Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Small ; 19(44): e2301439, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37420326

RESUMO

Although the chemo- and immuno-therapies have obtained good responses for several solid tumors, including those with brain metastasis, their clinical efficacy in glioblastoma (GBM) is disappointing. The lack of safe and effective delivery systems across the blood-brain barrier (BBB) and the immunosuppressive tumor microenvironment (TME) are two main hurdles for GBM therapy. Herein, a Trojan-horse-like nanoparticle system is designed, which encapsulates biocompatible PLGA-coated temozolomide (TMZ) and IL-15 nanoparticles (NPs) with cRGD-decorated NK cell membrane (R-NKm@NP), to elicit the immunostimulatory TME for GBM chemo-immunotherapy. Taking advantage of the outer NK cell membrane cooperating with cRGD, the R-NKm@NPs effectively traversed across the BBB and targeted GBM. In addition, the R-NKm@NPs exhibited good antitumor ability and prolonged the median survival of GBM-bearing mice. Notably, after R-NKm@NPs treatment, the locally released TMZ and IL-15 synergistically stimulated the proliferation and activation of NK cells, leading to the maturation of dendritic cells and infiltration of CD8+ cytotoxic T cells, eliciting an immunostimulatory TME. Lastly, the R-NKm@NPs not only effectively prolonged the metabolic cycling time of the drugs in vivo, but also has no noticeable side effects. This study may offer valuable insights for developing biomimetic nanoparticles to potentiate GBM chemo- and immuno-therapies in the future.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Nanopartículas , Camundongos , Animais , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Interleucina-15/uso terapêutico , Microambiente Tumoral , Biomimética , Linhagem Celular Tumoral , Temozolomida/uso terapêutico , Imunoterapia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia
2.
Proc Natl Acad Sci U S A ; 117(49): 31448-31458, 2020 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-33229571

RESUMO

Adult neural stem cells (NSC) serve as a reservoir for brain plasticity and origin for certain gliomas. Lineage tracing and genomic approaches have portrayed complex underlying heterogeneity within the major anatomical location for NSC, the subventricular zone (SVZ). To gain a comprehensive profile of NSC heterogeneity, we utilized a well-validated stem/progenitor-specific reporter transgene in concert with single-cell RNA sequencing to achieve unbiased analysis of SVZ cells from infancy to advanced age. The magnitude and high specificity of the resulting transcriptional datasets allow precise identification of the varied cell types embedded in the SVZ including specialized parenchymal cells (neurons, glia, microglia) and noncentral nervous system cells (endothelial, immune). Initial mining of the data delineates four quiescent NSC and three progenitor-cell subpopulations formed in a linear progression. Further evidence indicates that distinct stem and progenitor populations reside in different regions of the SVZ. As stem/progenitor populations progress from neonatal to advanced age, they acquire a deficiency in transition from quiescence to proliferation. Further data mining identifies stage-specific biological processes, transcription factor networks, and cell-surface markers for investigation of cellular identities, lineage relationships, and key regulatory pathways in adult NSC maintenance and neurogenesis.


Assuntos
Envelhecimento/genética , Linhagem da Célula , Ventrículos Laterais/anatomia & histologia , Ventrículos Laterais/citologia , Nicho de Células-Tronco/genética , Transcriptoma/genética , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Animais , Biomarcadores/metabolismo , Linhagem da Célula/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Camundongos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Transgenes
3.
J Clin Lab Anal ; 36(9): e24635, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35908795

RESUMO

OBJECTIVE: To discover novel serodiagnostic candidates for the serological diagnosis of syphilis. METHODS: Two recombinant Treponema pallidum proteins Tp0100 and Tp1016 were expressed, purified, and identified by Western Blotting. A total of 600 clinical serum samples were tested with the Tp0100-based ELISA, the Tp1016-based ELISA, and the commercial LICA Syphilis TP kit (ChIVD, Beijing, China). The sensitivities were determined by testing 340 samples from individuals with clinically diagnosed primary, secondary, latent, and tertiary syphilis. The specificities were determined by screening 260 samples from healthy controls and individuals with potentially cross-reactive infections, including leptospirosis, Lyme disease, hepatitis B, tuberculosis, rheumatoid arthritis, systemic lupus erythematosus. Kappa (κ) values were applied to compare the agreement between clinical syphilis diagnosis and the Tp0100-based ELISA, the Tp1016-based ELISA, or the LICA Syphilis TP test. RESULTS: Using clinical syphilis diagnosis as the gold standard, Tp0100 exhibited an overall sensitivity of 95.6% and specificity of 98.1% for testing IgG antibody while Tp1016 demonstrated only an overall sensitivity of 75.0% and specificity of 79.6%. In contrast, the LICA Syphilis TP test revealed an overall sensitivity of 97.6% and specificity of 96.2%. In addition, the overall percent agreement and corresponding κ values were 96.7% (95% CI 95.6%-97.8%) and 0.93 for the Tp0100-based ELISA, 77.0% (95% CI 74.3%-79.7%) and 0.54 for the Tp1016-based ELISA, and 97.0% (95% CI 96.0%-98.0%) and 0.94 for the LICA Syphilis TP test, respectively. CONCLUSION: The recombinant T. pallidum protein Tp0100 shows promise as a novel diagnostic antigen in the serological tests for syphilis.


Assuntos
Sífilis , Treponema pallidum , Anticorpos Antibacterianos , Antígenos de Bactérias , Ensaio de Imunoadsorção Enzimática , Humanos , Proteínas Recombinantes , Sensibilidade e Especificidade , Testes Sorológicos , Sífilis/diagnóstico , Sorodiagnóstico da Sífilis , Treponema pallidum/genética
4.
J Cell Mol Med ; 25(19): 9350-9363, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34469035

RESUMO

Patients with malignant glioma often suffered from depression, which leads to an increased risk of detrimental outcomes. Imipramine, an FDA-approved tricyclic antidepressant, has been commonly used to relieve depressive symptoms in the clinic. Recently, imipramine has been reported to participate in the suppression of tumour progression in several human cancers, including prostate cancer, colon cancer and lymphomas. However, the effect of imipramine on malignant glioma is largely unclear. Here, we show that imipramine significantly retarded proliferation of immortalized and primary glioma cells. Mechanistically, imipramine suppressed tumour proliferation by inhibiting yes-associated protein (YAP), a recognized oncogene in glioma, independent of Hippo pathway. In addition to inhibiting YAP transcription, imipramine also promoted the subcellular translocation of YAP from nucleus into cytoplasm. Consistently, imipramine administration significantly reduced orthotopic tumour progression and prolonged survival of tumour-bearing mice. Moreover, exogenous overexpression of YAP partially restored the inhibitory effect of imipramine on glioma progression. Most importantly, compared with imipramine or temozolomide (TMZ) monotherapy, combination therapy with imipramine and TMZ exhibited enhanced inhibitory effect on glioma growth both in vitro and in vivo, suggesting the synergism of both agents. In conclusion, we found that tricyclic antidepressant imipramine impedes glioma progression by inhibiting YAP. In addition, combination therapy with imipramine and TMZ may potentially serve as promising anti-glioma regimens, thus predicting a broad prospect of clinical application.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Ciclo Celular/metabolismo , Imipramina/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Temozolomida/farmacologia , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Glioma , Humanos , Camundongos , Prognóstico , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Cell Physiol Biochem ; 47(6): 2445-2457, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29990993

RESUMO

BACKGROUND/AIMS: Golgi phosphoprotein 3 (GOLPH3) plays pro-malignancy roles in several types of cancer. However, the molecular mechanism underlying GOLPH3 promoting tumor progression remains poorly understood. METHODS: The expression of GOLPH3 and Wntless (Wls) in glioma tissues was examined by western blotting and immunohistochemistry. EdU incorporation assay and colony formation assay was used to examine the cell growth ability. The effect of GOLPH3 on Wls recycling, Wnt secretion and ß-catenin activity was detected using western blotting, immunofluorescence, RT-PCR, ELISA or luciferase assay. RESULTS: The protein levels of GOLPH3 and Wls were upregulated and positively correlated with each other in human glioma tissues. The promoting effect of GOLPH3 on glioma cell proliferation was partially mediated by Wls. In addition, GOLPH3 interacted with Wls and GOLPH3 down-regulation drove Wls into lysosome for degradation, inhibiting its recycling to golgi and the plasma membrane. Importantly, GOLPH3 down-regulation inhibited Wnt2b secretion and decreased ß-catenin level and transcription activity. CONCLUSIONS: This study provides a brand new evidence that GOLPH3 promotes glioma cell proliferation by facilitating Wls recycling and Wnt/ß-catenin signaling. Our findings suggest a rationale for targeting the GOLPH3-Wls-Wnt axis as a promising therapeutic approach for glioblastoma.


Assuntos
Proliferação de Células , Glioma/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , Linhagem Celular Tumoral , Glioma/patologia , Células HEK293 , Humanos
6.
J Neurooncol ; 139(2): 269-279, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29713848

RESUMO

INTRODUCTION: Our recent work reported that GOLPH3 promotes glioma progression via inhibiting endocytosis and degradation of EGFR. The current study aimed to explore the potential regulating mechanism of GOLPH3 on JAK2-STAT3 signaling, a downstream effector of EGFR, in glioma progression. METHODS: The expression of JAK2, STAT3 and GOLPH3 in glioma tissues was detected by western blotting, tissue microarray and immunohistochemistry. The U251 and U87 cells with GOLPH3 down-regulation or over-expression were generated by lentivirus system. The effects of GOLPH3 on the activity of JAK2 and STAT3 were detected by western blotting and reverse transcription polymerase chain reaction. Co-immunoprecipitation was used to detect the association of GOLPH3 with JAK2 and STAT3. Cell proliferation was detected by CCK8 and EdU assay. RESULTS: The level of JAK2, STAT3 and GOLPH3 were significantly up-regulated and exhibited pairwise correlation in human glioma tissues. The level of p-JAK2 and p-STAT3, as well as the mRNA and protein levels of cyclin D1 and c-myc, two target genes of STAT3, decreased after GOLPH3 down-regulation, while they increased after GOLPH3 over-expression both in U251 and U87 cells. Interestingly, GOLPH3, JAK2 and STAT3 existed in the same protein complex and GOLPH3 affected the interaction of JAK2 and STAT3. Importantly, down-regulation of STAT3 partially abolished cell proliferation induced by GOLPH3 over-expression. CONCLUSIONS: GOLPH3 may act as a scaffold protein to regulate JAK2-STAT3 interaction and then its activation, which therefore mediates the effect of GOLPH3 on cell proliferation.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Janus Quinase 2/metabolismo , Proteínas de Membrana/metabolismo , Fator de Transcrição STAT3/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Glioma/patologia , Humanos , RNA Mensageiro/metabolismo , Transdução de Sinais
7.
Molecules ; 23(5)2018 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-29772722

RESUMO

Genus Cynanchum L. belongs to the family Asclepiadaceae, which comprise more than 200 species distributed worldwide. In Chinese medical practice, numerous drugs (such as tablets and powders) containing different parts of plants of this genus are used to treat snake bites, bruises, osteoblasts, rheumatoid arthritis and tumors. A search for original articles published on the cynanchum genus was performed by using several resources, including Flora of China Official Website and various scientific databases, such as PubMed, SciFinder, the Web of Science, Science Direct, and China Knowledge Resource Integrated (CNKI). Advances in the botanical, ethnomedicinal, phytochemical, and pharmacological studies of this genus are reviewed in this paper. Results showed that more than 440 compounds, including C21 steroids, steroidal saponins, alkaloids, flavonoids and terpene, have been isolated and identified from Cynanchum plants up to now. In vivo and in vitro studies have shown that plants possess an array of biological activities, including anti-tumor, neuroprotective and anti-fungal effects. Popular traditional prescription of Cynanchum sp. was also summed up in this paper. However, many Cynanchum species have received little or no attention. Moreover, few reports on the clinical use and toxic effects of Cynanchum sp. are available. Further attention should be focused on the study of these species to gather information on their respective toxicology data and relevant quality-control measures and clinical value of the crude extracts, active compounds, and bioactive metabolites from this genus. Further research on Cynanchum sp. should be conducted, and bioactivity-guided isolation strategies should be emphasized. In addition, systematic studies of the chemical composition of plants should be enhanced.


Assuntos
Cynanchum/química , Medicamentos de Ervas Chinesas/farmacologia , Compostos Fitoquímicos/farmacologia , Animais , Antifúngicos/química , Antifúngicos/farmacologia , Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/farmacologia , Cynanchum/classificação , Medicamentos de Ervas Chinesas/química , Etnobotânica , Etnofarmacologia , Flavonoides/química , Flavonoides/farmacologia , Humanos , Medicina Tradicional , Estrutura Molecular , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/farmacologia , Compostos Fitoquímicos/química , Fitosteróis/química , Fitosteróis/farmacologia
8.
Molecules ; 22(4)2017 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-28420183

RESUMO

A cytotoxicity-guided phytochemical investigation of Anemone vitifolia roots led to the isolation of six oleanane saponins (1-6), which were reported from the species for the first time. Their structures were determined by comparing its MS and NMR data with those in literature. Compounds 1-4 showed significant inhibitory effects on the proliferation of hepatocellular carcinoma HepG2 cells with IC50 values ranging from 2.0 to 8.5 µM, compared to positive control methotrexate with IC50 value of 15.8 µM. Flow cytometry analysis revealed that compounds 1-4 exerted anti-proliferative effects through a pro-apoptotic way of hepatocellular carcinoma cells.


Assuntos
Anemone/química , Antineoplásicos Fitogênicos/farmacologia , Extratos Vegetais/farmacologia , Raízes de Plantas/química , Antineoplásicos Fitogênicos/química , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Hep G2 , Humanos , Espectroscopia de Ressonância Magnética , Estrutura Molecular , Extratos Vegetais/química , Relação Estrutura-Atividade
9.
Zhongguo Zhong Yao Za Zhi ; 42(12): 2398-2407, 2017 Jun.
Artigo em Zh | MEDLINE | ID: mdl-28822199

RESUMO

Rheumatoid arthritis(RA) is a chronic autoimmune disease, belonging to the "Arthromyodynia (Bi Zheng)" category in traditional Chinese medicine. However, the ethnomedicine has a unique understanding of RA, with a long-term clinical experience accumulation and significant efficacy in the treatment of RA, and it has now become one of the important means in treatment of RA. On the basis of literature research, the understanding of RA and commonly used Tibetan medicine, Mongolian medicine, Hui medicine and other herbs and preparations were reviewed in this paper, with the aim of providing a reference for its clinical treatment of RA and research and development of innovative drugs.


Assuntos
Artrite Reumatoide/tratamento farmacológico , Medicina Tradicional , Humanos , Medicina Tradicional Chinesa , Medicina Tradicional da Mongólia , Medicina Tradicional Tibetana , Plantas Medicinais
10.
Tumour Biol ; 2016 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-27718125

RESUMO

Dysregulation of Hippo/YAP signaling leads to aberrant cell growth and neoplasia. Although the roles and regulation of Hippo/YAP signaling were extensively studied in cancer biology recently, study systematically checking the expression pattern of core components of this pathway at the tumor tissue level remains lacking. In this study, we thoroughly examined the profile of core components of Hippo/YAP signaling in patient specimens both at the mRNA and at protein levels. We found that the mRNA level of YAP1/TAZ and their target genes, CRY61, CTGF, and BIRC5, was remarkably amplified in glioma tissues. Consistently, the protein level of YAP1/TAZ increased and meanwhile those of p-YAP1/p-TAZ and LATS1/2 decreased in gliomas. Unexpectedly, both the mRNA and protein levels of MST1/2 increased in the glioma tissues, inconsistent with its presumed tumor suppressor identity. In addition, over-expression of LATS2 decreased, while over-expression of YPA1 increased the cell proliferation ability. Furthermore, based on the data from the free public database, YAP1/TAZ and BIRC5 were positively associated with the prognosis of glioma patients, while LATS1/2 exhibited negative correlation with the glioma patient prognosis. Collectively, we deduce that, in glioma tissue context, MST1/2 may not be the essential component of the hippo/YAP pathway. Moreover, our findings uncover a new evidence supporting that YAP1/TAZ-BIRC5 might be abnormally activated due to LATS1/2 down-regulation, which in turn promote the occurrence and development of gliomas, paving the way to identify the potential therapeutic molecular target for gliomas.

11.
Hippocampus ; 25(3): 373-84, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25330763

RESUMO

Geranylgeranyltransferase I (GGT), a protein prenyltransferase, is responsible for the posttranslational lipidation of Rho GTPases, such as Rac, Rho and Cdc42, all of which play an important role in neuronal synaptogenesis. We previously demonstrated that GGT promotes dendritic morphogenesis in cultured hippocampal neurons and cerebellar slices. We report here that inhibiting GGT activity decreases basal- and activity-dependent changes in spine density as well as in learning and memory ability of mice in vivo. We found that KCl- or bicuculline-induced dendritic spine density increases was abolished by specific GGT inhibitor GGTi-2147 treatment in cultured hippocampal neurons. GGTi-2147 lateral ventricular injection reduced GGT activity and membrane association of Rac and decreased the density of dendritic spines in the mouse hippocampus, frontal cortex and cerebellum. GGTi-2147 administration also impaired learning and memory ability of mice. More importantly, mice exposed to environmental enrichment (EE) showed increased spine density and learning and memory ability, which were significantly reversed by GGTi-2147 administration. These data demonstrate that inhibiting GGT activity prevents both basal- and activity-dependent changes in spine density in central nervous system both in vitro and in vivo. Manipulating GGT activity may be a promising strategy for the therapies of neurodevelopmental disorders, such as autism, depression, and schizophrenia.


Assuntos
Alquil e Aril Transferases/metabolismo , Sistema Nervoso Central/citologia , Sistema Nervoso Central/enzimologia , Espinhas Dendríticas/patologia , Animais , Bicuculina/farmacologia , Células Cultivadas , Sistema Nervoso Central/efeitos dos fármacos , Espinhas Dendríticas/efeitos dos fármacos , Embrião de Mamíferos , Inibidores Enzimáticos/farmacologia , Antagonistas de Receptores de GABA-A/farmacologia , Hipocampo/citologia , Imidazóis/farmacologia , Aprendizagem/efeitos dos fármacos , Leucina/análogos & derivados , Leucina/farmacologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Neurogênese/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/ultraestrutura , Cloreto de Potássio/farmacologia , Ratos , Ratos Sprague-Dawley , Proteínas rac de Ligação ao GTP/metabolismo
12.
Biochem Biophys Res Commun ; 456(1): 494-9, 2015 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-25490384

RESUMO

Previously, we found that brain expressed X-linked gene 2 (Bex2) regulates the invasion/migration ability of glioma cells. However, the mechanism of this effect remains unknown. In current study, we reported that Bex2 down-regulation inhibited glioma cell migration and invasion by decreasing the nucleus and cytoplasm protein level of ß-catenin. We found that the protein levels of Bex2 and ß-catenin were up-regulated and showed direct correlation in glioma tissues. Bex2 down-regulation significantly decreased ß-catenin protein levels but not its mRNA levels. Furthermore, the decreased protein level of ß-catenin was located in the nucleus and cytoplasm but not in the cell membrane. Further study found that the effects of Bex2 down-regulation on the invasion and migration of glioma cell could be reversed by ß-catenin over-expression. Taken together, Bex2 affects the invasion and migration ability of glioma cells by regulating ß-catenin.


Assuntos
Regulação Neoplásica da Expressão Gênica , Glioma/metabolismo , Proteínas do Tecido Nervoso/metabolismo , beta Catenina/metabolismo , Western Blotting , Encéfalo/metabolismo , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Movimento Celular , Citoplasma/metabolismo , Glioma/patologia , Humanos , Invasividade Neoplásica , RNA Interferente Pequeno/metabolismo , Cicatrização
13.
Mol Carcinog ; 54(11): 1252-63, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25156912

RESUMO

The identification of genes involved in carcinogenesis and tumor progression is of great interest, since these genes might be possible as candidates for new tumor targeted therapy strategies. Our previous study shows that Golgi phosphoprotein 3 (GOLPH3) is involved in glioma cell migration and invasion, the critical characteristics of malignant gliomas. In this study, we explored the mechanism of GOLPH3 affecting cell migration and invasion and found that GOLPH3 promotes glioblastoma (GBM) cell migration and invasion via the mammalian target of rapamycin(mTOR)-Y-box binding protein-1 (YB1) pathway in vitro. Both the protein levels of GOLPH3 and YB1 were up-regulated in human glioma tissues and they exhibited direct correlation with each other. In addition, down-regulation of GOLPH3 inhibited glioma cell migration and invasion, while over-expression of GOLPH3 enhanced them. Meanwhile, GOLPH3 down-regulation led to a significant decrease of YB1 level as well as mTOR activity, both required for glioma cell migration and invasion. On the contrary, YB1 level and mTOR activity increased after GOLPH3 over-expression. YB1 down-regulation or mTOR ATP site inhibitor INK128 treatment inhibited cell migration and invasion, similar to the effect of GOLPH3 down-regulation. Furthermore, over-expression of GOLPH3 induced glioma cell migration and invasion was blocked by INK128 and YB1 down-regulation. Taken together, these results show that GOLPH3 promotes glioblastoma cell migration and invasion via the mTOR-YB1pathway, indicating that GOLPH3-mTOR-YB1 pathway might be a new therapeutic target for glioma treatment.


Assuntos
Movimento Celular/genética , Glioblastoma/genética , Proteínas de Membrana/genética , Invasividade Neoplásica/genética , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/genética , Proteína 1 de Ligação a Y-Box/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Regulação para Baixo/genética , Regulação Neoplásica da Expressão Gênica/genética , Glioblastoma/patologia , Humanos , Regulação para Cima/genética
14.
J Neurooncol ; 121(2): 279-88, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25373346

RESUMO

Mammalian sterile 20-like 1 (Mst1), an upstream serine/threonine-specific protein kinase of the Hippo pathway, is reported to play important roles in tumor suppression and organ size regulation in mammals via regulating cell proliferation and survival. However, whether it is involved in the pathogenesis of malignant gliomas remains poorly understood. Therefore, in the present work, we examined the effect and mechanism of Mst1 on the proliferation and apoptosis of malignant glioma cells. The cell proliferation and growth of glioma cells were examined by EdU incorporation and CCK-8 assay. In addition, the cell apoptosis was assessed by flow cytometry. We found that down-regulation of Mst1 promoted glioma cell proliferation and growth, but inhibited the cell apoptosis. Consistent with this, over-expression of Mst1 inhibited glioma cell proliferation and growth. Interestingly, Mst1 did not affect the phosphorylation of YAP1, the key downstream molecule of Hippo pathway. However, Mst1 was found to bind to AKT in glioma cell and negatively regulated AKT and mTOR activity. Finally, the increased cell proliferation rate induced by Mst1 down-regulation was partially abolished by down-regulation of AKT1. Meanwhile, glioma cell growth inhibition induced by Mst1 over-expression was partially rescued by over-expression of AKT1. Taken together, these findings suggest that Mst1 regulates proliferation of glioma cells via AKT/mTOR signaling pathway.


Assuntos
Neoplasias Encefálicas/metabolismo , Proliferação de Células/fisiologia , Glioma/fisiopatologia , Fator de Crescimento de Hepatócito/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Apoptose/fisiologia , Encéfalo/metabolismo , Caspase 3/metabolismo , Caspase 9/metabolismo , Linhagem Celular Tumoral , Humanos , Transdução de Sinais
15.
J Neurooncol ; 121(3): 469-77, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25528634

RESUMO

Plumbagin, a natural quinonoid constituent isolated from the root of medicinal plant Plumbago zeylanica L, has exhibited anti-tumor and anti-proliferative activities in various tumor cell lines as well as in animal tumor models. However, its anticancer effects and the mechanisms underlying its suppression of glioma cell growth have not been elucidated. Oncogenic transcription factor Forkhead Box M1 (FOXM1) has garnered particular interest in recent years as a potential target for the prevention and/or therapeutic intervention in glioma, nevertheless, less information is currently available regarding FOXM1 inhibitor. Here, we reported that plumbagin could effectively inhibit cell proliferation, migration and invasion and induce apoptosis of glioma cells. Cell cycle assay showed that plumbagin induced G2/M arrest. Interestingly, we found that plumbagin decreased the expression of FOXM1 both at mRNA level and protein level. Plumbagin also inhibited the transactivation ability of FOXM1, resulting in down-regulating the expression of FOXM1 downstream target genes, such as cyclin D1, Cdc25B, survivin, and increasing the expression of p21(CIP1) and p27(KIP1). Most importantly, down-regulation of FOXM1 by siFOXM1 transfection enhanced plumbagin-induced change in viability. On the contrary, over-expression of FOXM1 by cDNA transfection reduced plumbagin-induced glioma cell growth inhibition. These results suggest that plumbagin exhibits its anticancer activity partially by inactivation of FOXM1 signaling pathway in glioma cells. Our findings indicate that plumbagin may be considered as a potential natural FOXM1 inhibitor, which could contribute to the development of new anticancer agent for therapy of gliomas.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Fatores de Transcrição Forkhead/biossíntese , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioma/metabolismo , Naftoquinonas/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Regulação para Baixo , Proteína Forkhead Box M1 , Humanos , RNA Interferente Pequeno , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
16.
J Neurooncol ; 125(1): 113-21, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26341367

RESUMO

Drosophila lethal (2) giant larvae (lgl) has been reported as a tumor suppressor and could regulate the Drosophila hippo signaling. Human giant larvae-1(Hugl-1), one human homologue of Drosophila lgl, also has been reported to be involved in the development of some human cancers. However, whether Hugl-1 is associated with the pathogenesis of malignant gliomas remains poorly understood. In the present work, we examined the effect of Hugl-1 on glioma cell growth both in vitro and in vivo. Firstly, we found that Hugl-1 protein levels decreased in the human glioma tissues, suggesting that Hugl-1 is involved in glioma progression. Unfortunately, either stably or transiently over-expressing Hugl-1 did not affect glioma cell proliferation in vitro. In addition, Hugl-1 over-expression did not regulate hippo signaling pathway. Interestingly, over-expression of Hugl-1 not only inhibited gliomagenesis but also markedly inhibited cell proliferation and promoted the apoptosis of U251 cells in an orthotopic model of nude mice. Taken together, this study provides the evidence that Hugl-1 inhibits glioma cell growth in intracranial model of nude mice, suggesting that Hugl-1 might be a potential tumor target for glioma therapy.


Assuntos
Astrocitoma/patologia , Neoplasias Encefálicas/patologia , Proteínas do Citoesqueleto/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Animais , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Proteínas do Citoesqueleto/genética , Modelos Animais de Doenças , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Antígeno Ki-67/metabolismo , Masculino , Camundongos , Camundongos Nus , Compostos de Fenilureia/metabolismo , Sincalida/metabolismo , Transfecção
17.
Cell Biol Int ; 39(3): 283-90, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25262972

RESUMO

Bmi-1 is involved in the development of several human cancers; however, its significance in glioma progression remains largely unknown. We report that downregulation of Bmi-1 clearly reduces glioma cell migration and invasion. Downregulation of Bmi-1 promotes the expression of the tumor suppressor p16, which is important in glioma cell motility. Reduction in glioma cell invasion due to downregulation of Bmi-1 could be rescued by p16 downregulation. These results show that Bmi-1 contributes to the motility of glioma cells by regulating the expression of p16.


Assuntos
Neoplasias Encefálicas/patologia , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Glioma/patologia , Complexo Repressor Polycomb 1/metabolismo , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Inibidor p16 de Quinase Dependente de Ciclina/antagonistas & inibidores , Inibidor p16 de Quinase Dependente de Ciclina/genética , Regulação para Baixo , Glioma/metabolismo , Humanos , Metaloproteinase 2 da Matriz/metabolismo , Invasividade Neoplásica , Complexo Repressor Polycomb 1/antagonistas & inibidores , Complexo Repressor Polycomb 1/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo
18.
J Pharmacol Sci ; 128(3): 131-6, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26154848

RESUMO

Plumbagin is a natural compound that is isolated from the root of the medicinal plant Plumbago zeylanica L. Based on a previous in vitro study performed by our group, which demonstrated the effectiveness of plumbagin against glioma cells, we further ascertained whether plumbagin exhibits the same effectiveness against glioma cell xenografts in nude mice. Our results revealed that tumor volume was reduced by 54.48% in the plumbagin-treated group compared with the controls. Furthermore, there were no obvious signs of toxicity as assessed by the organ sizes and cell morphologies of the mice that were treated with plumbagin. Immunofluorescence assays further revealed that plumbagin significantly inhibited glioma cell proliferation and induced cell apoptosis. Importantly, we also determined that the expressions of FOXM1 and its downstream target effectors, including cyclin D1 and Cdc25B, were down-regulated in the treated group, while the expressions of p21 and p27 were increased; the latter findings corroborate the results of our previous in vitro study. Taken together, these findings indicate that plumbagin may be a natural downregulator of FOXM1 with potential therapeutic effectiveness for the treatment of gliomas.


Assuntos
Antineoplásicos Fitogênicos , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Expressão Gênica/efeitos dos fármacos , Glioma/genética , Glioma/patologia , Terapia de Alvo Molecular , Naftoquinonas/farmacologia , Naftoquinonas/uso terapêutico , Fitoterapia , Plumbaginaceae/química , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Proteína Forkhead Box M1 , Expressão Gênica/genética , Glioma/tratamento farmacológico , Humanos , Camundongos Nus , Naftoquinonas/isolamento & purificação , Transplante de Neoplasias
19.
J Neurooncol ; 119(1): 49-58, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24792491

RESUMO

The Fyn related kinase (FRK) is a noteworthy member of the Src non-receptor tyrosine kinase family for its distinctive tumor suppressive function. Recently, we have shown that FRK plays a protective role against the progression of glioma by suppressing cell migration and invasion. However, it is unclear whether the cell growth of glioma is also regulated by FRK and by which mechanism FRK alters its specific biological functions. In the current study, we found that FRK over-expression significantly suppressed the proliferation of glioma cells. In contrast, FRK knockdown by siRNA promoted glioma cell growth. In addition, FRK over-expression caused G1 phase arrest as well as apoptosis of glioma cells. Further investigation disclosed that FRK-induced G1 arrest was accompanied by down-regulation of hyperphosphorylated retinoblastoma protein (pRb), which led to the consequent suppression of E2F1. More importantly, we found that over-expression of FRK inhibited proper cyclin D1 accumulation in the nucleus of proliferating cells. Taken together, our results demonstrate a combined mechanism for the anti-proliferative effects of FRK by inhibiting cyclin D1 nucleus accumulation and pRb phosphorylation in glioma cells.


Assuntos
Neoplasias Encefálicas/metabolismo , Núcleo Celular/metabolismo , Proliferação de Células/fisiologia , Ciclina D1/metabolismo , Glioma/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Tirosina Quinases/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Regulação para Baixo , Fase G1 , Glioma/patologia , Humanos , Fosforilação , Proteína do Retinoblastoma/metabolismo , Transdução de Sinais/fisiologia
20.
Cell Biol Int ; 38(3): 326-34, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24293123

RESUMO

Ras-oncogenic pathway contributes to the pathogenesis of various tumours in humans, in which mutations of three canonical genes including H-Ras, N-Ras and K-Ras are the most common events. Dysregulation of Ras signalling can be tumourigenic, especially gliomas of the central nervous system. Rap proteins are members of the small GTPase superfamily that involved in many biological processes. However, it remains largely unclear as to whether and how Rap proteins are involved in the development of multiple gliomas. We found that the levels of the protein Rap2a and the activity of Rap2a (GTP-Rap2a) were weakly expressed in glioma tissues. Overexpressed Rap2a significantly inhibited the migration and invasion of glioma cells with an increase of GTP-Rap2a. Overexpression of the dominant-active (DA-Rap2a), but not the dominant-negative (DN-Rap2a) form of Rap2a, also similarly inhibited the migration and invasion of glioma cells by reducing the phosphorylation level of AKT. In contrast, downregulation of Rap2a promoted glioma migration and invasion, and raised the phosphorylation level of AKT, whereas these effects were inhibited by PI3K-specific inhibitor, LY294002. Thus unlike the other family members of Ras, Rab2a probably serves as a tumour suppressor in the pathogenesis of glioma.


Assuntos
Neoplasias Encefálicas/genética , Movimento Celular/genética , Regulação Neoplásica da Expressão Gênica/genética , Glioma/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas rap de Ligação ao GTP/genética , Linhagem Celular Tumoral , Regulação para Baixo/genética , Regulação para Baixo/fisiologia , Humanos , Invasividade Neoplásica , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/genética , Transdução de Sinais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA