Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
J Neurooncol ; 166(1): 113-127, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38191954

RESUMEN

PURPOSE: Endosome associated trafficking regulator 1 (ENTR1) is a novel endosomal protein, which can affect multiple cellular biological behavior by remodeling plasma membrane structures. However, little is known regarding its function and underlying mechanisms in glioblastoma multiforme. METHODS: Expression profile and clinical signature were obtained from The Public Database of human tumor. Immunohistochemical staining and western blotting assays were used to measure ENTR1 expression level. Human primary GBM tumor cells and human GBM cell lines A172, U87 and U251 were used to clarify the precise role of ENTR1. CCK-8 assays, wound healing and transwell invasion assays were designed to investigate cell viability, invasion and migration of GBM cells, respectively. Underlying molecular mechanisms of ENTR1 were determined via RNA-seq analysis. Tumor formation assay was used to validate the influence of ENTR1 in vivo. RESULTS: Compared with normal brain tissues, ENTR1 was highly expressed in gliomas and correlated with malignant grades of gliomas and poor overall survival time. The proliferation and invasion of GBM cells could be weaken and the sensitivity to temozolomide (TMZ) chemotherapy increased after knocking down ENTR1. Overexpression of ENTR1 could reverse this effect. RNA-seq analysis showed that tumor necrosis factor (TNF) signaling pathway might be a putative regulatory target of ENTR1. Tumor formation assay validated that ENTR1 was a significant factor in tumor growth. CONCLUSION: Our results indicated that ENTR1 played an important role in cell proliferation, invasion and chemotherapeutic sensitivity of GBM, suggesting that ENTR1 might be a novel prognostic marker and significant therapeutic target for GBM.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Humanos , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular , Endosomas/metabolismo , Endosomas/patología , Regulación Neoplásica de la Expresión Génica , Glioblastoma/patología , Transducción de Señal
2.
Int J Clin Oncol ; 25(6): 1055-1066, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32103382

RESUMEN

BACKGROUND: Filamin A is the most widely expressed isoform of filamin in mammalian tissues. It can be hydrolyzed by Calpain, producing a 90-kDa carboxyl-terminal fragment (ABP90). Calpeptin is a chemical inhibitor of Calpain, which can inhibit this effect. It has been shown that ABP90 acts as a transcription factor which is involved in mediating cell signaling. However, the significance of ABP90 and its clinical signature with underlying mechanisms have not been well studied in glioblastoma multiforme (GBM). METHODS: ABP90 protein was measured in 36 glioma patients by Western blot. Human GBM cell lines U87 and A172 were used to clarify the precise role of ABP90. CCK-8 assay was used to analyze the cell viability. Transwell invasion assay and wound healing assay were used to analyze the migration and invasion. Expression of matrix metalloproteinase 2/tissue inhibitors of metalloproteinase 2 (MMP2/TIMP2) protein was analyzed by Western blot. RESULTS: ABP90 protein expression was lower in GBM tissues. The patients with low ABP90 protein expression had a shorter OS time (p = 0.046). After being treated with Calpain, the expression of ABP90 was upregulated, which led to a decline of cell viability, enhanced the efficacy of temozolomide and restrained the cell invasion. Calpeptin could inhibit the effect. The mechanism might be involved in the balance of MMP2/TIMP2. CONCLUSIONS: Our present data suggest that ABP90 expression is a significant prognostic factor and may play an important role in cell viability, chemotherapeutic sensitivity and invasion of GBM.


Asunto(s)
Neoplasias Encefálicas/patología , Calpaína/farmacología , Proliferación Celular/efectos de los fármacos , Filaminas/metabolismo , Glioblastoma/patología , Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/metabolismo , Calpaína/antagonistas & inhibidores , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Dipéptidos/farmacología , Glioblastoma/metabolismo , Humanos , Metaloproteinasa 2 de la Matriz/metabolismo , Invasividad Neoplásica , Pronóstico , Temozolomida/farmacología , Inhibidor Tisular de Metaloproteinasa-2/metabolismo
3.
Biochem Biophys Res Commun ; 505(1): 20-28, 2018 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-30227940

RESUMEN

The splicing factor SPF45 (RBM17) is a well-known component of the spliceosome that is involved in alternative splicing. RBM17 is frequently overexpressed in many tumors and plays a crucial role in cancer progression and drug resistance. However, the role of RBM17 in the development of glioma has not been thoroughly elucidated to date. In the present study, we found that RBM17 was overexpressed in glioma and that a high level of expression of RBM17 was closely associated with a poor prognosis in glioma patients. We investigated the effect of RBM17 on apoptosis, cell growth and cell cycle indexes and the activation of apoptosis signaling by shRNA in human U87 and U251 glioma cells. The downregulated expression of RBM17 mRNA was accompanied by the induction of cell cycle arrest, and apoptosis, reduced cell proliferation in the two cell lines, and reduced cell survival, as measured by the increased activation of caspase-3, caspase-9, and PARP (poly ADP-ribose polymerase). Furthermore, in subcutaneous U87 cell xenograft tumors in nude mice, intradermal administration of an shRNA targeting RBM17 significantly downregulated RBM17 expression in vivo and was accompanied by the suppressed growth of glioma. To the best of our knowledge, our results are the first to confirm that RBM17 functions in promoting cell proliferation, affecting the cell cycle, and inducing apoptosis in human glioma cells both in vitro and in vivo. These results indicate that RBM17 may be a therapeutic target in the clinical management of glioma.


Asunto(s)
Apoptosis/genética , Neoplasias Encefálicas/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Glioma/genética , Factores de Empalme de ARN/genética , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Puntos de Control del Ciclo Celular/genética , Línea Celular Tumoral , Progresión de la Enfermedad , Glioma/metabolismo , Glioma/patología , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Pronóstico , Interferencia de ARN , Factores de Empalme de ARN/metabolismo , Análisis de Supervivencia , Trasplante Heterólogo
4.
Front Pharmacol ; 13: 1076958, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36506527

RESUMEN

Paclitaxel is an herbal active ingredient used in clinical practice that shows anti-tumor effects. However, its biological activity, mechanism, and cancer cell-killing effects remain unknown. Information on the chemical gene interactions of paclitaxel was obtained from the Comparative Toxicogenomics Database, SwishTargetPrediction, Binding DB, and TargetNet databases. Gene expression data were obtained from the GSE4290 dataset. Differential gene analysis, Kyoto Encyclopedia of Genes and Genomes, and Gene Ontology analyses were performed. Gene set enrichment analysis was performed to evaluate disease pathway activation; weighted gene co-expression network analysis with diff analysis was used to identify disease-associated genes, analyze differential genes, and identify drug targets via protein-protein interactions. The Molecular Complex Detection (MCODE) analysis of critical subgroup networks was conducted to identify essential genes affected by paclitaxel, assess crucial cluster gene expression differences in glioma versus standard samples, and perform receiver operator characteristic mapping. To evaluate the pharmacological targets and signaling pathways of paclitaxel in glioblastoma, the single-cell GSE148196 dataset was acquired from the Gene Expression Omnibus database and preprocessed using Seurat software. Based on the single-cell RNA-sequencing dataset, 24 cell clusters were identified, along with marker genes for the two different cell types in each cluster. Correlation analysis revealed that the mechanism of paclitaxel treatment involves effects on neurons. Paclitaxel may affect glioblastoma by improving glucose metabolism and processes involved in modulating immune function in the body.

5.
Front Neurol ; 13: 822735, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35250826

RESUMEN

BACKGROUND: Glioblastoma (GBM) is one of the most malignant types of tumors in the central nervous system, and the 5-year survival remains low. Several studies have shown that preoperative peripheral blood tests and preoperative conventional Magnetic Resonance Imaging (MRI) examinations affect the prognosis of GBM patients. Therefore, it is necessary to construct a risk score based on a preoperative peripheral blood test and conventional MRI and develop a multielement prognostic nomogram for GBM. METHODS: This study retrospectively analyzed 131 GBM patients. Determination of the association between peripheral blood test variables and conventional MRI variables and prognosis was performed by univariate Cox regression. The nomogram model, which was internally validated using a cohort of 56 GBM patients, was constructed by multivariate Cox regression. RNA sequencing data from Gene Expression Omnibus (GEO) and Chinese Glioma Genome Atlas (CGGA datasets were used to determine peripheral blood test-related genes based on GBM prognosis. RESULTS: The constructed risk score included the neutrophil/lymphocyte ratio (NLR), lymphocyte/monocyte ratio (LMR), albumin/fibrinogen (AFR), platelet/lymphocyte ratio (PLR), and center point-to-ventricle distance (CPVD). A final nomogram was developed using factors associated with prognosis, including age, sex, the extent of tumor resection, IDH mutation status, radiotherapy status, chemotherapy status, and risk. The Area Under Curve (AUC) values of the receiver operating characteristic curve (ROC) curve were 0.876 (12-month ROC), 0.834 (24-month ROC) and 0.803 (36-month ROC) in the training set and 0.906 (12-month ROC), 0.800 (18-month ROC) and 0.776 (24-month ROC) in the validation set. In addition, vascular endothelial growth factor A (VEGFA) was closely associated with NLR and LMR and identified as the most central negative gene related to the immune microenvironment and influencing immune activities. CONCLUSION: The risk score was established as an independent predictor of GBM prognosis, and the nomogram model exhibit appropriate predictive power. In addition, VEGFA is the key peripheral blood test-related gene that is significantly associated with poor prognosis.

