Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Heliyon ; 8(10): e11022, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36281400

RESUMO

Gliomas typically have unfavorable prognosis, due to late detection and interventions. However, effective biomarkers for early glioma diagnosis based on 5-hydroxymethylcytosines (5 hm C) in circulating cell-free DNA (cfDNA) are not currently available. 5 hm C profiles in GSE132118 set were subjected for establishment of diagnostic model using the LASSO (least absolute shrinkage and selection operator) algorithm. The 5 hm C-based models demonstrated great potency in differentiating healthy subjects from gliomas, with area under the curves (AUCs) > 0.91 in the training and validation sets. Moreover, the indicator performed well in combination with clinicopathological characteristics to differentiate glioblastomas (GBMs) from lower grade glioma (LGGs). Enrichment analysis on 5 hm C profiles displayed great correlation with glioma pathophysiology. The 5 hm C-derived biomarker might act as an effective and non-invasive measure in glioma screening.

2.
Front Immunol ; 13: 865020, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36119086

RESUMO

MS4A6A has been recognized as being associated with aging and the onset of neurodegenerative disease. However, the mechanisms of MS4A6A in glioma biology and prognosis are ill-defined. Here, we show that MS4A6A is upregulated in glioma tissues, resulting in unfavorable clinical outcomes and poor responses to adjuvant chemotherapy. Multivariate Cox regression analysis suggested that MS4A6A expression can act as a strong and independent predictor for glioma outcomes (CGGA1: HR: 1.765, p < 0.001; CGGA2: HR: 2.626, p < 0.001; TCGA: HR: 1.415, p < 0.001; Rembrandt: HR: 1.809, p < 0.001; Gravendeel: HR: 1.613, p < 0.001). A protein-protein interaction (PPI) network revealed that MS4A6A might be coexpressed with CD68, CD163, and macrophage-specific signatures. Enrichment analysis showed the innate immune response and inflammatory response to be markedly enriched in the high MS4A6A expression group. Additionally, single-cell RNA sequencing (scRNA-seq) analysis revealed distinctive expression features for MS4A6A in macrophages in the glioma immune microenvironment (GIME). Immunofluorescence staining confirmed colocalization of CD68/MS4A6A and CD163/MS4A6A in macrophages. Correlation analysis revealed that MS4A6A expression is positively related to the tumor mutation burden (TMB) of glioma, displaying the high potential of applying MS4A6A to evaluate responsiveness to immunotherapy. Altogether, our research indicates that MS4A6A upregulation may be used as a promising and effective indicator for adjuvant therapy and prognosis assessment.


Assuntos
Neoplasias Encefálicas , Glioma , Doenças Neurodegenerativas , Biomarcadores Tumorais/genética , Neoplasias Encefálicas/patologia , Glioma/patologia , Humanos , Macrófagos/metabolismo , Doenças Neurodegenerativas/metabolismo , Prognóstico , Microambiente Tumoral
3.
Drug Deliv ; 29(1): 2414-2427, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35904177

RESUMO

Polymersomes possess the self-assembly vesicular structure similar to liposomes. Although a variety of comparisons between polymersomes and liposomes in the aspects of physical properties, preparation and applications have been elaborated in many studies, few focus on their differences in drug encapsulation, delivery and release in vitro and in vivo. In the present work, we have provided a modified direct hydration method to encapsulate anti-cancer drug paclitaxel (PTX) into PEG-b-PCL constituted polymersomes (PTX@PS). In addition to advantages including narrow particle size distribution, high colloid stability and moderate drug-loading efficiency, we find that the loaded drug aggregate in small clusters and reside through the polymersome membrane, representing a unique core-satellite structure which might facilitate the sustained drug release. Compared with commercial liposomal PTX formulation (Lipusu®), PTX@PS exhibited superb tumor cell killing ability underlain by multiple pro-apoptotic mechanisms. Moreover, endocytic process of PTX@PS significantly inhibits drug transporter P-gp expression which could be largely activated by free drug diffusion. In glioma mice models, it has also confirmed that PTX@PS remarkably eradicate tumors, which renders polymersomes as a promising alternative to liposomes as drug carriers in clinic.


Assuntos
Antineoplásicos , Lipossomos , Animais , Linhagem Celular Tumoral , Portadores de Fármacos/química , Liberação Controlada de Fármacos , Camundongos , Paclitaxel/química , Polietilenoglicóis/química
4.
Cell Death Dis ; 13(4): 339, 2022 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-35418179

RESUMO

Glioblastoma (GBM) is the most common and aggressive primary malignant brain tumor. The unregulated expression of Claudin-4 (CLDN4) plays an important role in tumor progression. However, the biological role of CLDN4 in GBM is still unknown. This study aimed to determine whether CLDN4 mediates glioma malignant progression, if so, it would further explore the molecular mechanisms of carcinogenesis. Our results revealed that CLDN4 was significantly upregulated in glioma specimens and cells. The inhibition of CLND4 expression could inhibit mesenchymal transformation, cell invasion, cell migration and tumor growth in vitro and in vivo. Moreover, combined with in vitro analysis, we found that CLDN4 can modulate tumor necrosis factor-α (TNF-α) signal pathway. Meanwhile, we also validated that the transforming growth factor-ß (TGF-ß) signal pathway can upregulate the expression of CLDN4, and promote the invasion ability of GBM cells. Conversely, TGF-ß signal pathway inhibitor ITD-1 can downregulate the expression of CLDN4, and inhibit the invasion ability of GBM cells. Furthermore, we found that TGF-ß can promote the nuclear translocation of CLDN4. In summary, our findings indicated that the TGF-ß/CLDN4/TNF-α/NF-κB signal axis plays a key role in the biological progression of glioma. Disrupting the function of this signal axis may represent a new treatment strategy for patients with GBM.


Assuntos
Claudina-4 , Glioblastoma , Glioma , Fator de Crescimento Transformador beta , Linhagem Celular Tumoral , Claudina-4/genética , Claudina-4/metabolismo , Glioblastoma/genética , Glioblastoma/metabolismo , Glioma/metabolismo , Humanos , NF-kappa B/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Regulação para Cima
5.
J Nanobiotechnology ; 20(1): 39, 2022 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-35062946

RESUMO

BACKGROUND: Glioblastoma (GBM) is the most common and fatal primary tumor in the central nervous system (CNS). Due to the existence of blood-brain barrier (BBB), most therapeutics cannot efficiently reach tumors in the brain, and as a result, they are unable to be used for effective GBM treatment. Accumulating evidence shows that delivery of therapeutics in form of nanoparticles (NPs) may allow crossing the BBB for effective GBM treatment. METHODS: Betulinic acid NPs (BA NPs) were synthesized by the standard emulsion approach and characterized by electron microscopy and dynamic light scattering analysis. The resulting NPs were characterized for their anti-tumor effects by cell viability assay, EdU-DNA synthesis assay, cell cycle assay, mitochondrial membrane potential, and PI-FITC apoptosis assay. Further mechanistic studies were carried out through Western Blot and immunostaining analyses. Finally, we evaluated BA NPs in vivo for their pharmacokinetics and antitumor effects in intracranial xenograft GBM mouse models. RESULTS: BA NPs were successfully prepared and formed into rod shape. BA NPs could significantly suppress glioma cell proliferation, induce apoptosis, and arrest the cell cycle in the G0/G1 phase in vitro. Furthermore, BA NPs downregulated the Akt/NFκB-p65 signaling pathway in a concentration dependent manner. We found that the observed anti-tumor effect of BA NPs was dependent on the function of CB1/CB2 receptors. Moreover, in the intracranial GBM xenograft mouse models, BA NPs could effectively cross the BBB and greatly prolong the survival time of the mice. CONCLUSIONS: We successfully synthesized BA NPs, which could cross the BBB and demonstrated a strong anti-tumor effect. Therefore, BA NPs may potentially be used for effective treatment of GBM.


Assuntos
Antineoplásicos Fitogênicos , Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Nanopartículas/química , Triterpenos Pentacíclicos , Animais , Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Humanos , Camundongos , Triterpenos Pentacíclicos/química , Triterpenos Pentacíclicos/farmacologia , Receptores de Canabinoides/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Ácido Betulínico
6.
Front Cell Dev Biol ; 9: 717601, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34650972

RESUMO

The tumor immune microenvironment (TIME) has been recognized to be associated with sensitivity to immunotherapy and patient prognosis. Recent research demonstrates that assessing the TIME patterns on large-scale samples will expand insights into TIME and will provide guidance to formulate immunotherapy strategies for tumors. However, until now, thorough research has not yet been reported on the immune infiltration landscape of glioma. Herein, the CIBERSORT algorithm was used to unveil the TIME landscape of 1,975 glioma observations. Three TIME subtypes were established, and the TIMEscore was calculated by least absolute shrinkage and selection operator (LASSO)-Cox analysis. The high TIMEscore was distinguished by an elevated tumor mutation burden (TMB) and activation of immune-related biological process, such as IL6-JAK-STAT3 signaling and interferon gamma (IFN-γ) response, which may demonstrate that the patients with high TIMEscore were more sensitive to immunotherapy. Multivariate analysis revealed that the TIMEscore could strongly and independently predict the prognosis of gliomas [Chinese Glioma Genome Atlas (CGGA) cohort: hazard ratio (HR): 2.134, p < 0.001; Gravendeel cohort: HR: 1.872, p < 0.001; Kamoun cohort: HR: 1.705, p < 0.001; The Cancer Genome Atlas (TCGA) cohort: HR: 2.033, p < 0.001; the combined cohort: HR: 1.626, p < 0.001], and survival advantage was evident among those who received chemotherapy. Finally, we validated the performance of the signature in human tissues from Wuhan University (WHU) dataset (HR: 15.090, p = 0.008). Our research suggested that the TIMEscore could be applied as an effective predictor for adjuvant therapy and prognosis assessment.

7.
Front Pharmacol ; 12: 659511, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34381355

RESUMO

The most common primary central nervous system tumor in adults is glioblastoma multiforme (GBM). The high invasiveness of GBM cells is an important factor leading to inevitable tumor recurrence and a poor prognosis of patients. GNE-477, a novel PI3K/mTOR inhibitor, has been reported to exert antiproliferative effects on other cancer cells. However, researchers have not clearly determined whether GNE-477 produces antitumor effects on GBM. In the present study, GNE-477 significantly inhibited the proliferation, migration and invasion of U87 and U251 cells. In addition, GNE-477 also induced apoptosis of GBM cells, arresting the cell cycle in G0/G1 phase. More importantly, GNE-477 also reduced the levels of AKT and mTOR phosphorylation in the AKT/mTOR signaling pathway in a concentration-dependent manner. An increase in AKT activity induced by SC79 rescued the GNE-477-mediated inhibition of GBM cell proliferation and apoptosis. The antitumor effects of GNE-477 and the regulatory effects on related molecules were further confirmed in vivo using a nude mouse intracranial xenograft model. In conclusion, our study indicated that GNE-477 exerted significant antitumor effects on GBM cells in vitro and in vivo by downregulating the AKT/mTOR pathway.

8.
J Cancer ; 12(10): 2912-2920, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33854592

RESUMO

Background: ADPRH is a modulator of CD8+ T cell functions, and dysregulation of ADPRH has been identified to involve in carcinogenesis of cancers. However, the association of ADPRH with low grade glioma (LGG) remains unclear. Methods: The expression of ADPRH in LGG was first analyzed in GLIOVIS and GEPIA databases and then validated by real-time PCR (rt-PCR), immunochemistry and human protein atlas (HPA). Univariate and multivariate Cox analysis and Kaplan-Meier plots were designed to assess the prognostic value of ADPRH in LGG. The correlation of ADPRH and immune infiltration was evaluated by data in TIMER and ESTIMATE databases. Gene set enrichment analysis was conducted to detect biological processes associated with ADPRH. Results: ADPRH was significantly upregulated in LGG in comparison to non-tumor brain samples in transcriptomic and proteomic levels. The high ADPRH expression indicated unfavorable overall survival (OS) and progression-free survival (PFS) in patients with LGG using Kaplan-Meier plots. And multivariate Cox analysis demonstrated the expression level of ADPRH was an independent prognosis-predicting index for OS and PFS of LGG patients in all cohorts separately. Gene Set Enrichment Analysis (GSEA) indicated that high expression of ADPRH was involved in the upregulation of P53 signaling pathway, KRAS signaling pathway, IL6/JAK-STAT3 signaling and TNF-beta signaling pathways. By TIMER and ESTIMATE databases, we identified ADPRH expression had strong correlation with tumor immune infiltrating cells (TIICs). Conclusions: In summary, our findings demonstrated that ADPRH might be a potential prognostic biomarker and correlated with TIICs in LGG.

9.
J Cancer ; 12(9): 2756-2767, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33854635

RESUMO

Serum amyloid A1 (SAA1) is an inflammatory associated high-density lipoprotein. And It is also considered as a predictor and prognostic marker of cancer risk. However, its role and mechanisms in glioblastoma (GBM) still unclear. In this study, we validate that SAA1 is up-regulated in GBM, and its high expression predicts poor prognosis. SAA1 knockdown promotes the apoptosis of GBM cell. Mechanistically, SAA1 knockdown can inhibit serine/threonine protein kinase B (AKT) phosphorylation, thereby regulating the expression of apoptosis-related proteins such as Bcl2 and Bax, leading to GBM cell death. Moreover, Gliomas with low SAA1 expression have increased sensitivity to Temozolomide (TMZ). Low SAA1 expression segregated glioma patients who were treated with Temozolomide (TMZ) or with high MGMT promoter methylation into survival groups in TCGA and CGGA dataset. Our study strongly suggested that SAA1 was a regulator of cells apoptosis and acted not only as a prognostic marker but also a novel biomarker of sensitivity of glioma to TMZ.

10.
BMC Cancer ; 21(1): 251, 2021 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-33750353

RESUMO

BACKGROUND: To accurately predict the prognosis of glioma patients. METHODS: A total of 541 samples from the TCGA cohort, 181 observations from the CGGA database and 91 samples from our cohort were included in our study. Long non-coding RNAs (LncRNAs) associated with glioma WHO grade were evaluated by weighted gene co-expression network analysis (WGCNA). Five lncRNA features were selected out to construct prognostic signatures based on the Cox regression model. RESULTS: By weighted gene co-expression network analysis (WGCNA), 14 lncRNAs related to glioma grade were identified. Using univariate and multivariate Cox analysis, five lncRNAs (CYTOR, MIR155HG, LINC00641, AC120036.4 and PWAR6) were selected to develop the prognostic signature. The Kaplan-Meier curve depicted that the patients in high risk group had poor prognosis in all cohorts. The areas under the receiver operating characteristic curve of the signature in predicting the survival of glioma patients at 1, 3, and 5 years were 0.84, 0.92, 0.90 in the CGGA cohort; 0.8, 0.85 and 0.77 in the TCGA set and 0.72, 0.90 and 0.86 in our own cohort. Multivariate Cox analysis demonstrated that the five-lncRNA signature was an independent prognostic indicator in the three sets (CGGA set: HR = 2.002, p < 0.001; TCGA set: HR = 1.243, p = 0.007; Our cohort: HR = 4.457, p = 0.008, respectively). A nomogram including the lncRNAs signature and clinical covariates was constructed and demonstrated high predictive accuracy in predicting 1-, 3- and 5-year survival probability of glioma patients. CONCLUSION: We established a five-lncRNA signature as a potentially reliable tool for survival prediction of glioma patients.


Assuntos
Neoplasias Encefálicas/mortalidade , Biologia Computacional/métodos , Glioma/mortalidade , RNA Longo não Codificante/análise , Neoplasias Encefálicas/genética , Redes Reguladoras de Genes , Glioma/genética , Humanos , Nomogramas , Prognóstico , Modelos de Riscos Proporcionais
11.
Front Oncol ; 11: 769033, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35047393

RESUMO

The Fc Fragment of IgG Binding Protein (FCGBP) has been proven to participate in intestinal tumor immunity. However, the biological role of FCGBP has remained unclear in glioma. The differential expression of FCGBP was explored by Oncomine and GEPIA databases. The effect of FCGBP on prognosis was analyzed via Kaplan-Meier plotter and GEPIA. The Tumor Immune Estimation Resource (TIMER) tool was used to determine the correlations of FCGBP expression with tumor immune infiltration. Firstly, FCGBP was highly expressed in glioma and correlated with a worse prognosis. Gene Ontology (GO) and KEGG pathway enrichment analyses revealed that the differentially expressed genes (DEGs) and co-expression genes of FCGBP were mainly involved in the immune response. Furthermore, FCGBP expression was positively associated with multiple immune cells infiltrates as well as the expression levels of multiple immune markers in glioma. FCGBP co-expression networks mostly participated in the regulation of immune response. Finally, immunohistochemistry (IHC) assays were conducted to explore the expression of FCGBP, PD-L1, CCL2 and CD8 in glioma and correlations between them. We found that PDL1 and FCGBP were synchronously upregulated in glioma tissues. These findings revealed a new mechanism by which FCGBP participates in the immune tolerance of glioma, and implied the potential of FCGBP as a therapeutic target or predictive marker for patients.

12.
J Clin Neurosci ; 82(Pt A): 20-25, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33317732

RESUMO

OBJECTIVE: The optimal timing for glioma patients to stop taking antiepileptic drugs (AEDs) and the risk factors of seizure relapse have not been determined. Here, we explored the short-term outcomes and risk factors of seizure relapse in glioma patients after withdrawal of AEDs. METHODS: 91 patients with gliomas who had no seizures at least 2 years after surgery were enrolled in the study. The patients were followed up for 1 year or until the relapse of seizure after AEDs withdrawal. The risk factors of seizure relapse were analyzed by univariate and multivariate analysis. The optimal discrimination point was determined by plotting a receiver operating characteristic (ROC) curve to explore the relationship between the number of risk factors and seizure relapse. RESULTS: 28 patients (30.8%) relapsed during the follow-up period while 63 patients (69.2%) remained seizure-free. Of the 28 relapsed patients, 20 (71.4%) relapsed within the first 6 months after the AEDs withdrawal. Multivariate analyses revealed that subtotal resection (p = 0.026), IDH1 mutation (p = 0.019), and combined use of AEDs (p = 0.037) were independent risk factors for seizure relapse in glioma patients. ROC curve based on the seizure relapse showed that the sensitivity was 0.821 and 1-specificity was 0.238, corresponding to 1.5 independent risk factors for each patient. CONCLUSION: To obtain a favorable outcome for glioma patients with preoperative seizures, only patients with less than two independent risk factors for seizure relapse should consider discontinuing AEDs.


Assuntos
Anticonvulsivantes/uso terapêutico , Neoplasias Encefálicas/complicações , Redução da Medicação , Glioma/complicações , Convulsões , Adolescente , Adulto , Neoplasias Encefálicas/cirurgia , Feminino , Glioma/cirurgia , Humanos , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Recidiva , Estudos Retrospectivos , Fatores de Risco , Convulsões/epidemiologia , Convulsões/etiologia , Convulsões/prevenção & controle , Fatores de Tempo , Adulto Jovem
13.
Aging (Albany NY) ; 12(21): 22122-22138, 2020 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-33186124

RESUMO

Glioma is the most common malignant tumor in the central nervous system. Evidence shows that clinical efficacy of immunotherapy is closely related to the tumor microenvironment. This study aims to establish a microenvironment-related genes (MRGs) model to predict the prognosis of patients with Grade II/III gliomas. Gene expression profile and clinical data of 459 patients with Grade II/III gliomas were extracted from The Cancer Genome Atlas. Then according to the immune/stromal scores generated by the ESTIMATE algorithm, the patients were scored one by one. Weighted gene co-expression network analysis (WGCNA) was used to construct a gene co-expression network to identify potential biomarkers for predicting the prognosis of patients. When adjusting clinical features including age, histology, grading, IDH status, we found that these features were independently associated with survival. The predicted value of the prognostic model was then verified in 440 samples in CGGA part B dataset and 182 samples in CGGA part C dataset by univariate and multivariate cox analysis. The clinical samples of 10 patients further confirmed our signature. Our findings suggested the eight-MRGs signature identified in this study are valuable prognostic predictors for patients with Grade II/III glioma.


Assuntos
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Glioma/genética , Glioma/patologia , Microambiente Tumoral/genética , Algoritmos , Biomarcadores Tumorais/genética , Feminino , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Masculino , Prognóstico , Transcriptoma
14.
Front Pharmacol ; 11: 1102, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32848734

RESUMO

Glioblastoma multiforme (GBM) is the most common intracranial malignancy in adults with the highest degree of malignancy and mortality. Due to its nature of diffuse invasiveness and high migration, GBM lacks an effective treatment strategy and is associated with poor prognosis. SC66 is a novel AKT inhibitor that has been reported to exert antiproliferative activity in many types of cancer cells. However, it remains unclear whether SC66 has antitumor effects in GBM. In this study, we found SC66 obviously suppressed U87 and U251 cell proliferation and EMT- mediated cell migration and invasion. Moreover, SC66 induced GBM cells apoptosis and arrested cell cycle in G0/G1 phase. Furthermore, SC66 also downregulated AKT signaling pathway in a concentration dependent manner. We also found the level of ß-catenin nuclear translocation was prominently downregulated after SC66 treatment. Meanwhile, TCF/LEF luciferase report assay indicated that the activity of TCF/LEF was remarkably suppressed. Elevating ß-catenin activity by using IM12 rescued SC66 inhibition-mediated GBM cell proliferation and metastasis. In addition, SC66 showed significantly suppressed the tumorigenicity compared to the control group in the xenograft mouse model. In conclusion, our study demonstrated that SC66 exerts prominently antitumor efficiency in GBM cells in vivo and in vitro by downregulated AKT/ß-catenin pathway.

15.
Oncol Rep ; 43(3): 773-782, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32020210

RESUMO

Glioblastoma (GBM) is the most common type of primary central nervous system tumor in adults, which has high mortality and morbidity rates, and short survival time, namely <15 months after the diagnosis and application of standard therapy, which includes surgery, radiation therapy and chemotherapy; thus, novel therapeutic strategies are imperative. The activation of the PI3K/AKT signaling pathway plays an important role in GBM. In the present study, U87 and U251 GBM cells were treated with the PI3K/mTORC1/2 inhibitor PQR309, and its effect on glioma cells was investigated. Cell Counting Kit­8 assay, 5­ethynyl­2'­deoxyuridine and colony formation assays revealed dose­ and time­dependent cytotoxicity in glioma cells that were treated with PQR309. Flow cytometry and western blotting revealed that PQR309 can significantly induce tumor cell apoptosis and arrest the cell cycle in the G1 phase. Furthermore, the expression levels of AKT, phosphorylated (p)­AKT, Bcl­2, Bcl­xL, Bad, Bax, cyclin D1, cleaved caspase­3, MMP­9 and MMP­2 were altered. In addition, the migration and invasion of glioma cells, as detected by wound healing, migration and Transwell invasion assays, exhibited a marked suppression after treating the cells with PQR309. These results indicated that PQR309 exerts an antitumor effect by inhibiting proliferation, inducing apoptosis, inducing G1 cell cycle arrest, and inhibiting invasion and migration in human glioma cells. The present study provides evidence supportive of further development of PQR309 for adjuvant therapy of GBM.


Assuntos
Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Inibidores de Fosfoinositídeo-3 Quinase/farmacologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina/antagonistas & inibidores , Alvo Mecanístico do Complexo 2 de Rapamicina/antagonistas & inibidores , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Proteínas de Neoplasias/genética , Fosfatidilinositol 3-Quinases/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais/efeitos dos fármacos
16.
J Clin Neurosci ; 72: 163-168, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31937499

RESUMO

To explore the predictive factors and short-term outcomes of post-surgical seizures in patients with supratentorial low-grade gliomas (LGGs). A consecutive series of 70 supratentorial LGG patients with seizures were reviewed to determine the predictors and short-term outcomes of seizures. Univariate analyses and multivariate logistic regression analyses were performed to determine the predictive factors associated with postoperative seizure outcomes. We identified the preoperative seizure frequency threshold by plotting a receiver operating characteristic curve. A Kaplan-Meier curve was constructed to illustrate the seizure-free survival rate of our cohort over time. 54 patients who remained seizure -free post-surgery were classified into the Engel class I, and the other 16 patients whose seizures relapsed were classified into Engel classes II-IV. Univariate and multivariate logistic regression analyses showed that the preoperative seizure frequency (X2 = 16.069, P = 0.001), extent of resection (x2 = 5.031, P = 0.025), IDH1 mutation (x2 = 4.435, P = 0.035) and adjuvant chemotherapy of temozolomide (X2 = 4.081, P = 0.043) were related to the postoperative short-term seizure outcome. The ROC curve indicated that the area under the curve for the preoperative seizure frequency test was 0.805 (95% confidence interval 0.690-0.920, p < 0.05), which corresponded to an optimal threshold of 2 preoperative seizures. The IDH1WT status and adjuvant chemotherapy with temozolomide were related to a better post-operative seizure outcome. Within the first year after the surgical resection, seizures reoccurred among 16 patients (22.9%) with a mean time of 10.8 months. The preoperative seizure frequency, extent of resection, IDH1 status, and adjuvant chemotherapy with temozolomide were predictive factors of short-term postoperative seizure outcomes for supratentorial LGGs. To obtain a favorable seizure outcome, early intervention and removal are warranted. IDH1 mutation is the predictive biomarker of postoperative seizure outcomes. The adjuvant chemotherapy with temozolomide appears to be associated with better seizure outcomes, and it may be useful in helping to control the postoperative seizures.


Assuntos
Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/cirurgia , Glioma/diagnóstico por imagem , Glioma/cirurgia , Complicações Pós-Operatórias/diagnóstico por imagem , Convulsões/diagnóstico por imagem , Adulto , Estudos de Coortes , Eletroencefalografia/tendências , Feminino , Humanos , Imageamento por Ressonância Magnética/tendências , Masculino , Pessoa de Meia-Idade , Complicações Pós-Operatórias/fisiopatologia , Valor Preditivo dos Testes , Estudos Retrospectivos , Convulsões/tratamento farmacológico , Convulsões/fisiopatologia , Temozolomida/uso terapêutico , Fatores de Tempo , Resultado do Tratamento
17.
Front Oncol ; 9: 1176, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31824836

RESUMO

Glioblastoma (GBM) is the most malignant tumor in the central nervous system and the treatment is still unsatisfactory because the mechanism of the disease remains unclear. The abnormal expression of miRNAs and its target proteins play a crucial role in the development of glioblastoma. In this study, we demonstrated that high expression of miR-9-5p and low expression of forkhead box P2 (FOXP2) were related with better outcome in patients with GBM, and down regulated FOXP2 expression was able to inhibit glioma cells proliferation by cell cycle arrest. Furthermore, we found that FOXP2 was the target protein of miR-9-5p in luciferase assay. The results of this study suggest a novel regulatory mechanism that miR-9-5p can inhibit glioma cells proliferation by downregulating FOXP2.

18.
Cancer Lett ; 446: 1-14, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30639532

RESUMO

Tumor necrosis factor alpha-induced protein 8-like 3 (TIPE3) is closely related to tumourigenesis and development. However, its role in human glioblastoma (GBM) and the underlying mechanisms remain unclear. In this study, we demonstrate that TIPE3 is upregulated in GBM, and its high expression predicts poor prognosis. TIPE3 depletion induces GBM cell apoptosis both in vitro and in vivo. Mechanism studies reveal that TIPE3 inhibits p38 phosphorylation and negatively regulates the p38 MAPK pathway. TIPE3 associates with p38. The nuclear translocation of p38 is blocked by TIPE3 overexpression. And p38 phosphorylation could regulate TIPE3-mediated p38 nuclear-cytopalsmic translocation but does not affect TIPE3-p38 association. Rescue experiments confirm that TIPE3 inhibits GBM cell apoptosis via the p38 MAPK pathway. In conclusion, TIPE3 inhibits p38 phosphorylation and blocks p38 nuclear translocation. This action thus negatively regulates the p38 MAPK pathway and results in GBM cell survival.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Apoptose , Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Animais , Proteínas Reguladoras de Apoptose/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos Endogâmicos BALB C , Camundongos Nus , Técnicas de Transferência Nuclear , Fosforilação , Transdução de Sinais , Regulação para Cima , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
19.
Oncol Rep ; 38(5): 2836-2842, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29048665

RESUMO

Glioblastoma is the most common type of primary brain tumor in adults, with high mortality and morbidity rates. More effective therapeutic strategies are imperative. Previous studies have shown that the known p110-ß-selective inhibitor TGX-221 blocks the activation of PKB/Akt in PTEN-deficient cells. We treated U87 and U251 glioblastoma cells with TGX-221 to determine the effect of TGX-221. We performed a Cell Counting Kit-8 (CCK-8) test, EDU staining and cell cycle distribution analysis and found that TGX-221 inhibited glioblastoma cell proliferation. Next, the effect of TGX-221 on cell apoptosis was investigated using flow cytometry. These results demonstrated that TGX-221 induced apoptosis in glioblastoma cells. Moreover, migration and invasion assays revealed that TGX-221 inhibited human glioblastoma cell migration and invasion. Collectively, our study revealed that TGX-221 could inhibit proliferation and induce apoptosis in glioblastoma cells.


Assuntos
Neoplasias Encefálicas/metabolismo , Classe I de Fosfatidilinositol 3-Quinases/metabolismo , Glioblastoma/metabolismo , Morfolinas/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Pirimidinonas/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Proteínas de Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Humanos , Transdução de Sinais/efeitos dos fármacos
20.
Oncol Rep ; 37(2): 1084-1092, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28035411

RESUMO

Ellagic acid (EA) is present in various fruits and plants and has recently been found to possess anticarcinogenic effects. The objective of this study was to investigate the anti­glioblastoma effect of EA and its mechanisms in vitro and in vivo. We first studied the anticancer activity of EA in U87 and U118 human glioblastoma cell lines. The cell viability and cell proliferation were examined by Cell Counting Kit-8 (CCK-8) assay and 5-ethynyl-2'-deoxyuridine staining respectively. The cell cycle was detected with propidium iodide staining method by flow cytometry and the DNA damage of the cells caused by EA exposure was evaluated by detection of γ-H2AX foci. Then we examined the effect of EA on tumor growth in glioblastoma xenografted mice, and expression of Akt and Notch signaling and their target gene products were detected by immunohistochemistry and western blot analysis. As a result, we found that the cell viability and proliferation of glioblastoma cells treated with EA were significantly suppressed compared with the control; EA significantly increased the proportion of cells in the S phase accompanied by a decrease in the population in the G1 and G2/M phase in both cell lines. Meanwhile, the level of DNA damage in the EA-treated group was significantly higher than that of the control. Treatment of glioblastoma in xenografted mice by EA led to a significant suppression in tumor growth. EA upregulated the expression of E-cadherin and inhibited the expression of Snail, matrix metalloproteinase (MMP)-2 and MMP-9. EA also inhibited the expression of Bcl-2, cyclin D1, cyclin-dependent kinase (CDK)2 and CDK6 in U87 xenograft tissues. In addition, significant suppression of Akt and Notch was found in the xenografts of the tumor-bearing mice treated with EA. These data indicate that EA can suppress glioblastoma proliferation and invasion by inhibiting the Akt and Notch signaling pathways, which suggests that EA may be beneficial for the treatment of glioblastoma.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Neoplasias do Sistema Nervoso Central/tratamento farmacológico , Ácido Elágico/farmacologia , Glioblastoma/tratamento farmacológico , Animais , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Neoplasias do Sistema Nervoso Central/metabolismo , Neoplasias do Sistema Nervoso Central/patologia , Dano ao DNA/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Glioblastoma/metabolismo , Glioblastoma/patologia , Histonas/metabolismo , Humanos , Camundongos Endogâmicos BALB C , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA