451
|
Vuong HG, Nguyen TQ, Ngo TNM, Nguyen HC, Fung KM, Dunn IF. The interaction between TERT promoter mutation and MGMT promoter methylation on overall survival of glioma patients: a meta-analysis. BMC Cancer 2020; 20:897. [PMID: 32957941 PMCID: PMC7504655 DOI: 10.1186/s12885-020-07364-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 08/31/2020] [Indexed: 12/22/2022] Open
Abstract
Background There are controversial results concerning the prognostic implication of TERT promoter mutation in glioma patients concerning MGMT status. In this meta-analysis, we investigated whether there are any interactions of these two genetic markers on the overall survival (OS) of glioma patients. Methods Electronic databases including PubMed and Web of Science were searched for relevant studies. Hazard ratio (HR) and its 95% confidence interval (CI) for OS adjusted for selected covariates were calculated from the individual patient data (IPD), Kaplan-Meier curve (KMC), or directly obtained from the included studies. Results A total of nine studies comprising 2819 glioma patients were included for meta-analysis. Our results showed that TERT promoter mutation was associated with a superior outcome in MGMT-methylated gliomas (HR = 0.73; 95% CI = 0.55–0.98; p-value = 0.04), whereas this mutation was associated with poorer survival in gliomas without MGMT methylation (HR = 1.86; 95% CI = 1.54–2.26; p-value < 0.001). TERT-mutated glioblastoma (GBM) patients with MGMT methylation benefited from temozolomide (TMZ) treatment (HR = 0.33; 95% CI = 0.23–0.47; p-value < 0.001). MGMT methylation was not related with any improvement in OS in TERT-wild type GBMs (HR = 0.80; 95% CI = 0.56–1.15; p-value = 0.23). Conclusions The prognostic value of TERT promoter mutation may be modulated by MGMT methylation status. Not all MGMT-methylated GBM patients may benefit from TMZ; it is possible that only TERT-mutated GBM with MGMT methylation, in particular, may respond.
Collapse
Affiliation(s)
- Huy Gia Vuong
- Department of Pathology, Oklahoma University Health Sciences Center, Oklahoma City, OK, 73104, USA.,Stephenson Cancer Center, Oklahoma University Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Thu Quynh Nguyen
- Faculty of Medicine, Pham Ngoc Thach University of Medicine, Ho Chi Minh City, 700-000, Vietnam
| | - Tam N M Ngo
- Faculty of Medicine, Pham Ngoc Thach University of Medicine, Ho Chi Minh City, 700-000, Vietnam
| | - Hoang Cong Nguyen
- Faculty of Medicine, Pham Ngoc Thach University of Medicine, Ho Chi Minh City, 700-000, Vietnam
| | - Kar-Ming Fung
- Department of Pathology, Oklahoma University Health Sciences Center, Oklahoma City, OK, 73104, USA.,Stephenson Cancer Center, Oklahoma University Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Ian F Dunn
- Department of Neurosurgery, Oklahoma University Health Sciences Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
452
|
Diagnostic and Prognostic Potentials of Long Noncoding RNA ELF3-AS1 in Glioma Patients. DISEASE MARKERS 2020; 2020:8871746. [PMID: 33014189 PMCID: PMC7519982 DOI: 10.1155/2020/8871746] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 08/30/2020] [Accepted: 09/02/2020] [Indexed: 12/13/2022]
Abstract
Objective Accumulating evidence implies that long noncoding RNAs (lncRNAs) play a crucial role in predicting survival for glioma patients. However, the potential function of lncRNA ELF3-antisense RNA 1 (ELF3-AS1) in tumors remained largely unclear. The aim of this study was to explore the expression of lncRNA ELF3-antisense RNA 1 (ELF3-AS1) and evaluate its functions in glioma patients. Patients and Methods. ELF3-AS1 expressions were examined by RT-PCR in 182 pairs of glioma specimens and adjacent normal tissues. The receiver operating characteristic (ROC) curve was performed to estimate the diagnostic value of ELF3-AS1. The chi-square tests were used to examine the associations between ELF3-AS1 expression and the clinicopathological characters. The overall survival (OS) and disease-free survival (DFS) were analyzed by log-rank test, and survival curves were plotted according to Kaplan-Meier. The prognostic value of the ELF3-AS1 expression in glioma patients was further analyzed using univariate and multivariate Cox regression analyses. Loss-of-function assays were performed to determine the potential function of ELF3-AS1 on the proliferation and invasion of glioma cells. Results The ELF3-AS1 expression level was significantly higher in glioma specimens compared with adjacent nontumor specimens (p < 0.01). A high expression of ELF3-AS1 was shown to be associated with the WHO grade (p = 0.023) and KPS score (p = 0.012). ROC assays revealed that high ELF3-AS1 expression had an AUC value of 0.8073 (95% CI: 0.7610 to 0.8535) for glioma. Using the Kaplan-Meier analysis, we found that patients with a high ELF3-AS1 expression had significantly poor OS (p = 0.006) and DFS (p = 0.0002). In a multivariate Cox model, we confirmed that ELF3-AS1 expression was an independent poor prognostic factor for glioma patients. The functional assay revealed that knockdown of ELF3-AS1 suppressed the proliferation and invasion of glioma cells. Conclusions Our findings confirmed that ELF3-AS1 functions as an oncogene in glioma and indicated that ELF3-AS1 is not only an important prognostic marker but also a potential therapy target for glioma.
Collapse
|
453
|
Wang Z, Sun D, Chen YJ, Xie X, Shi Y, Tabar V, Brennan CW, Bale TA, Jayewickreme CD, Laks DR, Alcantara Llaguno S, Parada LF. Cell Lineage-Based Stratification for Glioblastoma. Cancer Cell 2020; 38:366-379.e8. [PMID: 32649888 PMCID: PMC7494533 DOI: 10.1016/j.ccell.2020.06.003] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/21/2020] [Accepted: 06/02/2020] [Indexed: 12/29/2022]
Abstract
Glioblastoma, the predominant adult malignant brain tumor, has been computationally classified into molecular subtypes whose functional relevance remains to be comprehensively established. Tumors from genetically engineered glioblastoma mouse models initiated by identical driver mutations in distinct cells of origin portray unique transcriptional profiles reflective of their respective lineage. Here, we identify corresponding transcriptional profiles in human glioblastoma and describe patient-derived xenografts with species-conserved subtype-discriminating functional properties. The oligodendrocyte lineage-associated glioblastoma subtype requires functional ERBB3 and harbors unique therapeutic sensitivities. These results highlight the importance of cell lineage in glioblastoma independent of driver mutations and provide a methodology for functional glioblastoma classification for future clinical investigations.
Collapse
Affiliation(s)
- Zilai Wang
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Daochun Sun
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yu-Jung Chen
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Louis V. Gerstner, Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Xuanhua Xie
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yufeng Shi
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Viviane Tabar
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Cameron W Brennan
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Tejus A Bale
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Chenura D Jayewickreme
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Biochemistry, Cell and Molecular Biology Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY 10065, USA
| | - Dan R Laks
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sheila Alcantara Llaguno
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Luis F Parada
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
454
|
Bakirci E, Schaefer N, Dahri O, Hrynevich A, Strissel P, Strick R, Dalton PD, Villmann C. Melt Electrowritten In Vitro Radial Device to Study Cell Growth and Migration. ACTA ACUST UNITED AC 2020; 4:e2000077. [PMID: 32875734 DOI: 10.1002/adbi.202000077] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/24/2020] [Indexed: 12/14/2022]
Abstract
The development of in vitro assays for 3D microenvironments is essential for understanding cell migration processes. A 3D-printed in vitro competitive radial device is developed to identify preferred Matrigel concentration for glioblastoma migration. Melt electrowriting (MEW) is used to fabricate the structural device with defined and intricate radial structures that are filled with Matrigel. Controlling the printing path is necessary to account for the distance lag in the molten jet, the applied electric field, and the continuous direct-writing nature of MEW. Circular printing below a diameter threshold results in substantial inward tilting of the MEW fiber wall. An eight-chamber radial device with a diameter of 9.4 mm is printed. Four different concentrations of Matrigel are dispensed into the radial chambers. Glioblastoma cells are seeded into the center and grow into all chambers within 8 days. The cell spreading area demonstrates that 6 and 8 mg mL-1 of Matrigel are preferred over 2 and 4 mg mL-1 . Furthermore, topographical cues via the MEW fiber wall are observed to promote migration even further away from the cell seeding depot. Previous studies implement MEW to fabricate cell invasive scaffolds whereas here it is applied to 3D-print in vitro tools to study cell migration.
Collapse
Affiliation(s)
- Ezgi Bakirci
- Department of Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute, University Hospital of Würzburg, Pleicherwall 2, Würzburg, 97070, Germany
| | - Natascha Schaefer
- Department of Clinical Neurobiology, University Hospital of Würzburg, Versbacherstr. 5, Würzburg, 97078, Germany
| | - Ouafa Dahri
- Department of Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute, University Hospital of Würzburg, Pleicherwall 2, Würzburg, 97070, Germany
| | - Andrei Hrynevich
- Department of Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute, University Hospital of Würzburg, Pleicherwall 2, Würzburg, 97070, Germany
| | - Pamela Strissel
- Department of Gynecology and Obstetrics, Laboratory for Molecular Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, University Str. 21-23, Erlangen, 91054, Germany
| | - Reiner Strick
- Department of Gynecology and Obstetrics, Laboratory for Molecular Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, University Str. 21-23, Erlangen, 91054, Germany
| | - Paul D Dalton
- Department of Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute, University Hospital of Würzburg, Pleicherwall 2, Würzburg, 97070, Germany
| | - Carmen Villmann
- Department of Clinical Neurobiology, University Hospital of Würzburg, Versbacherstr. 5, Würzburg, 97078, Germany
| |
Collapse
|
455
|
Long non-coding RNA LBX2-AS1 enhances glioma proliferation through downregulating microRNA-491-5p. Cancer Cell Int 2020; 20:411. [PMID: 32863770 PMCID: PMC7448496 DOI: 10.1186/s12935-020-01433-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/08/2020] [Accepted: 07/18/2020] [Indexed: 12/16/2022] Open
Abstract
Background Dysregulation of lncRNAs is frequent in glioma and has emerged as an important mechanism involved in tumorigenesis. Previous analysis of Chinese Glioma Genome Atlas (CGGA) database indicated that LBX2-AS1 expression is one of differentially expression lncRNA between lower grade glioma (LGG) (grade II and III) and glioblastoma multiforme (GBM). However, the function and mechanism of LBX2-AS1 in glioma has not been evaluated yet. Methods Here, we analyzed the expression of LBX2-AS1 in GTEx data (normal brain), TCGA-LGG and TCGA-GBM. RT-PCR was performed to detect LBX2-AS1 in surgery obtained normal brain and glioma. CCK-8 kit and Annexin V-FITC-PI Apoptosis Detection Kit were used to study the function of LBX2-AS1 on glioma proliferation and apoptosis. Bioinformatic analysis, RNA immunoprecipitation, RT-PCR, western blotting and dual luciferase reporter assay were carried out to investigate the target miRNA of LBX2-AS1. The discovered mechanism was validated by the rescue assay. Results Following study of GTEx and TCGA data, LBX2-AS1 was significantly elevated in glioma compared with normal brain and in GBM compared with LGG. Higher expression of LBX2-AS1 was associated with poor prognosis of patients with glioma. Expression of LBX2-AS1 was positively correlated with pathology classification of glioma. Knockdown of LBX2-AS1 inhibited cell proliferation and induced cell apoptosis in glioma. LBX2-AS1 have complimentary binding site for tumor suppressor miR-491-5p and we showed that LBX2-AS1 sponged miR-491-5p to upregulate TRIM28 expression in glioma cells. TRIM28 overexpression attenuated the effect of LBX2-AS1 knockdown on glioma cells. Conclusions In conclusion, LBX2-AS1 was an increased lncRNA in glioma. Mechanistically, LBX2-AS1 promoted glioma cell proliferation and resistance to cell apoptosis via sponging miR-491-5p.
Collapse
|
456
|
Liu Y, Lang F, Chou FJ, Zaghloul KA, Yang C. Isocitrate Dehydrogenase Mutations in Glioma: Genetics, Biochemistry, and Clinical Indications. Biomedicines 2020; 8:biomedicines8090294. [PMID: 32825279 PMCID: PMC7554955 DOI: 10.3390/biomedicines8090294] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/13/2020] [Accepted: 08/17/2020] [Indexed: 12/22/2022] Open
Abstract
Mutations in isocitrate dehydrogenase (IDH) are commonly observed in lower-grade glioma and secondary glioblastomas. IDH mutants confer a neomorphic enzyme activity that converts α-ketoglutarate to an oncometabolite D-2-hydroxyglutarate, which impacts cellular epigenetics and metabolism. IDH mutation establishes distinctive patterns in metabolism, cancer biology, and the therapeutic sensitivity of glioma. Thus, a deeper understanding of the roles of IDH mutations is of great value to improve the therapeutic efficacy of glioma and other malignancies that share similar genetic characteristics. In this review, we focused on the genetics, biochemistry, and clinical impacts of IDH mutations in glioma.
Collapse
Affiliation(s)
- Yang Liu
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA; (Y.L.); (F.L.); (F.-J.C.)
| | - Fengchao Lang
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA; (Y.L.); (F.L.); (F.-J.C.)
| | - Fu-Ju Chou
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA; (Y.L.); (F.L.); (F.-J.C.)
| | - Kareem A. Zaghloul
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Chunzhang Yang
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA; (Y.L.); (F.L.); (F.-J.C.)
- Correspondence: ; Tel.: +1-240-760-7083
| |
Collapse
|
457
|
Yin W, Jiang X, Tan J, Xin Z, Zhou Q, Zhan C, Fu X, Wu Z, Guo Y, Jiang Z, Ren C, Tang G. Development and Validation of a Tumor Mutation Burden-Related Immune Prognostic Model for Lower-Grade Glioma. Front Oncol 2020; 10:1409. [PMID: 32974146 PMCID: PMC7468526 DOI: 10.3389/fonc.2020.01409] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 07/03/2020] [Indexed: 12/29/2022] Open
Abstract
Tumor mutation burden (TMB) is a useful biomarker to predict prognosis and the efficacy of immune checkpoint inhibitors (ICIs). In this study, we aimed to explore the prognostic value of TMB and the potential association between TMB and immune infiltration in lower-grade gliomas (LGGs). Somatic mutation and RNA-sequencing (RNA-seq) data were downloaded from the Cancer Genome Atlas (TCGA) database. TMB was calculated and patients were divided into high- and low-TMB groups. After performing differential analysis between high- and low-risk groups, we identified six hub TMB and immune-related genes that were correlated with overall survival in LGGs. Then, Gene Set Enrichment Analysis was performed to screen significantly enriched GO terms between the two groups. Moreover, an immune-related risk score system was developed by LASSO Cox analysis based on the six hub genes and was validated with the Chinese Glioma Genome Atlas dataset. Using the TIMER database, we further systematically analyzed the relationships between mutants of the six hub genes and immune infiltration levels, as well as the relationships between the immune-related risk score system and the immune microenvironment in LGGs. The results showed that TMB was negatively correlated with OS and high TMB might inhibit immune infiltration in LGGs. Furthermore, the risk score system could effectively stratify patients into low- and high-risk groups in both the training and validation datasets. Multivariate Cox analysis demonstrated that TMB was not an independent prognostic factor, but the risk score was. Higher infiltration of immune cells (B cells, CD4+ T cells, CD8+ T cells, neutrophils, macrophages, and dendritic cells) and higher levels of immune checkpoints (PD-1, CTLA-4, LAG-3, and TIM-3) were found in patients in the high-risk group. Finally, a novel nomogram model was constructed and evaluated to estimate the overall survival of LGG patients. In summary, our study provided new insights into immune infiltration in the tumor microenvironment and immunotherapies for LGGs.
Collapse
Affiliation(s)
- Wen Yin
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, China
| | - Xingjun Jiang
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, China
| | - Jun Tan
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, China
| | - Zhaoqi Xin
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, China
| | - Quanwei Zhou
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, China
| | - Chaohong Zhan
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, China
| | - Xianyong Fu
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, China
| | - Zhaoping Wu
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, China
| | - Youwei Guo
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, China
| | - Zhipeng Jiang
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, China
| | - Caiping Ren
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University, Changsha, China
| | - Guihua Tang
- Department of Clinical Laboratory, Hunan Provincial People's Hospital (First Affiliated Hospital of Hunan Normal University), Changsha, China
| |
Collapse
|
458
|
Li S, Li W, Chen G, Huang J, Li W. MiRNA-27a-3p induces temozolomide resistance in glioma by inhibiting NF1 level. Am J Transl Res 2020; 12:4749-4756. [PMID: 32913547 PMCID: PMC7476123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 07/19/2020] [Indexed: 06/11/2023]
Abstract
To elucidate the role of miRNA-27a-3p in inducing Temozolomide (TMZ) resistance in glioma and the underlying mechanism. Relative levels of miRNA-27a-3p and NF1 in glioma tissues and peritumoral ones were detected. In addition, their levels in TMZ-sensitive and TMZ-resistant glioma tissues were examined as well. TMZ-resistant glioma cell lines SHG44-TG and U87-TR were used for in vitro experiments. Relative levels of miRNA-27a-3p and NF1 in TMZ-resistant glioma cells and their parental cells were determined. Subsequently, regulatory effects of miRNA-27a-3p on IC50 value of TMZ, viability and migratory ability in SHG44-TG and U87-TR cells were assessed. The interaction between miRNA-27a-3p and NF1 was evaluated by luciferase assay and Western blot. MiRNA-27a-3p was upregulated in glioma tissues than controls, especially TMZ-resistant ones, whereas NF-1 displayed a negative correlation to miRNA-27a-3p as its downstream target. Overexpression of miRNA-27a-3p increased IC50 value of TMZ, viability and migratory ability in SHG44-TG and U87-TR cells. Regulatory effects of miRNA-27a-3p on phenotypes of TMZ-resistant glioma cells were abolished by overexpression of NF1. MiRNA-27a-3p induces TMZ resistance in glioma by negatively regulating NF1 level.
Collapse
Affiliation(s)
- Shenglan Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical UniversityBeijing, China
| | - Wei Li
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical UniversityBeijing, China
| | - Gang Chen
- Beijing Qinglian Biotech Co., Ltd.Beijing, China
| | - Jing Huang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical UniversityBeijing, China
| | - Wenbin Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical UniversityBeijing, China
| |
Collapse
|
459
|
Ji X, Zhang H, Cui Q. A Panel of Synapse-Related Genes as a Biomarker for Gliomas. Front Neurosci 2020; 14:822. [PMID: 32848578 PMCID: PMC7431624 DOI: 10.3389/fnins.2020.00822] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 07/14/2020] [Indexed: 01/08/2023] Open
Abstract
Gliomas are the most common primary brain cancers. In recent years, IDH mutation and 1p/19q codeletion have been suggested as biomarkers for the diagnosis, treatment, and prognosis of gliomas. However, these biomarkers are only effective for a part of glioma patients, and thus more biomarkers are still emergently needed. Recently, an electrochemical communication between normal neurons and glioma cells by neuro-glioma synapse has been reported. Moreover, it was discovered that breast-to-brain metastasis tumor cells have pseudo synapses with neurons, and these synapses were indicated to promote tumor progression and metastasis. Based on the above observations, we first curated a panel of 17 synapse-related genes and then proposed a metric, synapse score to quantify the "stemness" for each sample of 12 glioma gene expression datasets from TCGA, CGGA, and GEO. Strikingly, synapse score showed excellent predictive ability for the prognosis, diagnosis, and grading of gliomas. Moreover, being compared with the two established biomarkers, IDH mutation and 1p/19q codeletion, synapse score demonstrated independent and better predictive performance. In conclusion, this study proposed a quantitative method, synapse score, as an efficient biomarker for monitoring gliomas.
Collapse
Affiliation(s)
- Xiangwen Ji
- Department of Biomedical Informatics, Center for Non-coding RNA Medicine, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, Beijing, China.,Department of Physiology and Pathophysiology, Center for Non-coding RNA Medicine, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Hongwei Zhang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Qinghua Cui
- Department of Biomedical Informatics, Center for Non-coding RNA Medicine, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, Beijing, China.,Department of Physiology and Pathophysiology, Center for Non-coding RNA Medicine, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, Beijing, China
| |
Collapse
|
460
|
Zheng C, Berger NA, Li L, Xu R. Epigenetic age acceleration and clinical outcomes in gliomas. PLoS One 2020; 15:e0236045. [PMID: 32692766 PMCID: PMC7373289 DOI: 10.1371/journal.pone.0236045] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 06/27/2020] [Indexed: 12/03/2022] Open
Abstract
Epigenetic age acceleration-the difference between an individual's DNA methylation age and chronological age-is associated with many diseases including cancer. This study aims to evaluate epigenetic age acceleration as a prognostic biomarker for gliomas. DNA methylation data of gliomas patients (516 low-grade and intermediate-grade gliomas and 140 glioblastoma) were obtained from The Cancer Genome Atlas (TCGA) and patient epigenetic ages were computed using Horvath's age prediction model. We used multivariate linear regression to assess the association of epigenetic age acceleration with tumor molecular subtypes, including Codel, Classic-like, G-CIMP-high, G-CIMP-low, Mesenchymal-like and PA-like. Compared with Codel subtype, epigenetic ages in other molecular subtypes show deceleration after controlling age and race. Age deceleration for Classic-like, G-CIMP-high, G-CIMP-low, Mesenchymal-like and PA-like were 15.42 years (CI: 7.98-22.86, p = 5.38E-05), 25.00 years (CI: 20.79-29.22, p = 4.06E-28), 28.56 years (CI: 14.37-42.74, p = 8.75E-05), 45.34 years (CI: 38.80-51.88, p = 2.15E-36), and 53.58 years (CI: 44.90-62.26, p = 4.81E-30), respectively. Then, Cox proportional hazards regression was used to assess the association of epigenetic age acceleration with patient overall survival. Our results show epigenetic age acceleration is positively associated with patient overall survival (per 10-year age acceleration, HR = 0.89; 95%CI: 0.82-0.97; p = 9.04E-03) in multivariate analysis. When stratified by molecular subtypes, epigenetic age acceleration remains positively associated with patient survival after adjusting age and tumor grade. In conclusion, epigenetic age acceleration is significantly associated with molecular subtypes and patient overall survival in gliomas, indication that epigenetic age acceleration has potential as a quantitative prognostic biomarker for gliomas.
Collapse
Affiliation(s)
- Chunlei Zheng
- Center for Artificial Intelligence in Drug Discovery, School of Medicine, Case Western Reserve University, Cleveland, OH, United States of America
| | - Nathan A. Berger
- Center for Science, Health, and Society, School of Medicine, Case Western Reserve University, Cleveland, OH, United States of America
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, United States of America
| | - Li Li
- Department of Family Medicine, School of Medicine, University of Virginia, Charlottesville, VA, United States of America
| | - Rong Xu
- Center for Artificial Intelligence in Drug Discovery, School of Medicine, Case Western Reserve University, Cleveland, OH, United States of America
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, United States of America
| |
Collapse
|
461
|
Madurga R, García-Romero N, Jiménez B, Collazo A, Pérez-Rodríguez F, Hernández-Laín A, Fernández-Carballal C, Prat-Acín R, Zanin M, Menasalvas E, Ayuso-Sacido Á. Normal tissue content impact on the GBM molecular classification. Brief Bioinform 2020; 22:5868069. [PMID: 32632447 DOI: 10.1093/bib/bbaa129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 05/18/2020] [Accepted: 05/26/2020] [Indexed: 12/19/2022] Open
Abstract
Molecular classification of glioblastoma has enabled a deeper understanding of the disease. The four-subtype model (including Proneural, Classical, Mesenchymal and Neural) has been replaced by a model that discards the Neural subtype, found to be associated with samples with a high content of normal tissue. These samples can be misclassified preventing biological and clinical insights into the different tumor subtypes from coming to light. In this work, we present a model that tackles both the molecular classification of samples and discrimination of those with a high content of normal cells. We performed a transcriptomic in silico analysis on glioblastoma (GBM) samples (n = 810) and tested different criteria to optimize the number of genes needed for molecular classification. We used gene expression of normal brain samples (n = 555) to design an additional gene signature to detect samples with a high normal tissue content. Microdissection samples of different structures within GBM (n = 122) have been used to validate the final model. Finally, the model was tested in a cohort of 43 patients and confirmed by histology. Based on the expression of 20 genes, our model is able to discriminate samples with a high content of normal tissue and to classify the remaining ones. We have shown that taking into consideration normal cells can prevent errors in the classification and the subsequent misinterpretation of the results. Moreover, considering only samples with a low content of normal cells, we found an association between the complexity of the samples and survival for the three molecular subtypes.
Collapse
Affiliation(s)
- Rodrigo Madurga
- Biostatistics and Bioinformatics at Fundación de Investigación HM Hospitales and Professor at the Faculty of Experimental Sciences of the Universidad Francisco de Vitoria
| | - Noemí García-Romero
- Molecular Biology at Fundación Vithas and professor at Francisco de Vitoria University
| | | | | | | | | | | | | | | | - Ernestina Menasalvas
- Department of Computer Systems Languages and Software Engineering at the Faculty of Computer Science of Universidad Politecnica de Madrid
| | - Ángel Ayuso-Sacido
- Brain Tumour Laboratory, Scientific Director at Vithas Hospitals, Managing Director at Fundación Vithas and Professor at the Medial School of Francisco de Vitoria University
| |
Collapse
|
462
|
Mirchia K, Richardson TE. Beyond IDH-Mutation: Emerging Molecular Diagnostic and Prognostic Features in Adult Diffuse Gliomas. Cancers (Basel) 2020; 12:E1817. [PMID: 32640746 PMCID: PMC7408495 DOI: 10.3390/cancers12071817] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/03/2020] [Accepted: 07/04/2020] [Indexed: 12/19/2022] Open
Abstract
Diffuse gliomas are among the most common adult central nervous system tumors with an annual incidence of more than 16,000 cases in the United States. Until very recently, the diagnosis of these tumors was based solely on morphologic features, however, with the publication of the WHO Classification of Tumours of the Central Nervous System, revised 4th edition in 2016, certain molecular features are now included in the official diagnostic and grading system. One of the most significant of these changes has been the division of adult astrocytomas into IDH-wildtype and IDH-mutant categories in addition to histologic grade as part of the main-line diagnosis, although a great deal of heterogeneity in the clinical outcome still remains to be explained within these categories. Since then, numerous groups have been working to identify additional biomarkers and prognostic factors in diffuse gliomas to help further stratify these tumors in hopes of producing a more complete grading system, as well as understanding the underlying biology that results in differing outcomes. The field of neuro-oncology is currently in the midst of a "molecular revolution" in which increasing emphasis is being placed on genetic and epigenetic features driving current diagnostic, prognostic, and predictive considerations. In this review, we focus on recent advances in adult diffuse glioma biomarkers and prognostic factors and summarize the state of the field.
Collapse
Affiliation(s)
- Kanish Mirchia
- Department of Pathology, State University of New York, Upstate Medical University, Syracuse, NY 13210, USA;
| | | |
Collapse
|
463
|
CHEN Y, LI Q, ZHANG J, GU R, LI K, ZHAO G, YUAN H, FENG T, OU D, LIN P. [Increased TRIM5 is associated with a poor prognosis and immune infiltration in glioma patients]. SHENG WU YI XUE GONG CHENG XUE ZA ZHI = JOURNAL OF BIOMEDICAL ENGINEERING = SHENGWU YIXUE GONGCHENGXUE ZAZHI 2020; 37:469-479. [PMID: 32597089 PMCID: PMC10319560 DOI: 10.7507/1001-5515.202004064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 05/11/2020] [Indexed: 02/05/2023]
Abstract
Tripartite motif 5 (TRIM5) plays a significant function in autophagy and involves in immune and tumor processes. While the function of TRIM5 remains poorly understood in glioma. We purpose to evaluate the possible prognostic role of TRIM5 in glioma via bioinformatics analyses. The database clinical samples of glioma in this study included low grade glioma (LGG) and glioblastoma multiforme (GBM). TRIM5 expression in glioma tissues were explored in Oncomine, GEPIA and The Cancer Genome Atlas (TCGA) databases. Survival analysis and the multivariate Cox regression analysis of TRIM5 based on TCGA were used to evaluate the prognostic role of TRIM5. The protein networks of TRIM5 was detected by STRING database. KEGG enrichment analyses were performed to predict the potential molecular pathways of TRIM5 in glioma. In addition, immune infiltration analysis was conducted by CIBERSORT and TIMER databases. We found that TRIM5 was strongly increased in glioma samples compared with normal samples in Oncomine, GEPIA and TCGA databases. Higher TRIM5 was significantly contributed to worse overall survival (OS) in LGG+GBM patients and LGG patients, while was no correlated with OS of GBM patients. Interaction networks analysis identified that IRF3, IRF7, OAS1, OAS2, OAS3, OASL, GBP1, PML, BTBD1 and BTBD2 proteins were contacted with TRIM5. Moreover, KEGG revealed that apoptosis and cancer- and immune-related pathways were enriched with elevated TRIM5. Specifically, TRIM5 could influence the immune infiltration levels, such as activated NK cells, monocytes, activated mast cells and macrophages in glioma. In conclusion, our data indicated that TRIM5 was upregulated in glioma tissues and associated with poor prognosis and immune infiltration. TRIM5 may be acted as a biomarker in prognosis and immunotherapy guidance of glioma.
Collapse
Affiliation(s)
- Yue CHEN
- Lab of Experimental Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, P.R.China
| | - Qin LI
- Lab of Experimental Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, P.R.China
| | - Jie ZHANG
- Lab of Experimental Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, P.R.China
| | - Rui GU
- Lab of Experimental Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, P.R.China
| | - Kai LI
- Lab of Experimental Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, P.R.China
| | - Gang ZHAO
- Lab of Experimental Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, P.R.China
| | - Hang YUAN
- Lab of Experimental Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, P.R.China
| | - Tianyu FENG
- Lab of Experimental Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, P.R.China
| | - Deqiong OU
- Lab of Experimental Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, P.R.China
| | - Ping LIN
- Lab of Experimental Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, P.R.China
| |
Collapse
|
464
|
Chavda V, Patel V, Yadav D, Shah J, Patel S, Jin JO. Therapeutics and Research Related to Glioblastoma: Advancements and Future Targets. Curr Drug Metab 2020; 21:186-198. [DOI: 10.2174/1389200221666200408083950] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 11/28/2019] [Accepted: 03/27/2020] [Indexed: 12/19/2022]
Abstract
Glioblastoma, the most common primary brain tumor, has been recognized as one of the most lethal and
fatal human tumors. It has a dismal prognosis, and survival after diagnosis is less than 15 months. Surgery and radiotherapy
are the only available treatment options at present. However, numerous approaches have been made to upgrade
in vivo and in vitro models with the primary goal of assessing abnormal molecular pathways that would be
suitable targets for novel therapeutic approaches. Novel drugs, delivery systems, and immunotherapy strategies to
establish new multimodal therapies that target the molecular pathways involved in tumor initiation and progression in
glioblastoma are being studied. The goal of this review was to describe the pathophysiology, neurodegeneration
mechanisms, signaling pathways, and future therapeutic targets associated with glioblastomas. The key features have
been detailed to provide an up-to-date summary of the advancement required in current diagnosis and therapeutics
for glioblastoma. The role of nanoparticulate system graphene quantum dots as suitable therapy for glioblastoma has
also been discussed.
Collapse
Affiliation(s)
- Vishal Chavda
- Department of Pharmacology, Nirma University, Ahmadabad, Gujarat, 382481, India
| | - Vimal Patel
- Department of Pharmaceutics, Nirma University, Ahmadabad, Gujarat, 382481, India
| | - Dhananjay Yadav
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 712-749, Korea
| | - Jigar Shah
- Department of Pharmaceutics, Nirma University, Ahmadabad, Gujarat, 382481, India
| | - Snehal Patel
- Department of Pharmacology, Nirma University, Ahmadabad, Gujarat, 382481, India
| | - Jun-O Jin
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 712-749, Korea
| |
Collapse
|
465
|
Hao C, Chen G, Zhao H, Li Y, Chen J, Zhang H, Li S, Zhao Y, Chen F, Li W, Jiang WG. PD-L1 Expression in Glioblastoma, the Clinical and Prognostic Significance: A Systematic Literature Review and Meta-Analysis. Front Oncol 2020; 10:1015. [PMID: 32670884 PMCID: PMC7326811 DOI: 10.3389/fonc.2020.01015] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 05/22/2020] [Indexed: 01/11/2023] Open
Abstract
Background: The clinical and prognostic value of programmed death-ligand 1, PD-L1, in glioblastoma remains controversial. The present study aimed to identify the expression of PD-L1 for its prognostic value in glioblastoma. Methods: A comprehensive literature search was performed using the PubMed and CNKI databases. The overall survival (OS) and disease-free survival (DFS) of GBM was analyzed based on Hazard ratios (HRs) and 95% confidence intervals (CIs). Furthermore, Odds ratios (ORs) and 95% CIs were summarized for clinicopathological parameters. The statistical analysis was using RevMan 5.3 software. Results: The meta-analysis was performed by using total nine studies including 806 patients who had glioblastoma. The pooled results indicated that PD-L1 expression in tumor tissues was significantly related to a poor OS (HR = 1.63, 95%CI: 1.19–2.24, P = 0.003, random effects model) with heterogeneity (I2 = 51%). In subgroup analyses, PD-L1 positivity was significantly associated with a worse OS for patients of American and Asian regions, but not for those of European regions. Moreover, PD-L1 expression implied a trend toward the mutation status of the IDH1 gene [coding the Isocitrate Dehydrogenase (NADP(+))-1 protein] (HR = 9.92, 95%CI: 1.85–53.08, P = 0.007, fixed effects model). However, the prediction overall survival (OS) of the patients showed that PD-L1 expression was independent from other clinicopathological features, such as gender and age. Conclusions: Our analyses indicated that high expression of PD-L1 in glioblastoma tumor tissues is associated with poor survival of patients, and PD-L1 may act as a prognostic predictor and an effective therapeutic target for glioblastoma.
Collapse
Affiliation(s)
- Chengcheng Hao
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Gang Chen
- Beijing Qinglian Biotech, Co., Ltd., Beijing, China
| | - Huishan Zhao
- Reproductive Medicine Centre, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Yan Li
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Jianxin Chen
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Hongmei Zhang
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Shan Li
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yuze Zhao
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Feng Chen
- Department of Neuro-Oncology, Neurosurgery Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wenbin Li
- Department of Neuro-Oncology, Neurosurgery Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wen G Jiang
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff, United Kingdom
| |
Collapse
|
466
|
Yuan T, Ying J, Zuo Z, Gui S, Gao Z, Li G, Zhang Y, Li C. Structural plasticity of the bilateral hippocampus in glioma patients. Aging (Albany NY) 2020; 12:10259-10274. [PMID: 32507763 PMCID: PMC7346025 DOI: 10.18632/aging.103212] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/17/2020] [Indexed: 01/26/2023]
Abstract
This study investigates the structural plasticity and neuronal reaction of the hippocampus in glioma patient pre-surgery. Ninety-nine glioma patients without bilateral hippocampus involvement (low-grade, n=52; high-grade, n=47) and 80 healthy controls with 3D T1 images and resting-fMRI were included. Hippocampal volume and dynamic amplitude of low-frequency fluctuation (dALFF) were analyzed among groups. Relationships between hippocampal volume and clinical characteristics were assessed. We observed remote hippocampal volume increases in low- and high-grade glioma and a greater response of the ipsilateral hippocampus than the contralesional hippocampus. The bilateral hippocampal dALFF was significantly increased in high-grade glioma. Tumor-associated epilepsy and the IDH-1 mutation did not affect hippocampal volume in glioma patients. No significant relationship between hippocampal volume and age was observed in high-grade glioma. The Kaplan-Meier curve and log-rank test revealed that large hippocampal volume was associated with shorter overall survival (OS) compared with small hippocampal volume (p=0.007). Multivariate Cox regression analysis revealed that large hippocampal volume was an independent predictor of unfavorable OS (HR=3.597, 95% CI: 1.160-11.153, p=0.027) in high-grade glioma. Our findings suggest that the hippocampus has a remarkable degree of plasticity in response to pathological stimulation of glioma and that the hippocampal reaction to glioma may be related to tumor malignancy.
Collapse
Affiliation(s)
- Taoyang Yuan
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Jianyou Ying
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Zhentao Zuo
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Songbai Gui
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhixian Gao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Guilin Li
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yazhuo Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Beijing Institute for Brain Disorders Brain Tumor Center, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Chuzhong Li
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Beijing Institute for Brain Disorders Brain Tumor Center, Beijing, China
| |
Collapse
|
467
|
Shen J, Xiong J, Shao X, Cheng H, Fang X, Sun Y, Di G, Mao J, Jiang X. Knockdown of the long noncoding RNA XIST suppresses glioma progression by upregulating miR-204-5p. J Cancer 2020; 11:4550-4559. [PMID: 32489472 PMCID: PMC7255366 DOI: 10.7150/jca.45676] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 04/26/2020] [Indexed: 12/18/2022] Open
Abstract
Background: Gliomas are the most prevalent primary malignant tumors of the central nervous system. Our previous study showed that miR-204-5p is a tumor suppressor gene in glioma. Bioinformatic analyses suggest that long noncoding RNA (lncRNA) X-inactive specific transcript (XIST) is a potential target gene of miR-204-5p. Methods: We analyzed the expression of XIST and miR-204-5p in glioma tissues and the correlation with glioma grade. A series of in vitro experiments were carried out to elucidate the role of XIST in glioma progression. A mouse xenograft model was established to detect whether knockdown of XIST can inhibit glioma growth. A luciferase assay was performed to determine whether XIST can bind to miR-204-5p and the binding specificity. Cells stably expressing shXIST or shNC were transfected with anti-miR-204-5p or anti-miR-204-5p-NC to evaluate whether XIST mediates the tumor-suppressive effects of miR-204-5p. Results: XIST was upregulated in glioma tissues compared with normal brain tissues (NBTs), while miR-204-5p expression was significantly decreased in glioma tissues compared with NBTs. Both XIST and miR-204-5p expression levels were clearly related to glioma grade, and the expression of XIST was obviously negatively correlated with miR-204-5p expression. Knockdown of XIST inhibited glioma cell proliferation, migration, and invasion, promoted apoptosis of glioma cells, inhibited tumor growth and increased the survival time in nude mice. miR-204-5p could directly bind to XIST and negatively regulate XIST expression. XIST mediated glioma progression by targeting miR-204-5p in glioma cells. XIST crosstalk with miR-204-5p regulated Bcl-2 expression to promote apoptosis. Conclusion: Our results provide evidence that XIST, miR-204-5p and Bcl-2 form a regulatory axis that controls glioma progression and can serve as a potential therapeutic target for glioma.
Collapse
Affiliation(s)
- Jun Shen
- Department of Neurosurgery, The First Affiliated Hospital (Yijishan Hospital) of Wannan Medical College, Wuhu, P.R. China
| | - Jianhua Xiong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, P.R. China
| | - Xuefei Shao
- Department of Neurosurgery, The First Affiliated Hospital (Yijishan Hospital) of Wannan Medical College, Wuhu, P.R. China
| | - Hao Cheng
- Department of Neurosurgery, The First Affiliated Hospital (Yijishan Hospital) of Wannan Medical College, Wuhu, P.R. China
| | - Xinyun Fang
- Department of Neurosurgery, The First Affiliated Hospital (Yijishan Hospital) of Wannan Medical College, Wuhu, P.R. China
| | - Yongkang Sun
- Department of Neurosurgery, The First Affiliated Hospital (Yijishan Hospital) of Wannan Medical College, Wuhu, P.R. China
| | - Guangfu Di
- Department of Neurosurgery, The First Affiliated Hospital (Yijishan Hospital) of Wannan Medical College, Wuhu, P.R. China
| | - Jie Mao
- Department of Neurosurgery, Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, P.R. China
| | - Xiaochun Jiang
- Department of Neurosurgery, The First Affiliated Hospital (Yijishan Hospital) of Wannan Medical College, Wuhu, P.R. China
| |
Collapse
|
468
|
Zheng D, Chen D, Lin F, Wang X, Lu L, Luo S, Chen J, Xu X. LncRNA NNT-AS1 promote glioma cell proliferation and metastases through miR-494-3p/PRMT1 axis. Cell Cycle 2020; 19:1621-1631. [PMID: 32420808 DOI: 10.1080/15384101.2020.1762037] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) are key players in cancer progression. However, the function of lncRNA NNT-AS1 on glioma is unclear. In the present study, a total of 73 tumor tissues and matched adjacent non-tumor tissues were collected, and glioma cell lines were cultured in vitro. mRNA expression was tested using RT-qPCR. The protein expression level was determined using the western blot assay, cell proliferation was measured using the CCK-8 and BrdU proliferation assay, and the cell cycle, cell migration and invasion were determined using flow cytometry analysis, the wound healing assay and transwell, respectively. The results showed that lncNNT-AS1 is significantly up-regulated during the early stages of glioma. In particular, high levels of NNT-AS1 are observed in glioma cell lines compared to human astrocyte (HA) cells. Furthermore, the inhibition of lnc-NNT-AS1 by siRNA interfere attenuates the cell viability, proliferation, migration and invasion of glioma cell lines. Mechanistically, the inhibition of NNT-AS1 directly interacted with miRNA-494-3p, and positively regulated the downstream target PRMT1 in vitro. Further study proved that the overexpression of miRNA-494-3p and the inhibition of PRMT1 could attenuate both glioma cell proliferation and metastases. Collectively, our results indicated that the miR-494-3p-PRMT1 axis is involved the tumor-suppressive effects of NNT-AS1 inhibition, which sheds new light on lncRNA-directed diagnostics and the therapeutics of glioma.
Collapse
Affiliation(s)
- Dahai Zheng
- Department of Neurosurgery, Shunde Hospital of Southern Medical University (The First People's Hospital of Shunde Foshan) , Foshan, Guangdong, China
| | - Daliang Chen
- Department of Neurosurgery, Shunde Hospital of Southern Medical University (The First People's Hospital of Shunde Foshan) , Foshan, Guangdong, China
| | - Famu Lin
- Department of Neurosurgery, Shunde Hospital of Southern Medical University (The First People's Hospital of Shunde Foshan) , Foshan, Guangdong, China
| | - Xiang Wang
- Department of Neurosurgery, Shunde Hospital of Southern Medical University (The First People's Hospital of Shunde Foshan) , Foshan, Guangdong, China
| | - Lenian Lu
- Department of Neurosurgery, Shunde Hospital of Southern Medical University (The First People's Hospital of Shunde Foshan) , Foshan, Guangdong, China
| | - Shi Luo
- Department of Neurosurgery, Shunde Hospital of Southern Medical University (The First People's Hospital of Shunde Foshan) , Foshan, Guangdong, China
| | - Jianmin Chen
- Department of Neurosurgery, Shunde Hospital of Southern Medical University (The First People's Hospital of Shunde Foshan) , Foshan, Guangdong, China
| | - Xiaobing Xu
- Department of Neurosurgery, Shunde Hospital of Southern Medical University (The First People's Hospital of Shunde Foshan) , Foshan, Guangdong, China
| |
Collapse
|
469
|
Li L, Wang Y, Li Y, Fang S, Jiang T. Role of molecular biomarkers in glioma resection: a systematic review. Chin Neurosurg J 2020; 6:18. [PMID: 32922947 PMCID: PMC7398179 DOI: 10.1186/s41016-020-00198-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 04/24/2020] [Indexed: 12/13/2022] Open
Abstract
New discoveries based on genetic and epigenetic evidence have significantly expanded the understanding of diffuse gliomas. Molecular biomarkers detected in diffuse gliomas are not only potential targets for radiotherapy, chemotherapy, and immunotherapy, but are also able to guide surgical treatment. Previous studies have suggested that the optimal extent of resection of diffuse gliomas varies according to the expression of specific molecular biomarkers. However, the specific guiding role of these biomarkers in the resection of diffuse gliomas has not been systemically analyzed. This review summarizes several critical molecular biomarkers of tumorigenesis and progression in diffuse gliomas and discusses different strategies of tumor resection in the context of varying genetic expression. With ongoing study and advances in technology, molecular biomarkers will play a more important role in glioma resection and maximize the survival benefit from surgery for diffuse gliomas.
Collapse
Affiliation(s)
- Lianwang Li
- Beijing Neurosurgical Institute, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, Beijing, 10070 China
| | - Yinyan Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, Beijing, 10070 China
| | - Yiming Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, Beijing, 10070 China
| | - Shengyu Fang
- Beijing Neurosurgical Institute, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, Beijing, 10070 China
| | - Tao Jiang
- Beijing Neurosurgical Institute, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, Beijing, 10070 China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, Beijing, 10070 China
| |
Collapse
|
470
|
Lin N, Li W, Wang X, Hou S, Yu D, Zhao X, Jin C, Yao G, Yan W, You Y. Upregulation of miR-340 Inhibits Tumor Growth and Mesenchymal Transition via Targeting c-MET in Glioblastoma. Cancer Manag Res 2020; 12:3343-3352. [PMID: 32494198 PMCID: PMC7229788 DOI: 10.2147/cmar.s250772] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 04/22/2020] [Indexed: 12/12/2022] Open
Abstract
Background Epithelial-mesenchymal Transition (EMT) is involved in various cancers including glioblastoma. Our previous study has shown that miR-340 negatively correlated with EMT process in glioblastoma. Purpose In the present study, we aim to explore the underlying molecular mechanisms of miR-340 in EMT process of glioblastomas. Materials and Methods Using RT-qPCR assay, we analyzed the expression of miR-340 in glioma cell lines and normal human glia (NHA) cell line. Using CCK8, Colony formation assays, transwell and Western blot assays, we investigated tumor growth and EMT process. Using luciferase reporter assay, we confirmed a target of miR-340. Results Our results showed that miR-340 was down-regulated in glioma cell lines (U87, U251 and LN229) compared to NHA cells. MiR-340 overexpression remarkably inhibited cell proliferation and invasion as well as up-regulated E-cadherin expression and down-regulated N-cadherin, Vimentin, ZEB1, Slug and Snail expressions in U251 and LN229 cells. Further studies have confirmed c-MET as a target gene of miR-340. The EMT-inhibitory effect of miR-340 was lost after c-MET expression was restored. We also identified the antitumorigenic activity of miR-340 in vivo. Conclusion These results demonstrated that miR-340 functioned as a tumor suppressor via targeting EMT process and could be a potential therapeutic candidate for treating glioblastomas.
Collapse
Affiliation(s)
- Ning Lin
- Department of Neurosurgery, Chuzhou Clinical College of Anhui Medical University, The First People's Hospital Chuzhou, Chuzhou, People's Republic of China
| | - Wentao Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Xiefeng Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Shiqiang Hou
- Department of Neurosurgery, Chuzhou Clinical College of Anhui Medical University, The First People's Hospital Chuzhou, Chuzhou, People's Republic of China
| | - Dong Yu
- Department of Neurosurgery, Chuzhou Clinical College of Anhui Medical University, The First People's Hospital Chuzhou, Chuzhou, People's Republic of China
| | - Xingyuan Zhao
- Department of Neurosurgery, Chuzhou Clinical College of Anhui Medical University, The First People's Hospital Chuzhou, Chuzhou, People's Republic of China
| | - Chunjing Jin
- Laboratory Medicine Center, Affiliated Hospital of Nantong University, Nantong, People's Republic of China
| | - Guoquan Yao
- Department of Neurosurgery, Chuzhou Clinical College of Anhui Medical University, The First People's Hospital Chuzhou, Chuzhou, People's Republic of China
| | - Wei Yan
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Yongping You
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| |
Collapse
|
471
|
Ruan X, Zheng J, Liu X, Liu Y, Liu L, Ma J, He Q, Yang C, Wang D, Cai H, Li Z, Liu J, Xue Y. lncRNA LINC00665 Stabilized by TAF15 Impeded the Malignant Biological Behaviors of Glioma Cells via STAU1-Mediated mRNA Degradation. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 20:823-840. [PMID: 32464546 PMCID: PMC7256440 DOI: 10.1016/j.omtn.2020.05.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/23/2020] [Accepted: 05/01/2020] [Indexed: 12/11/2022]
Abstract
Glioma is a brain cancer characterized by strong invasiveness with limited treatment options and poor prognosis. Recently, dysregulation of long non-coding RNAs (lncRNAs) has emerged as an important component in cellular processes and tumorigenesis. In this study, we demonstrated that TATA-box binding protein associated factor 15 (TAF15) and long intergenic non-protein coding RNA 665 (LINC00665) were both downregulated in glioma tissues and cells. TAF15 overexpression enhanced the stability of LINC00665, inhibiting malignant biological behaviors of glioma cells. Both metal regulatory transcription factor 1 (MTF1) and YY2 transcription factor (YY2) showed high expression levels in glioma tissues and cells, and their knockdown inhibited malignant progression. Mechanistically, overexpression of LINC00665 was confirmed to destabilize MTF1 and YY2 mRNA by interacting with STAU1, and knockdown of STAU1 could rescue the MTF1 and YY2 mRNA degradation caused by LINC00665 overexpression. G2 and S-phase expressed 1 (GTSE1) was identified as an oncogene in glioma, and knockdown of MTF1 or YY2 decreased the mRNA and protein expression levels of GTSE1 through direct binding to the GTSE1 promoter region. Our study highlights a key role of the TAF15/LINC00665/MTF1(YY2)/GTSE1 axis in modulating the malignant biological behaviors of glioma cells, suggesting novel mechanisms by which lncRNAs affect STAU1-mediated mRNA stability, which can inform new molecular therapies for glioma.
Collapse
Affiliation(s)
- Xuelei Ruan
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang 110122, China; Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, China; Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China
| | - Jian Zheng
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Xiaobai Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Libo Liu
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang 110122, China; Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, China; Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China
| | - Jun Ma
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang 110122, China; Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, China; Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China
| | - Qianru He
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang 110122, China; Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, China; Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China
| | - Chunqing Yang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Di Wang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Heng Cai
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Zhen Li
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Jing Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Yixue Xue
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang 110122, China; Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, China; Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China.
| |
Collapse
|
472
|
Liu X, Huang H, Li X, Zheng X, Zhou C, Xue B, He J, Zhang Y, Liu L. Knockdown of ADAMDEC1 inhibits the progression of glioma in vitro. Histol Histopathol 2020; 35:997-1005. [PMID: 32378728 DOI: 10.14670/hh-18-227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Glioma is one of the most lethal malignant tumors all over the world. The prognosis of patients with high‑grade glioma remains very poor. Therefore, it is urgent to find a novel strategy for the treatment of glioma. It has been reported that ADAMDEC1 could regulate the progression of multiple diseases, including cancers. However, the role of ADAMDEC1 during the tumorigenesis of glioma remains largely unknown. Methods, Gene expression of ADAMDEC1 in glioma tissues or in cells was detected by qRT-PCR. Western blot was performed to measure the protein expressions of p53, active caspase3, active caspase9, CDK2 and Cyclin A in glioma cells. Cell proliferation was detected by CCK-8 assay. Cell apoptosis or cycle was tested by flow cytometry. Transwell was used to test the invasion of glioma cells. RESULTS The expression of ADAMDEC1 in glioma tissues or cells was significantly upregulated. In addition, downregulation of ADAMDEC1 notably inhibited the proliferation and induced apoptosis of glioma cells through upregulation of active caspase 3 and active caspase 9. Besides, silencing of ADAMDEC1 obviously induced G1 arrest in glioma cells via modulation of cell cycle-related proteins. Finally, knockdown of ADAMDEC1 significantly inhibited the migration and invasion of glioma cells. In contrast, overexpression of ADAMDEC1 promoted cell proliferation, migration and invasion of glioma cells. CONCLUSION Downregulation of ADAMDEC1 could significantly inhibit the tumorigenesis of glioma in vitro, which may serve as a novel target for the treatment of glioma.
Collapse
Affiliation(s)
- Xueliang Liu
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Hao Huang
- Department of Neurosurgery, Guang'an People's Hospital, Guangan, Sichuan, China
| | - Xuehan Li
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaomei Zheng
- Department of Neurology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Chong Zhou
- Department of Neurology, Jianyang People's Hospital, Jianyang, Sichuan, China
| | - Bin Xue
- Department of Neurosurgery, Nanbu People's Hospital, Nanchong, Sichuan, China
| | - Jimin He
- Department of Neurosurgery, Suining Central Hospital, Suining, Sichuan, China
| | - Ye Zhang
- Department of Neurosurgery, Mianyang Central Hospital, Mianyang, Sichuan, China
| | - Liang Liu
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
473
|
Ferraris C, Cavalli R, Panciani PP, Battaglia L. Overcoming the Blood-Brain Barrier: Successes and Challenges in Developing Nanoparticle-Mediated Drug Delivery Systems for the Treatment of Brain Tumours. Int J Nanomedicine 2020; 15:2999-3022. [PMID: 32431498 PMCID: PMC7201023 DOI: 10.2147/ijn.s231479] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 04/14/2020] [Indexed: 12/14/2022] Open
Abstract
High-grade gliomas are still characterized by a poor prognosis, despite recent advances in surgical treatment. Chemotherapy is currently practiced after surgery, but its efficacy is limited by aspecific toxicity on healthy cells, tumour cell chemoresistance, poor selectivity, and especially by the blood–brain barrier (BBB). Thus, despite the large number of potential drug candidates, the choice of effective chemotherapeutics is still limited to few compounds. Malignant gliomas are characterized by high infiltration and neovascularization, and leaky BBB (the so-called blood–brain tumour barrier); surgical resection is often incomplete, leaving residual cells that are able to migrate and proliferate. Nanocarriers can favour delivery of chemotherapeutics to brain tumours owing to different strategies, including chemical stabilization of the drug in the bloodstream; passive targeting (because of the leaky vascularization at the tumour site); inhibition of drug efflux mechanisms in endothelial and cancer cells; and active targeting by exploiting carriers and receptors overexpressed at the blood–brain tumour barrier. Within this concern, a suitable nanomedicine-based therapy for gliomas should not be limited to cytotoxic agents, but also target the most important pathogenetic mechanisms, including cell differentiation pathways and angiogenesis. Moreover, the combinatorial approach of cell therapy plus nanomedicine strategies can open new therapeutical opportunities. The major part of attempted preclinical approaches on animal models involves active targeting with protein ligands, but, despite encouraging results, a few number of nanomedicines reached clinical trials, and most of them include drug-loaded nanocarriers free of targeting ligands, also because of safety and scalability concerns.
Collapse
Affiliation(s)
- Chiara Ferraris
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Roberta Cavalli
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Pier Paolo Panciani
- Clinic of Neurosurgery, Spedali Civili and University of Brescia, Brescia, Italy
| | - Luigi Battaglia
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| |
Collapse
|
474
|
Lee H, Suh M, Choi H, Ha S, Paeng JC, Cheon GJ, Kang KW, Lee DS. A pan-cancer analysis of the clinical and genetic portraits of somatostatin receptor expressing tumor as a potential target of peptide receptor imaging and therapy. EJNMMI Res 2020; 10:42. [PMID: 32335823 PMCID: PMC7183516 DOI: 10.1186/s13550-020-00632-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 04/14/2020] [Indexed: 12/19/2022] Open
Abstract
Purpose Although somatostatin receptor (SST) is a promising theranostic target and is widely expressed in tumors of various organs, the indication for therapies targeting SST is limited to typical gastroenteropancreatic neuroendocrine tumors (NETs). Thus, broadening the scope of the current clinical application of peptide receptor radiotherapy (PRRT) can be supported by a better understanding of the landscape of SST-expressing tumors. Methods SST expression levels were assessed in data from The Cancer Genome Atlas across 10,701 subjects representing 32 cancer types. As the major target of PRRT is SST subtype 2 (SST2), correlation analyses between the pan-cancer profiles, including clinical and genetic features, and SST2 level were conducted. The median SST2 expression level of pheochromocytoma and paraganglioma (PCPG) samples was used as the threshold to define “high-SST2 tumors.” The prognostic value of SST2 in each cancer subtype was evaluated by using Cox proportional regression analysis. Results We constructed a resource of SST expression patterns associated with clinicopathologic features and genomic alterations. It provides an interactive tool to analyze SST expression patterns in various cancer types. As a result, eight of the 31 cancer subtypes other than PCPG had more than 5% of tumors with high-SST2 expression. Low-grade glioma (LGG) showed the highest proportion of high-SST2 tumors, followed by breast invasive carcinoma (BRCA). LGG showed different SST2 levels according to tumor grade and histology. IDH1 mutation was significantly associated with high-SST2 status. In BRCA, the SST2 level was different according to the hormone receptor status. High-SST2 status was significantly associated with good prognosis in LGG patients. High-SST2 status showed a trend for association with poor prognosis in triple-negative breast cancer subjects. Conclusion A broad range of SST2 expression was observed across diverse cancer subtypes. The SST2 expression level showed a significant association with genomic and clinical aspects across cancers, especially in LGG and BRCA. These findings extend our knowledge base to diversify the indications for PRRT as well as SST imaging.
Collapse
Affiliation(s)
- Hyunjong Lee
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul National University College of Medicine, 28 Yongon-Dong, Jongno-Gu, Seoul, 110-744, Republic of Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Minseok Suh
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul National University College of Medicine, 28 Yongon-Dong, Jongno-Gu, Seoul, 110-744, Republic of Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Hongyoon Choi
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul National University College of Medicine, 28 Yongon-Dong, Jongno-Gu, Seoul, 110-744, Republic of Korea.
| | - Seunggyun Ha
- Division of Nuclear Medicine, Department of Radiology, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jin Chul Paeng
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul National University College of Medicine, 28 Yongon-Dong, Jongno-Gu, Seoul, 110-744, Republic of Korea
| | - Gi Jeong Cheon
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul National University College of Medicine, 28 Yongon-Dong, Jongno-Gu, Seoul, 110-744, Republic of Korea.,Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Keon Wook Kang
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul National University College of Medicine, 28 Yongon-Dong, Jongno-Gu, Seoul, 110-744, Republic of Korea.,Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Dong Soo Lee
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul National University College of Medicine, 28 Yongon-Dong, Jongno-Gu, Seoul, 110-744, Republic of Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
475
|
Chen Y, Li N, Wang H, Wang N, Peng H, Wang J, Li Y, Liu M, Li H, Zhang Y, Wang Z. Amentoflavone suppresses cell proliferation and induces cell death through triggering autophagy-dependent ferroptosis in human glioma. Life Sci 2020; 247:117425. [PMID: 32057904 DOI: 10.1016/j.lfs.2020.117425] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 01/27/2020] [Accepted: 02/09/2020] [Indexed: 01/07/2023]
Abstract
AIMS Glioma is the most common type of malignant tumor of the nervous system, and aggressiveness and recurrence are major obstacles for treatment. This study is designed to explore the effects of amentoflavone (AF) on glioma, and to investigate the underlying mechanism of the anti-cancer activities of AF. METHODS Cell morphology was recorded under microscopy. Cell viability and cell death ratio were determined by CCK-8 assay and lactate dehydrogenase (LDH) release assay, respectively. Cell cycle progression was assessed by flow cytometry. The levels of iron, MDA (malondialdehyde), lipid ROS, and GSH (reduced glutathione) were assessed by ELISA kit. The cycle-related proteins, ferroptosis-related protein, autophagy-related protein, and the phosphorylation of AMPK, mTOR and p70S6K were analyzed by western blotting. The autophagic flux was observed by transfecting cells with mRFP-GFP-LC3 plasmids. The xenograft murine models were established to analyze the effects of amentoflavone in vivo. The immunohistochemistry assay was performed to analyze the expression of LC3B, Beclin1, ATG5, ATG7, and ferritin heavy chain (FTH). RESULTS Our results showed that AF treatment led to reduction in cell viability and cell death. In addition, AF was found to block cell cycle progression in a dose-dependent manner in vitro. Following treatment with AF, the intracellular levels of iron, MDA, and lipid OS were increased, and the levels of GSH and the mitochondrial membrane potential were reduced. In addition, our results showed that AF promoted the autophagic by regulating autophagy-relevant proteins. Our results also showed that the autophagy-induction by AF was associated with regulation of AMPK/mTOR signaling. Mechanistically, the inhibition effects of AF on glioma cell were reversed by DFO, ferreostatin-1 as well as upregulation of FTH. Meanwhile, the FTH levels were increased by compound C and knockdown of ATG7. Moreover, both autophagy inhibitor Baf A1 and knockdown of ATG7 were able to compromising AF-induce ferroptosis and cell death. In vivo, the tumor growth was suppressed by AF in a dose-dependent manner. The level of MDA in the tumor tissue was increased while the level of GSH in tumor tissue was decreased by AF in a dose-dependent manner. Furthermore, the expression of LC3B, Beclin1, ATG5, ATG7 were increased, and the expression of FTH were decreased by AF in a dose-dependent manner in vivo. Conclusion These results demonstrate that AF triggered ferroptosis in autophagy-dependent manner. Our results suggest that AF has the potential to be considered as a novel treatment agent in glioma.
Collapse
Affiliation(s)
- Yan Chen
- The Affiliated Qingdao Hiser Hospital of Qingdao University, Qingdao, Shandong, China
| | - Ning Li
- The Affiliated Qingdao Hiser Hospital of Qingdao University, Qingdao, Shandong, China
| | - Haijing Wang
- The Affiliated Qingdao Hiser Hospital of Qingdao University, Qingdao, Shandong, China
| | - Ningning Wang
- The Affiliated Qingdao Hiser Hospital of Qingdao University, Qingdao, Shandong, China
| | - Hui Peng
- The Affiliated Qingdao Hiser Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jing Wang
- The Affiliated Qingdao Hiser Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yihong Li
- The Affiliated Qingdao Hiser Hospital of Qingdao University, Qingdao, Shandong, China
| | - Mingdi Liu
- The Affiliated Qingdao Hiser Hospital of Qingdao University, Qingdao, Shandong, China
| | - Hui Li
- The Affiliated Qingdao Hiser Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yu Zhang
- The Affiliated Qingdao Hiser Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zhaohui Wang
- The Affiliated Qingdao Hiser Hospital of Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
476
|
Transcriptomic Analysis of Glioma Based on IDH Status Identifies ACAA2 as a Prognostic Factor in Lower Grade Glioma. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1086792. [PMID: 32280672 PMCID: PMC7115055 DOI: 10.1155/2020/1086792] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/05/2020] [Indexed: 01/01/2023]
Abstract
Background Glioma is the most common and lethal tumor in the central nervous system (CNS). More than 70% of WHO grade II/III gliomas were found to harbor isocitrate dehydrogenase (IDH) mutations which generated targetable metabolic vulnerabilities. Focusing on the metabolic vulnerabilities, some targeted therapies, such as NAMPT, have shown significant effects in preclinical and clinical trials. Methods We explored the TCGA as well as CGGA database and analyzed the RNA-seq data of lower grade gliomas (LGG) with the method of weighted correlation network analysis (WGCNA). Differential expressed genes were screened, and coexpression relationships were grouped together by performing average linkage hierarchical clustering on the topological overlap. Clinical data were used to conduct Kaplan–Meier analysis. Results In this study, we identified ACAA2 as a prognostic factor in IDH mutation lower grade glioma with the method of weighted correlation network analysis (WGCNA). The difference of ACAA2 gene expressions between the IDH wild-type (IDH-WT) group and the IDH mutant (IDH-MUT) group suggested that there may be different potential targeted therapies based on the fatty acid metabolic vulnerabilities, which promoted the personalized treatment for LGG patients.
Collapse
|
477
|
Zhu F, Cheng C, Qin H, Wang H, Yu H. A novel circular RNA circENTPD7 contributes to glioblastoma progression by targeting ROS1. Cancer Cell Int 2020; 20:118. [PMID: 32308563 PMCID: PMC7147020 DOI: 10.1186/s12935-020-01208-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 04/06/2020] [Indexed: 12/24/2022] Open
Abstract
Background Circular RNAs (circRNAs) are identified to play an important role in many human cancers, such as glioblastoma. However, the potential mechanisms underlying the relationship between circRNAs and glioblastoma pathogenesis are still elusive. This study is designed to investigate the role of circRNAs in glioblastoma progression. Methods The present study is designed to investigate the mechanism by which circRNAs involves in glioblastoma pathogenesis. By using circRNAs microarray, we detected the dysregulated circRNAs and identified an up-regulated circRNA, circENTPD7 in glioblastoma tissues. Cell proliferation was measured using a CCK-8 assay. Cell clone formation ability was assessed with a clone formation test. We used the bioinformatics website to predict circRNA–miRNA and miRNA–mRNA interactions. CircRNA–miRNA interaction was confirmed by dual-luciferase reporter assays and RNA–RNA pulldown assay. Results circENTPD7 (hsa_circ_0019421) was upregulated in glioblastoma tissues. Kaplan–Meier survival analysis indicated that glioblastoma patients had a poor overall survival when circENTPD7 expression levels were high. Knockdown of circENTPD7 inhibited the motility and proliferation of glioblastoma cells. Moreover, we demonstrated that circENTPD7 acted as a sponge of miR-101-3p to regulate the expression of ROS1 further promoted the proliferation and motility of glioblastoma cells. Conclusions Taken together, these findings indicate that circRNA circENTPD7 promotes glioblastoma cell proliferation and motility by regulating miR-101-3p/ROS1.
Collapse
Affiliation(s)
- Fei Zhu
- Department of Neuro Surgery, The Affiliated Hospital of Yangzhou University, No. 45, Taizhou Road, Yangzhou, Jiangsu China
| | - Cheng Cheng
- Department of Neuro Surgery, The Affiliated Hospital of Yangzhou University, No. 45, Taizhou Road, Yangzhou, Jiangsu China
| | - Hong Qin
- Department of Neuro Surgery, The Affiliated Hospital of Yangzhou University, No. 45, Taizhou Road, Yangzhou, Jiangsu China
| | - Hongsheng Wang
- Department of Neuro Surgery, The Affiliated Hospital of Yangzhou University, No. 45, Taizhou Road, Yangzhou, Jiangsu China
| | - Hailong Yu
- Department of Neuro Surgery, The Affiliated Hospital of Yangzhou University, No. 45, Taizhou Road, Yangzhou, Jiangsu China
| |
Collapse
|
478
|
Wang Z, Tang W, Yuan J, Qiang B, Han W, Peng X. Integrated Analysis of RNA-Binding Proteins in Glioma. Cancers (Basel) 2020; 12:E892. [PMID: 32272554 PMCID: PMC7226056 DOI: 10.3390/cancers12040892] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 03/21/2020] [Accepted: 03/24/2020] [Indexed: 02/07/2023] Open
Abstract
RNA-binding proteins (RBPs) play important roles in many cancer types. However, RBPs have not been thoroughly and systematically studied in gliomas. Global analysis of the functional impact of RBPs will provide a better understanding of gliomagenesis and new insights into glioma therapy. In this study, we integrated a list of the human RBPs from six sources-Gerstberger, SONAR, Gene Ontology project, Poly(A) binding protein, CARIC, and XRNAX-which covered 4127 proteins with RNA-binding activity. The RNA sequencing data were downloaded from The Cancer Genome Atlas (TCGA) (n = 699) and Chinese Glioma Genome Atlas (CGGA) (n = 325 + 693). We examined the differentially expressed genes (DEGs) using the R package DESeq2, and constructed a weighted gene co-expression network analysis (WGCNA) of RBPs. Furthermore, survival analysis was also performed based on the univariate and multivariate Cox proportional hazards regression models. In the WGCNA analysis, we identified a key module involved in the overall survival (OS) of glioblastomas. Survival analysis revealed eight RBPs (PTRF, FNDC3B, SLC25A43, ZC3H12A, LRRFIP1, HSP90B1, HSPA5, and BNC2) are significantly associated with the survival of glioblastoma patients. Another 693 patients within the CGGA database were used to validate the findings. Additionally, 3564 RBPs were classified into canonical and non-canonical RBPs depending on the domains that they contain, and non-canonical RBPs account for the majority (72.95%). The Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis showed that some non-canonical RBPs may have functions in glioma. Finally, we found that the knockdown of non-canonical RBPs, PTRF, or FNDC3B can alone significantly inhibit the proliferation of LN229 and U251 cells. Simultaneously, RNA Immunoprecipitation (RIP) analysis indicated that PTRF may regulate cell growth and death- related pathways to maintain tumor cell growth. In conclusion, our findings presented an integrated view to assess the potential death risks of glioblastoma at a molecular level, based on the expression of RBPs. More importantly, we identified non-canonical RNA-binding proteins PTRF and FNDC3B, showing them to be potential prognostic biomarkers for glioblastoma.
Collapse
Affiliation(s)
- Zhixing Wang
- State Key Laboratory of Medical Molecular Biology, Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100730, China
| | - Wanjun Tang
- State Key Laboratory of Medical Molecular Biology, Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100730, China
| | - Jiangang Yuan
- State Key Laboratory of Medical Molecular Biology, Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100730, China
| | - Boqin Qiang
- State Key Laboratory of Medical Molecular Biology, Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100730, China
| | - Wei Han
- State Key Laboratory of Medical Molecular Biology, Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100730, China
| | - Xiaozhong Peng
- State Key Laboratory of Medical Molecular Biology, Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100730, China
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Kunming 650031, China
| |
Collapse
|
479
|
Rodríguez Pérez Á, Campillo-Davo D, Van Tendeloo VFI, Benítez-Ribas D. Cellular immunotherapy: a clinical state-of-the-art of a new paradigm for cancer treatment. Clin Transl Oncol 2020; 22:1923-1937. [PMID: 32266674 DOI: 10.1007/s12094-020-02344-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 03/19/2020] [Indexed: 12/31/2022]
Abstract
Cancer immunotherapy has opened a new chapter in Medical Oncology. Many novel therapies are under clinical testing and some have already been approved and implemented in cancer treatment protocols. In particular, cellular immunotherapies take advantage of the antitumor capabilities of the immune system. From dendritic cell-based vaccines to treatments centered on genetically engineered T cells, this form of personalized cancer therapy has taken the field by storm. They commonly share the ex vivo genetic modification of the patient's immune cells to generate or induce tumor antigen-specific immune responses. The latest clinical trials and translational research have shed light on its clinical effectiveness as well as on the mechanisms behind targeting specific antigens or unique tumor alterations. This review gives an overview of the clinical developments in immune cell-based technologies predominantly for solid tumors and on how the latest discoveries are being incorporated within the standard of care.
Collapse
Affiliation(s)
- Á Rodríguez Pérez
- Laboratory of Molecular and Translational Oncology-CELLEX, University of Barcelona, 08035, Barcelona, Spain.,Medical Oncology Department, University Hospital "Fundación Jiménez Díaz", Autonomous University of Madrid, 28040, Madrid, Spain
| | - D Campillo-Davo
- Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - V F I Van Tendeloo
- Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - D Benítez-Ribas
- Department of Immunology, Hospital Clinic, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), University of Barcelona, Carrer Villarroel, 170. 08036, Barcelona, Spain.
| |
Collapse
|
480
|
Zheng W, Chen Q, Wang C, Yao D, Zhu L, Pan Y, Zhang J, Bai Y, Shao C. Inhibition of Cathepsin D (CTSD) enhances radiosensitivity of glioblastoma cells by attenuating autophagy. Mol Carcinog 2020; 59:651-660. [PMID: 32253787 DOI: 10.1002/mc.23194] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/21/2020] [Accepted: 03/26/2020] [Indexed: 12/12/2022]
Abstract
Postoperative radiotherapy combined with chemotherapy is a commonly used treatment for glioblastoma (GBM) but radiotherapy often fails to achieve the expected results mainly due to tumor radioresistance. In this study, we established a radioresistant subline from human glioma cell line U251 and found that Cathepsin D (CTSD), a gene closely related to the clinical malignancy and prognosis in glioma, had higher expression level in radioresistant clones than that in parental cells, and knocking down CTSD by small interfering RNA (siRNA) or its inhibitor Pepstatin-A increased the radiosensitivity. The level of autophagy was enhanced in the radioresistant GBM cells compared with its parent cells, and silencing autophagy by light chain 3 (LC3) siRNA significantly sensitized GBM cells to ionizing radiation (IR). Moreover, the protein expression level of CTSD was positively correlated with the autophagy marker LC3 II/I and negatively correlated with P62 after IR in radioresistant cells. As expected, through the combination of Western blot and immunofluorescence assays, inhibition of CTSD increased the formation of autophagosomes, while decreased the formation of autolysosomes, which indicating an attenuated autophagy level, leading to radiosensitization ultimately. Our results revealed for the first time that CTSD regulated the radiosensitivity of glioblastoma by affecting the fusion of autophagosomes and lysosomes. In significance, CTSD might be a potential molecular biomarker and a new therapeutic target in glioblastoma.
Collapse
Affiliation(s)
- Wang Zheng
- Department of Radiation Biology, Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qianping Chen
- Department of Radiation Biology, Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chen Wang
- Department of Radiation Biology, Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Dan Yao
- Department of Radiation Biology, Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lin Zhu
- Department of Radiation Biology, Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan Pan
- Department of Radiation Biology, Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianghong Zhang
- Department of Radiation Biology, Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yang Bai
- Department of Radiation Biology, Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chunlin Shao
- Department of Radiation Biology, Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
481
|
Advances in Noninvasive Neurodiagnostics. World Neurosurg 2020; 139:1-3. [PMID: 32194266 DOI: 10.1016/j.wneu.2020.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 03/02/2020] [Indexed: 11/21/2022]
|
482
|
Yang B, Pan YB, Ma YB, Chu SH. Integrated Transcriptome Analyses and Experimental Verifications of Mesenchymal-Associated TNFRSF1A as a Diagnostic and Prognostic Biomarker in Gliomas. Front Oncol 2020; 10:250. [PMID: 32257943 PMCID: PMC7090130 DOI: 10.3389/fonc.2020.00250] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 02/13/2020] [Indexed: 12/12/2022] Open
Abstract
Gliomas are the most prevalent malignant primary brain tumors with poor outcome, and four different molecular subtypes (Mesenchymal, Proneural, Neural, and Classical) are popularly applied in scientific researches and clinics of gliomas. Public databases contain an abundant genome-wide resource to explore the potential biomarker and molecular mechanisms using the informatics analysis. The aim of this study was to discover the potential biomarker and investigate its effect in gliomas. Weighted gene co-expression network analysis (WGCNA) was used to construct the co-expression modules and explore the biomarker among the dataset CGGA mRNAseq_693 carrying 693 glioma samples. Functional annotations, ROC, correlation, survival, univariate, and multivariate Cox regression analyses were implemented to investigate the functional effect in gliomas, and molecular experiments in vitro were performed to study the biological effect on glioma pathogenesis. The brown module was found to be strongly related to WHO grade of gliomas, and KEGG pathway analysis demonstrated that TNFRSF1A was enriched in MAPK signaling pathway and TNF signaling pathway. Overexpressed TNFRSF1A was strongly related to clinical features such as WHO grade, and functioned as an independent poor prognostic predictor of glioma patients. Notably, TNFRSF1A was preferentially upregulated in the Mesenchymal subtype gliomas (Mesenchymal-associated). Knockdown of TNFRSF1A inhibited proliferation and migration of glioma cell lines in vitro. Our findings provide a further understanding of the progression of gliomas, and Mesenchymal-associated TNFRSF1A might be a promising target of diagnosis, therapy, and prognosis of gliomas.
Collapse
Affiliation(s)
- Biao Yang
- Department of Neurosurgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan-Bo Pan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yan-Bin Ma
- Department of Neurosurgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sheng-Hua Chu
- Department of Neurosurgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
483
|
Sun H, Long S, Wu B, Liu J, Li G. NKCC1 involvement in the epithelial-to-mesenchymal transition is a prognostic biomarker in gliomas. PeerJ 2020; 8:e8787. [PMID: 32211242 PMCID: PMC7081783 DOI: 10.7717/peerj.8787] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 02/23/2020] [Indexed: 12/16/2022] Open
Abstract
Background Gliomas are the most prevalent type of intracranial tumors. NKCC1 is an important regulator in tumor cell volume. We noticed that abnormally high NKCC1 expression resulted in changes in the shape and adhesion of glioma cells. However, little is known about the role of NKCC1 in the epithelial-mesenchymal transition (EMT) of gliomas. This study aims to clarify the biological function of NKCC1 in glioblastoma multiforme (GBM) progression. Methods Using data from The Cancer Genome Atlas (TCGA), we performed a Kaplan–Meier analysis on NKCC1 expression levels to estimate the rate of survival of mesenchymal GBM patients. The correlation between NKCC1 and EMT-related proteins was analyzed from the Gene Expression Profiling Interactive Analysis (GEPIA) server. We conducted Gene Set Enrichment Analysis (GSEA) to verify molecular signatures and pathways. We then studied the expression of NKCC1 in grade I–IV glioma tissue samples collected from patients using immunohistochemistry (IHC). Finally, we evaluated the effects of NKCC1 migration and invasion on the cellular behaviors of U251 cells using the transwell assay and western blots. Results High NKCC1 expression was associated with poor prognoses in mesenchymal GBM. Our results suggest a correlation between NKCC1 and EMT-protein markers: CDH2 and VIM. GSEA showed that gliomas, TGF-beta signaling and EMT were enriched in the NKCC1 high expression phenotype. Higher expression levels of NKCC1 in gliomas correlate with higher glioma grades. Transwell assay and western blot results demonstrated that the knockdown of NKCC1 led to a reduction in migration and invasion, while also inhibiting MMP-2 and MMP-9 expression in U251. Conclusion These results suggest that high expression of NKCC1 regulates EMT in gliomas, providing a new therapeutic strategy for addressing the spread of gliomas by inhibiting the spread of intracranial tumors.
Collapse
Affiliation(s)
- Huaiyu Sun
- Department of Neurosurgery, First Hospital of China Medical University, Shenyang, China
| | - Shengrong Long
- Department of Neurosurgery, First Hospital of China Medical University, Shenyang, China
| | - Bingbing Wu
- Department of Neurosurgery, First Hospital of China Medical University, Shenyang, China
| | - Jia Liu
- Department of Neurosurgery, First Hospital of China Medical University, Shenyang, China
| | - Guangyu Li
- Department of Neurosurgery, First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
484
|
Liu W, Chai Y, Hu L, Wang J, Pan X, Yuan H, Zhao Z, Song Y, Zhang Y. Polyphyllin VI Induces Apoptosis and Autophagy via Reactive Oxygen Species Mediated JNK and P38 Activation in Glioma. Onco Targets Ther 2020; 13:2275-2288. [PMID: 32214827 PMCID: PMC7078907 DOI: 10.2147/ott.s243953] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Accepted: 03/02/2020] [Indexed: 12/16/2022] Open
Abstract
Background Polyphyllin VI (PPVI), a bioactive component derived from a traditional Chinese herb Paris polyphylla, exhibits potential antitumor activity against hepatocellular carcinoma, as well as breast and lung cancers. However, its effect on glioma remains unknown. Methods Five glioma cell lines (U251, U343, LN229, U87 and HEB) and an animal model were employed in the study. Anti-proliferation effects of PPVI were first determined using CCK-8 cell proliferation and clone formation assays, then reactive oxygen species (ROS), cell cycle progression and apoptosis effects measured by flow cytometry. The effect of PPVI on protein expression was quantified by Western blot analysis. Results Data showed that PPVI inhibited the proliferation of glioma cell lines by modulating the G2/M phase. Additionally, incubation of cells with PPVI promoted apoptosis, autophagy, increased accumulation of ROS and activated ROS-modulated JNK and p38 pathways. On the other hand, N-acetyl cysteine, a ROS inhibitor, attenuated PPVI-triggered effects. Furthermore, JNK and p38 inhibitors ameliorated PPVI-triggered autophagy and apoptosis in glioma cells. In vivo assays showed that PPVI inhibited tumor growth of U87 cell line in nude mice. Conclusion Overall, these data suggested that PPVI might be an effective therapeutic agent for glioma.
Collapse
Affiliation(s)
- Wei Liu
- School of Clinical Medicine, Tsinghua University, Beijing 10084, People's Republic of China
| | - Yi Chai
- School of Clinical Medicine, Tsinghua University, Beijing 10084, People's Republic of China
| | - Libo Hu
- School of Clinical Medicine, Tsinghua University, Beijing 10084, People's Republic of China
| | - Junhua Wang
- Department of Neurosurgery, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040, People's Republic of China
| | - Xin Pan
- Department of Neurosurgery, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040, People's Republic of China
| | - Hongyu Yuan
- State Key Laboratory of Molecular Oncology, National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, People's Republic of China
| | - Zitong Zhao
- State Key Laboratory of Molecular Oncology, National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, People's Republic of China
| | - Yongmei Song
- State Key Laboratory of Molecular Oncology, National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, People's Republic of China
| | - Yuqi Zhang
- School of Clinical Medicine, Tsinghua University, Beijing 10084, People's Republic of China
| |
Collapse
|
485
|
Hu X, Deng Q, Ma L, Li Q, Chen Y, Liao Y, Zhou F, Zhang C, Shao L, Feng J, He T, Ning W, Kong Y, Huo Y, He A, Liu B, Zhang J, Adams R, He Y, Tang F, Bian X, Luo J. Meningeal lymphatic vessels regulate brain tumor drainage and immunity. Cell Res 2020; 30:229-243. [PMID: 32094452 PMCID: PMC7054407 DOI: 10.1038/s41422-020-0287-8] [Citation(s) in RCA: 236] [Impact Index Per Article: 47.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 01/21/2020] [Indexed: 11/14/2022] Open
Abstract
Recent studies have shown that meningeal lymphatic vessels (MLVs), which are located both dorsally and basally beneath the skull, provide a route for draining macromolecules and trafficking immune cells from the central nervous system (CNS) into cervical lymph nodes (CLNs), and thus represent a potential therapeutic target for treating neurodegenerative and neuroinflammatory diseases. However, the roles of MLVs in brain tumor drainage and immunity remain unexplored. Here we show that dorsal MLVs undergo extensive remodeling in mice with intracranial gliomas or metastatic melanomas. RNA-seq analysis of MLV endothelial cells revealed changes in the gene sets involved in lymphatic remodeling, fluid drainage, as well as inflammatory and immunological responses. Disruption of dorsal MLVs alone impaired intratumor fluid drainage and the dissemination of brain tumor cells to deep CLNs (dCLNs). Notably, the dendritic cell (DC) trafficking from intracranial tumor tissues to dCLNs decreased in mice with defective dorsal MLVs, and increased in mice with enhanced dorsal meningeal lymphangiogenesis. Strikingly, disruption of dorsal MLVs alone, without affecting basal MLVs or nasal LVs, significantly reduced the efficacy of combined anti-PD-1/CTLA-4 checkpoint therapy in striatal tumor models. Furthermore, mice bearing tumors overexpressing VEGF-C displayed a better response to anti-PD-1/CTLA-4 combination therapy, and this was abolished by CCL21/CCR7 blockade, suggesting that VEGF-C potentiates checkpoint therapy via the CCL21/CCR7 pathway. Together, the results of our study not only demonstrate the functional aspects of MLVs as classic lymphatic vasculature, but also highlight that they are essential in generating an efficient immune response against brain tumors.
Collapse
Affiliation(s)
- Xueting Hu
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, 100871, Beijing, China
| | - Qiuping Deng
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, 100871, Beijing, China
| | - Lu Ma
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, 100871, Beijing, China
| | - Qingqing Li
- Biodynamic Optical Imaging Center, College of Life Sciences, Peking University, 100871, Beijing, China
| | - Yidong Chen
- Biodynamic Optical Imaging Center, College of Life Sciences, Peking University, 100871, Beijing, China
| | - Yuhan Liao
- Biodynamic Optical Imaging Center, College of Life Sciences, Peking University, 100871, Beijing, China
| | - Fan Zhou
- State Key Laboratory of Proteomics, Translational Medicine Center of Stem Cells, 307-Ivy Translational Medicine Center, Laboratory of Oncology, Affiliated Hospital, Academy of Military Medical Sciences, 100071, Beijing, China
| | - Chen Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, 100069, Beijing, China
| | - Linlin Shao
- Eye Hospital of China Academy of Chinese Medical Sciences, 100040, Beijing, China
| | - Jun Feng
- Eye Hospital of China Academy of Chinese Medical Sciences, 100040, Beijing, China
| | - Tubao He
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, 100871, Beijing, China
| | - Weihai Ning
- Sanbo Brain Hospital, Capital Medical University, 100093, Beijing, China
| | - Yan Kong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Cell Biology, Peking University Cancer Hospital and Institute, 100142, Beijing, China
| | - Yingqing Huo
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, 100871, Beijing, China
| | - Aibin He
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, 100871, Beijing, China
- Peking-Tsinghua Center for Life Sciences, 100871, Beijing, China
| | - Bing Liu
- State Key Laboratory of Proteomics, Translational Medicine Center of Stem Cells, 307-Ivy Translational Medicine Center, Laboratory of Oncology, Affiliated Hospital, Academy of Military Medical Sciences, 100071, Beijing, China
| | - Jingjing Zhang
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Ralf Adams
- Max-Planck-Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, and University of Münster, Faculty of Medicine, Münster, D-48149, Germany
| | - Yulong He
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Fuchou Tang
- Biodynamic Optical Imaging Center, College of Life Sciences, Peking University, 100871, Beijing, China
- Peking-Tsinghua Center for Life Sciences, 100871, Beijing, China
| | - Xiuwu Bian
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of the Ministry of Education, Southwest Hospital, Army Medical University (Third Military Medical University), 400038, Chongqing, China
| | - Jincai Luo
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, 100871, Beijing, China.
| |
Collapse
|
486
|
Peng L, Liang Y, Zhong X, Liang Z, Tian Y, Li S, Liang J, Wang R, Zhong Y, Shi Y, Zhang X. Aptamer-Conjugated Gold Nanoparticles Targeting Epidermal Growth Factor Receptor Variant III for the Treatment of Glioblastoma. Int J Nanomedicine 2020; 15:1363-1372. [PMID: 32184591 PMCID: PMC7053811 DOI: 10.2147/ijn.s238206] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 02/21/2020] [Indexed: 12/13/2022] Open
Abstract
Purpose In this study, we constructed novel brain-targeting complexes (U2-AuNP) by conjugating aptamer U2 to the gold nanoparticle (AuNPs) surface as a promising option for GBM therapy. Materials and Methods The properties of the U2-AuNP complexes were thoroughly characterized. Then, we detected the in vitro effects of U2-AuNP in U87-EGFRvIII cell lines and the in vivo antitumor effects of U2-AuNP in GBM-bearing mice. Furthermore, we explored the inhibition mechanism of U2-AuNP in U87-EGFRvIII cell lines. Results We found that U2-AuNP inhibits the proliferation and invasion of U87-EGFRvIII cell lines and prolongs the survival time of GBM-bearing mice. We found that U2-AuNP can inhibit the EGFR-related pathway and prevent DNA damage repair in GBM cells. Conclusion These results reveal the promising potential of U2-AuNP as a drug candidate for targeted therapy in GBM.
Collapse
Affiliation(s)
- Li Peng
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, People's Republic of China.,The Fifth Affiliated Hospital, Southern Medical University, Guangzhou 510900, People's Republic of China
| | - Yanling Liang
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Xinxin Zhong
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Zhiman Liang
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, People's Republic of China.,The Fifth Affiliated Hospital, Southern Medical University, Guangzhou 510900, People's Republic of China
| | - Yinghong Tian
- Experiment Teaching & Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Shuji Li
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Jingxue Liang
- The First Affiliated Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Ransheng Wang
- The First Affiliated Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Yuqi Zhong
- The First Affiliated Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Yusheng Shi
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Xingmei Zhang
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, People's Republic of China
| |
Collapse
|
487
|
Xiao Y, Zhu Z, Li J, Yao J, Jiang H, Ran R, Li X, Li Z. Expression and prognostic value of long non-coding RNA H19 in glioma via integrated bioinformatics analyses. Aging (Albany NY) 2020; 12:3407-3430. [PMID: 32081833 PMCID: PMC7066912 DOI: 10.18632/aging.102819] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 01/27/2020] [Indexed: 12/16/2022]
Abstract
Numerous discoveries have elucidated that long noncoding RNAs (lncRNAs) play a critical role in cancer malignant progression. However, their potential involvement in gliomas remains to be explored. Herein, the expression level of lncRNA H19 in glioma tissues, and its relevance with clinical characteristics were analyzed through Oncomine. The results showed that H19 was highly expressed in glioma tissues and its expression increased with the increase of malignancy. Next, GTEx and TCGA data were downloaded for differently expressed genes (DEGs) identification, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses, and the correlation analyses between H19 expression and clinic features. Radiation therapy had a good effect on glioblastoma multiforme (GBM), but didn't have a good effect on low grade glioma (LGG). Meanwhile, the expression level of H19 could act as an indicator molecule indicating the effect of radiotherapy. Finally, gene set enrichment analysis (GSEA) and immune infiltration analysis were conducted. It was found that H19 could affect the immune infiltration level of glioma through copy number variations, thus affecting the prognosis of glioma patients. Collectively, H19 may be involved in the occurrence and development of glioma, and has potential reference value for the relief and immunotherapy of glioma.
Collapse
Affiliation(s)
- Yilei Xiao
- Department of Neurosurgery, Liaocheng People’s Hospital, Liaocheng 250000, Shandong Province, P.R. China
| | - Zipeng Zhu
- Department of Neurosurgery, Liaocheng People’s Hospital, Liaocheng 250000, Shandong Province, P.R. China
| | - Jianxiong Li
- Department of Chemotherapy, Chinese PLA General Hospital, Beijing 100036, P.R. China
| | - Jie Yao
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, P.R. China
| | - Haitao Jiang
- Department of Neurosurgery, Liaocheng People’s Hospital, Liaocheng 250000, Shandong Province, P.R. China
| | - Ran Ran
- Department of Neurosurgery, Liaocheng People’s Hospital, Liaocheng 250000, Shandong Province, P.R. China
| | - Xueyuan Li
- Department of Neurosurgery, Liaocheng People’s Hospital, Liaocheng 250000, Shandong Province, P.R. China
| | - Zhiqiang Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, P.R. China
| |
Collapse
|
488
|
Yuan T, Zuo Z, Ying J, Jin L, Kang J, Gui S, Wang R, Li C. Structural and Functional Alterations in the Contralesional Medial Temporal Lobe in Glioma Patients. Front Neurosci 2020; 14:10. [PMID: 32153348 PMCID: PMC7044242 DOI: 10.3389/fnins.2020.00010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 01/08/2020] [Indexed: 01/13/2023] Open
Abstract
Background The human brain has an extraordinary ability to functionally change or reorganize its structure in response to disease. The aim of this study is to assess the structural and functional plasticity of contralesional medial temporal lobe (MTL) in patients with unilateral MTL glioma. Methods Sixty-eight patients with unilateral MTL glioma (left MTL glioma, n = 33; right MTL glioma, n = 35) and 40 healthy controls were recruited and scanned with 3D T1 MRI and rest-fMRI. We explored the structure of the contralesional MTL using voxel-based morphometry (VBM) and assessed the memory networks of the contralesional hemisphere using resting-state functional connectivity (rs-FC). The association between FC and cognitive function was assessed with partial correlation analysis. Results Compared with healthy controls, both patient groups exhibited (1) a large cluster of voxels with gray matter (GM) volume decrease in the contralesional MTL using region of interest (ROI)-based VBM analysis (cluster level p < 0.05, FDR corrected); and (2) decreased intrahemispheric FC between the posterior hippocampus (pHPC) and posterior cingulate cortex (PCC) (p < 0.01, Bonferroni corrected). Intrahemispheric FC between the pHPC and PCC was positively correlated with cognitive function in both patient groups. Conclusion Using multi-modality brain imaging tools, we found structural and functional changes in the contralesional MTL in patients with unilateral MTL glioma. These findings suggest that the contralesional cortex may have decompensation of structure and function in patients with unilateral glioma, except for compensatory structural and functional adaptations. Our study provides additional insight into the neuroanatomical and functional network changes in the contralesional cortex in patients with glioma.
Collapse
Affiliation(s)
- Taoyang Yuan
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Zhentao Zuo
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,Sino-Danish College, University of Chinese Academy of Sciences, Beijing, China
| | - Jianyou Ying
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Lu Jin
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jie Kang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Songbai Gui
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Rui Wang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Chuzhong Li
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Beijing Institute for Brain Disorders Brain Tumour Center, Beijing, China
| |
Collapse
|
489
|
Gong H, Gao S, Yu C, Li M, Liu P, Zhang G, Song J, Zheng J. Effect and mechanism of YB-1 knockdown on glioma cell growth, migration, and apoptosis. Acta Biochim Biophys Sin (Shanghai) 2020; 52:168-179. [PMID: 32047913 DOI: 10.1093/abbs/gmz161] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Indexed: 12/13/2022] Open
Abstract
Y-box binding protein 1 (YB-1) is manifested as its involvement in cell proliferation and differentiation and malignant cell transformation. Overexpression of YB-1 is associated with glioma progression and patient survival. The aim of this study is to investigate the influence of YB-1 knockdown on glioma cell progression and reveal the mechanisms of YB-1 knockdown on glioma cell growth, migration, and apoptosis. It was found that the knockdown of YB-1 decreased the mRNA and protein levels of YB-1 in U251 glioma cells. The knockdown of YB-1 significantly inhibited cell proliferation, colony formation, and migration in vitro and tumor growth in vivo. Proteome and phosphoproteome data revealed that YB-1 is involved in glioma progression through regulating the expression and phosphorylation of major proteins involved in cell cycle, adhesion, and apoptosis. The main regulated proteins included CCNB1, CCNDBP1, CDK2, CDK3, ADGRG1, CDH-2, MMP14, AIFM1, HO-1, and BAX. Furthermore, it was also found that YB-1 knockdown is associated with the hypo-phosphorylation of ErbB, mTOR, HIF-1, cGMP-PKG, and insulin signaling pathways, and proteoglycans in cancer. Our findings indicated that YB-1 plays a key role in glioma progression in multiple ways, including regulating the expression and phosphorylation of major proteins associated with cell cycle, adhesion, and apoptosis.
Collapse
Affiliation(s)
- Huilin Gong
- Department of Pathology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Shan Gao
- Department of Kidney Transplant, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Chenghuan Yu
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou 310013, China
| | - Meihe Li
- Department of Kidney Transplant, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Ping Liu
- Department of Dermatology, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Guanjun Zhang
- Department of Pathology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Jinning Song
- Department of Neurosurgery, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Jin Zheng
- Department of Kidney Transplant, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| |
Collapse
|
490
|
Mirabdaly S, Elieh Ali Komi D, Shakiba Y, Moini A, Kiani A. Effects of temozolomide on U87MG glioblastoma cell expression of CXCR4, MMP2, MMP9, VEGF, anti-proliferatory cytotoxic and apoptotic properties. Mol Biol Rep 2020; 47:1187-1197. [PMID: 31897867 DOI: 10.1007/s11033-019-05219-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 11/30/2019] [Indexed: 12/13/2022]
Abstract
Temozolomide is an alkylating agent which is used in glioblastoma treatment. We aimed to investigate the effects of different concentrations of temozolomide and exposure time on U87MG glioblastoma cell expression of CXCR4, MMP2, MMP9 and VEGF. U87MG cells were cultured in different temozolomide concentrations and incubation time and the effects of temozolomide on inducing apoptosis was investigated. The levels of VEGF and CXCR4 expression were measured by RT-PCR and flowcytometry. Moreover, MMP2 and MMP9 activity and expression were assessed by ELISA and zymography. CXCR4 and VEGF expression levels decreased upon applying higher concentration of temozolomide. MMP2 and MMP-9 had lower activity in cells with longer exposure time or higher doses of temozolomide. Temozolomide induces the apoptosis in U87MG glioblastoma cells at therapeutic or higher dose. It is capable of decreasing their expression levels of VEGF and CXCR4.
Collapse
Affiliation(s)
- Seyedsaber Mirabdaly
- Students Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Daniel Elieh Ali Komi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Shakiba
- Regenerative Medicine Research Center, Kermanshah University of Medical Sciences, PO-Box: 6714869914, Kermanshah, Iran
| | - Ali Moini
- Department of Internal Medicine Imam, Reza Hospital Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Amir Kiani
- Regenerative Medicine Research Center, Kermanshah University of Medical Sciences, PO-Box: 6714869914, Kermanshah, Iran. .,Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
491
|
In silico Design of Novel Histone Deacetylase 4 Inhibitors: Design Guidelines for Improved Binding Affinity. Int J Mol Sci 2019; 21:ijms21010219. [PMID: 31905609 PMCID: PMC6981887 DOI: 10.3390/ijms21010219] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/19/2019] [Accepted: 12/25/2019] [Indexed: 02/02/2023] Open
Abstract
Histone deacetylases (HDAC) are being targeted for a number of diseases such as cancer, inflammatory disease, and neurological disorders. Within this family of 18 isozymes, HDAC4 is a prime target for glioma, one of the most aggressive brain tumors reported. Thus, the development of HDAC4 inhibitors could present a novel therapeutic route for glioma. In this work, molecular docking studies on cyclopropane hydroxamic acid derivatives identified five novel molecular interactions to the HDAC4 receptor that could be harnessed to enhance inhibitor binding. Thus, design guidelines for the optimization of potent HDAC4 inhibitors were developed which can be utilized to further the development of HDAC4 inhibitors. Using the developed guidelines, eleven novel cyclopropane hydroxamic acid derivatives were designed that outcompeted all original cyclopropane hydroxamic acids HDAC4 inhibitors studied in silico. The results of this work will be an asset to paving the way for further design and optimization of novel potent HDAC4 inhibitors for gliomas.
Collapse
|
492
|
Gomez-Zepeda D, Taghi M, Scherrmann JM, Decleves X, Menet MC. ABC Transporters at the Blood-Brain Interfaces, Their Study Models, and Drug Delivery Implications in Gliomas. Pharmaceutics 2019; 12:pharmaceutics12010020. [PMID: 31878061 PMCID: PMC7022905 DOI: 10.3390/pharmaceutics12010020] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/13/2019] [Accepted: 12/20/2019] [Indexed: 12/22/2022] Open
Abstract
Drug delivery into the brain is regulated by the blood-brain interfaces. The blood-brain barrier (BBB), the blood-cerebrospinal fluid barrier (BCSFB), and the blood-arachnoid barrier (BAB) regulate the exchange of substances between the blood and brain parenchyma. These selective barriers present a high impermeability to most substances, with the selective transport of nutrients and transporters preventing the entry and accumulation of possibly toxic molecules, comprising many therapeutic drugs. Transporters of the ATP-binding cassette (ABC) superfamily have an important role in drug delivery, because they extrude a broad molecular diversity of xenobiotics, including several anticancer drugs, preventing their entry into the brain. Gliomas are the most common primary tumors diagnosed in adults, which are often characterized by a poor prognosis, notably in the case of high-grade gliomas. Therapeutic treatments frequently fail due to the difficulty of delivering drugs through the brain barriers, adding to diverse mechanisms developed by the cancer, including the overexpression or expression de novo of ABC transporters in tumoral cells and/or in the endothelial cells forming the blood-brain tumor barrier (BBTB). Many models have been developed to study the phenotype, molecular characteristics, and function of the blood-brain interfaces as well as to evaluate drug permeability into the brain. These include in vitro, in vivo, and in silico models, which together can help us to better understand their implication in drug resistance and to develop new therapeutics or delivery strategies to improve the treatment of pathologies of the central nervous system (CNS). In this review, we present the principal characteristics of the blood-brain interfaces; then, we focus on the ABC transporters present on them and their implication in drug delivery; next, we present some of the most important models used for the study of drug transport; finally, we summarize the implication of ABC transporters in glioma and the BBTB in drug resistance and the strategies to improve the delivery of CNS anticancer drugs.
Collapse
Affiliation(s)
- David Gomez-Zepeda
- Inserm, UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France; (M.T.); (J.-M.S.); (X.D.)
- Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France
- Sorbonne Paris Cité, Université Paris Diderot, 75013 Paris, France
- Correspondence: (D.G.-Z.); (M.-C.M.)
| | - Méryam Taghi
- Inserm, UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France; (M.T.); (J.-M.S.); (X.D.)
- Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France
- Sorbonne Paris Cité, Université Paris Diderot, 75013 Paris, France
| | - Jean-Michel Scherrmann
- Inserm, UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France; (M.T.); (J.-M.S.); (X.D.)
- Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France
- Sorbonne Paris Cité, Université Paris Diderot, 75013 Paris, France
| | - Xavier Decleves
- Inserm, UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France; (M.T.); (J.-M.S.); (X.D.)
- Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France
- Sorbonne Paris Cité, Université Paris Diderot, 75013 Paris, France
- UF Biologie du médicament et toxicologie, Hôpital Cochin, AP HP, 75006 Paris, France
| | - Marie-Claude Menet
- Inserm, UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France; (M.T.); (J.-M.S.); (X.D.)
- Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France
- Sorbonne Paris Cité, Université Paris Diderot, 75013 Paris, France
- UF Hormonologie adulte, Hôpital Cochin, AP HP, 75006 Paris, France
- Correspondence: (D.G.-Z.); (M.-C.M.)
| |
Collapse
|
493
|
Qu C, Yan C, Cao W, Li F, Qu Y, Guan K, Si C, Yu Z, Qu Z. miR-128-3p contributes to mitochondrial dysfunction and induces apoptosis in glioma cells via targeting pyruvate dehydrogenase kinase 1. IUBMB Life 2019; 72:465-475. [PMID: 31828927 DOI: 10.1002/iub.2212] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 11/30/2019] [Indexed: 12/13/2022]
Abstract
Glioma, like most cancers, possesses a unique bioenergetic state of aerobic glycolysis known as the Warburg effect, which is a dominant phenotype of most tumor cells. Glioma tumors exhibit high glycolytic metabolism with increased lactate production. Data derived from the gene expression profiling interactive analysis (GEPIA) database show that pyruvate dehydrogenase kinase 1 (PDK1) is significantly highly expressed in glioma tissues compared with corresponding normal tissues. PDK1 is a key enzyme in the transition of glycolysis to tricarboxylic acid cycle, via inactivating PDH and converting oxidative phosphorylation to Warburg effect, resulting in increment of lactate production. Silencing of PDK1 expression resulted in reduced lactate and ATP, accumulation of ROS, mitochondrial damage, decreased cell growth, and increased cell apoptosis. Aberrant expression of miR-128 has been observed in many human malignancies. Mechanistically, we discover that overexpressed miR-128-3p disturbs the Warburg effect in glioma cells via reducing PDK1. Our experiments confirmed that the miR-128-3p/PDK1 axis played a pivotal role in cancer cell metabolism and growth. Collectively, these findings suggest that therapeutic strategies to modulate the Warburg effect, such as targeting of PDK1, might act as a potential therapeutic target for glioma treatment.
Collapse
Affiliation(s)
- Changda Qu
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chang Yan
- Department of Anesthesiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Weifan Cao
- College of Life Science, Northeast Forestry University, Harbin, China
| | - Fangqin Li
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yewei Qu
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ke Guan
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chengqing Si
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ze Yu
- Guangzhou Institute of Oncology, Cancer Center of Guangzhou Medical University, Guangzhou, China
| | - Zhangyi Qu
- Department of Microbiology, Public Health College, Harbin Medical University, Harbin, China
| |
Collapse
|
494
|
Smith-Cohn M, Davidson C, Colman H, Cohen AL. Challenges of targeting BRAF V600E mutations in adult primary brain tumor patients: a report of two cases. CNS Oncol 2019; 8:CNS48. [PMID: 31818130 PMCID: PMC6912849 DOI: 10.2217/cns-2019-0018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 11/19/2019] [Indexed: 12/18/2022] Open
Abstract
Aim: Therapeutic targeting of BRAF alterations in primary brain tumor patients has demonstrated clinical activity in case reports and early trials; however, there is limited high-level evidence of the efficacy. Patients & results: Targeting BRAF V600E mutations with concurrent dabrafenib and trametinib in anaplastic pleomorphic xanthoastrocytoma resulted in a transient radiographic and clinical response and no therapeutic benefit in a patient with an epithelioid glioblastoma. Conclusion:BRAF/MEK inhibition did not produce a durable treatment effect in glioblastoma or pleomorphic xanthoastrocytoma with BRAF V600E alterations. Heterogenicity of related cases in the literature makes an evaluation of efficacy BRAF targeting therapies in gliomas difficult and requires additional investigation.
Collapse
Affiliation(s)
- Matthew Smith-Cohn
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Christian Davidson
- Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - Howard Colman
- Department of Neurosurgery, Huntsman Cancer Institute, & Clinical Neuroscience Center, University of Utah, Salt Lake City, UT 84112, USA
| | - Adam L Cohen
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
495
|
Sood D, Tang-Schomer M, Pouli D, Mizzoni C, Raia N, Tai A, Arkun K, Wu J, Black LD, Scheffler B, Georgakoudi I, Steindler DA, Kaplan DL. 3D extracellular matrix microenvironment in bioengineered tissue models of primary pediatric and adult brain tumors. Nat Commun 2019; 10:4529. [PMID: 31586101 PMCID: PMC6778192 DOI: 10.1038/s41467-019-12420-1] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 08/27/2019] [Indexed: 12/15/2022] Open
Abstract
Dynamic alterations in the unique brain extracellular matrix (ECM) are involved in malignant brain tumors. Yet studies of brain ECM roles in tumor cell behavior have been difficult due to lack of access to the human brain. We present a tunable 3D bioengineered brain tissue platform by integrating microenvironmental cues of native brain-derived ECMs and live imaging to systematically evaluate patient-derived brain tumor responses. Using pediatric ependymoma and adult glioblastoma as examples, the 3D brain ECM-containing microenvironment with a balance of cell-cell and cell-matrix interactions supports distinctive phenotypes associated with tumor type-specific and ECM-dependent patterns in the tumor cells' transcriptomic and release profiles. Label-free metabolic imaging of the composite model structure identifies metabolically distinct sub-populations within a tumor type and captures extracellular lipid-containing droplets with potential implications in drug response. The versatile bioengineered 3D tumor tissue system sets the stage for mechanistic studies deciphering microenvironmental role in brain tumor progression.
Collapse
Affiliation(s)
- Disha Sood
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Min Tang-Schomer
- Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA.,Connecticut Children's Medical Center, Harford, CT, 06106, USA
| | - Dimitra Pouli
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA.,Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Craig Mizzoni
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Nicole Raia
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Albert Tai
- Genomics Core, Tufts University School of Medicine, Boston, MA, 02111, USA
| | - Knarik Arkun
- Department of Pathology and Laboratory Medicine, Tufts Medical Center, Boston, MA, 02111, USA
| | - Julian Wu
- Department of Neurosurgery, Tufts Medical Center, Boston, MA, 02111, USA
| | - Lauren D Black
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Bjorn Scheffler
- Department of Neuroscience, University of Florida, McKnight Brain Institute, Gainesville, FL, 32610, USA.,DKFZ-Division of Translational Oncology/ Neurooncology, German Cancer Consortium (DKTK), Heidelberg & University Hospital Essen, Essen, Germany
| | - Irene Georgakoudi
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Dennis A Steindler
- Department of Neuroscience, University of Florida, McKnight Brain Institute, Gainesville, FL, 32610, USA.,Neuroscience and Aging Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, 02111, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA.
| |
Collapse
|
496
|
Discovery of a Ruthenium Complex for the Theranosis of Glioma through Targeting the Mitochondrial DNA with Bioinformatic Methods. Int J Mol Sci 2019; 20:ijms20184643. [PMID: 31546801 PMCID: PMC6770666 DOI: 10.3390/ijms20184643] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 09/05/2019] [Accepted: 09/17/2019] [Indexed: 12/11/2022] Open
Abstract
Glioma is the most aggressive and lethal brain tumor in humans. Mutations of mitochondrial DNA (mtDNA) are commonly found in tumor cells and are closely associated with tumorigenesis and progress. However, glioma-specific inhibitors that reflect the unique feature of tumor cells are rare. Here we uncover RC-7, a ruthenium complex with strong red fluorescence, could bind with glioma mtDNA and then inhibited the growth of human glioma cells but not that of neuronal cells, liver, or endothelial cells. RC-7 significantly reduced energy production and increased the oxidative stress in the glioma cells. Administration of RC-7 into mice not only could be observed in the glioma mass of brain by fluorescence imaging, but also obviously prevented the growth of xenograft glioma and prolonged mouse survival days. The findings suggested the theranostic application of a novel type of complex through targeting the tumor mtDNA.
Collapse
|
497
|
Abstract
PURPOSE OF REVIEW This article discusses the diagnosis and management of neoplasms that affect the spinal cord as well as spinal cord disorders that can occur due to cancer treatments. RECENT FINDINGS Neoplastic myelopathies are uncommon neurologic disorders but cause significant morbidity when they occur. Primary spinal cord tumors can be classified into intramedullary, intradural extramedullary, or extradural tumors. Diffuse gliomas and ependymal tumors are the most common intramedullary tumors. Diffuse gliomas include the World Health Organization (WHO) grade II and grade III astrocytomas, the grade II and grade III oligodendrogliomas, the grade IV glioblastomas, and newly recognized pediatric diffuse midline gliomas with H3 K27M mutation. The majority of diffuse and anaplastic astrocytomas are IDH-mutant tumors, whereas only 10% of glioblastomas are IDH-mutant. Oligodendrogliomas are typically IDH-mutant and are characterized by the molecular signature of 1p/19q codeletion. Nine distinct molecular subgroups of ependymomas have been identified based on their genetic features and location. NF2 mutations are frequently found in spinal cord ependymomas. Metastatic tumors are the most common tumors of the spine and can be extradural, leptomeningeal, or, rarely, intramedullary. Extradural metastatic spinal cord compression is a neurologic emergency and should be promptly diagnosed as pretreatment neurologic status dictates the posttreatment outcome. SUMMARY Neoplastic myelopathies encompass many diagnoses ranging from benign and malignant spinal tumors to paraneoplastic syndromes heralding cancers. The knowledge of the clinical features and management of neoplastic myelopathies is essential to practicing neurologists as early diagnosis and treatment can prevent devastating neurologic sequelae.
Collapse
|
498
|
Samudra N, Zacharias T, Plitt A, Lega B, Pan E. Seizures in glioma patients: An overview of incidence, etiology, and therapies. J Neurol Sci 2019; 404:80-85. [PMID: 31352293 DOI: 10.1016/j.jns.2019.07.026] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/24/2019] [Accepted: 07/18/2019] [Indexed: 12/19/2022]
Abstract
Gliomas are fatal brain tumors, and even low-grade gliomas (LGGs) have an average survival of less than a decade. Seizures are a common presentation of gliomas, particularly LGGs, and substantially impact quality of life. Glioma-related seizures differ from other focal epilepsies in their pathogenesis and in the likelihood of refractory epilepsy. We review factors that predict seizure activity and response to treatment, optimal pharmacologic and surgical management of glioma-related epilepsy, and the benefit of using newer anti-seizure medications in patients with gliomas. As surgery is so often beneficial with seizure reduction, we discuss oncologic and epilepsy surgery perspectives. Treatment of gliomas has the potential to ameliorate seizures and increase rates of seizure freedom. Prospective, well-powered studies are needed to provide more definitive answers for practitioners taking care of glioma patients with seizures.
Collapse
Affiliation(s)
- Niyatee Samudra
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Tresa Zacharias
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Aaron Plitt
- Department of Neurosurgery, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Bradley Lega
- Department of Neurosurgery, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Edward Pan
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA.
| |
Collapse
|
499
|
Bailly C, Vidal A, Bonnemaire C, Kraeber-Bodéré F, Chérel M, Pallardy A, Rousseau C, Garcion E, Lacoeuille F, Hindré F, Valable S, Bernaudin M, Bodet-Milin C, Bourgeois M. Potential for Nuclear Medicine Therapy for Glioblastoma Treatment. Front Pharmacol 2019; 10:772. [PMID: 31354487 PMCID: PMC6637301 DOI: 10.3389/fphar.2019.00772] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 06/14/2019] [Indexed: 12/17/2022] Open
Abstract
Glioblastoma is the most common malignant adult brain tumor and has a very poor patient prognosis. The mean survival for highly proliferative glioblastoma is only 10 to 14 months despite an aggressive current therapeutic approach known as Stupp's protocol, which consists of debulking surgery followed by radiotherapy and chemotherapy. Despite several clinical trials using anti-angiogenic targeted therapies, glioblastoma medical care remains without major progress in the last decade. Recent progress in nuclear medicine, has been mainly driven by advances in biotechnologies such as radioimmunotherapy, radiopeptide therapy, and radionanoparticles, and these bring a new promising arsenal for glioblastoma therapy. For therapeutic purposes, nuclear medicine practitioners classically use β- particle emitters like 131I, 90Y, 186/188Re, or 177Lu. In the glioblastoma field, these radioisotopes are coupled with nanoparticles, monoclonal antibodies, or peptides. These radiopharmaceutical compounds have resulted in a stabilization and/or improvement of the neurological status with only transient side effects. In nuclear medicine, the glioblastoma-localized and targeted internal radiotherapy proof-of-concept stage has been successfully demonstrated using β- emitting isotopes. Similarly, α particle emitters like 213Bi, 211At, or 225Ac appear to be an innovative and interesting alternative. Indeed, α particles deliver a high proportion of their energy inside or at close proximity to the targeted cells (within a few micrometers from the emission point versus several millimeters for β- particles). This physical property is based on particle-matter interaction differences and results in α particles being highly efficient in killing tumor cells with minimal irradiation of healthy tissues and permits targeting of isolated tumor cells. The first clinical trials confirmed this idea and showed good therapeutic efficacy and less side effects, thus opening a new and promising era for glioblastoma medical care using α therapy. The objective of this literature review is focused on the developing field of nuclear medicine and aims to describe the various parameters such as targets, vectors, isotopes, or injection route (systemic and local) in relation to the clinical and preclinical results in glioblastoma pathology.
Collapse
Affiliation(s)
- Clément Bailly
- Nuclear Medicine, Centre Hospitalier Universitaire (CHU) de Nantes, Nantes, France.,CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | | | | | - Françoise Kraeber-Bodéré
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Nuclear Medecine, Centre Hospitalier Universitaire (CHU) de Nantes, Nantes, France
| | - Michel Chérel
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Institut de Cancérologie de l'Ouest (ICO), Angers, France
| | - Amandine Pallardy
- Nuclear Medicine, Centre Hospitalier Universitaire (CHU) de Nantes, Nantes, France
| | | | - Emmanuel Garcion
- Team 17-Design and Application of Innovative Local Treatments in Glioblastoma, INSERM U1232 Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), Nantes, France
| | - Franck Lacoeuille
- Team 17-Design and Application of Innovative Local Treatments in Glioblastoma, INSERM U1232 Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), Nantes, France.,Nuclear Medicine, Centre Hospitalier Universitaire d'Angers, Angers, France
| | - François Hindré
- Team 17-Design and Application of Innovative Local Treatments in Glioblastoma, INSERM U1232 Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), Nantes, France
| | | | | | - Caroline Bodet-Milin
- Nuclear Medicine, Centre Hospitalier Universitaire (CHU) de Nantes, Nantes, France.,CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Mickaël Bourgeois
- Nuclear Medicine, Centre Hospitalier Universitaire (CHU) de Nantes, Nantes, France.,CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Arronax, Saint-Herblain, France
| |
Collapse
|
500
|
Zhao Z, Liang T, Feng S. Silencing of HAS2-AS1 mediates PI3K/AKT signaling pathway to inhibit cell proliferation, migration, and invasion in glioma. J Cell Biochem 2019; 120:11510-11516. [PMID: 30790335 DOI: 10.1002/jcb.28430] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 11/22/2018] [Accepted: 11/29/2018] [Indexed: 01/24/2023]
Abstract
Hyaluronan synthase 2 (HAS2)-AS1 (natural antisense transcript of HAS2) functions as oncogenic long noncoding RNA (lncRNA) in oral squamous cell carcinoma, breast cancer, and osteosarcoma. The role of HAS2-AS1 in glioma remains unknown. In our research, HAS2-AS1 expression was elevated in glioma tissues compared with normal brain tissues. Moreover, high levels of HAS2-AS1 expression was observed in patients with glioma with high WHO grade (III-IV) or large tumor size ( > 4 cm). The survival analysis from The Cancer Genome Atlas showed glioma cases with high HAS2-AS1 expression that had shorter disease-free survival time and overall survival time than those with low HAS2-AS1 expression. In vitro studies suggested that knocking down HAS2-AS1 expression inhibited glioma cell viability, migration, and invasion through phosphoinositide 3-kinase/protein kinase B signaling pathway. In conclusion, HAS2-AS1 may be considered as a predictor for clinical outcome and a potential therapeutic target in glioma.
Collapse
Affiliation(s)
- Zhenyi Zhao
- Department of Neurosurgery, Tianjin Baodi Hospital, Baodi Clinical College of Tianjin Medical University, Tianjin, China
| | - Tiansong Liang
- Department of Radiotherpy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shijun Feng
- Department of Neurosurgery, The First Hospital Attached to Baotou Medical College of Inner Mongolia University of Science & Technology, Baotou, Inner Mongolia, China
| |
Collapse
|