6.
Front Neurol ; 13: 902402, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35968275

RESUMEN

Background: Glioblastoma multiforme (GBM) is a common malignant brain tumor with high mortality. It is urgently necessary to develop a new treatment because traditional approaches have plateaued. Purpose: Here, we identified an immune-related gene (IRG)-based prognostic signature to comprehensively define the prognosis of GBM. Methods: Glioblastoma samples were selected from the Chinese Glioma Genome Atlas (CGGA). We retrieved IRGs from the ImmPort data resource. Univariate Cox regression and LASSO Cox regression analyses were used to develop our predictive model. In addition, we constructed a predictive nomogram integrating the independent predictive factors to determine the one-, two-, and 3-year overall survival (OS) probabilities of individuals with GBM. Additionally, the molecular and immune characteristics and benefits of ICI therapy were analyzed in subgroups defined based on our prognostic model. Finally, the proteins encoded by the selected genes were identified with liquid chromatography-tandem mass spectrometry and western blotting (WB). Results: Six IRGs were used to construct the predictive model. The GBM patients were categorized into a high-risk group and a low-risk group. High-risk group patients had worse survival than low-risk group patients, and stronger positive associations with multiple tumor-related pathways, such as angiogenesis and hypoxia pathways, were found in the high-risk group. The high-risk group also had a low IDH1 mutation rate, high PTEN mutation rate, low 1p19q co-deletion rate and low MGMT promoter methylation rate. In addition, patients in the high-risk group showed increased immune cell infiltration, more aggressive immune activity, higher expression of immune checkpoint genes, and less benefit from immunotherapy than those in the low-risk group. Finally, the expression levels of TNC and SSTR2 were confirmed to be significantly associated with patient prognosis by protein mass spectrometry and WB. Conclusion: Herein, a robust predictive model based on IRGs was developed to predict the OS of GBM patients and to aid future clinical research.

7.
Seizure ; 93: 120-126, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34740141

RESUMEN

PURPOSE: This study aim to identify the clinical risk factors of and to develop a radiomics-based predictive model for early postoperative seizure. METHODS: We retrospectively assessed 322 operative patients with meningioma who met the inclusion criteria from January 2014 to December 2016 at The First Affiliated Hospital of Wenzhou Medical University. Univariate and multivariate analyses were performed to determine the predictive value of clinical variables. Magnetic resonance imaging (MRI) was performed to obtain the radiomic score (Rscore) for early postoperative seizure. Radiological features were evaluated using the AK software. The minimal redundancy (mRMR) and least absolute shrinkage and selection operator (LASSO) methods were used to assess for radiomic features, and the Rscore was obtained based on radiomic characteristics using a specific formula. RESULTS: In total, 260 patients who met the inclusion criteria were finally enrolled in this study. Among them, 20 experienced early postoperative seizure. Logistic regression analysis showed that Rscore was associated with a significantly high risk of seizure (p<0.000). Receiver operating characteristic (ROC) curve analysis revealed that the area under the ROC curve of the Rscore was 0.92 (95% confidence interval: 0.853-0.987). The model had a high accuracy for predicting early postoperative seizure. CONCLUSIONS: The Rscore was found to be associated with a high risk of early postoperative seizures. Thus, a higher Rscore (>-1.644) can identify high-risk patients requiring intensive care.


Asunto(s)
Neoplasias Meníngeas , Meningioma , Humanos , Imagen por Resonancia Magnética , Neoplasias Meníngeas/complicaciones , Neoplasias Meníngeas/diagnóstico por imagen , Neoplasias Meníngeas/cirugía , Meningioma/complicaciones , Meningioma/diagnóstico por imagen , Meningioma/cirugía , Estudios Retrospectivos , Factores de Riesgo , Convulsiones/diagnóstico por imagen , Convulsiones/etiología
8.
Front Pharmacol ; 12: 775680, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34970146

RESUMEN

Brusatol (Bru), a Chinese herbal extract, has a variety of anti-tumor effects. However, little is known regarding its role and underlying mechanism in glioblastoma cells. Here, we found that Bru could inhibit the proliferation of glioblastoma cells in vivo and in vitro. Besides, it also had an inhibitory effect on human primary glioblastoma cells. RNA-seq analysis indicated that Bru possibly achieved these effects through inhibiting the expression of extracellular matrix protein 1 (ECM1). Down-regulating the expression of ECM1 via transfecting siRNA could weaken the proliferation and invasion of glioblastoma cells and promote the inhibitory effect of Bru treatment. Lentivirus-mediated overexpression of ECM1 could effectively reverse this weakening effect. Our findings indicated that Bru could inhibit the proliferation and invasion of glioblastoma cells by suppressing the expression of ECM1, and Bru might be a novel effective anticancer drug for glioblastoma cells.

9.
Int J Endocrinol ; 2021: 9930059, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34504526

RESUMEN

OBJECTIVE: Dopamine agonists (DAs) are recommended as the first-line treatment for prolactinomas; however, tumour recurrence after drug withdrawal remains a clinical problem. Recent studies have reported high remission rates via surgery in microprolactinomas. The aim of this systematic review and meta-analysis was to compare the clinical result of DA treatment with surgery as initial therapy in patients with treatment-naive microprolactinoma. METHODS: A comprehensive literature search for studies and reports regarding microprolactinoma patients treated with DAs and/or surgery published between January 1970 and November 2020 was conducted using four electronic databases (PubMed, Embase, Google Scholar, and the Cochrane Library). Clinical treatment outcome was evaluated by the biochemical remission of serum prolactin level to normal after treatment. The I 2 statistic was used to quantify heterogeneity. Pooled data were analysed according to a random effect model. RESULTS: Eighteen studies with 661 patients were included for analysis. The DA treatment group achieved a higher remission rate at ≥12 months follow-up (96% vs. 86%; P=0.019). Surgery showed a higher remission rate than the DA treatment group after the treatment withdrawal (78% vs. 44%; P=0.003). Patients with preoperative prolactin level of ≤200 ng/mL had a higher remission rate than patients with preoperative prolactin level of >200 ng/mL (92% vs. 40%; P=0.029). CONCLUSION: Surgery showed a high remission rate in treatment-naive microprolactinoma patients after treatment withdrawal and may be an alternative first-line treatment strategy in addition to DAs, particularly in patients with a preoperative prolactin level of ≤200 ng/mL.

10.
Front Oncol ; 10: 585313, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33123487

RESUMEN

Aim: This study aimed to identify the independent risk factors of recurrence in patients undergoing primary resection of meningioma and construct a scoring system for the prediction of the risk of postoperative recurrence. Materials and Methods: The clinical data of 591 patients who underwent primary surgical resection for meningioma at the First Affiliated Hospital of Wenzhou Medical University between November 2010 and December 2016 were retrospectively reviewed. The clinical, radiological, and pathological characteristics were evaluated, and the independent risk factors for recurrence were identified via receiver operating characteristic (ROC) curve and logistic analyses. A scoring system that included these independent risk factors was used to construct a risk-predicting model that was evaluated via a ROC curve analysis. The recurrences of different subgroups were observed by Kaplan-Meier's curves. Results: The clinical data of 392 patients with meningioma were used to construct the scoring system. The logistic analysis showed that sex (OR = 2.793, 95% CI = 1.076-7.249, P = 0.035), heterogeneous tumor enhancement (OR = 4.452, 95% CI = 1.714-11.559, P = 0.002), brain invasion (OR = 2.650, 95% CI = 1.043-6.733, P = 0.041), Simpson's removal grade (OR = 5.139, 95% CI = 1.355-19.489, P = 0.016), and pathological grade (OR = 3.282, 95% CI = 1.123-9.595, P = 0.030) were independent risk factors for recurrence. A scoring system was developed and used to divide the patients into the following four subgroups: subgroup 1 with scores of 0-75 (n = 249), subgroup 2 with scores of 76-154 (n = 88), subgroup 3 with scores of 155-215 (n = 46), and subgroup 4 with scores of 216-275 (n = 9). The incidences of recurrence in each subgroup were as follows: subgroup 1, 1.2%; subgroup 2, 5.7%; subgroup 3, 26.1%; and subgroup 4, 66.7% (P < 0.001). The scoring system reliably predicted the postoperative recurrence of meningioma with a high area under the ROC curve. Conclusions: Our scoring system is a simple and reliable instrument for identifying meningioma patients at risk of postoperative recurrence and could help in optimizing individualized clinical treatment.

11.
Mol Med Rep ; 19(3): 1849-1857, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30592284

RESUMEN

Insights into the roles of microRNAs (miRNAs/miRs) in development and disease, particularly in cancer, have made miRNAs attractive tools and targets for novel therapeutic approaches in the treatment of glioma. miR­34a, as a well­known tumor suppressor miRNA, is closely related with cellular senescence. Mesenchymal stem cells (MSCs) are a major component of the tumor microenvironment and possess the ability to deliver exogenous miRs to glioma cells to exert anti­tumor effects. The present study investigated whether modified MSCs with miR­34a possess an anti­tumor function in glioma cells. A Transwell system was used to co­culture U87 glioma cells and MSCs overexpressing miR­34a, and cell proliferation and senescence assessed. The expression of senescence­related genes p53, Cdkn1a, and Cdkn2c were tested using reverse transcription­quantitative polymerase chain reaction and protein expression levels of sirtuin 1 (SIRT1) and γ­H2A histone family, member X were detected by western blotting. Telomerase activity of U87 cells was examined using the Telo TAGGG Telomerase PCR ELISA PLUS kit. The results demonstrated that the delivered exogenous miR­34a from MSCs significantly decreased expression of the target gene SIRT1. In addition, the delivered miR­34a decreased the proliferation of glioma cells and provoked the expression of senescence­related genes p53, Cdkn1a, and Cdkn2c. In addition, upregulation of miR­34a induced DNA damage, shortened telomere length and impaired telomerase activity. However, these pro­senescent effects were reversed by forced SIRT1 upregulation. In conclusion, the results demonstrated a novel role for miR­34a, inducing glioma cell senescence, whereas miR­34a modulation of SIRT1, inducing DNA damage, is crucial for miRNA replacement therapy in glioma treatment.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Senescencia Celular , Daño del ADN , Glioma/genética , Glioma/patología , Células Madre Mesenquimatosas/metabolismo , MicroARNs/metabolismo , Línea Celular Tumoral , Senescencia Celular/genética , Técnicas de Cocultivo , Regulación hacia Abajo/genética , Humanos , MicroARNs/genética , Sirtuina 1/genética , Sirtuina 1/metabolismo , Telómero/metabolismo
12.
Int J Mol Med ; 42(2): 1054-1063, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29749459

RESUMEN

Stroke is the second most common cause of death worldwide, and thus, it imposes great financial burdens on both individuals and society. Mesenchymal stem cell (MSC) therapy is a promising approach for ischemic brain injury. However, MSC treatment potential is progressively reduced with age, limiting their therapeutic efficacy for brain repair post­stroke. C1q and tumor necrosis factor­related protein 9 (CTRP9) is a novel cytoprotective cytokine with antioxidant effects, which is highly expressed in brain tissue. The present study tested the hypothesis that CTRP9 might act as an antisenescence factor to promote the rejuvenation of aged MSCs. MSCs were isolated from the bone marrow of young (8­weeks­old) and aged (18­months­old) male C57BL/6 mice. Cell proliferation was measured by Cell Counting Kit­8 assay and cell viability was determined by MTT assay. Gene expression levels of interleukin (IL)­6 and IL­10 were evaluated with reverse transcription­quantitative polymerase chain reaction, and secretion of vascular endothelial growth factor, basic fibroblast growth factor, hepatocyte growth factor, and insulin­like growth factor were measured by ELISA. The expression levels of proteins in the peroxisome proliferator­activated receptor γcoactivator (PGC)­1α/AMP­activated protein kinase (AMPK) signaling pathway were investigated with western blotting. Oxidative stress was evaluated by detecting mitochondrial membrane potential, reactive oxygen species, superoxide dismutase activity and malondialdehyde. MSCs isolated from aged mice exhibited reduced proliferation and viability, and impaired immunoregulatory and paracrine abilities, compared with MSCs from younger mice. CTRP9 had a significant antisenescence effect in aged MSCs by activating PGC­1α/AMPK signaling and decreasing the oxidative response. Silencing either PGC­1α or AMPK abolished the above effects of CTRP9. These results suggest that CTRP9 may have a critical role in cellular senescence by facilitating stem cell rejuvenation, and may therefore have the potential to enhance the efficacy of stem cell therapy.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Adiponectina/metabolismo , Senescencia Celular , Glicoproteínas/metabolismo , Células Madre Mesenquimatosas/citología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Transducción de Señal , Animales , Células Cultivadas , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA