Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
BMC Public Health ; 24(1): 1238, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38711042

ABSTRACT

BACKGROUND: We conducted this meta-analysis to investigate the potential association between maternal smoking, alcohol and caffeinated beverages consumption during pregnancy and the risk of childhood brain tumors (CBTs). METHODS: A thorough search was carried out on PubMed, Embase, Web of Science, Cochrane Library, and China National Knowledge Internet to identify pertinent articles. Fixed or random effects model was applied to meta-analyze the data. RESULTS: The results suggested a borderline statistically significant increased risk of CBTs associated with maternal smoking during pregnancy (OR 1.04, 95% CI 0.99-1.09). We found that passive smoking (OR 1.12, 95% CI 1.03-1.20), rather than active smoking (OR 1.00, 95% CI 0.93-1.07), led to an increased risk of CBTs. The results suggested a higher risk in 0-1 year old children (OR 1.21, 95% CI 0.94-1.56), followed by 0-4 years old children (OR 1.12, 95% CI 0.97-1.28) and 5-9 years old children (OR 1.11, 95% CI 0.95-1.29). This meta-analysis found no significant association between maternal alcohol consumption during pregnancy and CBTs risk (OR 1.00, 95% CI 0.80-1.24). An increased risk of CBTs was found to be associated with maternal consumption of caffeinated beverages (OR 1.16, 95% CI 1.07-1.26) during pregnancy, especially coffee (OR 1.18, 95% CI 1.00-1.38). CONCLUSIONS: Maternal passive smoking, consumption of caffeinated beverages during pregnancy should be considered as risk factors for CBTs, especially glioma. More prospective cohort studies are warranted to provide a higher level of evidence.


Subject(s)
Alcohol Drinking , Brain Neoplasms , Caffeine , Observational Studies as Topic , Prenatal Exposure Delayed Effects , Humans , Pregnancy , Female , Alcohol Drinking/adverse effects , Alcohol Drinking/epidemiology , Prenatal Exposure Delayed Effects/epidemiology , Brain Neoplasms/epidemiology , Brain Neoplasms/chemically induced , Brain Neoplasms/etiology , Child , Child, Preschool , Caffeine/adverse effects , Infant , Infant, Newborn , Smoking/epidemiology , Smoking/adverse effects , Risk Factors , Beverages/adverse effects
3.
Cell Death Dis ; 15(1): 45, 2024 01 13.
Article in English | MEDLINE | ID: mdl-38218875

ABSTRACT

Interferon-induced transmembrane protein 3 (IFITM3) has been previously verified to be an endosomal protein that prevents viral infection. Recent findings suggested IFITM3 as a key factor in tumor invasion and progression. To clarify the role and molecular mechanism of IFITM3 in Glioblastoma multiforme (GBM) progression, we investigated the expression of IFITM3 in glioma datasets culled from The Cancer Genome Atlas (TCGA) and Chinese Glioma Genome Atlas (CGGA). Primary GBM stem cells (GSCs) were cultured and identified in vitro. Loss-of-function and gain-of-function experiments were established by using shRNAs and lentiviral vectors targeting IFITM3. Co-culture system of GSCs and vascular endothelial cells was constructed in a Transwell chamber. Tube formation and spheroid-based angiogenesis assays were performed to determine the angiogenic capacity of endothelial cells. Results revealed that IFITM3 is elevated in GBM samples and predictive of adverse outcome. Mechanistically, GSCs-derived IFITM3 causes activation of Jak2/STAT3 signaling and leads to robust secretion of bFGF into tumor environment, which eventually results in enhanced angiogenesis. Taken together, these evidence indicated IFITM3 as an essential factor in GBM angiogenesis. Our findings provide a new insight into mechanism by which IFITM3 modulates GBM angiogenesis.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Humans , Glioblastoma/pathology , Endothelial Cells/metabolism , Angiogenesis , Glioma/genetics , Signal Transduction , Stem Cells/metabolism , Brain Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
4.
J Neurooncol ; 165(1): 79-90, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37819535

ABSTRACT

BACKGROUND: The efficacy of current immunotherapeutic strategies for patients with glioblastoma multiforme (GBM) remains unsatisfactory. The purpose of this study was to investigate the correlation between tumor necrosis factor alpha-induced protein 2 (TNFAIP2) and immunogenic cell death (ICD) in GBM, and to examine the effect of TNFAIP2 knockdown and anti-PD-1 combination treatment in a mouse glioma model. METHODS: The CGGA and TCGA databases were used to explore the possible function of TNFAIP2 in GBM. Multiplex immunohistochemistry (mIHC) staining was performed to detect the immune infiltration of tissues. Western blot, quantitative real-time polymerase chain reaction (qRT-PCR), flow cytometry, and enzyme linked immunosorbent assay (ELISA) were utilized to detect the release of damage-associated molecular patterns (DAMPs) and the activation of the immune response. A mouse glioma model was applied to examine the induction of immune response. RESULTS: In vitro and in vivo studies demonstrated that TNFAIP2 knockdown increased the surface exposure of calreticulin (CALR), heat shock protein 70 kDa (HSP70), and heat shock protein 90 kDa (HSP90) in GBM cell lines, thereby inducing immunogenic cell death (ICD). Importantly, the study found that TNFAIP2 knockdown in combination with anti-PD-1 therapy significantly improved the overall survival of glioma in a mouse model. CONCLUSIONS: TNFAIP2 knockdown induces ICD by downregulating TNFAIP2 in GBM. In addition, TNFAIP2 knockdown sensitized glioma to anti-PD-1 therapy. Hence, targeting TNFAIP2 alone or in combination with anti-PD-1 therapy may be a potential strategy for GBM treatment through ICD.


Subject(s)
Glioblastoma , Glioma , Animals , Mice , Humans , Glioblastoma/pathology , Immunogenic Cell Death , Glioma/pathology , Cell Line , Disease Models, Animal , Cell Line, Tumor , Cytokines
5.
Int J Pharm ; 636: 122851, 2023 Apr 05.
Article in English | MEDLINE | ID: mdl-36931535

ABSTRACT

The postoperative thrombus attached to the damaged blood vessels severely obstructs drugs from crossing the damaged blood-brain barrier (BBB) and targeting residual glioma cells around surgical margins, leading to glioblastoma (GBM) recurrence. A thrombus-bypassing, BBB-crossing, and surgical margin-targeted nanodrug is needed to address this phenomenon. Encouraged by the intrinsic damaged vascular endothelium chemotaxis of platelets, a platelet membrane-coated nanodrug (PM-HDOX) delivering doxorubicin (DOX) for postoperative GBM treatment is proposed and systematically investigated. Because surgery damages the vascular endothelium on the BBB around the surgical margin, the platelet membrane coating endows PM-HDOX with its inherent capacity to cross the broken BBB and target the surgical margin. Moreover, preoperative administration combined with fast-targeted PM-HDOX can realize the potential of bypassing thrombus. In GBM resection models, PM-HDOX with preoperative administration demonstrated significantly enhanced BBB-crossing and surgical margin-targeted efficacy. In particular, the PM-HDOX intensities around the surgical margins of the preoperative administration group were more than twice that of the postoperative administration group due to bypassing the thrombus formed in the broken BBB. In the antitumor experiment, the preoperative administration of PM-HDOX significantly inhibited the growth of postoperative residual tumors and prolonged the median survival time of mice. In conclusion, preoperative administration of a biomimetic platelet nanodrug can be an efficient and promising drug delivery strategy for residual GBM after surgery.


Subject(s)
Brain Neoplasms , Glioblastoma , Nanoparticles , Thrombosis , Mice , Animals , Margins of Excision , Blood Platelets/pathology , Biomimetics , Brain Neoplasms/drug therapy , Brain Neoplasms/surgery , Brain Neoplasms/pathology , Drug Delivery Systems , Blood-Brain Barrier , Glioblastoma/drug therapy , Glioblastoma/surgery , Glioblastoma/pathology , Thrombosis/drug therapy , Nanoparticles/therapeutic use , Cell Line, Tumor
6.
Am J Transl Res ; 13(8): 8611-8631, 2021.
Article in English | MEDLINE | ID: mdl-34539983

ABSTRACT

Glioma, a highly aggressive neuroepithelial malignant brain tumor, is associated with high disability and recurrence rates. Enhancer RNA (eRNA) plays a significant role in tumor proliferation and metastasis; however, their functions in gliomas need further evaluation. We used the computational pipeline, PreSTIGE, to predict tissue-specific enhancer-derived RNAs and the underlying regulatory genes. Using data retrieved from the TCGA and CGGA databases, a LASSO regression analysis and multiCox proportional hazards regression analyses were performed to determine the hub eRNAs associated with glioma prognosis. Quantitative reverse transcription PCR was performed on the glioma samples to evaluate the expression characteristics of the identified hub eRNAs. To construct a risk signature, we selected three eRNAs, including CRNDE, MRPS31P5, and LINC00844, for their significant prognostic values. The predictive value of the risk signature was validated using the CGGA and Rembrandt cohorts. Apart from the risk signature, the nomogram performed well at predicting OS in glioma patients. An eRNA-target gene regulatory network was established, which we evaluated using a target gene enrichment analysis. Pathway and gene ontology (GO) analyses demonstrated that the risk signature is associated with mRNA processing and spliceosome in glioma. Furthermore, we found that hub eRNAs potentially regulate the expressions of numerous splicing factors, such as MOV10 and SEC31B, and are correlated with prognosis-associated alteration splicing (AS). In conclusion, we established a risk signature that comprises three eRNAs, which can accurately be utilized as targets to predict prognosis in glioma patients.

7.
Biosci Rep ; 41(5)2021 05 28.
Article in English | MEDLINE | ID: mdl-33969375

ABSTRACT

Synapse and synapse-associated proteins (SAPs) play critical roles in various neurodegeneration diseases and brain tumors. However, in lower-grade gliomas (LGG), SAPs have not been explored systematically. Herein, we are going to explore SAPs expression profile and its clinicopathological significance in LGG which can offer new insights to glioma therapy. In the present study, we integrate a list of SAPs that covered 231 proteins with synaptogenesis activity and post synapse formation. The LGG RNA-seq data were downloaded from GEO, TCGA and CGGA database. The prognosis associated SAPs in key modules of PPI (protein-protein interaction networks) was regarded as hub SAPs. Western blot, quantitative reverse transcription PCR (qRT-PCR) and immunochemistry results from HPA database were used to verify the expression of hub SAPs. There were 68 up-regulated SAPs and 44 down-regulated SAPs in LGG tissue compared with normal brain tissue. Data from function enrichment analysis revealed functions of differentially expressed SAPs in synapse organization and glutamatergic receptor pathway in LGGs. Survival analysis revealed that four SAPs, GRIK2, GABRD, GRID2 and ARC were correlate with the prognosis of LGG patients. Interestingly, we found that GABRD were up-regulated in LGG patients with seizures, indicating that SAPs may link to the pathogenesis of seizures in glioma patients. The four-SAPs signature was revealed as an independent prognostic factor in gliomas. Our study presented a novel strategy to assess the prognostic risks of LGGs, based on the expression of SAPs.


Subject(s)
Biomarkers, Tumor/standards , Brain Neoplasms/genetics , Glioma/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Gene Regulatory Networks , Glioma/metabolism , Glioma/pathology , Humans , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Predictive Value of Tests , Protein Interaction Maps , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Receptors, Glutamate/genetics , Receptors, Glutamate/metabolism , Receptors, Kainic Acid/genetics , Receptors, Kainic Acid/metabolism , GluK2 Kainate Receptor
8.
Genet Test Mol Biomarkers ; 25(5): 334-345, 2021 May.
Article in English | MEDLINE | ID: mdl-33970702

ABSTRACT

Background: Vascular endothelial growth factors (VEGFs) are important for glioblastoma multiforme (GBM) growth and development. However, the effects of VEGF-targeting drugs in primary GBM remain poorly understood. Aim: We aimed to explore the key genes correlated with VEGF expression and prognosis and elucidate their potential implications in GBM anti-VEGF therapy. Materials and Methods: RNA-seq data with the corresponding clinicopathological information was retrieved from The Cancer Genome Atlas and the Chinese Glioma Genome Atlas. Weighted gene coexpression network analyses was performed on differentially expressed genes to construct coexpression modules and investigate their correlation with VEGFs. Functional enrichment analyses were performed based on the coexpressed genes from the most promising modules. CytoHubba and Kaplan-Meier analyses were implemented to identify the key genes in the modules of interest. The oncomine database, quantitative reverse transcription PCR, and the Human Protein Atlas were used to investigate the expression characteristics of the identified key genes. Results: Four modules (cyan, green, purple, and tan) correlated significantly with VEGF expression. Enrichment analyses suggested that extracellular matrix-receptor interaction, growth factor binding, and the PI3K-Akt pathways were involved in VEGF expression. Four hub genes (COL6A1, SNRPG, COL3A1, and AHI1) associated with VEGF were identified. Among them, COL6A1 was regarded as the key gene associated with anti-VEGF therapy. Further, COL6A1 was upregulated in GBM compared to that in normal brain tissues. COL6A1 overexpression was associated with a poor prognosis. Conclusion: COL6A1 was identified as the key gene associated with anti-VEGF therapy and may provide novel insight into GBM targeted therapy.


Subject(s)
Collagen Type VI/metabolism , Glioblastoma/genetics , Vascular Endothelial Growth Factors/antagonists & inhibitors , Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , China , Collagen Type VI/genetics , Databases, Genetic , Endothelial Growth Factors/genetics , Gene Expression/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/genetics , Gene Regulatory Networks , Glioblastoma/metabolism , Glioma/genetics , Humans , Kaplan-Meier Estimate , Phosphatidylinositol 3-Kinases/genetics , Prognosis , Protein Interaction Maps , Transcriptome/genetics , Vascular Endothelial Growth Factors/drug effects , Vascular Endothelial Growth Factors/metabolism , snRNP Core Proteins/genetics
9.
Br J Neurosurg ; 35(4): 444-450, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33305648

ABSTRACT

Introduction: Several studies explored the effects of exposure to radiofrequency-electromagnetic field (RF-EMF) and extremely low frequency (ELF) EMF emitted from mobile phones on meningioma among adults. However, the results could not reach an agreement. This meta-analysis was conducted to confirm the relationship between adult meningioma risk and the use of a wireless phone. Methods: Pertinent studies were identified by searching PubMed and Embase up to August 2018. The random- or fixed-effects model was used to combine the results depending on the heterogeneity of the analysis. The publication bias was evaluated using Egger's regression asymmetry test. The subgroup analysis was performed by time since the first use of wireless phone and laterality (ipsilateral/contralateral). Results: Eight studies were enrolled in this meta-analysis. The pooled results suggested that the ever use of wireless phone led to a borderline decreased adult meningioma risk [odds ratio (OR) 0.90; 95% confidence interval (CI) 0.83-0.99] with no heterogeneity (I2 = 5.3%; p = 0.391). A decreased risk of meningioma was seen in short-term (OR = 0.85; 95% CI = 0.77-0.94) users. Neither decreased nor increased risk of meningioma was observed in mid-term (OR = 0.93, 95% CI = 0.75-1.16) and long-term (OR = 1.05, 95% CI = 0.93-1.19) users. Neither ipsilateral (OR = 1.05, 95% CI = 0.90-1.22) nor contralateral (OR = 0.86, 95% CI = 0.62-1.18) wireless phone use was associated with the risk of meningioma. Conclusions: This meta-analysis suggested a relationship between decreased meningioma risk and wireless phone use. However, the findings need further validation.


Subject(s)
Cell Phone , Meningeal Neoplasms , Meningioma , Adult , Electromagnetic Fields/adverse effects , Humans , Meningeal Neoplasms/epidemiology , Meningeal Neoplasms/etiology , Meningioma/epidemiology , Meningioma/etiology , Radio Waves
10.
Biosci Rep ; 40(10)2020 10 30.
Article in English | MEDLINE | ID: mdl-33030522

ABSTRACT

CD44 has been considered as a cancer stem cell marker in various tumors. With great enthusiasm, we read an article written by Wu et al. entitled "Expression of CD44 and the survival in glioma: a meta-analysis" published in Bioscience Reports. The authors performed meta-analyses to study the prognostic significance of CD44 in gliomas, and drew the conclusion that high expression of CD44 may predict poor survival in glioma, particularly in WHO grade II-III gliomas. However, two major defects exist in the present study, which made the meta-analysis on the prognostic significance of CD44 in all gliomas unreliable. In this commentary, we discussed the limitations and significance of the present study.


Subject(s)
Brain Neoplasms , Glioma , Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Glioma/genetics , Humans , Hyaluronan Receptors/genetics , Neoplastic Stem Cells
11.
World Neurosurg ; 132: e76-e88, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31518750

ABSTRACT

OBJECTIVE: The tumor microenvironment greatly influences tumor formation, invasion, and progression. The ESTIMATE (Estimation of STromal and Immune cells in MAlignant Tumor tissues) algorithm quantifies stromal and immune components in a tumor, reflecting the tumor microenvironment. This study aimed to explore key prognostic genes in a grade II/III glioma microenvironment. METHODS: We obtained stromal/immune scores for the Cancer Genome Atlas (TCGA) grade II/III glioma cohort from the online ESTIMATE portal. The associations of stromal/immune scores with clinicopathologic characteristics and overall survival of patients with grade II/III glioma were assessed by the Mann-Whitney U test and the Kaplan-Meier method, respectively. Functional enrichment analysis and protein-protein interaction network assessments were employed to analyze differentially expressed genes (DEGs). The top 7 genes with 5 or more edges in the protein-protein interaction network were selected. For validation, CGGA grade II/III glioma data were analyzed. RESULTS: The results showed that elevated stromal/immune/ESTIMATE score was significantly associated with poor survival of patients with TCGA grade II/III glioma. Functional enrichment analysis showed that DEGs were associated with immune cell regulation, extracellular matrix, cytokine activation, and receptor binding. The selected DEGs (interleukin-10, beta-2 microglobulin, C-C motif chemokine ligand 5, cluster of differentiation 74, human leukocyte antigen-DRA, lymphocyte cytosolic protein 2, and myxovirus resistance protein 1) showed prognostic values in patients with grade II/III glioma of the TCGA and CGGA database. CONCLUSIONS: Stromal/immune/ESTIMATE scores have prognostic values in patients with grade II/III glioma. The selected DEGs, including interleukin-10, beta-2 microglobulin, C-C motif chemokine ligand 5, cluster of differentiation 74, human leukocyte antigen-DRA, lymphocyte cytosolic protein 2, and myxovirus resistance protein 1, associated with tumor immunity and microenvironment, have prognostic values in grade II/III glioma. Further investigation of these genes could provide novel insights into the tumor microenvironment of glioma.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Glioma/genetics , Glioma/pathology , Tumor Microenvironment , Adult , Aged , Aged, 80 and over , Algorithms , Biomarkers, Tumor , Brain Neoplasms/immunology , Cohort Studies , Data Mining , Databases, Factual , Female , Gene Expression Regulation, Neoplastic/genetics , Glioma/immunology , Humans , Immunity, Cellular , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis , Protein Interaction Maps , Survival Analysis
12.
J Neurooncol ; 145(2): 201-210, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31506754

ABSTRACT

PURPOSE: Overexpression of CD44 has been detected in many types of tumor tissues. Moreover, CD44 is recognized as a cancer stem cell marker for many cancers. However, the prognostic value of CD44 for glioma patients has not yet been clarified. The authors tried to explore the impact of CD44 expression on grade II/III glioma patients. METHODS: To assess the RNA expression levels of CD44 in glioma tissues and normal brain tissues, meta-analyses were conducted in the online Oncomine database. The mRNA expression levels of CD44, CD44s, and CD44v2-v10 in 112 grade II/III glioma patients in Hokkaido University Hospital (HUH) were detected by qPCR. The RNA-seq data and clinical data of grade II/III glioma patients were obtained from The Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA) databases. RESULTS: Based on the Oncomine database, CD44 has significantly high expression in glioma tissues as compared with normal tissues. We explored the clinical relevance of CD44 mRNA expression based on the HUH cohorts, the TCGA cohorts, and the CGGA cohorts. In survival analysis, high mRNA expression of CD44 was correlated with poor overall survival and poor progression-free survival in grade II/III glioma patients. Multivariate Cox regression analyses confirmed CD44 as an independent prognostic factor for grade II/III glioma patients. CONCLUSIONS: The present study suggests that overexpression of CD44 is associated with a poor prognosis for grade II/III glioma patients. Moreover, our findings suggest that CD44 could serve as a prognostic biomarker in grade II/III glioma patients.


Subject(s)
Brain Neoplasms/diagnosis , Brain Neoplasms/metabolism , Glioma/diagnosis , Glioma/metabolism , Hyaluronan Receptors/metabolism , Biomarkers, Tumor/metabolism , Brain Neoplasms/pathology , Female , Glioma/pathology , Humans , Male , Middle Aged , Neoplasm Grading , Prognosis , RNA, Messenger/metabolism
13.
World Neurosurg ; 130: e775-e785, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31295606

ABSTRACT

OBJECTIVE: Secreted phosphoprotein 1 (SPP1) is an important extracellular glycoprotein that is associated with immune regulation, tumorigenesis, and cell signaling. However, the prognostic value of SPP1 in patients with glioma has not yet been clarified, especially in lower-grade gliomas. The objective of this study is to evaluate the prognostic merit of SPP1 in lower-grade gliomas. METHODS: The messenger RNA (mRNA) expression of SPP1 in about 1000 cancer cell lines was explored by using the data from the Cancer Cell Line Encyclopedia database. The Oncomine database was mined to evaluate the mRNA expression of SPP1 in lower-grade glioma, glioblastoma, and normal brain tissues. The correlation between SPP1 mRNA expression and overall survival of patients with glioma from The Cancer Genome Atlas database was analyzed. RESULTS: SPP1 mRNA expression of glioma was ranked as the eighth highest of all cancer cell lines in the Cancer Cell Line Encyclopedia database. The data from the Oncomine database suggested that SPP1 expression was significantly high in glioblastoma compared with normal brain tissues but was not significantly high in lower-grade glioma compared with normal brain tissue. Analysis of the RNA-Seq data from The Cancer Genome Atlas database showed that the increased SPP1 mRNA expression in lower-grade glioma was significantly associated with poor survival outcomes in patients with lower-grade glioma. Multivariate Cox regression analysis showed that SPP1 might be considered as an independent prognostic factor in lower-grade gliomas. CONCLUSIONS: The present study showed that SPP1 overexpression is related to worse overall survival in patients with lower-grade glioma. Moreover, SPP1 could be considered as an independent factor in lower-grade gliomas.


Subject(s)
Biomarkers, Tumor/metabolism , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/metabolism , Glioma/diagnostic imaging , Glioma/metabolism , Osteopontin/metabolism , Adult , Aged , Brain Neoplasms/surgery , Female , Glioma/surgery , Humans , Male , Middle Aged , Neoplasm Grading/methods , Prognosis
14.
J Neurooncol ; 143(2): 197-206, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30929128

ABSTRACT

PURPOSE: 5-aminolevulinic acid (5-ALA) fluorescence-guided surgery (FGS) appears to be a promising treatment for glioma. However, 5-ALA-mediated fluorescence cannot always be detected in grade II/III gliomas. We hypothesized that gene expression patterns in the Protoporphyrin IX (PpIX) synthesis pathway may be associated with intraoperative fluorescence status of grade II/III gliomas, and then attempted to identify the key molecule of 5-ALA-mediated fluorescence. METHODS: Using 50 surgically obtained specimens, which were diagnosed as grade II and III gliomas, we analyzed gene expression within the PpIX synthesis pathway to identify candidate molecules according to intraoperative 5-ALA-mediated fluorescence status. The most likely candidate gene was selected and confirmed by protein expression analysis. To evaluate the biological function of the molecule in PpIX synthesis, functional analysis was performed using specific, small interference (si)RNA in the SW-1783 human grade III glioma cell line. RESULTS: Among the genes involved in the porphyrin synthesis pathway, the mRNA expression of Peptide transporter 2 (PEPT2) in FGS fluorescence-positive gliomas was significantly higher than that in fluorescence-negative gliomas. Protein expression of PEPT2 was also significantly higher in the fluorescence-positive gliomas, which was confirmed by western blot analysis and immunofluorescence analysis. The siRNA-mediated downregulation of the mRNA and protein expression of PEPT2 led to decreased PpIX fluorescence intensity, as confirmed by fluorescence spectrum analysis. CONCLUSIONS: The results suggest PEPT2 is an important candidate molecule in 5-ALA-mediated FGS in grade II/III gliomas. As the overexpression of PEPT2 was associated with higher PpIX fluorescence intensity, PEPT2 may improve fluorescence-guided resection in grade II/III gliomas.


Subject(s)
Aminolevulinic Acid/metabolism , Biomarkers, Tumor/metabolism , Brain Neoplasms/surgery , Glioma/surgery , Photosensitizing Agents/metabolism , Surgery, Computer-Assisted/methods , Symporters/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Fluorescence , Follow-Up Studies , Glioma/metabolism , Glioma/pathology , Humans , Neoplasm Grading , Neurosurgical Procedures
15.
World Neurosurg ; 115: e629-e636, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29709736

ABSTRACT

BACKGROUND: Wireless phone use has been increasing rapidly and is associated with the risk of glioma. Many studies have been conducted on this association without reaching agreement. The aim of this meta-analysis was to determine the possible association between wireless phone use and risk of adult glioma. METHODS: Eligible studies were identified by searching PubMed and Embase up to July 2017. Random-effects or fixed-effects model was used to combine the results depending on the heterogeneity of the analysis. Publication bias was evaluated using Begg's funnel plot and Egger's regression asymmetry test. Subgroup analysis was performed to evaluate possible influence of these variables. RESULTS: Ten studies on the association of wireless phone use and risk of glioma were included. The combined odds ratio of adult gliomas associated with ever use of wireless phones was 1.03 (95% confidence interval [CI], 0.92-1.16) with high heterogeneity (I2 = 54.2%, P = 0.013). In subgroup analyses, no significant association was found between tumor location in the temporal lobe and adult glioma risk, with odds ratios of 1.26 (95% CI, 0.87-1.84), 0.93 (95% CI, 0.69-1.24), and 1.61 (95% CI, 0.78-3.33). A significant association with risk of glioma was found in long-term users (≥10 years) with odds ratio of 1.33 (95% CI, 1.05-1.67). CONCLUSIONS: Ever use of wireless phones was not significantly associated with risk of adult glioma, but there could be increased risk in long-term users.


Subject(s)
Brain Neoplasms/epidemiology , Cell Phone/trends , Electromagnetic Fields/adverse effects , Glioma/epidemiology , Adult , Brain Neoplasms/diagnosis , Case-Control Studies , Glioma/diagnosis , Humans , Publication Bias/trends , Risk Factors , Temporal Lobe , Time Factors
16.
Oncol Rep ; 36(5): 2544-2552, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27633132

ABSTRACT

Temozolomide (TMZ) is approved for use as first-line treatment for glioblastoma multiforme (GBM). However, GBM shows chemoresistance shortly after the initiation of treatment. In order to detect whether silencing of human protein phosphatase 1D magnesium dependent (PPM1D) gene could increase the effects of TMZ in glioma cells, glioma cells U87-MG were infected with lentiviral shRNA vector targeting PPM1D silencing. After PPM1D silencing was established, cells were treated with TMZ. The multiple functions of human glioma cells after PPM1D silencing and TMZ chemotherapy were detected by flow cytometry and MTT assay. Significantly differentially expressed genes were distinguished by microarray-based gene expression profiling and analyzed by gene pathway enrichment analysis and ontology assessment. Western blotting was used to establish the protein expression of the core genes. PPM1D gene silencing improves TMZ induced cell proliferation and induces cell apoptosis and cell cycle arrest. When PPM1D gene silencing combined with TMZ was performed in glioma cells, 367 genes were upregulated and 444 genes were downregulated compared with negative control. The most significant differential expression pathway was pathway in cancer and IGFR1R, PIK3R1, MAPK8 and EP300 are core genes in the network. Western blotting showed that MAPK8 and PIK3R1 protein expression levels were upregulated and RB1 protein expression was decreased. It was consistent with that detected in gene expression profiling. In conclusion, PPM1D gene silencing combined with TMZ eradicates glioma cells through cell apoptosis and cell cycle arrest. PIK3R1/AKT pathway plays a role in the multiple functions of glioma cells after PPM1D silencing and TMZ chemotherapy.


Subject(s)
Dacarbazine/analogs & derivatives , Glioma/drug therapy , Glioma/genetics , Protein Phosphatase 2C/genetics , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Class Ia Phosphatidylinositol 3-Kinase , Combined Modality Therapy , Dacarbazine/administration & dosage , Flow Cytometry , Gene Expression Regulation, Neoplastic , Gene Silencing , Genetic Therapy , Glioma/pathology , Humans , Lentivirus/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Oncogene Protein v-akt/biosynthesis , Oncogene Protein v-akt/genetics , Phosphatidylinositol 3-Kinases/biosynthesis , Phosphatidylinositol 3-Kinases/genetics , Protein Phosphatase 2C/antagonists & inhibitors , Temozolomide
SELECTION OF CITATIONS
SEARCH DETAIL
...