , Melanoma/genética , Melanoma/patologia , Camundongos
6.
In Vitro Cell Dev Biol Anim ; 52(1): 20-6, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26487427

RESUMO

The African savannah elephant (Loxodonta africana) is one of the critically endangered animals. Conservation of genetic and cellular resources is important for the promotion of wild life-related research. Although primary cultured cells are a useful model for the physiology and genomics of the wild-type animals, their distribution is restricted due to the limited number of cell divisions allowed in them. Here, we tried to immortalize a primary cell line of L. africana with by overexpressing human mutant form of cyclin-dependent kinase 4 (CDK4R24C), cyclin D, and telomerase (TERT). It has been shown before that the combination of human CDK4R24C, cyclin D, and TERT induces the efficient cellular immortalization of cells derived from humans, bovine, swine, and monkeys. Interestingly, although the combination of these three genes extended the cellular proliferation of the L. africana-derived cells, they did not induce cellular immortalization. This study suggest that control of cellular senescence in L. africana-derived cells would be different molecular mechanisms compared to those governing human, bovine, swine, and monkey cells.


Assuntos
Proteínas de Ciclo Celular/biossíntese , Proliferação de Células/genética , Senescência Celular/genética , Elefantes/genética , Cultura Primária de Células , Animais , Bovinos , Conservação dos Recursos Naturais , Ciclina D/biossíntese , Ciclina D/genética , Quinase 4 Dependente de Ciclina/genética , Espécies em Perigo de Extinção , Regulação da Expressão Gênica , Humanos , Telomerase/biossíntese , Telomerase/genética
7.
Oncol Rep ; 35(3): 1711-7, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26707131

RESUMO

In previous studies, we demonstrated that rhein lysinate (RHL), the salt of rhein and lysine that is easily dissolved in water, inhibited the growth of tumor cells derived from breast and ovarian cancer, hepatocellular carcinoma, cervical cancer and lung carcinoma. Based on these observations, human glioma U87 cells and a xenograft model in BALB/c nude mice were used to examine the antitumor activity of RHL against human glioma. Notably, RHL statistically significantly suppressed the growth of human glioma U87 xenografts in BALB/c nude mice. In vitro, there was a significant reduction in cell proliferation after treatment with RHL in a dose- and time-dependent manner. The overall growth inhibition was correlated with the increase in reactive oxygen species (ROS) production and cell apoptosis. The apoptosis- and cell cycle-related proteins including BAX and Bim were increased, whereas Bcl-2 and cyclin D were decreased in the RHL-treated cells. The results demonstrated that RHL is highly effective against the growth of human glioma U87 xenografts in BALB/c nude mice. The potent antitumor activity of RHL may be mediated through downregulation of Bcl-2 and cyclin D expression and upregulation of BAX and Bim expression.


Assuntos
Antraquinonas/administração & dosagem , Proteínas Reguladoras de Apoptose/biossíntese , Ciclina D/biossíntese , Glioma/tratamento farmacológico , Lisina/análogos & derivados , Proteínas de Membrana/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteínas Proto-Oncogênicas/biossíntese , Proteína X Associada a bcl-2/biossíntese , Animais , Antineoplásicos Fitogênicos/administração & dosagem , Apoptose/efeitos dos fármacos , Proteína 11 Semelhante a Bcl-2 , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioma/genética , Glioma/patologia , Humanos , Lisina/administração & dosagem , Camundongos , Espécies Reativas de Oxigênio , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Oncol Rep ; 34(3): 1549-56, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26151768

RESUMO

Purine analogue roscovitine, a cyclin-dependent kinase (CDK) inhibitor, has shown strong anti-proliferative and pro-apoptotic effects in solid and hematologic cancers such as non small-cell lung cancer and lymphomas. It targets CDK2, 7 and 9 preferentially, which are also overexpressed in glioblastoma. Τherefore, the biological effects of roscovitine in glioblastoma cell lines were investigated. Glioblastoma A172 and G28 cell lines were incubated with serial concentrations of roscovitine for 24-120 h. Proliferation was measured using the xCELLigence Real-Time Cell Analyzer, an impedance­based cell viability system. Cell cycle distribution was assessed by flow cytometry and gene expression was quantified by quantitative RT-PCR and western blot analysis. Roscovitine exhibited a clear dose-dependent anti­proliferative and pro­apoptotic effect in the A172 cell line, while G28 cells showed a anti-proliferative effect only at 100 µM. The results of the flow cytometric (FACS) analysis revealed a dose-dependent increase of the G2/M and sub-G1 fractions in A172 cells, while G28 cells responded with an elevated sub-G1 fraction only at the highest concentration. Roscovitine led to a dose­dependent decrease of transcripts of p53, CDK 7 and cyclins A and E and an increase of >4-fold of p21 in A172 cells. In G28 cells, a dose­dependent induction of CDK2, p21 and cyclin D was observed between 10 and 50 µM roscovitine after 72 h, however, at the highest concentration of 100 µM, all investigated genes were downregulated. Roscovitine exerted clear dose-dependent anti-proliferative and pro-apoptotic effects in A172 cells and less distinct effects on G28 cells. In A172 cells, roscovitine led to G2/M arrest and induced apoptosis, an effect accompanied by induced p21 and a reduced expression of CDK2, 7 and 9 and cyclins A and E. These effects requre further studies on a larger scale to confirm whether roscovitine can be used as a therapeutic agent against glioblastoma.


Assuntos
Ciclina D/biossíntese , Quinase 2 Dependente de Ciclina/biossíntese , Inibidor de Quinase Dependente de Ciclina p21/biossíntese , Glioblastoma/tratamento farmacológico , Purinas/administração & dosagem , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ciclina D/genética , Quinase 2 Dependente de Ciclina/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Proteínas de Neoplasias/biossíntese , Roscovitina
9.
Cell Cycle ; 14(13): 2129-41, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25928155

RESUMO

The role of the G1-phase Cyclin D-CDK 4/6 regulatory module in linking germline stem cell (GSC) proliferation to nutrition is evolutionarily variable. In invertebrate Drosophila and C. elegans GSC models, G1 is nearly absent and Cyclin E is expressed throughout the cell cycle, whereas vertebrate spermatogonial stem cells have a distinct G1 and Cyclin D1 plays an important role in GSC renewal. In the invertebrate, chordate, Oikopleura, where germline nuclei proliferate asynchronously in a syncytium, we show a distinct G1-phase in which 2 Cyclin D variants are co-expressed. Cyclin Dd, present in both somatic endocycling cells and the germline, localized to germline nuclei during G1 before declining at G1/S. Cyclin Db, restricted to the germline, remained cytoplasmic, co-localizing in foci with the Cyclin-dependent Kinase Inhibitor, CKIa. These foci showed a preferential spatial distribution adjacent to syncytial germline nuclei at G1/S. During nutrient-restricted growth arrest, upregulated CKIa accumulated in arrested somatic endoreduplicative nuclei but did not do so in germline nuclei. In the latter context, Cyclin Dd levels gradually decreased. In contrast, the Cyclin Dbß splice variant, lacking the Rb-interaction domain and phosphodegron, was specifically upregulated and the number of cytoplasmic foci containing this variant increased. This upregulation was dependent on stress response MAPK p38 signaling. We conclude that under favorable conditions, Cyclin Dbß-CDK6 sequesters CKIa in the cytoplasm to cooperate with Cyclin Dd-CDK6 in promoting germline nuclear proliferation. Under nutrient-restriction, this sequestration function is enhanced to permit continued, though reduced, cycling of the germline during somatic growth arrest.


Assuntos
Núcleo Celular/metabolismo , Proliferação de Células/fisiologia , Ciclina D/biossíntese , Variação Genética/fisiologia , Células Germinativas/metabolismo , Células Gigantes/metabolismo , Sequência de Aminoácidos , Animais , Núcleo Celular/genética , Cordados não Vertebrados , Ciclina D/genética , Regulação da Expressão Gênica , Dados de Sequência Molecular
10.
Oral Dis ; 20(1): 100-8, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23444918

RESUMO

OBJECTIVE: Porphyromonas gingivalis is a major pathogen in the development and progression of periodontal disease. The interactions or cross-talk between bacteria and gingival epithelial cells drive bacteria to manipulate the cell cycle to favor bacterial survival and virulence expression within the host. This study aims to dissect the effects of P. gingivalis on the cell cycle in human gingival epithelial cells. MATERIALS AND METHODS: We established a model of P. gingivalis invading IHGE cells. The cell cycle distribution of human gingival epithelial cells was analyzed by flow cytometry. Cyclin D and cyclin E mRNA and protein were detected by real-time PCR and Western blot, respectively. RESULTS: Porphyromonas gingivalis-induced facilitation of cell growth was correlated with the acceleration of G1 phase of cell cycle. Cyclin D1 mRNA levels were significantly upregulated from 6 to 12 h after infection. Cyclin E protein and mRNA levels were elevated at 10 and 12 h after invasion. CONCLUSIONS: We confirmed that P. gingivalis significantly enhances IHGE cell proliferation by promoting the G1/S transition, involving the up-regulation of cyclin D and cyclin E.


Assuntos
Ciclo Celular , Células Epiteliais/citologia , Células Epiteliais/microbiologia , Gengiva/citologia , Porphyromonas gingivalis , Proliferação de Células , Células Cultivadas , Ciclina D/biossíntese , Ciclina E/biossíntese , Células Epiteliais/metabolismo , Humanos , Regulação para Cima
11.
Int J Hematol ; 97(3): 313-23, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23456262

RESUMO

Multiple myeloma is divided into two distinct genetic subtypes based on chromosome content. Hyperdiploid myeloma is characterized by multiple trisomies of chromosomes 3, 5, 7, 9 11, 15, 19 and 21, and lacks recurrent immunoglobulin gene translocations. Non-hyperdiploid myeloma in contrast is characterized by chromosome translocations t(4;14), t(14;16), t(14;20), t(6;14) and t(11;14). A unifying event in the pathogenesis of multiple myeloma is the dysregulated expression of a cyclin D gene, either directly by juxtaposition to an immunoglobulin enhancer, as a result of ectopic expression of a MAF family transcription factor, or indirectly by as yet unidentified mechanisms. Secondary genetic events include rearrangements of MYC, activating mutations of NRAS, KRAS or BRAF, a promiscuous array of mutations that activate NFkB and deletions of 17p. Among the poor-risk genetic features are t(4;14), t(14;16), t(14;20), del 17p and gains of 1q. Available evidence supports the use of a risk-stratified approach to the treatment of patients with multiple myeloma, with the early and prolonged use of bortezomib particularly in patients with t(4;14) and del 17p.


Assuntos
Cromossomos Humanos , Mieloma Múltiplo , Translocação Genética , Trissomia , Animais , Antineoplásicos/uso terapêutico , Ácidos Borônicos/uso terapêutico , Bortezomib , Proliferação de Células , Cromossomos Humanos/genética , Cromossomos Humanos/metabolismo , Ciclina D/biossíntese , Ciclina D/genética , Intervalo Livre de Doença , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Imunoglobulina M/genética , Imunoglobulina M/metabolismo , Incidência , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/mortalidade , Mieloma Múltiplo/patologia , Proteínas Proto-Oncogênicas B-raf/biossíntese , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas c-myc/biossíntese , Proteínas Proto-Oncogênicas c-myc/genética , Pirazinas/uso terapêutico , Fatores de Risco , Fatores Sexuais , Hipermutação Somática de Imunoglobulina/genética , Taxa de Sobrevida , Estados Unidos/epidemiologia , Proteínas ras/biossíntese , Proteínas ras/genética
12.
Mol Cell Biochem ; 374(1-2): 173-80, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23229232

RESUMO

Active mutations of Notch1 play pivotal roles during leukemogenesis, but the downstream targets and molecular mechanisms of activated Notch1 signaling have not yet been fully clarified. In this study, we detected the overexpression of the high mobility group A1 (HMGA1) and activation of Notch1 signaling in mouse thymic lymphomas. A direct regulation of Notch1 on HMGA1 transcription was demonstrated and two Notch1/RBPJ cobinding sites of T/CTCCCACA were found in HMGA1 promoter regions. It was the first time demonstrated that HMGA1 was the downstream target of Notch1 signaling. Moreover, knockdown of HMGA1 resulted in significantly impaired cell growth and decreased expressions of cyclin D and cyclin E in human T leukemia cells. The formation of complexes was also observed between HMGA1 and retinoblastoma (RB) protein indicating a mechanism of cell cycle regulation. These findings suggest that activated HMGA1 regulates cell proliferation through the Notch1 signaling pathway, which represents an important molecular pathway leading to leukemogenesis.


Assuntos
Proliferação de Células , Proteína HMGA1a/metabolismo , Leucemia de Células T/metabolismo , Receptor Notch1/metabolismo , Animais , Sequência de Bases , Linhagem Celular Tumoral , Ciclina D/biossíntese , Ciclina E/biossíntese , Feminino , Regulação Neoplásica da Expressão Gênica , Proteína HMGA1a/genética , Humanos , Leucemia de Células T/induzido quimicamente , Leucemia de Células T/genética , Leucemia de Células T/patologia , Linfoma/induzido quimicamente , Metilnitrosoureia , Camundongos , Camundongos Endogâmicos C57BL , Complexos Multiproteicos , Interferência de RNA , RNA Interferente Pequeno , Receptor Notch1/genética , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo , Análise de Sequência de DNA , Transdução de Sinais/genética
13.
Mol Cell Endocrinol ; 371(1-2): 166-73, 2013 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-23219869

RESUMO

The Adrenocorticotropic hormone (ACTH) and Pro-opimelanocortin (POMC) 1-28N-terminal peptide (N-POMC(1-28)) have been shown to act as an adrenal mitogen in vivo. A possible role for cyclin E in the zona glomerulosa (ZG) proliferation, following ACTH and/or N-POMC(1-28) administration, has been previously demonstrated. In this study, we investigated the effect of ACTH and N-POMC(1-28) on the expression of adrenal cortex proteins related to cell cycle control such as cyclins D and P27(kip1). The administration of N-POMC upregulated cyclin D1 and D2 expression in the outer zone of the adrenal cortex; cyclin D3 expression was upregulated in the cortex inner zone even after administration of ACTH. Both ACTH and N-POMC peptides induced a decrease in the P27(kip1) expression in the ZG. These novel findings suggest that the POMC-derivate peptides, ACTH and N-POMC, promote proliferation in the adrenal cortex by upregulating the D2 and D3 cyclins and downregulating the P27(kip1) expression.


Assuntos
Córtex Suprarrenal/metabolismo , Hormônio Adrenocorticotrópico/farmacologia , Ciclina D/biossíntese , Inibidor de Quinase Dependente de Ciclina p27/biossíntese , Fragmentos de Peptídeos/farmacologia , Pró-Opiomelanocortina/farmacologia , Córtex Suprarrenal/efeitos dos fármacos , Hormônio Adrenocorticotrópico/metabolismo , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Pontos de Checagem do Ciclo Celular , Proteínas de Ciclo Celular/biossíntese , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Ciclina D1/biossíntese , Ciclina D2/biossíntese , Ciclina D3/biossíntese , Regulação para Baixo , Masculino , Fragmentos de Peptídeos/metabolismo , Pró-Opiomelanocortina/metabolismo , Ratos , Ratos Sprague-Dawley , Regulação para Cima , Zona Glomerulosa/metabolismo
14.
Oncol Rep ; 28(5): 1889-93, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22948717

RESUMO

The constitutive activation of the Src family kinases (SFKs) has been established as a poor prognostic factor in malignant mesothelioma (MM), however, the family member(s) which contribute to the malignancy have not been defined. This study aimed to identify the SFK member(s) contributing to cell growth using RNA interference in various MM cell lines. Silencing of Yes but not of c-Src or Fyn in MM cells leads to cell growth suppression. This suppressive effect caused by Yes silencing mainly depends on G1 cell cycle arrest and partly the induction of apoptosis. Also, the knockout of Yes induces the inactivation of ß-catenin signaling and subsequently decreases the levels of cyclin D necessary for G1-S transition in the cell cycle. In addition, Yes knockout has less effect on cell growth suppression in ß-catenin-deficient H28 MM cells compared to other MM cells which express the catenin. Overall, we conclude that Yes is a central mediator for MM cell growth that is not shared with other SFKs such as c-Src.


Assuntos
Mesotelioma/metabolismo , Mesotelioma/patologia , Proteínas Proto-Oncogênicas c-yes/metabolismo , Apoptose/genética , Divisão Celular , Linhagem Celular Tumoral , Proliferação de Células , Ciclina D/biossíntese , Pontos de Checagem da Fase G1 do Ciclo Celular , Humanos , Mesotelioma/genética , Proteínas Proto-Oncogênicas c-fyn/genética , Proteínas Proto-Oncogênicas c-yes/genética , Interferência de RNA , RNA Interferente Pequeno , beta Catenina/metabolismo , Quinases da Família src/genética
15.
Mol Cell Biol ; 32(15): 3121-31, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22645312

RESUMO

Constitutive NF-κB activation by proinflammatory cytokines plays a major role in cancer progression. However, the underlying mechanism is still unclear. We report here that histone methyltransferase NSD2 (also known as MMSET or WHSC1), a target of bromodomain protein ANCCA/ATAD2, acts as a strong coactivator of NF-κB by directly interacting with NF-κB for activation of target genes, including those for interleukin-6 (IL-6), IL-8, vascular endothelial growth factor A (VEGFA), cyclin D, Bcl-2, and survivin, in castration-resistant prostate cancer (CRPC) cells. NSD2 is recruited to the target gene promoters upon induction and mediates NF-κB activation-associated elevation of histone H3K36me2 and H3K36me3 marks at the promoter, which involves its methylase activity. Interestingly, we found that NSD2 is also critical for cytokine-induced recruitment of NF-κB and acetyltransferase p300 and histone hyperacetylation. Importantly, NSD2 is overexpressed in prostate cancer tumors, and its overexpression correlates with NF-κB activation. Furthermore, NSD2 expression is strongly induced by tumor necrosis factor alpha (TNF-α) and IL-6 via NF-κB and plays a crucial role in tumor growth. These results identify NSD2 to be a key chromatin regulator of NF-κB and mediator of the cytokine autocrine loop for constitutive NF-κB activation and emphasize the important roles played by NSD2 in cancer cell proliferation and survival and tumor growth.


Assuntos
Cromatina/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , NF-kappa B/metabolismo , Proteínas Repressoras/metabolismo , Acetilação , Animais , Comunicação Autócrina , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Ciclina D/biossíntese , Ciclina D/metabolismo , Regulação Neoplásica da Expressão Gênica , Histona-Lisina N-Metiltransferase/biossíntese , Histonas/biossíntese , Histonas/metabolismo , Humanos , Proteínas Inibidoras de Apoptose/biossíntese , Proteínas Inibidoras de Apoptose/metabolismo , Interleucina-6/biossíntese , Interleucina-6/metabolismo , Interleucina-8/biossíntese , Interleucina-8/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Regiões Promotoras Genéticas , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Proteínas Repressoras/biossíntese , Transdução de Sinais , Survivina , Transplante Heterólogo , Fator de Necrose Tumoral alfa/metabolismo , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fatores de Transcrição de p300-CBP/metabolismo
16.
J Immunol ; 187(4): 1733-44, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21742971

RESUMO

The interaction of cancer within a microenvironment is an important factor determining cancer development. This study analyzed the soluble factors secreted by tumor-associated dendritic cells (TADCs), which are responsible for increasing lung cancer growth, migration, invasion, and epithelial-to-mesenchymal transition. Addition of amphiregulin, present in large amounts in TADC-conditioned medium (CM), mimicked the inductive effect of TADC-CM on lung cancer progression, supported by the enhancement of cell proliferation, migration, and invasion as well as osteolytic bone metastases phenotypes. In contrast, neutralization of amphiregulin from TADC-CM decreased the advanced malignancy-inductive properties of TADC-CM. Significant upregulation of amphiregulin has been seen in tumor-infiltrating CD11c(+) DCs in human lung cancer samples and patients' sera. The enhancement of amphiregulin in TADCs has also been noted in mice transplanted with lung cancer cells. Induction of lung cancer progression by TADC-derived amphiregulin is associated with increased STAT3 and AKT activation, which subsequently increases the expression of cyclin D, Twist, and Snail. Blocking AKT significantly decreases TADC-CM and amphiregulin-mediated migration by decreasing the upregulation of Snail, whereas inhibition of STAT3 reduced the modulation of TADC-derived amphiregulin on Twist and cyclin D expression, suggesting that cooperation of STAT3 and AKT plays a critical role in TADC-mediated cancer progression. Moreover, mice treated with anti-amphiregulin Abs showed decreased incidence of cancer development and increased survival rates. Our study suggests that inhibition of amphiregulin or amphiregulin-related signaling is an attractive therapeutic target in lung cancer patients.


Assuntos
Carcinoma Pulmonar de Lewis/imunologia , Movimento Celular , Proliferação de Células , Células Dendríticas/imunologia , Glicoproteínas/imunologia , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Neoplasias Pulmonares/imunologia , Anfirregulina , Animais , Neoplasias Ósseas/imunologia , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/secundário , Neoplasias Ósseas/terapia , Antígeno CD11c/imunologia , Antígeno CD11c/metabolismo , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patologia , Carcinoma Pulmonar de Lewis/terapia , Linhagem Celular Tumoral , Ciclina D/biossíntese , Ciclina D/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Família de Proteínas EGF , Regulação Neoplásica da Expressão Gênica/imunologia , Glicoproteínas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/terapia , Camundongos , Invasividade Neoplásica , Metástase Neoplásica , Transplante de Neoplasias , Proteínas Nucleares/biossíntese , Proteínas Nucleares/imunologia , Proteínas Proto-Oncogênicas c-akt/imunologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Transcrição STAT3/imunologia , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/imunologia , Proteína 1 Relacionada a Twist/biossíntese , Proteína 1 Relacionada a Twist/imunologia
17.
J Cell Mol Med ; 15(2): 296-306, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19912438

RESUMO

Our previous works revealed that human ribosomal protein S13 (RPS13) was up-regulated in multidrug-resistant gastric cancer cells and overexpression of RPS13 could protect gastric cancer cells from drug-induced apoptosis. The present study was designed to explore the role of RPS13 in tumorigenesis and development of gastric cancer. The expression of RPS13 in gastric cancer tissues and normal gastric mucosa was evaluated by immunohistochemical staining and Western blot analysis. It was found RPS13 was expressed at a higher level in gastric cancer tissues than that in normal gastric mucosa. RPS13 was then genetically overexpressed in gastric cancer cells or knocked down by RNA interference. It was demonstrated that up-regulation of RPS13 accelerated the growth, enhanced in vitro colony forming and soft agar cologenic ability and promoted in vivo tumour formation potential of gastric cancer cells. Meanwhile, down-regulation of RPS13 in gastric cancer cells resulted in complete opposite effects. Moreover, overexpression of RPS13 could promote G1 to S phase transition whereas knocking down of RPS13 led to G1 arrest of gastric cancer cells. It was further demonstrated that RPS13 down-regulated p27(kip1) expression and CDK2 kinase activity but did not change the expression of cyclin D, cyclin E, CDK2, CDK4 and p16(INK4A). Taken together, these data indicate that RPS13 could promote the growth and cell cycle progression of gastric cancer cells at least through inhibiting p27(kip1) expression.


Assuntos
Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Proteínas Ribossômicas/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Animais , Apoptose , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Ciclina D/biossíntese , Ciclina E/biossíntese , Quinase 2 Dependente de Ciclina/biossíntese , Quinase 2 Dependente de Ciclina/metabolismo , Quinase 4 Dependente de Ciclina/biossíntese , Inibidor p16 de Quinase Dependente de Ciclina/biossíntese , Inibidor de Quinase Dependente de Ciclina p27/biossíntese , Inibidor de Quinase Dependente de Ciclina p27/genética , Regulação para Baixo , Pontos de Checagem da Fase G1 do Ciclo Celular , Mucosa Gástrica/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Interferência de RNA , Proteínas Ribossômicas/biossíntese
18.
Mol Cancer Res ; 8(10): 1375-87, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20736297

RESUMO

Smad3, a component of the transforming growth factor ß signaling cascade, contributes to G(1) arrest in breast cancer cells. Cyclin D1/cyclin-dependent kinase 4 (CDK4) promotes G(1)-S-phase transition, and CDK phosphorylation of Smad3 has been associated with inhibition of Smad3 activity. We hypothesized that overexpression of cyclin D1 exerts tumorigenic effects in breast cancer cells through CDK4-mediated phosphorylation and inhibition of Smad3 and release of G(1) arrest. Real-time quantitative reverse transcription-PCR and immunoblotting were used to evaluate expression of study proteins in cyclin D1-overexpressing breast cancer cells. Smad3 transcriptional activity and cell cycle control were examined in cells transfected with wild-type (WT) Smad3 or Smad3 with single or multiple CDK phosphorylation site mutations (M) in the presence or absence of the CDK4 inhibitor or cotransfection with cdk4 small interfering RNA (siRNA). Transfection of the Smad3 5M construct resulted in decreased c-myc and higher p15(INK4B) expression. Compared with WT Smad3, overexpression of the Smad3 T8, T178, 4M, or 5M mutant constructs resulted in higher Smad3 transcriptional activity. Compared with cells transfected with WT Smad3, Smad3 transcriptional activity was higher in cells overexpressing Smad3 mutant constructs and treated with the CDK4 inhibitor or transfected with cdk4 siRNA. Cells transfected with Smad3 T8 or T178 and treated with the CDK4 inhibitor showed an increase in the G(1) cell population. Inhibition of CDK-mediated Smad3 phosphorylation released cyclin D1-regulated blockade of Smad3 transcriptional activity and recovered cell cycle arrest in breast cancer cells. Targeted inhibition of CDK4 activity may have a role in the treatment of cyclin D-overexpressing breast cancers.


Assuntos
Neoplasias da Mama/enzimologia , Ciclina D/biossíntese , Quinase 4 Dependente de Ciclina/fisiologia , Proteína Smad3/antagonistas & inibidores , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Ciclo Celular/genética , Linhagem Celular Tumoral , Ciclina D/genética , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Quinase 4 Dependente de Ciclina/genética , Ativação Enzimática/genética , Feminino , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Fosforilação/genética , Proteína Smad3/genética , Proteína Smad3/metabolismo
19.
Anticancer Drugs ; 21(1): 53-64, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19829098

RESUMO

In our laboratory, a novel therapeutic probe, T11TS, a membrane glycoprotein, was isolated which had antineoplastic activity against experimental glioma. Development of a novel therapeutic strategy with T11TS has unearthed a newer dimension of its mechanism of action: modulation of the cell cycle. In this study, we have presented evidence to support the finding that T11TS induces G1 cell cycle arrest of rat glioma cells. Results of flow cytometric studies showed that the treatment produced a marked increase in the proportion of cells in the G1 phase. Flow cytometry, immunoblotting, immunoprecipitation, and kinase assays were performed for investigating the involvement of G1 cell cycle regulators. T11TS induces downregulation of the cyclin-D (1 and 3) expression with the concurrent upregulation of p21 and p27 and their concomitant association with cyclin-dependent kinase 4, proliferating cell nuclear antigen and cyclin E respectively leading to a decrease in cyclin-dependent kinase 4 kinase activity. A transient rise in retinoblastoma protein level and coordinated binding of retinoblastoma protein with E2F coincided with the accumulation of cells in G1 phase. Thus, our observations have uncovered an antiproliferative pathway for T11TS, causing retardation of glioma cell cycle.


Assuntos
Antineoplásicos/farmacologia , Ciclina D/biossíntese , Inibidor de Quinase Dependente de Ciclina p21/biossíntese , Inibidor de Quinase Dependente de Ciclina p27/biossíntese , Fase G1/efeitos dos fármacos , Glicoproteínas/farmacologia , Animais , Animais Recém-Nascidos , Antineoplásicos/isolamento & purificação , Neoplasias Encefálicas/induzido quimicamente , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Ciclina D1/biossíntese , Ciclina D3/biossíntese , Regulação para Baixo , Membrana Eritrocítica/química , Etilnitrosoureia , Feminino , Citometria de Fluxo , Glioma/induzido quimicamente , Glioma/metabolismo , Glioma/patologia , Glicoproteínas/isolamento & purificação , Masculino , Ratos , Ratos Endogâmicos , Ovinos , Células Tumorais Cultivadas , Regulação para Cima
20.
Zhonghua Shao Shang Za Zhi ; 19(4): 209-12, 2003 Aug.
Artigo em Chinês | MEDLINE | ID: mdl-14514399

RESUMO

OBJECTIVE: To explore the influence of glucagon-like peptide-2 (GLP-2) on the proliferation of the intestinal mucosal cells in scalded rats. METHODS: Fifty-five Wistar rats were employed in the study and were randomly divided into normal control (C), simple scald (S) and scald with GLP-2 treatment (G) groups. The rats in G group received GLP-2 introperitoneally in a dose of 200 micro g/kg two times a day. The rats in S and G groups were sacrificed at 6 postburn hours (PBHs), 12 PBHs, 1 postburn day (PBD1), PBD3 and PBD5 and the rats in C group were also sacrificed. Plasma diamine oxidase (DAO) activity, cell cycle protein cyclin D expression and the proliferating cell nuclear antigen (PCNA) in all groups were determined. And the histological change in the intestinal mucosal tissue was observed simultaneously. with all the above determinations. RESULTS: Compared with those in C group, the PCNA expression at 6 and 12 PBHs in S group was enhanced slightly and weakened at PBD1, reaching the lowest level at PBD3 and it was still lower than that in C group at PBD5. Changes in PCNA in G group were similar to that in S group, except that the expression at PBD3 and PBD5 was stronger than that in S group. The intestinal mucosal cyclin D protein expression was increased at 6 and 12 PBHs in S group, but decreased by 40% before injury at PBD1. Nevertheless, the cyclin D protein expression in G group was much higher than that in S group at PBD1, PBD3 and PBD5. The plasma DAO activity increased significantly in rats after burn injury. But the activity decreased obviously after GLP-2 treatment for 5 days (P < 0.01). It was observed histologically in G group that the lining of Exogenous intestinal villi was regular and well arranged without evident epithelial exfoliation. CONCLUSION: Exogenous GLP-2 might ameliorate intestinal mucosal injury in scalded rats, and promotion of the expression of PCNA and cyclin D, resulting in proliferation of injured intestinal mucosal cells, might be the underlying mechanisms.


Assuntos
Queimaduras/patologia , Proliferação de Células/efeitos dos fármacos , Peptídeo 2 Semelhante ao Glucagon/farmacologia , Mucosa Intestinal/patologia , Animais , Queimaduras/metabolismo , Ciclina D/biossíntese , Feminino , Mucosa Intestinal/metabolismo , Masculino , Antígeno Nuclear de Célula em Proliferação/biossíntese , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA