Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Oncol Rep ; 45(1): 390-400, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33200225

RESUMO

The present study aimed to analyze the compensatory signaling pathways induced by forkhead domain inhibitor­6 (FDI­6), which is a forkhead box protein M1 (FOXM1) inhibitor, in ovarian cancer cells and evaluate the effectiveness of simultaneous inhibition of FOXM1 and the compensatory signaling pathway in decreasing the survival of ovarian cancer cells. The present study identified the proteins involved in the compensatory mechanism activated by FDI­6 in HeyA8 ovarian cancer cells using western blot analysis and a reverse­phase protein array. In addition, a cell viability assay was performed to determine the effects of FDI­6 and the compensatory signaling pathway on cancer cell viability. All experiments were performed in three­dimensional cell cultures. The present study observed that FDI­6 stimulated the upregulation of N­Ras, phosphoprotein kinase Cδ (p­PKCδ) (S664) and HER3 in HeyA8 cells. Tipifarnib as an N­Ras inhibitor, rottlerin as a p­PKCδ (S664) inhibitor and sapitinib as a HER3 inhibitor were selected. The combination of FDI­6 with tipifarnib attenuated the upregulation of N­Ras induced by FDI­6 and the combination of FDI­6 with sapitinib also attenuated HER3 downstream signaling pathway in HeyA8 cells, as shown by on western blot analysis. Rottlerin downregulated p­PKCδ (S664) by inhibiting the activity of a Src­related tyrosine kinase that transfers a phosphate group to PKCδ. Compared with FDI­6 alone, the addition of tipifarnib or rottlerin to FDI­6 was significantly more effective in reducing the growth of HeyA8 cells. However, the combination of FDI­6 and sapitinib did not induce a significant decrease in survival of HeyA8 cells. In conclusion, the addition of tipifarnib or rottlerin to inhibit N­Ras or p­PKCδ (S664), respectively, inhibited the compensatory signaling pathway response induced by FDI­6 in HeyA8 cells. These inhibitors increased the efficacy of FDI­6, which inhibits FOXM1, in reducing ovarian cancer cell viability.


Assuntos
Proteína Forkhead Box M1/antagonistas & inibidores , Neoplasias Ovarianas/tratamento farmacológico , Piridinas/farmacologia , Tiofenos/farmacologia , Acetofenonas/farmacologia , Benzopiranos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Feminino , Proteína Forkhead Box M1/análise , GTP Fosfo-Hidrolases/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Neoplasias Ovarianas/patologia , Quinazolinas/farmacologia , Quinolonas/farmacologia , Transdução de Sinais/efeitos dos fármacos
2.
Cell Mol Gastroenterol Hepatol ; 9(3): 425-446, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31669262

RESUMO

BACKGROUND & AIMS: Liver inflammation has been recognized as a hallmark of hepatocarcinogenesis. Although Forkhead Box M1 (FoxM1) is a well-defined oncogenic transcription factor that is overexpressed in hepatocellular carcinoma (HCC), its role in liver inflammation has never been explored. METHODS: We generated hepatocyte-specific FoxM1 conditional transgenic (TG) mice by using the Cre-loxP and Tetracycline (Tet)-on systems to induce FoxM1 expression in a hepatocyte-specific and time-dependent manner. RESULTS: After treatment of Tet-derivatives doxycycline (DOX) to induce FoxM1, TG mice exhibited spontaneous development of hepatocyte death with elevated serum alanine aminotransferase levels and hepatic infiltration of macrophages. The removal of DOX in TG mice completely removed this effect, suggesting that spontaneous inflammation in TG mice occurs in a hepatocyte FoxM1-dependent manner. In addition, liver inflammation in TG mice was associated with increased levels of hepatic and serum chemokine (C-C motif) ligand 2 (CCL2). In vitro transcriptional analysis confirmed that CCL2 is a direct target of FoxM1 in murine hepatocytes. After receiving FoxM1 induction since birth, all TG mice exhibited spontaneous HCC with liver fibrosis at 12 months of age. Hepatic expression of FoxM1 was significantly increased in liver injury models. Finally, pharmacologic inhibition of FoxM1 reduced liver inflammation in models of liver injury. CONCLUSIONS: Hepatocyte FoxM1 acts as a crucial regulator to orchestrate liver inflammation linking to hepatocarcinogenesis. Thus, hepatocyte FoxM1 may be a potential target not only for the treatment of liver injury but also for the prevention toward HCC.


Assuntos
Carcinogênese/imunologia , Proteína Forkhead Box M1/metabolismo , Hepatite/patologia , Neoplasias Hepáticas/patologia , Animais , Apoptose/imunologia , Tetracloreto de Carbono/administração & dosagem , Tetracloreto de Carbono/toxicidade , Células Cultivadas , Quimiocina CCL2/metabolismo , Modelos Animais de Doenças , Proteína Forkhead Box M1/análise , Proteína Forkhead Box M1/genética , Hepatectomia , Hepatite/diagnóstico , Hepatite/imunologia , Hepatócitos/imunologia , Hepatócitos/patologia , Humanos , Fígado/imunologia , Fígado/patologia , Fígado/cirurgia , Cirrose Hepática Experimental/induzido quimicamente , Cirrose Hepática Experimental/imunologia , Cirrose Hepática Experimental/patologia , Testes de Função Hepática , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/cirurgia , Camundongos , Camundongos Transgênicos , Cultura Primária de Células
3.
Medicina (Kaunas) ; 56(1)2019 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-31861475

RESUMO

Background and Objectives: Intrahepatic cholangiocarcinoma (iCCA) is a pernicious tumor characterized by a dismal outcome and scarce therapeutic options. To substantially improve the prognosis of iCCA patients, a better understanding of the molecular mechanisms responsible for development and progression of this disease is imperative. In the present study, we aimed at elucidating the role of the maternal embryonic leucine zipper kinase (MELK) protooncogene in iCCA. Materials and Methods: We analyzed the expression of MELK and two putative targets, Forkhead Box M1 (FOXM1) and Enhancer of Zeste Homolog 2 (EZH2), in a collection of human iCCA by real-time RT-PCR and immunohistochemistry (IHC). The effects on iCCA growth of both the multi-kinase inhibitor OTSSP167 and specific small-interfering RNA (siRNA) against MELK were investigated in iCCA cell lines. Results: Expression of MELK was significantly higher in tumors than in corresponding non-neoplastic liver counterparts, with highest levels of MELK being associated with patients' shorter survival length. In vitro, OTSSP167 suppressed the growth of iCCA cell lines in a dose-dependent manner by reducing proliferation and inducing apoptosis. These effects were amplified when OTSSP167 administration was coupled to the DNA-damaging agent doxorubicin. Similar results, but less remarkable, were obtained when MELK was silenced by specific siRNA in the same cells. At the molecular level, siRNA against MELK triggered downregulation of MELK and its targets. Finally, we found that MELK is a downstream target of the E2F1 transcription factor. Conclusion: Our results indicate that MELK is ubiquitously overexpressed in iCCA, where it may represent a prognostic indicator and a therapeutic target. In particular, the combination of OTSSP167 (or other, more specific MELK inhibitors) with DNA-damaging agents might be a potentially effective therapy for human iCCA.


Assuntos
Proliferação de Células/efeitos dos fármacos , Colangiocarcinoma/tratamento farmacológico , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Idoso , Animais , Colangiocarcinoma/genética , Colangiocarcinoma/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste/análise , Feminino , Proteína Forkhead Box M1/análise , Humanos , Imunoquímica/métodos , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Pessoa de Meia-Idade , Proteínas Serina-Treonina Quinases/análise , Coelhos , Reação em Cadeia da Polimerase em Tempo Real/métodos
4.
J Microbiol Biotechnol ; 29(8): 1281-1287, 2019 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-31370114

RESUMO

Nattokinase (NK, E.C. 3.4.21.62) is a serine protease produced by Bacillus subtilis natto that shows promise for the treatment of thrombotic disease. In this study, we assessed the effects of NK on the development of hepatocellular carcinoma (HCC), a principal malignancy of the liver that causes morbidity and mortality worldwide. Crude extracts of NK (NCE) were isolated from fermentation medium by centrifugation and separated into three fractions (<10 K, 100~30 K and >30K). Orthotopic HCC mouse models were established and NCE was administered by oral gavage. H&E staining was performed to examine the pathology of HCC livers. Immunohistochemistry and immunofluorescence were used to evaluate FOXM1, CD31, CD44 and vimentin expression in the liver. Compared to PBS groups, NCE increased the survival rates of HCC-bearing mice to 31% and decreased ascites. Low-intensity ultrasound imaging showed that the hypoechoic mass area was lower in NCE-treated mice and that tumor growth significantly decreased. IHC staining showed that the expression of FOXM1 was inhibited by NCE treatment. Immunofluorescence results revealed lower levels of CD31, CD44 and vimentin in the NCE groups. Taken together, these data demonstrate that NCE from Bacillus subtilis natto improves survival and inhibits tumor growth in HCC mice.


Assuntos
Carcinoma Hepatocelular/tratamento farmacológico , Misturas Complexas/farmacologia , Neoplasias Hepáticas/tratamento farmacológico , Subtilisinas/farmacologia , Animais , Bacillus subtilis/enzimologia , Carcinoma Hepatocelular/patologia , Misturas Complexas/isolamento & purificação , Modelos Animais de Doenças , Fermentação , Fibrinolíticos/farmacologia , Proteína Forkhead Box M1/análise , Receptores de Hialuronatos/análise , Fígado/metabolismo , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Endogâmicos C57BL , Molécula-1 de Adesão Celular Endotelial a Plaquetas/análise , Subtilisinas/isolamento & purificação , Vimentina/análise
5.
Braz J Med Biol Res ; 52(4): e7728, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30994730

RESUMO

Pituitary adenoma is one of the most common tumors in the neuroendocrine system. This study investigated the effects of long non-coding RNAs (lncRNAs) highly up-regulated in liver cancer (HULC) on rat secreting pituitary adenoma GH3 cell viability, migration, invasion, apoptosis, and hormone secretion, as well as the underlying potential mechanisms. Cell transfection and qRT-PCR were used to change and measure the expression levels of HULC, miR-130b, and FOXM1. Cell viability, migration, invasion, and apoptosis were assessed using trypan blue staining assay, MTT assay, two-chamber transwell assay, Guava Nexin assay, and western blotting. The concentrations of prolactin (PRL) and growth hormone (GH) in culture supernatant of GH3 cells were assessed using ELISA. The targeting relationship between miR-130b and FOXM1 was verified using dual luciferase activity. Finally, the expression levels of key factors involved in PI3K/AKT/mTOR and JAK1/STAT3 pathways were evaluated using western blotting. We found that HULC was highly expressed in GH3 cells. Overexpression of HULC promoted GH3 cell viability, migration, invasion, PRL and GH secretion, as well as activated PI3K/AKT/mTOR and JAK1/STAT3 pathways. Knockdown of HULC had opposite effects and induced cell apoptosis. HULC negatively regulated the expression of miR-130b, and miR-130b participated in the effects of HULC on GH3 cells. FOXM1 was a target gene of miR-130b, which was involved in the regulation of GH3 cell viability, migration, invasion, and apoptosis, as well as PI3K/AKT/mTOR and JAK1/STAT3 pathways. In conclusion, HULC tumor-promoting roles in secreting pituitary adenoma might be via down-regulating miR-130b, up-regulating FOXM1, and activating PI3K/AKT/mTOR and JAK1/STAT3 pathways.


Assuntos
Adenoma/patologia , Neoplasias Hipofisárias/patologia , RNA Longo não Codificante/fisiologia , Adenoma/genética , Adenoma/metabolismo , Animais , Apoptose/fisiologia , Western Blotting , Linhagem Celular Tumoral , Ensaios de Migração Celular , Movimento Celular/fisiologia , Sobrevivência Celular/fisiologia , Ensaio de Imunoadsorção Enzimática , Proteína Forkhead Box M1/análise , Proteína Forkhead Box M1/metabolismo , Humanos , Janus Quinase 1/análise , Janus Quinase 1/metabolismo , Luciferases , MicroRNAs/análise , MicroRNAs/metabolismo , Fosfatidilinositol 3-Quinases/análise , Fosfatidilinositol 3-Quinases/metabolismo , Neoplasias Hipofisárias/genética , Neoplasias Hipofisárias/metabolismo , Proteínas Proto-Oncogênicas c-akt/análise , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Longo não Codificante/análise , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT3/análise , Fator de Transcrição STAT3/metabolismo , Serina-Treonina Quinases TOR/análise , Serina-Treonina Quinases TOR/metabolismo , Transfecção
6.
Braz. j. med. biol. res ; 52(4): e7728, 2019. graf
Artigo em Inglês | LILACS | ID: biblio-1001506

RESUMO

Pituitary adenoma is one of the most common tumors in the neuroendocrine system. This study investigated the effects of long non-coding RNAs (lncRNAs) highly up-regulated in liver cancer (HULC) on rat secreting pituitary adenoma GH3 cell viability, migration, invasion, apoptosis, and hormone secretion, as well as the underlying potential mechanisms. Cell transfection and qRT-PCR were used to change and measure the expression levels of HULC, miR-130b, and FOXM1. Cell viability, migration, invasion, and apoptosis were assessed using trypan blue staining assay, MTT assay, two-chamber transwell assay, Guava Nexin assay, and western blotting. The concentrations of prolactin (PRL) and growth hormone (GH) in culture supernatant of GH3 cells were assessed using ELISA. The targeting relationship between miR-130b and FOXM1 was verified using dual luciferase activity. Finally, the expression levels of key factors involved in PI3K/AKT/mTOR and JAK1/STAT3 pathways were evaluated using western blotting. We found that HULC was highly expressed in GH3 cells. Overexpression of HULC promoted GH3 cell viability, migration, invasion, PRL and GH secretion, as well as activated PI3K/AKT/mTOR and JAK1/STAT3 pathways. Knockdown of HULC had opposite effects and induced cell apoptosis. HULC negatively regulated the expression of miR-130b, and miR-130b participated in the effects of HULC on GH3 cells. FOXM1 was a target gene of miR-130b, which was involved in the regulation of GH3 cell viability, migration, invasion, and apoptosis, as well as PI3K/AKT/mTOR and JAK1/STAT3 pathways. In conclusion, HULC tumor-promoting roles in secreting pituitary adenoma might be via down-regulating miR-130b, up-regulating FOXM1, and activating PI3K/AKT/mTOR and JAK1/STAT3 pathways.


Assuntos
Humanos , Animais , Ratos , Neoplasias Hipofisárias/patologia , Adenoma/patologia , RNA Longo não Codificante/fisiologia , Ensaio de Imunoadsorção Enzimática , Transfecção , Adenoma/genética , Adenoma/metabolismo , Movimento Celular/fisiologia , Sobrevivência Celular/fisiologia , Western Blotting , Apoptose/fisiologia , MicroRNAs/análise , Linhagem Celular Tumoral , Fator de Transcrição STAT3/análise , Janus Quinase 1/análise , Janus Quinase 1/metabolismo , Ensaios de Migração Celular , Proteína Forkhead Box M1/análise , Proteína Forkhead Box M1/metabolismo , Luciferases
7.
MedicalExpress (São Paulo, Online) ; 4(5)Sept.-Oct. 2017. graf
Artigo em Inglês | LILACS | ID: biblio-894363

RESUMO

OBJECTIVE: To analyze the associated expression of STMN1, MELK and FOXM1 in search of alternative drugable target in glioblastoma (GBM) and to review relevant functional roles of STMN1 in cancer biology. METHOD: STMN1, MELK and FOXM1 expressions were studied by quantitative PCR and their coexpressions were analyzed in two independent glioblastoma cohorts. A review of articles in indexed journals that addressed the multiple functional aspects of STMN1 was conducted, focusing on the most recent reports discussing its role in cancer, in chemoresistance and in upstream pathways involving MELK and FOXM1. RESULTS: Significant associated expressions of MELK and FOXM1 were observed with STMN1 in GBM. Additionally, the literature review highlighted the relevance of STMN1 in cancer progression. CONCLUSION: STMN1 is very important to induce events in cancer development and progression, as cellular proliferation, migration, and drug resistance. Therefore, STMN1 can be an important therapeutic target for a large number of human cancers. In glioblastoma, the most aggressive brain tumor, the MELK/FOXM1/STMN1 presented significant associated expressions, thus pointing MELK and FOXM1 as alternative targets for therapy instead of STMN1, which is highly expressed in normal brain tissue. Continuous functional research to understand the STMN1 signaling pathway is worthwhile to improve the therapeutic approaches in cancer.


OBJETIVO: Analisar as expressões associadas de STMN1, MELK e FOXM1 na procura de alvos alternativos de tratamento em glioblastoma (GBM) e revisar os papeis funcionais relevantes de STMN1 na biologia do câncer. MÉTODO: As expressões de STMN1, MELK e FOXM1 foram estudadas por PCR quantitativo e suas coexpressões foram analisadas em dois coortes independentes de GBM. A revisão dos artigos publicados em revistas indexadas na procura dos aspectos funcionais múltiplos de STMN1 foi conduzida focando-se nos estudos mais recentes discutindo o seu papel em câncer, quimiorresistência e vias de sinalização envolvendo MELK e FOXM1. RESULTADOS: Observou-se expressões associadas significantes de MELK e FOXM1 com STMN1. Adicionalmente, a revisão da literatura salientou a relevância do STMN1 na progressão do câncer. CONCLUSÃO: STMN1 é muito importante nos eventos relacionados ao desenvolvimento e progressão do câncer, como proliferação celular, migração e resistência ao tratamento. Desta forma, STMN1 pode ser um forte alvo terapêutico em um grande número de cânceres humanos. Em GBM, o tumor cerebral mais agressivo, MELK/FOXM1/STMN1 apresentaram significativa associação em suas expressões gênicas, indicando, portanto, MELK e FOXM1 como alvos alternativos para terapia em substituição ao STMN1, que apresenta alta expressão no tecido cerebral normal. Perseverar nos estudos funcionais para o entendimento da via de sinalização do STMN1 é relevante para melhorar os esquemas terapêuticos para câncer.


Assuntos
Humanos , Glioblastoma/terapia , Estatmina/análise , Proteína Forkhead Box M1/análise , Citoesqueleto , Microtúbulos
8.
Br J Cancer ; 117(7): 974-983, 2017 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-28850563

RESUMO

BACKGROUND: Tyrosine kinase inhibitors (TKIs) have demonstrated clinical benefits in the treatment of several tumour types. However, the emergence of TKI resistance restricts the therapeutic effect. This study uses non-small cell lung cancer (NSCLC) to explore the mechanisms contributing to TKI resistance in tumours. METHODS: Biological phenotypes and RNA microarray expression data were analysed in NSCLC cells with and without TKI pretreatment. Specific inhibitors and siRNAs were used to validate the direct involvement of an AKT/FOXM1/STMN1 pathway in TKI resistance. Patients' tissues were analysed to explore the clinical importance of FOXM1 and STMN1. RESULTS: In vitro and in vivo studies showed that TKIs induced the enrichment of cancer stem cells (CSC), promoted epithelial to mesenchymal transition (EMT), and conferred multidrug resistance on NSCLC cells in a cell type- and TKI class-dependent manner. Mechanistically, TKIs activated an AKT/FOXM1/STMN1 pathway. The crucial role of this pathway in TKI-induced enrichment of CSC and drug resistance was verified by silencing FOXM1 and STMN1 or blocking the AKT pathway. Additionally, overexpression of STMN1 was associated with upregulation of FOXM1 in advanced NSCLC patients, and STMN1/FOXM1 upregulation predicted a poor outcome. CONCLUSIONS: Our findings elucidate an additional common mechanism for TKI resistance and provide a promising therapeutic target for reversing TKI resistance in NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Proteína Forkhead Box M1/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Estatmina/metabolismo , Animais , Carcinoma Pulmonar de Células não Pequenas/química , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Proteína Forkhead Box M1/análise , Proteína Forkhead Box M1/genética , Gefitinibe , Inativação Gênica , Humanos , Neoplasias Pulmonares/química , Neoplasias Pulmonares/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Niacinamida/análogos & derivados , Niacinamida/farmacologia , Fenótipo , Compostos de Fenilureia/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Quinazolinas/farmacologia , RNA Neoplásico/análise , Transdução de Sinais/efeitos dos fármacos , Sorafenibe , Estatmina/análise , Estatmina/genética , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA