1
|
Li Z, Du L, Du B, Ullah Z, Zhang Y, Tu Y, Zhou Y, Guo B. Inorganic and hybrid nanomaterials for NIR-II fluorescence imaging-guided therapy of Glioblastoma and perspectives. Theranostics 2025; 15:5616-5665. [PMID: 40365286 PMCID: PMC12068291 DOI: 10.7150/thno.112204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 03/24/2025] [Indexed: 05/15/2025] Open
Abstract
Glioblastoma (GBM) is the most invasive and lethal brain tumor, with limited therapeutic options due to its highly infiltrative nature, resistance to conventional therapies, and blood-brain barriers. Recent advancements in near-infrared II (NIR-II) fluorescence imaging have facilitated greater tissue penetration, improved resolution, and real-time visualization of GBM, providing a promising approach for precise diagnosis and treatment. The inorganic and hybrid NIR-II fluorescent materials have developed rapidly for NIR-II fluorescence imaging-guided diagnosis and therapy of many diseases, including GBM. Herein, we offer a timely update to explore the contribution of inorganic/hybrid NIR-II fluorescent nanomaterials, such as quantum dots, rare-earth-doped nanoparticles, carbon-based nanomaterials, and metal nanoclusters in imaging-guided treatment for GBM. These nanomaterials provide high photostability, strong fluorescence intensity, and tunable optical properties, allowing for multimodal imaging and enhanced therapeutic efficacy. Additionally, their integration with modern therapeutic strategies, such as photothermal therapy, chemodynamic therapy, photodynamic therapy, sonodynamic therapy, and immunotherapy, has shown significant potential in overcoming the limitations of traditional treatments. Looking forward, future advancements including safe body clearance, long-term biocompatibility, efficient BBB penetration, and extended emission wavelengths beyond 1500 nm could enhance the theranostic outcomes. The integration of dual imaging with immunotherapy and AI-driven strategies will further enhance precision and accelerate the clinical translation of smart theranostic platforms for GBM treatment.
Collapse
Affiliation(s)
- Zhigang Li
- Department of Medical Imaging, Shenzhen Longhua District Central Hospital, Shenzhen Longhua District Key Laboratory of Neuroimaging, Shenzhen 518110, China
| | - Lixin Du
- Department of Medical Imaging, Shenzhen Longhua District Central Hospital, Shenzhen Longhua District Key Laboratory of Neuroimaging, Shenzhen 518110, China
| | - Binghua Du
- Department of Medical Imaging, Shenzhen Longhua District Central Hospital, Shenzhen Longhua District Key Laboratory of Neuroimaging, Shenzhen 518110, China
| | - Zia Ullah
- School of Science, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen 518055, China
| | - Yinghe Zhang
- School of Science, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen 518055, China
| | - Yanyang Tu
- Research Center, Huizhou Central People's Hospital, Guangdong Medical University, Huizhou City, Guangdong Province, China
| | - Ying Zhou
- Department of Pharmacy, Peking University First Hospital, Beijing, China
| | - Bing Guo
- School of Science, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen 518055, China
| |
Collapse
|
2
|
Norollahi SE, Yousefi B, Nejatifar F, Yousefzadeh-Chabok S, Rashidy-Pour A, Samadani AA. Practical immunomodulatory landscape of glioblastoma multiforme (GBM) therapy. J Egypt Natl Canc Inst 2024; 36:33. [PMID: 39465481 DOI: 10.1186/s43046-024-00240-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 09/21/2024] [Indexed: 10/29/2024] Open
Abstract
Glioblastoma multiforme (GBM) is the most common harmful high-grade brain tumor with high mortality and low survival rate. Importantly, besides routine diagnostic and therapeutic methods, modern and useful practical techniques are urgently needed for this serious malignancy. Correspondingly, the translational medicine focusing on genetic and epigenetic profiles of glioblastoma, as well as the immune framework and brain microenvironment, based on these challenging findings, indicates that key clinical interventions include immunotherapy, such as immunoassay, oncolytic viral therapy, and chimeric antigen receptor T (CAR T) cell therapy, which are of great importance in both diagnosis and therapy. Relatively, vaccine therapy reflects the untapped confidence to enhance GBM outcomes. Ongoing advances in immunotherapy, which utilizes different methods to regenerate or modify the resistant body for cancer therapy, have revealed serious results with many different problems and difficulties for patients. Safe checkpoint inhibitors, adoptive cellular treatment, cellular and peptide antibodies, and other innovations give researchers an endless cluster of instruments to plan profoundly in personalized medicine and the potential for combination techniques. In this way, antibodies that block immune checkpoints, particularly those that target the program death 1 (PD-1)/PD-1 (PD-L1) ligand pathway, have improved prognosis in a wide range of diseases. However, its use in combination with chemotherapy, radiation therapy, or monotherapy is ineffective in treating GBM. The purpose of this review is to provide an up-to-date overview of the translational elements concentrating on the immunotherapeutic field of GBM alongside describing the molecular mechanism involved in GBM and related signaling pathways, presenting both historical perspectives and future directions underlying basic and clinical practice.
Collapse
Affiliation(s)
- Seyedeh Elham Norollahi
- Cancer Research Center and, Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran
| | - Bahman Yousefi
- Cancer Research Center and, Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran
| | - Fatemeh Nejatifar
- Department of Hematology and Oncology, School of Medicine, Razi Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Shahrokh Yousefzadeh-Chabok
- Guilan Road Trauma Research Center, Trauma Institute, Guilan University of Medical Sciences, Rasht, Iran
- , Rasht, Iran
| | - Ali Rashidy-Pour
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran.
| | - Ali Akbar Samadani
- Guilan Road Trauma Research Center, Trauma Institute, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
3
|
Le MK, Tran NQV, Nguyen PT, Nguyen TA, Nakao A, Kondo T. Comprehensive analysis of distinct circadian clock subtypes of adult diffuse glioma and their associations with clinicopathological, genetic, and epigenetic profiles. J Neuropathol Exp Neurol 2024; 83:736-744. [PMID: 38964366 DOI: 10.1093/jnen/nlae055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/06/2024] Open
Abstract
The circadian clock (CC) has biological and clinical implications in gliomas. Most studies focused on CC effects on the tumor microenvironment and the application of chronotherapy. The present study focused on CC gene expression patterns and intracellular oncogenic activities. Glioma gene expression data were collected from The Human Cancer Genome Atlas (TCGA) project. After applying inclusion and exclusion criteria, we selected 666 patients from TCGA-GBM and TCGA-LGG projects and included important clinicopathological variables. The entire cohort was subjected to clustering analysis and divided into CC1 and CC2 subtypes based on statistical, biological, and clinical criteria. CC2 gliomas showed higher expression of BMAL1 and CRY1 and lower expression of CRY2 and PER2 (adjusted P < .001). CC2 gliomas had q higher activity of cell proliferation, metabolic reprogramming, angiogenesis, hypoxia, and many oncogenic signals (P < .001). The CC2 subtype contained a higher proportion of glioblastomas (P < .001) and had a worse prognosis (P < .001). Stratified Kaplan-Meier and multivariable Cox analyses illustrated that the CC subtype is an independent prognostic factor to clinicopathological characteristics (P < .001), genetic aberrations (P = .006), and biological processes (P < .001). Thus, this study shows statistical evidence of CC subtypes and their biological, and clinicopathological significance in adult gliomas.
Collapse
Affiliation(s)
- Minh-Khang Le
- Department of Human Pathology, University of Yamanashi, Yamanashi, Japan
| | | | - Phuc-Tan Nguyen
- Department of Immunology, University of Yamanashi, Yamanashi, Japan
| | - Thuy-An Nguyen
- Department of Immunology, University of Yamanashi, Yamanashi, Japan
| | - Atsuhito Nakao
- Department of Immunology, University of Yamanashi, Yamanashi, Japan
| | - Tetsuo Kondo
- Department of Human Pathology, University of Yamanashi, Yamanashi, Japan
| |
Collapse
|
4
|
Song B, Wang X, Qin L, Hussain S, Liang W. Brain gliomas: Diagnostic and therapeutic issues and the prospects of drug-targeted nano-delivery technology. Pharmacol Res 2024; 206:107308. [PMID: 39019336 DOI: 10.1016/j.phrs.2024.107308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/19/2024]
Abstract
Glioma is the most common intracranial malignant tumor, with severe difficulty in treatment and a low patient survival rate. Due to the heterogeneity and invasiveness of tumors, lack of personalized clinical treatment design, and physiological barriers, it is often difficult to accurately distinguish gliomas, which dramatically affects the subsequent diagnosis, imaging treatment, and prognosis. Fortunately, nano-delivery systems have demonstrated unprecedented capabilities in diagnosing and treating gliomas in recent years. They have been modified and surface modified to efficiently traverse BBB/BBTB, target lesion sites, and intelligently release therapeutic or contrast agents, thereby achieving precise imaging and treatment. In this review, we focus on nano-delivery systems. Firstly, we provide an overview of the standard and emerging diagnostic and treatment technologies for glioma in clinical practice. After induction and analysis, we focus on summarizing the delivery methods of drug delivery systems, the design of nanoparticles, and their new advances in glioma imaging and treatment in recent years. Finally, we discussed the prospects and potential challenges of drug-delivery systems in diagnosing and treating glioma.
Collapse
Affiliation(s)
- Baoqin Song
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China
| | - Xiu Wang
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China.
| | - Lijing Qin
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China
| | - Shehbaz Hussain
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China
| | - Wanjun Liang
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China.
| |
Collapse
|
5
|
Liu ZH, Xia Y, Ai S, Wang HL. Health risks of Bisphenol-A exposure: From Wnt signaling perspective. ENVIRONMENTAL RESEARCH 2024; 251:118752. [PMID: 38513750 DOI: 10.1016/j.envres.2024.118752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 03/23/2024]
Abstract
Human beings are routinely exposed to chronic and low dose of Bisphenols (BPs) due to their widely pervasiveness in the environment. BPs hold similar chemical structures to 17β-estradiol (E2) and thyroid hormone, thus posing threats to human health by rendering the endocrine system dysfunctional. Among BPs, Bisphenol-A (BPA) is the best-known and extensively studied endocrine disrupting compound (EDC). BPA possesses multisystem toxicity, including reproductive toxicity, neurotoxicity, hepatoxicity and nephrotoxicity. Particularly, the central nervous system (CNS), especially the developing one, is vulnerable to BPA exposure. This review describes our current knowledge of BPA toxicity and the related molecular mechanisms, with an emphasis on the role of Wnt signaling in the related processes. We also discuss the role of oxidative stress, endocrine signaling and epigenetics in the regulation of Wnt signaling by BPA exposure. In summary, dysfunction of Wnt signaling plays a key role in BPA toxicity and thus can be a potential target to alleviate EDCs induced damage to organisms.
Collapse
Affiliation(s)
- Zhi-Hua Liu
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, Anhui 230009, China; School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, 230009, China
| | - Yanzhou Xia
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, Anhui 230009, China; School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, 230009, China
| | - Shu Ai
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, Anhui 230009, China; School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, 230009, China
| | - Hui-Li Wang
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, Anhui 230009, China; School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, 230009, China.
| |
Collapse
|
6
|
Javanmard AR, Jahanbakhshi A, Nemati H, Mowla SJ, Soltani BM. ADAMTS9-AS1 Long Non‑coding RNA Sponges miR‑128 and miR-150 to Regulate Ras/MAPK Signaling Pathway in Glioma. Cell Mol Neurobiol 2023; 43:2309-2322. [PMID: 36449154 PMCID: PMC11412184 DOI: 10.1007/s10571-022-01311-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/18/2022] [Indexed: 12/03/2022]
Abstract
Glioma is a malignancy of the central nervous system with a poor prognosis. Therefore, the elaboration of its molecular features creates therapeutic opportunities. Looking for the regulatory non-coding RNAs (lncRNAs and miRNAs) that are involved in glioma incidence/progression, RNA-seq analysis introduced upregulated ADAMTS9-AS1 as a bona fide candidate that sponges miR-128 and miR-150 and shows the negative correlation of expression with them. Then, RT-qPCR verified the upregulation of ADAMTS9-AS1 in glioma tissues and cell lines. Furthermore, dual-luciferase assay supported that cytoplasmic ADAMTS9-AS1 is capable of sponging miR-128 and miR-150, which are known as regulators of Ras/MAPK, PI3K, and Wnt pathways. Following the overexpression of ADAMTS9-AS1 in 1321N1 and U87 glioma cells, tyrosine kinase receptors (IGF1R and TrkC), as well as Wnt receptors (Lrp6 and Fzd) were upregulated, detected by RT-qPCR. Furthermore, downstream genes of both Ras/MAPK and Wnt pathways were upregulated. Finally following the ADAMTS9-AS1 overexpression, upregulation of Ras/MAPK and Wnt signaling pathways was verified through western blotting and Top/Fop flash assay, respectively. At the cellular level, ADAMTS9-AS1 overexpression brought about reduced sub-G1 cell population, increased proliferation rate, reduced apoptosis level, increased migration rate, shortened Bax/Bcl2 ratio, induced EMT, and stemness characteristics of transfected cells, detected by flow cytometry, MTT assay, scratch test, and RT-qPCR. Overall, these results introduced ADAMTS9-AS1 as an oncogene that upregulates Ras/MAPK and Wnt pathways through sponging of the miR-128 and miR-150 in glioma cells. The outcome of ADAMTS9-AS1 expression is more aggression of the glioma cells through increased EMT and stemness characteristics. These features candidate ADAMTS9-AS1 locus for glioma therapy. As a result, we discovered the oncogenic properties of ADAMTS9-AS1 in glioma cancer. It sponges miR-128 and miR-150 and subsequently overstimulates RAS/MAPK and Wnt signaling pathways, particularly at the receptors level. Thus, ADAMTS9-AS1 increases proliferation, migration, and stemness in glioma cell lines. A schematic representation showing the functional effect of ADAMTS9-AS1.
Collapse
Affiliation(s)
- Amir-Reza Javanmard
- Genetics Department, Faculty of Biological Sciences, School of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amin Jahanbakhshi
- Stem Cell and Regenerative Medicine Research Centre, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| | - Hossein Nemati
- Genetics Department, Faculty of Biological Sciences, School of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Seyed Javad Mowla
- Genetics Department, Faculty of Biological Sciences, School of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Bahram M Soltani
- Genetics Department, Faculty of Biological Sciences, School of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
7
|
Liao YF, Pan HJ, Abudurezeke N, Yuan CL, Yuan YL, Zhao SD, Zhang DD, Huang S. Functional Axis of PDE5/cGMP Mediates Timosaponin-AIII-Elicited Growth Suppression of Glioblastoma U87MG Cells. Molecules 2023; 28:molecules28093795. [PMID: 37175205 PMCID: PMC10180307 DOI: 10.3390/molecules28093795] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/28/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Glioblastoma (GBM) is the most aggressive brain tumor, with high mortality. Timosaponin AIII (TIA), a steroidal saponin isolated from the medicinal plant Anemarrhena asphodeloides Bge., has been shown to possess anticancer properties in various cancer types. However, the effect of TIA on GBM is unknown. In this study, we reveal that TIA not only inhibited U87MG in vitro cell growth but also in vivo tumor development. Moreover, we found that the cause of TIA-induced cell growth suppression was apoptosis. When seeking to uncover antitumor mechanisms of TIA, we found that TIA diminished the expression of cGMP-specific phosphodiesterase 5(PDE5) while elevating the levels of guanylate cyclases (sGCβ), cellular cGMP, and phosphorylation of VASPser239. Following the knockdown of PDE5, PDE5 inhibitor tadalafil and cGMP analog 8-Bro-cGMP both inhibited cell growth and inactivated β-catenin; we reason that TIA elicited an antitumor effect by suppressing PDE5, leading to the activation of the cGMP signaling pathway, which, in turn, impeded β-catenin expression. As β-catenin is key for cell growth and survival in GBM, this study suggests that TIA elicits its anti-tumorigenic effect by interfering with β-catenin function through the activation of a PDE5/cGMP functional axis.
Collapse
Affiliation(s)
- Ya-Fang Liao
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
| | - Hui-Jun Pan
- Shanghai Skin Disease Hospital, Shanghai 200443, China
| | - Nuerziba Abudurezeke
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
| | - Chun-Lu Yuan
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
| | - Yan-Li Yuan
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
| | - Shu-Da Zhao
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
| | - Dan-Dan Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
| | - Shuang Huang
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
8
|
Xu J, Sun S, Zhang W, Dong J, Huang C, Wang X, Jia M, Yang H, Wang Y, Jiang Y, Cao L, Huang Z. Irigenin inhibits glioblastoma progression through suppressing YAP/β-catenin signaling. Front Pharmacol 2022; 13:1027577. [PMID: 36532767 PMCID: PMC9748621 DOI: 10.3389/fphar.2022.1027577] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 11/16/2022] [Indexed: 09/14/2024] Open
Abstract
Glioblastoma (GBM) is the most malignant glioma in brain tumors with low survival and high recurrence rate. Irigenin, as an isoflavone compound extracted from Shegan, has shown many pharmacological functions such as antioxidant, anti-inflammatory and anti-tumor. However, the effects of irigenin on GBM cells and the related molecular mechanisms remain unexplored. In this study, we found that irigenin inhibited the proliferation of GBM cells in a dose-dependent manner by several assays in vitro. Subsequently, we found that irigenin arrested cell cycle at G2/M phase and induced apoptosis of GBM cells in vitro. In addition, irigenin inhibited the migration of GBM cells. Mechanically, we found that irigenin treatment decreased the expression of YAP (yes-associated protein), suppressed β-catenin signaling. Furthermore, overexpression of YAP partially restored the anti-tumor effects of irigenin on GBM cells in vitro. Finally, we found that irigenin inhibited the growth of tumor in GBM xenograft mice model through inactivation of YAP. Taken together, these results suggest that irigenin exerts its anticancer effects on GBM via inhibiting YAP/β-catenin signaling, which may provide a new strategy for the treatment of GBM.
Collapse
Affiliation(s)
- Jiayun Xu
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Shanshan Sun
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Wei Zhang
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Jianhong Dong
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Changgang Huang
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Xin Wang
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Mengxian Jia
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Hao Yang
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Yongjie Wang
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Yuanyuan Jiang
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Liying Cao
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Laboratory of Aging and Cancer Biology of Zhejiang Province, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
| | - Zhihui Huang
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| |
Collapse
|
9
|
Xiang Z, Lv Q, Zhang Y, Chen X, Guo R, Liu S, Peng X. Long non-coding RNA DDX11-AS1 promotes the proliferation and migration of glioma cells by combining with HNRNPC. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 28:601-612. [PMID: 35614994 PMCID: PMC9109126 DOI: 10.1016/j.omtn.2022.04.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 04/23/2022] [Indexed: 12/11/2022]
Abstract
Glioma is a malignant tumor of the central nervous system with complex pathogenesis, difficult operation, and a high postoperative recurrence rate. At present, there is still a lack of effective treatment. Long non-coding RNA DDX11 antisense RNA 1 (DDX11-AS1) has been shown to promote tumor development, such as hepatocellular carcinoma, esophageal cancer, etc. However, its molecular mechanism in glioma is poorly understood. In this study, we found that the expression of DDX11-AS1 was elevated in glioma tissues, and patients with high expression of DDX11-AS1 had poor prognosis. DDX11-AS1 was a potential prognostic marker. Functionally, DDX11-AS1 promoted glioma cell proliferation and migration. Mechanistically, DDX11-AS1 interacted with RNA-binding protein heterogeneous nuclear ribonucleoprotein C (HNRNPC) to promote Wnt/β-catenin and AKT pathways and the epithelial-mesenchymal transition process. In summary, our study manifests that the DDX11-AS1/HNRNPC axis may play a vital part in the occurrence and development of glioma, which provides new ideas and therapeutic targets for the diagnosis, treatment, and prognosis of glioma.
Collapse
Affiliation(s)
- Zijin Xiang
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Qiaoli Lv
- Jiangxi Key Laboratory of Translational Cancer Research, Department of Head and Neck Surgery, Jiangxi Cancer Hospital of Nanchang University, Nanchang, China
| | - Yujun Zhang
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Xueru Chen
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Ren Guo
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Shikun Liu
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Xiangdong Peng
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| |
Collapse
|
10
|
Zhou Q, Fu Q, Shaya M, Kugeluke Y, Li S, Dilimulati Y. Knockdown of circ_0055412 promotes cisplatin sensitivity of glioma cells through modulation of CAPG and Wnt/β-catenin signaling pathway. CNS Neurosci Ther 2022; 28:884-896. [PMID: 35332692 PMCID: PMC9062567 DOI: 10.1111/cns.13820] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 01/20/2022] [Accepted: 02/09/2022] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION Glioma is the most frequent primary cerebral tumor in adults. Recent evidence has suggested that circular RNAs (circRNAs) are associated with the pathological processes in glioma. In our study, we aimed to investigate the function and mechanism of circ_CAPG (circ_0055412) in glioma. METHODS Firstly, circ_0055412 expression was examined through RT-qPCR analysis. Loss-of-function assays and animal experiments were implemented to evaluate the role of circ_0055412 on cisplatin resistance of glioma cells. Moreover, mechanism assays were done to probe into the regulatory mechanism of circ_0055412 in glioma cells. RESULTS Circ_0055412 was found to be notably upregulated in glioma cells. Moreover, depletion of circ_0055412 enhanced cisplatin sensitivity of glioma cells in vitro and in vivo. Moreover, circ_0055412 recruited eukaryotic translation initiation factor 4A3 (EIF4A3) protein to stabilize capping actin protein, gelsolin like (CAPG) mRNA. Furthermore, circ_0055412 served as a sponge for microRNA-330-3p (miR-330-3p) and regulated nuclear factor of activated T cells 3 (NFATC3) expression to activate the transcription of catenin beta 1 (CTNNB1), thus participating in the activation of Wnt/β-catenin signaling pathway. CONCLUSION Circ_0055412 contributed to cisplatin resistance of glioma cells via stabilizing CAPG mRNA and modulating Wnt/β-catenin signaling pathway. This finding might provide novel information for the treatment of glioma.
Collapse
Affiliation(s)
- Qingjiu Zhou
- Department of Neurosurgery, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Qiang Fu
- Department of Neurosurgery, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Mahati Shaya
- Department of Oncology, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Yalikun Kugeluke
- Department of Neurosurgery, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Shaoshan Li
- Department of Neurosurgery, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Yisireyili Dilimulati
- Department of Neurosurgery, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| |
Collapse
|
11
|
Yoo K, Kang J, Choi M, Suh Y, Zhao Y, Kim M, Chang JH, Shim J, Yoon S, Kang S, Lee S. Soluble ICAM-1 a Pivotal Communicator between Tumors and Macrophages, Promotes Mesenchymal Shift of Glioblastoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2102768. [PMID: 34813169 PMCID: PMC8805565 DOI: 10.1002/advs.202102768] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 10/18/2021] [Indexed: 06/13/2023]
Abstract
Despite aggressive clinical treatment, recurrence of glioblastoma multiforme (GBM) is unavoidable, and the clinical outcome is still poor. A convincing explanation is the phenotypic transition of GBM cells upon aggressive treatment such as radiotherapy. However, the microenvironmental factors contributing to GBM recurrence after treatment remain unexplored. Here, it is shown that radiation-treated GBM cells produce soluble intercellular adhesion molecule-1 (sICAM-1) which stimulates the infiltration of macrophages, consequently enriching the tumor microenvironment with inflammatory macrophages. Acting as a paracrine factor, tumor-derived sICAM-1 induces macrophages to secrete wingless-type MMTV integration site family, member 3A (WNT3A), which promotes a mesenchymal shift of GBM cells. In addition, blockade of either sICAM-1 or WNT3A diminishes the harmful effect of radiation on tumor progression. Collectively, the findings indicate that cellular crosstalk between GBM and macrophage through sICAM-1-WNT3A oncogenic route is involved in the mesenchymal shift of GBM cells after radiation, and suggest that radiotherapy combined with sICAM-1 targeted inhibition would improve the clinical outcome of GBM patients.
Collapse
Affiliation(s)
- Ki‐Chun Yoo
- Department of Life ScienceResearch Institute for Natural SciencesHanyang UniversitySeoul04763Korea
- Department of Lymphoma and MyelomaDivision of Cancer MedicineCenter for Cancer Immunology ResearchThe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Jae‐Hyeok Kang
- Department of Life ScienceResearch Institute for Natural SciencesHanyang UniversitySeoul04763Korea
| | - Mi‐Young Choi
- Department of Life ScienceResearch Institute for Natural SciencesHanyang UniversitySeoul04763Korea
| | - Yongjoon Suh
- Department of Life ScienceResearch Institute for Natural SciencesHanyang UniversitySeoul04763Korea
| | - Yi Zhao
- Department of Life ScienceResearch Institute for Natural SciencesHanyang UniversitySeoul04763Korea
| | - Min‐Jung Kim
- Laboratory of Radiation Exposure & TherapeuticsNational Radiation Emergency Medical CenterKorea Institute of Radiological and Medical SciencesSeoul01812Korea
| | - Jong Hee Chang
- Department of NeurosurgeryBrain Tumor CenterSeverance HospitalYonsei University College of MedicineSeoul03722Korea
| | - Jin‐Kyoung Shim
- Department of NeurosurgeryBrain Tumor CenterSeverance HospitalYonsei University College of MedicineSeoul03722Korea
| | - Seon‐Jin Yoon
- Department of NeurosurgeryBrain Tumor CenterSeverance HospitalYonsei University College of MedicineSeoul03722Korea
| | - Seok‐Gu Kang
- Department of NeurosurgeryBrain Tumor CenterSeverance HospitalYonsei University College of MedicineSeoul03722Korea
| | - Su‐Jae Lee
- Department of Life ScienceResearch Institute for Natural SciencesHanyang UniversitySeoul04763Korea
| |
Collapse
|
12
|
Ma S, Duan L, Dong H, Ma X, Guo X, Liu J, Li G, Yu Y, Xu Y, Yuan G, Zhao X, Tian G, Zhai S, Pan Y, Zhang Y. OLFML2A Downregulation Inhibits Glioma Proliferation Through Suppression of Wnt/β-Catenin Signaling. Front Oncol 2021; 11:717917. [PMID: 34650914 PMCID: PMC8506028 DOI: 10.3389/fonc.2021.717917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/02/2021] [Indexed: 11/13/2022] Open
Abstract
Glioma is a highly heterogeneous and lethal tumor with an extremely poor prognosis. Through analysis of TCGA data, we identified that OLFML2A is a key promotor of gliomagenesis. However, the molecular function of OLFML2A and its underlying mechanism of action in glioma remain unclear. In this study, we found that OLFML2A expression was significantly upregulated in glioma specimens and positively correlated with pathological grades in glioma patients. Moreover, Kaplan–Meier survival analysis of TCGA data revealed that glioma patients with higher OLFML2A expression had shorter overall survival. Importantly, OLFML2A knockdown in glioma cells inhibited cell proliferation and promoted apoptosis. Mechanistically, OLFML2A downregulation inhibits Wnt/β-catenin signaling by upregulating amyloid precursor protein (APP) expression and reducing stabilized β-catenin levels, leading to the repression of MYC, CD44, and CSKN2A2 expression. Furthermore, OLFML2A downregulation suppressed the growth of transplanted glioma subcutaneously and intracranially by inhibiting Wnt/β-catenin pathway-dependent cell proliferation. By uncovering the oncogenic effects in human and rodent gliomas, our data support OLFML2A as a potential therapeutic target for glioma.
Collapse
Affiliation(s)
- Shize Ma
- Department of Neurosurgery and Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China.,Second Clinical School, Lanzhou University, Lanzhou, China
| | - Lei Duan
- Department of Neurosurgery and Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China.,Second Clinical School, Lanzhou University, Lanzhou, China
| | - Huateng Dong
- Department of Pediatric Neurology, Gansu Provincial Maternity and Child-Care Hospital, Lanzhou, China
| | - Xiaodong Ma
- Department of Neurosurgery and Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China.,Second Clinical School, Lanzhou University, Lanzhou, China
| | - Xinyu Guo
- Department of Neurosurgery and Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China.,Second Clinical School, Lanzhou University, Lanzhou, China
| | - Jianli Liu
- Second Clinical School, Lanzhou University, Lanzhou, China.,Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China
| | - Guoqiang Li
- Department of Neurosurgery and Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China.,Second Clinical School, Lanzhou University, Lanzhou, China
| | - Yue Yu
- Department of Neurosurgery and Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China.,Second Clinical School, Lanzhou University, Lanzhou, China
| | - Yanlong Xu
- Department of Neurosurgery and Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China.,Second Clinical School, Lanzhou University, Lanzhou, China
| | - Guoqiang Yuan
- Department of Neurosurgery and Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China.,Second Clinical School, Lanzhou University, Lanzhou, China
| | - Xingkun Zhao
- Second Clinical School, Lanzhou University, Lanzhou, China
| | - Guopeng Tian
- Department of Neurosurgery and Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China.,Second Clinical School, Lanzhou University, Lanzhou, China
| | - Shijia Zhai
- Department of Neurosurgery and Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China.,Second Clinical School, Lanzhou University, Lanzhou, China
| | - Yawen Pan
- Department of Neurosurgery and Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China.,Second Clinical School, Lanzhou University, Lanzhou, China
| | - Yinian Zhang
- Department of Neurosurgery and Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China.,Second Clinical School, Lanzhou University, Lanzhou, China
| |
Collapse
|
13
|
Pyrvinium pamoate regulates MGMT expression through suppressing the Wnt/β-catenin signaling pathway to enhance the glioblastoma sensitivity to temozolomide. Cell Death Discov 2021; 7:288. [PMID: 34642308 PMCID: PMC8511032 DOI: 10.1038/s41420-021-00654-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 08/23/2021] [Accepted: 09/13/2021] [Indexed: 12/16/2022] Open
Abstract
Temozolomide (TMZ) is the mainstream chemotherapeutic drug for treating glioblastoma multiforme (GBM), but the intrinsic or acquired chemoresistance to TMZ has become the leading clinical concern, which is related to the repair of DNA alkylation sites by O6-methylguanine-DNA methyltransferase (MGMT). Pyrvinium pamoate (PP), the FDA-approved anthelminthic drug, has been reported to inhibit the Wnt/β-catenin pathway within numerous cancer types, and Wnt/β-catenin signaling pathway can modulate the expression of MGMT gene. However, whether PP affects the expression of MGMT and enhances TMZ sensitivity in GBM cells remains unclear. In the present study, we found that PP and TMZ had synergistic effect on inhibiting the viability of GBM cells, and PP induced inhibition of MGMT and enhanced the TMZ chemosensitivity of GBM cells through down-regulating Wnt/β-catenin pathway. Moreover, the overexpression of MGMT or β-catenin weakened the synergy between PP and TMZ. The mechanism of PP in inhibiting the Wnt pathway was indicated that PP resulted in the degradation of β-catenin via the AKT/GSK3β/β-catenin signaling axis. Moreover, Ser552 phosphorylation in β-catenin, which promotes its nuclear accumulation and transcriptional activity, is blocked by PP that also inhibits the Wnt pathway to some extent. The intracranial GBM mouse model also demonstrated that the synergy between PP and TMZ could be achieved through down-regulating β-catenin and MGMT, which prolonged the survival time of tumor-bearing mice. Taken together, our data suggest that PP may serve as the prospect medicine to improve the chemotherapeutic effect on GBM, especially for chemoresistant to TMZ induced by MGMT overexpression.
Collapse
|
14
|
Hu J, Wang Z, Chen J, Yu Z, Zhang J, Li W, Lin M, Yang X, Liu H. Overexpression of ICAT Inhibits the Progression of Colorectal Cancer by Binding with β-Catenin in the Cytoplasm. Technol Cancer Res Treat 2021; 20:15330338211041253. [PMID: 34569368 PMCID: PMC8485569 DOI: 10.1177/15330338211041253] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Inhibitor of β-catenin and T-cell factor (ICAT) was first found as a polypeptide that blocks β-catenin–TCF interaction. Abundant evidence has shown that ICAT has different functions in diverse cancers’ progression. Nevertheless, the roles it plays in colorectal cancer (CRC) have not been described. Here, we documented that ICAT expression was higher in CRC tissue than in the adjacent normal tissue and that prognosis was better in high-ICAT expression patients. The overexpression of ICAT inhibited CRC cell proliferation both in vitro and in vivo. Wnt pathway transcriptional activity was suppressed in the CRC cells with ICAT overexpression, where the CCND1 and MYC expression, which occurs downstream of the Wnt signaling pathway, was inhibited. Co-immunoprecipitation experiments showed that ICAT bound with β-catenin in stable overexpression cell lines; immunofluorescence showed the co-localization of ICAT and β-catenin in the cytoplasm. Overall, our study reveals that ICAT inhibits CRC cell proliferation by binding to cytoplasm-located β-catenin, and prevents its translocation, which results in Wnt signaling pathway inactivation. It may provide a scientific foundation for focusing on ICAT in treatments for CRC.
Collapse
Affiliation(s)
- Jiancong Hu
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zihan Wang
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Junxiong Chen
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhaoliang Yu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jingdan Zhang
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Weiqian Li
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Mengmeng Lin
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiangling Yang
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Huanliang Liu
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Huanliang Liu, Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, Guangdong, China.
| |
Collapse
|
15
|
AEG-1 silencing attenuates M2-polarization of glioma-associated microglia/macrophages and sensitizes glioma cells to temozolomide. Sci Rep 2021; 11:17348. [PMID: 34462446 PMCID: PMC8405821 DOI: 10.1038/s41598-021-96647-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 08/06/2021] [Indexed: 12/19/2022] Open
Abstract
Glioma is the most frequent primary malignancy in the brain; temozolomide (TMZ) is the first-line chemotherapeutic agent used to combat this tumor. We showed here that astrocyte elevated gene-1 (AEG-1) was overexpressed in glioma tissues and associated with a worse subtype and a poor prognosis. CCK-8 proliferation assays and clone formation experiments presented that AEG-1 knockdown sensitizes glioma cells to TMZ. The γH2AX foci formation assays indicated that AEG-1 silencing promotes TMZ-induced DNA damage in glioma cells. Glioma-associated microglia/macrophages (GAMs), the largest subpopulation infiltrating glioma, play important roles in the tumor microenvironment. Bioinformatics analyses and functional studies demonstrated that AEG-1 silencing decreased M2-polarization of HMC3 microglia and the secretion of tumor supportive cytokines IL-6 and TGF-β1. The expression of AEG-1 was positively associated with M2 markers in glioma tissues varified by IHC staining. Based on the results of Affymetrix microarray and GSEA analyses, Western blot and Co-Immunoprecipitation assays were conducted to show that AEG-1 activates Wnt/β-catenin signaling by directly interacting with GSK-3β. The co-localization of AEG-1 and GSK-3β in the cytoplasm of glioma cells was detected through immunofluorescence staining. This study raises the possibility that targeting AEG-1 might improve the efficiency of chemotherapy and reduce immunosuppressive M2 GAMs in glioma.
Collapse
|
16
|
Feng F, Zhao Z, Zhou Y, Cheng Y, Wu X, Heng X. CUX1 Facilitates the Development of Oncogenic Properties Via Activating Wnt/β-Catenin Signaling Pathway in Glioma. Front Mol Biosci 2021; 8:705008. [PMID: 34422906 PMCID: PMC8377541 DOI: 10.3389/fmolb.2021.705008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 07/26/2021] [Indexed: 11/30/2022] Open
Abstract
Background: Homeobox cut like 1 (CUX1), which often presents aberrated expression in many cancer cells, exerts a crucial role in tumorigenesis. Evidence describing CUX1 in gliomagenesis is scarce, and the effects of CUX1 on the Wnt/β-catenin pathway have not been reported. Our study aimed to explore the biological functions and molecular mechanisms involved in CUX1 activity in glioma. Methods: Datasets for bioinformatics analysis were obtained from the GEO, TCGA, CGGA, GTEX and CCLE databases. qRT-PCR, western blotting (WB), and immunohistochemistry (IHC) assays were used to investigate the expression patterns of CUX1 among glioma and brain tissues. CUX1 knockdown and overexpression vectors were transfected into glioma cell lines, the CCK-8, clone formation assay, wound healing, Transwell assay, and flow cytometry were performed to detect changes in cell viability, invasiveness, and the cell cycle. WB and immunofluorescence (IF) assays were used to explore changes in cell cycle-related and Wnt/β-catenin signaling protein levels. Results: Overexpression of CUX1 was identified in glioma tissues, and especially in glioblastoma (GBM), when compared to normal controls and correlated with poor prognosis. In comparison with untreated cells, TJ905 glioma cells overexpressing CUX1 showed higher proliferation and invasion abilities and S phase cell-cycle arrest, while the knockdown of CUX1 suppressed cell invasive ability and induced G1 phase arrest. Active Wnt/β-catenin signaling was enriched and clustered in a CUX1-associated GSEA/GSVA analysis. IF and WB assays indicated that CUX1 regulated the distribution of Axin2/β-catenin in glioma cells and regulated the expression of proteins downstream of the Wnt/β-catenin signaling pathway, suggesting that CUX1 served as an upstream positive regulator of the Wnt/β-catenin pathway. Finally, the knockdown of Axin2 or β-catenin could reverse the tumor-promoting effects caused by CUX1 overexpression, suggesting that CUX1 induced gliomagenesis and malignant phenotype by activating the Wnt/β-catenin signaling pathway. Conclusion: Our data suggested that the transcription factor CUX1 could be a novel therapeutic target for glioma with gene therapy.
Collapse
Affiliation(s)
- Fan Feng
- Institute of Clinical Medicine College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Institute of Brain Science and Brain-Like Intelligence, Linyi People's Hospital, Linyi, China.,Department of Neurosurgery, Linyi People's Hospital, Linyi, China
| | - Zongqing Zhao
- Institute of Brain Science and Brain-Like Intelligence, Linyi People's Hospital, Linyi, China.,Department of Neurosurgery, Linyi People's Hospital, Linyi, China
| | - Yunfei Zhou
- Department of Neurosurgery, Linyi People's Hospital, Linyi, China
| | - Yanhao Cheng
- Institute of Brain Science and Brain-Like Intelligence, Linyi People's Hospital, Linyi, China.,Department of Neurosurgery, Linyi People's Hospital, Linyi, China
| | - Xiujie Wu
- Institute of Brain Science and Brain-Like Intelligence, Linyi People's Hospital, Linyi, China.,Department of Neurosurgery, Linyi People's Hospital, Linyi, China
| | - Xueyuan Heng
- Institute of Brain Science and Brain-Like Intelligence, Linyi People's Hospital, Linyi, China.,Department of Neurosurgery, Linyi People's Hospital, Linyi, China
| |
Collapse
|
17
|
WNT Signaling as a Therapeutic Target for Glioblastoma. Int J Mol Sci 2021; 22:ijms22168428. [PMID: 34445128 PMCID: PMC8395085 DOI: 10.3390/ijms22168428] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/21/2021] [Accepted: 07/22/2021] [Indexed: 12/23/2022] Open
Abstract
The WNT (Wingless/Integrated) signaling pathway is implicated in various stages of glioblastoma, which is an aggressive brain tumor for which therapeutic options are limited. WNT has been recognized as a hallmark of therapeutic challenge due to its context-dependent role and critical function in healthy tissue homeostasis. In this review, we deeply scrutinize the WNT signaling pathway and its involvement in the genesis of glioblastoma as well as its acquired therapy resistance. We also provide an analysis of the WNT pathway in terms of its therapeutic importance in addition to an overview of the current targeted therapies under clinical investigation.
Collapse
|
18
|
Wang Y, Liu YY, Chen MB, Cheng KW, Qi LN, Zhang ZQ, Peng Y, Li KR, Liu F, Chen G, Cao C. Neuronal-driven glioma growth requires Gαi1 and Gαi3. Theranostics 2021; 11:8535-8549. [PMID: 34373757 PMCID: PMC8343996 DOI: 10.7150/thno.61452] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 07/08/2021] [Indexed: 12/17/2022] Open
Abstract
Neuroligin-3 (NLGN3) is necessary and sufficient to promote glioma cell growth. The recruitment of Gαi1/3 to the ligand-activated receptor tyrosine kinases (RTKs) is essential for mediating oncogenic signaling. Methods: Various genetic strategies were utilized to examine the requirement of Gαi1/3 in NLGN3-driven glioma cell growth. Results: NLGN3-induced Akt-mTORC1 and Erk activation was inhibited by decreasing Gαi1/3 expression. In contrast ectopic Gαi1/3 overexpression enhanced NLGN3-induced signaling. In glioma cells, NLGN3-induced cell growth, proliferation and migration were attenuated by Gαi1/3 depletion with shRNA, but facilitated with Gαi1/3 overexpression. Significantly, Gαi1/3 silencing inhibited orthotopic growth of patient-derived glioma xenografts in mouse brain, whereas forced Gαi1/3-overexpression in primary glioma xenografts significantly enhanced growth. The growth of brain-metastatic human lung cancer cells in mouse brain was largely inhibited with Gαi1/3 silencing. It was however expedited with ectopic Gαi1/3 overexpression. In human glioma Gαi3 upregulation was detected, correlating with poor prognosis. Conclusion: Gαi1/3 mediation of NLGN3-induced signaling is essential for neuronal-driven glioma growth.
Collapse
|
19
|
Wang M, Li C, Shi W. FAM84B acts as a tumor promoter in human glioma via affecting the Akt/GSK-3β/β-catenin pathway. Biofactors 2021; 47:600-611. [PMID: 33759248 DOI: 10.1002/biof.1727] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/03/2021] [Indexed: 01/23/2023]
Abstract
Family with sequence similarity 84, member B (FAM84B) has recently emerged as an oncoprotein in multiple types of cancer. However, whether FAM84B modulates the progression of glioma has not been determined. The goals of this work were to assess the possible relationship between FAM84B and glioma. Our data revealed high FAM84B level in glioma specimens and exhibited that the overexpression of FAM84B was correlated with a low survival rate in glioma patients. Cellular functional assays showed that silencing of FAM84B prohibited the proliferation and invasion, and induced the apoptosis of glioma cells. Further results determined that the knockdown of FAM84B remarkably decreased the levels of phosphorylated Akt and glycogen synthase kinase (GSK)-3β, and active β-catenin. Inhibition of Akt abolished the FAM84B-mediated promotion effects on Wnt/β-catenin pathway. The subcutaneous xenograft assay confirmed that the silencing of FAM84B significantly prohibited the tumorigenicity of glioma cells in vivo. Collectively, the findings from this work demonstrate that the downregulation of FAM84B exhibits a cancer-suppressive role in human glioma through the regulation of Akt/GSK-3β/β-catenin pathway.
Collapse
Affiliation(s)
- Minjuan Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Neurology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Chengliang Li
- Department of General Practice, The First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Wei Shi
- Department of Neurosurgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
20
|
Xia B, Chen C, Tao W. Neuroplasticity: A Key Player in the Antidepressant Action of Chinese Herbal Medicine. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2021; 49:1115-1133. [PMID: 34049476 DOI: 10.1142/s0192415x21500531] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Traditional Chinese medicine (TCM) is a systematic medicine. It provides alternative strategies for the treatment of depression with its clinical experience, comprehensive diagnosis, and treatment theory. Chinese herbal medicine (CHM) is the major form of TCM prescription, and numerous CHMs have been demonstrated to possess remarkable antidepressant-like properties. A diversity of mechanisms have been implicated in CHM-associated antidepressant property. This paper reviewed the neuroplastic mechanisms underlying the antidepressant actions of CHM, finding that CHM repairs neuroplasticity by improving neurogenesis, neurotrophic factors, synaptic spine morphology, cell signaling, glutamatergic system, monoamine neurotransmitters, and neural apoptosis. CHM thereby exerts an antidepressant effect, attempting to offer a better understanding of the mechanisms implicated in TCM-related antidepressant-like efficacy and laying a foundation for the scientific evaluation and development of TCM in treating depression.
Collapse
Affiliation(s)
- Baomei Xia
- Faculty of Rehabilitation Science, Nanjing Normal University of Special Education, Nanjing 210023, P. R. China.,School of Chinese Medicine & School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Chang Chen
- Department of Neurology, Nanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing 210022, P. R. China
| | - Weiwei Tao
- School of Chinese Medicine & School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| |
Collapse
|
21
|
Zhu H, Chen Z, Shen L, Tang T, Yang M, Zheng X. Long Noncoding RNA LINC-PINT Suppresses Cell Proliferation, Invasion, and EMT by Blocking Wnt/β-Catenin Signaling in Glioblastoma. Front Pharmacol 2021; 11:586653. [PMID: 33505307 PMCID: PMC7832092 DOI: 10.3389/fphar.2020.586653] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 11/16/2020] [Indexed: 12/12/2022] Open
Abstract
Background: Glioblastoma (GBM) represents the most aggressive glioma with high invasive potential. Recent studies proved the involvement of epithelial-mesenchymal transition (EMT) process in increasing the malignancy and invasiveness of GBM. LncRNAs have been verified to play pivotal roles in human disease including GBM. However, the molecular mechanisms of lncRNA-mediated EMT in GBM remain largely unknown. LINC-PINT, a LncRNA which has never been studied in GBM before, was predicted to be negatively associated with EMT in GBM. This study aimed to explore the biological function and the EMT relevance of LINC-PINT in GBM and further explore the molecular mechanism. Methods: The bioinformatic prediction data of LINC-PINT in GBM was derived from The Cancer Genome Atlas (TCGA) database by R software and GEPIA website. qRT-PCR assay was performed to detect the expression level of LINC-PINT in GBM cell lines. Cell counting kit-8 (CCK8), clone formation, transwell, and wound healing assays were performed to determine the biological function of LINC-PINT in vivo. Tumor xenograft experiment and tumor peritoneal metastasis experiments were performed to verify the in vivo function. Western blot and immunofluorescence staining assays were carried out to detect the relevance of LINC-PINT with EMT and Wnt/β-catenin signaling. Rescue assays were performed to check the regulation mechanism of LINC-PINT/Wnt signaling/EMT axis in GBM. Results: LINC-PINT was downregulated in GBM cell lines. LINC-PINT suppressed cell progression, invasion, and EMT in GBM. LINC-PINT blocked Wnt/β-catenin signaling in GBM. Conclusion: LINC-PINT suppressed cell proliferation, invasion, and EMT by blocking Wnt/β-catenin signaling in GBM.
Collapse
Affiliation(s)
- Hanshuo Zhu
- Department of Neurosurgery, XinHua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Zheng Chen
- Department of Neurosurgery, XinHua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Lin Shen
- Department of Neurosurgery, XinHua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Tianchi Tang
- Department of Neurosurgery, XinHua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Min Yang
- Department of Neurosurgery, XinHua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Xuesheng Zheng
- Department of Neurosurgery, XinHua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| |
Collapse
|
22
|
Zhang S, Zhang S, Wang H, Huang X, Wang J, Li J, Cheng D, Wang H, Lu D, Wang Y. Silencing myelin protein zero-like 1 expression suppresses cell proliferation and invasiveness of human glioma cells by inhibiting multiple cancer-associated signal pathways. JOURNAL OF NEURORESTORATOLOGY 2021. [DOI: 10.26599/jnr.2021.9040017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Glioma is the most common primary malignant tumor of the adult central nervous system. It has high morbidity and poor survival. Myelin protein zero-like protein 1 (MPZL1) is a cell surface glycoprotein that activates numerous adhesion-dependent signaling pathways. MPZL1 plays important roles in human cancers that include metastatic process; however, it is not clear if MPZL1 plays a role in human glioma. Therefore, this study aimed to determine if silencing MPZL1 impacted the cell proliferative features of human glioma cells. First, MPZL1 expression was investigated in human glioma samples and tumor cell lines. Then the effects of small interfering RNA (siRNA)-targeting MPZL1 were analyzed on proliferation, colony formation, cell cycle progression, and invasion of human glioma cells. The results from this study demonstrated that MPZL1 was highly expressed in human glioma tissues and glioma cell lines. In addition, knockdown of MPZL1 significantly inhibited cell proliferation, colony formation, and invasiveness of glioma cells, and effectively induced cell cycle arrest at the G1 phase. Western blotting analysis indicated that silencing MPZL1 expression downregulated the expression of matrix metalloproteinase-2 (MMP-2), WNT1, caspase-3, cyclin A1, epidermal growth factor receptor (EGFR), and signal transducer and activator of transcription 3 (STAT3), and upregulated p53. The results from this study suggest that MPZL1 might be a marker for tumors and could be a potential therapeutic target for human glioma.
Collapse
|
23
|
Sun J, Ma Q, Shu C, Xiong J, Li B, Wu J, Zhang S, Li J, Liu J, Wang J. MicroRNA‑301a/ZNRF3/wnt/β‑catenin signal regulatory crosstalk mediates glioma progression. Int J Oncol 2021; 58:45-56. [PMID: 33367931 PMCID: PMC7721082 DOI: 10.3892/ijo.2020.5145] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 10/23/2020] [Indexed: 12/03/2022] Open
Abstract
MicroRNA (miR)‑mediated mRNA and multiple signaling pathway dysregulations have been extensively implicated in several cancer types, including gliomas. Although previous studies have reported that miR‑301a acts as an oncogene, the underlying mechanisms of miR‑301a in the initiation and progression of glioma remain unknown. The present study aimed to investigate the involvement of miR‑301a‑mediated signaling pathway dysregulation in glioma. The results identified that miR‑301a was significantly upregulated in gliomas and was associated with a poor prognosis based on The Cancer Genome Atlas and Chinese Glioma Genome Atlas databases. Moreover, zinc and ring finger 3 (ZNRF3) exerted a critical role in the miR‑301a‑mediated effects on the malignant phenotype, such as by affecting proliferation and apoptosis. Mechanistically, the TOP/FOP luciferase assay, western blotting and immunofluorescence results demonstrated that miR‑301a knockdown inhibited the wnt/β‑catenin signaling pathway, at least partially via ZNRF3, while ZNRF3 was a direct functional target of miR‑301a, as indicated by luciferase reporter assay and western blot analysis. Furthermore, ZNRF3 could in turn repress miR‑301a expression, which was dependent on the wnt pathway. Collectively, the present study identified a novel miR‑301a/ZNRF3/wnt/β‑catenin signaling feedback loop that serves critical roles in glioma tumorigenesis, and that may represent a potential therapeutic target.
Collapse
Affiliation(s)
- Jikui Sun
- School of Medicine, Nankai University, Tianjin 300071
- Tianjin Cerebral Vascular and Neural Degenerative Disease Key Laboratory, Tianjin Neurosurgical Institute, Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin 300350
| | - Quanfeng Ma
- Tianjin Cerebral Vascular and Neural Degenerative Disease Key Laboratory, Tianjin Neurosurgical Institute, Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin 300350
| | - Chang Shu
- Tianjin Cerebral Vascular and Neural Degenerative Disease Key Laboratory, Tianjin Neurosurgical Institute, Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin 300350
| | - Jinbiao Xiong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Tianjin, 300052
| | - Banban Li
- Department of Hematology, Taian Central Hospital, Taian, Shandong 271000, P.R. China
| | - Jingchao Wu
- Tianjin Cerebral Vascular and Neural Degenerative Disease Key Laboratory, Tianjin Neurosurgical Institute, Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin 300350
| | - Shusheng Zhang
- Tianjin Cerebral Vascular and Neural Degenerative Disease Key Laboratory, Tianjin Neurosurgical Institute, Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin 300350
| | - Jialin Li
- Tianjin Cerebral Vascular and Neural Degenerative Disease Key Laboratory, Tianjin Neurosurgical Institute, Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin 300350
| | - Jun Liu
- Tianjin Cerebral Vascular and Neural Degenerative Disease Key Laboratory, Tianjin Neurosurgical Institute, Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin 300350
| | - Jinhuan Wang
- School of Medicine, Nankai University, Tianjin 300071
- Tianjin Cerebral Vascular and Neural Degenerative Disease Key Laboratory, Tianjin Neurosurgical Institute, Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin 300350
| |
Collapse
|
24
|
Wu B, Zhu J, Dai X, Ye L, Wang B, Cheng H, Wang W. Raddeanin A inhibited epithelial-mesenchymal transition (EMT) and angiogenesis in glioblastoma by downregulating β-catenin expression. Int J Med Sci 2021; 18:1609-1617. [PMID: 33746577 PMCID: PMC7976575 DOI: 10.7150/ijms.52206] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 01/14/2021] [Indexed: 02/06/2023] Open
Abstract
Raddeanin A (RA), an oleanane-type triterpenoid saponin derived from Anemone raddeana Regel, has been found to suppress the viability and metastasis of several cancers, including GBM, through various signaling pathways. However, the mechanisms underlying the anti-GBM properties of RA have not been fully elucidated. Epithelial to mesenchymal transition (EMT) and angiogenesis are important for the genesis and progression of GBM. These two crucial processes can be regulated by multiple molecular, including β-catenin, which has been demonstrated to act as a pro-tumorigenic molecular. In this study, we aimed to determine whether RA could suppress EMT and angiogenesis by inhibiting the action of β-catenin in GBM. We found that RA inhibited the proliferation, invasion and migratory properties of GBM cells. RA was also found to have downregulated the expressions of β-catenin and EMT-related biomarkers (N-cadherin, vimentin, and snail). In addition, the overexpression of β-catenin reversed the therapeutic effects of RA exerted on the EMT of GBM cells. RA restricted angiogenesis, as shown by the tube formation assay and CAM assay, while it downregulated VEGF levels in HUVECs. Moreover, massive β-catenin could reverse the suppression of angiogenesis induced by RA. Finally, we demonstrated that RA inhibited tumor growth and prolonged survival time in an intracranial U87 xenograft mouse model. Similar to the results in vitro, RA downregulated the expression of β-catenin, EMT makers and VEGF, and decreased vessel density in vivo. In summary, our results demonstrated that RA repressed GBM via downregulating β-catenin-mediated EMT and angiogenesis both in vitro and in vivo.
Collapse
Affiliation(s)
- Bingshan Wu
- Department of Neurosurgery, First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China 230032
| | - Jianwei Zhu
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu City, Sichuan Province, 611731, China
| | - Xingliang Dai
- Department of Neurosurgery, First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China 230032
| | - Lei Ye
- Department of Neurosurgery, First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China 230032
| | - Bin Wang
- Department of Neurosurgery, First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China 230032
| | - Hongwei Cheng
- Department of Neurosurgery, First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China 230032
| | - Weihong Wang
- Department of Neurosurgery, First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China 230032
| |
Collapse
|
25
|
Uddin MS, Mamun AA, Alghamdi BS, Tewari D, Jeandet P, Sarwar MS, Ashraf GM. Epigenetics of glioblastoma multiforme: From molecular mechanisms to therapeutic approaches. Semin Cancer Biol 2020; 83:100-120. [PMID: 33370605 DOI: 10.1016/j.semcancer.2020.12.015] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 02/07/2023]
Abstract
Glioblastoma multiforme (GBM) is the most common form of brain cancer and one of the most aggressive cancers found in humans. Most of the signs and symptoms of GBM can be mild and slowly aggravated, although other symptoms might demonstrate it as an acute ailment. However, the precise mechanisms of the development of GBM remain unknown. Due to the improvement of molecular pathology, current researches have reported that glioma progression is strongly connected with different types of epigenetic phenomena, such as histone modifications, DNA methylation, chromatin remodeling, and aberrant microRNA. Furthermore, the genes and the proteins that control these alterations have become novel targets for treating glioma because of the reversibility of epigenetic modifications. In some cases, gene mutations including P16, TP53, and EGFR, have been observed in GBM. In contrast, monosomies, including removals of chromosome 10, particularly q23 and q25-26, are considered the standard markers for determining the development and aggressiveness of GBM. Recently, amid the epigenetic therapies, histone deacetylase inhibitors (HDACIs) and DNA methyltransferase inhibitors have been used for treating tumors, either single or combined. Specifically, HDACIs are served as a good choice and deliver a novel pathway to treat GBM. In this review, we focus on the epigenetics of GBM and the consequence of its mutations. We also highlight various treatment approaches, namely gene editing, epigenetic drugs, and microRNAs to combat GBM.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh; Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Abdullah Al Mamun
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong Special Administrative Region
| | - Badrah S Alghamdi
- Department of Physiology, Neuroscience Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia; Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Devesh Tewari
- Department of Pharmacognosy, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Philippe Jeandet
- Research Unit, Induced Resistance and Plant Bioprotection, EA 4707, SFR Condorcet FR CNRS 3417, Faculty of Sciences, University of Reims Champagne-Ardenne, PO Box 1039, 51687, Reims Cedex 2, France
| | - Md Shahid Sarwar
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali-3814, Bangladesh
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.
| |
Collapse
|
26
|
Aslam N, Abusharieh E, Abuarqoub D, Ali D, Al-Hattab D, Wehaibi S, Al-Kurdi B, Jamali F, Alshaer W, Jafar H, Awidi AS. Anti-oncogenic activities exhibited by paracrine factors of MSCs can be mediated by modulation of KITLG and DKK1 genes in glioma SCs in vitro. MOLECULAR THERAPY-ONCOLYTICS 2020; 20:147-165. [PMID: 33575478 PMCID: PMC7851499 DOI: 10.1016/j.omto.2020.11.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 11/19/2020] [Indexed: 12/12/2022]
Abstract
Cancer stem cells (CSCs) use their stemness properties to perpetuate their lineage and survive chemotherapy. Currently cell-based and cell-free therapies are under investigation to develop novel anti-cancer treatment modalities. We designed this study to investigate how cell extracts of mesenchymal stem cells affect the growth of glioma stem cells in vitro. Gliospheres were generated from the U87MG cell line and treated with conditioned media of Wharton’s jelly and bone marrow mesenchymal stem cells. The effects were investigated at the functional and molecular levels. Our results showed that conditioned media from both types of mesenchymal stem cells changed the morphology of spheres and inhibited the proliferation, invasion, and self-renewal ability of glioma stem cells. At the molecular level, metabolism interruption at oxidative phosphorylation, cell cycle arrest, cell differentiation, and upregulation of the immune response were observed. Furthermore, this effect was mediated by the upregulation of the DKK1 gene inhibiting the Wnt pathway mediated by growth factor activity and downregulation of the KITLG gene activated by growth factor and cytokine activity, inhibiting multiple pathways. We conclude that different types of mesenchymal stem cells possess antitumor properties and their paracrine factors, in combination with anti-immune modalities, can provide practical therapeutic targets for glioblastoma treatment.
Collapse
Affiliation(s)
- Nazneen Aslam
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan
| | - Elham Abusharieh
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan.,Faculty of Pharmacy, The University of Jordan, Amman 11942, Jordan
| | - Duaa Abuarqoub
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan.,Department of Pharmacology and Biomedical Sciences, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman 11196, Jordan
| | - Dema Ali
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan
| | - Dana Al-Hattab
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan.,Laboratory for Nanomedicine, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - Suha Wehaibi
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan
| | - Ban Al-Kurdi
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan
| | - Fatima Jamali
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan
| | - Walhan Alshaer
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan
| | - Hanan Jafar
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan.,Department of Anatomy and Histology, School of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Abdalla S Awidi
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan.,Department of Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan.,Department of Hematology and Oncology, Jordan University Hospital, The University of Jordan, Amman 11942, Jordan
| |
Collapse
|
27
|
RPN2 is targeted by miR-181c and mediates glioma progression and temozolomide sensitivity via the wnt/β-catenin signaling pathway. Cell Death Dis 2020; 11:890. [PMID: 33087705 PMCID: PMC7578010 DOI: 10.1038/s41419-020-03113-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 10/08/2020] [Accepted: 10/09/2020] [Indexed: 12/12/2022]
Abstract
Accumulating evidence indicates that the dysregulation of the miRNAs/mRNA-mediated carcinogenic signaling pathway network is intimately involved in glioma initiation and progression. In the present study, by performing experiments and bioinformatics analysis, we found that RPN2 was markedly elevated in glioma specimens compared with normal controls, and its upregulation was significantly linked to WHO grade and poor prognosis. Knockdown of RPN2 inhibited tumor proliferation and invasion, promoted apoptosis, and enhanced temozolomide (TMZ) sensitivity in vitro and in vivo. Mechanistic investigation revealed that RPN2 deletion repressed β-catenin/Tcf-4 transcription activity partly through functional activation of glycogen synthase kinase-3β (GSK-3β). Furthermore, we showed that RPN2 is a direct functional target of miR-181c. Ectopic miR-181c expression suppressed β-catenin/Tcf-4 activity, while restoration of RPN2 partly reversed this inhibitory effect mediated by miR-181c, implying a molecular mechanism in which TMZ sensitivity is mediated by miR-181c. Taken together, our data revealed a new miR-181c/RPN2/wnt/β-catenin signaling axis that plays significant roles in glioma tumorigenesis and TMZ resistance, and it represents a potential therapeutic target, especially in GBM.
Collapse
|
28
|
Critical role of HOX transcript antisense intergenic RNA (HOTAIR) in gliomas. J Mol Med (Berl) 2020; 98:1525-1546. [PMID: 32978667 DOI: 10.1007/s00109-020-01984-x] [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: 06/05/2020] [Revised: 09/17/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023]
Abstract
Despite extensive research, gliomas are associated with high morbidity and mortality, mainly attributed to the rapid growth rate, excessive invasiveness, and molecular heterogeneity, as well as regenerative potential of cancer stem cells. Therefore, elucidation of the underlying molecular mechanisms and the identification of potential molecular diagnostic and prognostic biomarkers are of paramount importance. HOX transcript antisense intergenic RNA (HOTAIR) is a well-studied long noncoding RNA, playing an emerging role in tumorigenesis of several human cancers. A growing amount of preclinical and clinical evidence highlights the pro-oncogenic role of HOTAIR in gliomas, mainly attributed to the enhancement of proliferation and migration, as well as inhibition of apoptosis. In vitro and in vivo studies demonstrate that HOTAIR modulates the activity of specific transcription factors, such as MXI1, E2F1, ATF5, and ASCL1, and regulates the expression of cell cycle-associated genes along with related signaling pathways, like the Wnt/β-catenin axis. Moreover, it can interact with specific miRNAs, including miR-326, miR-141, miR-148b-3p, miR-15b, and miR-126-5p. Of importance, HOTAIR has been demonstrated to enhance angiogenesis and affect the permeability of the blood-tumor barrier, thus modulating the efficacy of chemotherapeutic agents. Herein, we provide evidence on the functional role of HOTAIR in gliomas and discuss the benefits of its targeting as a novel approach toward glioma treatment.
Collapse
|
29
|
Sun J, Chen Z, Xiong J, Wang Q, Tang F, Zhang X, Mo L, Wang C, Fan W, Wang J. MicroRNA‑422a functions as a tumor suppressor in glioma by regulating the Wnt/β‑catenin signaling pathway via RPN2. Oncol Rep 2020; 44:2108-2120. [PMID: 33000268 PMCID: PMC7550978 DOI: 10.3892/or.2020.7741] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 07/16/2020] [Indexed: 12/17/2022] Open
Abstract
MicroRNAs (miRs), which act as crucial regulators of oncogenes and tumor suppressors, have been confirmed to play a significant role in the initiation and progression of various malignancies, including glioma. The present study analyzed the expression and roles of miR‑422a in glioma, and reverse transcription‑quantitative PCR confirmed that miR‑422a expression was significantly lower in glioblastoma multiforme (GBM) samples and cell lines compared with the low‑grade glioma samples and the H4 cell line, respectively. miR‑422a overexpression suppressed proliferation and invasion, and induced apoptosis in LN229 and U87 cell lines. Luciferase reporter assay, western blotting and RNA immunoprecipitation analysis revealed that ribophorin II (RPN2) is a direct functional target of miR‑422a. Additionally, the overexpression of RPN2 partially reversed the miR‑422a‑mediated inhibitory effect on the malignant phenotype. Mechanistic investigation demonstrated that the upregulation of miR‑422a inhibited β‑catenin/transcription factor 4 transcriptional activity, at least partially through RPN2, as indicated by in vitro and in vivo experiments. Furthermore, RPN2 expression was inversely correlated with miR‑422a expression in GBM specimens and predicted patient survival in the Chinese Glioma Genome Atlas, UALCAN, Gene Expression Profiling Interactive Analysis databases. In conclusion, the present data reveal a new miR‑422a/RPN2/Wnt/β‑catenin signaling axis that plays critical roles in glioma tumorigenesis, and it represents a potential therapeutic target for GBM.
Collapse
Affiliation(s)
- Jikui Sun
- School of Medicine, Nankai University, Tianjin 300071, P.R. China
| | - Zhijuan Chen
- Clinical Medicine School, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Jinbiao Xiong
- Clinical Medicine School, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Qiong Wang
- Tianjin Cerebral Vascular and Neural Degenerative Disease Key Laboratory, Tianjin Neurosurgery Institute, Department of Neurosurgery, Tianjin Huan Hu Hospital, Tianjin 300350, P.R. China
| | - Fan Tang
- Pathology Department, Tianjin Huan Hu Hospital, Tianjin 300350, P.R. China
| | - Xuebin Zhang
- Pathology Department, Tianjin Huan Hu Hospital, Tianjin 300350, P.R. China
| | - Lidong Mo
- Tianjin Cerebral Vascular and Neural Degenerative Disease Key Laboratory, Tianjin Neurosurgery Institute, Department of Neurosurgery, Tianjin Huan Hu Hospital, Tianjin 300350, P.R. China
| | - Chen Wang
- Tianjin Cerebral Vascular and Neural Degenerative Disease Key Laboratory, Tianjin Neurosurgery Institute, Department of Neurosurgery, Tianjin Huan Hu Hospital, Tianjin 300350, P.R. China
| | - Weijia Fan
- Tianjin Cerebral Vascular and Neural Degenerative Disease Key Laboratory, Tianjin Neurosurgery Institute, Department of Neurosurgery, Tianjin Huan Hu Hospital, Tianjin 300350, P.R. China
| | - Jinhuan Wang
- School of Medicine, Nankai University, Tianjin 300071, P.R. China
| |
Collapse
|
30
|
Chen L, Li M, Li Q, Xu M, Zhong W. Knockdown of TRIM47 inhibits glioma cell proliferation, migration and invasion through the inactivation of Wnt/β-catenin pathway. Mol Cell Probes 2020; 53:101623. [PMID: 32603762 DOI: 10.1016/j.mcp.2020.101623] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 06/18/2020] [Indexed: 01/26/2023]
Abstract
Tripartite motif 47 (TRIM47), a member of the TRIM protein family, plays a crucial role in tumor development and progression. However, the role of TRIM47 in glioma has not been investigated. In the present study, we investigated the expression of TRIM47 in glioma and explored the role of TRIM47 in glioma proliferation and migration both in vitro and in vivo. Our results showed that TRIM47 expression was significantly increased in glioma tissues compared to the normal brain tissues. Knockdown of TRIM47 in U87 and U251 cells inhibited cell proliferation, as well as cell migration and invasion. TRIM47 knockdown caused significant increase in E-cadherin expression and remarkable decrease in N-cadherin and vimentin expressions in both U87 and U251 cells. In vivo assay proved that knockdown of TRIM47 prevented tumor growth of glioma. Furthermore, TRIM47 silencing significantly inhibited the activation of Wnt/β-catenin pathway. Additionally, treatment with LiCl reversed the inhibitory effects of TRIM47 knockdown on cell proliferation and migration in U87 cells. In conclusion, these findings indicated that knockdown of TRIM47 suppressed cell proliferation and metastasis of glioma both in vitro and in vivo. TRIM47 exerted an oncogenic role in glioma and might be a therapeutic target for the treatment of glioma.
Collapse
Affiliation(s)
- Lihong Chen
- International Medical Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| | - Mengdan Li
- Cardiovascular Department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Qi Li
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Min Xu
- International Medical Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Wenting Zhong
- International Medical Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| |
Collapse
|
31
|
Wu L, Zhu X, Song Z, Guo M, Liang J, Yan D. FGD5-AS1 facilitates glioblastoma progression by activation of Wnt/β-catenin signaling via regulating miR-129-5p/HNRNPK axis. Life Sci 2020; 256:117998. [PMID: 32585241 DOI: 10.1016/j.lfs.2020.117998] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 06/03/2020] [Accepted: 06/19/2020] [Indexed: 12/17/2022]
Abstract
AIMS Accumulating evidence elucidates the biological significance of long non-coding RNA (lncRNAs) in tumorigenesis and development. FGD5 antisense RNA 1 (FGD5-AS1) was previously revealed as an oncogene in several types of malignancies. However, the roles of FGD5-AS1 in glioblastoma (GBM) and its potential molecular mechanisms remain unclear. MATERIALS AND METHODS The expression of FGD5-AS1, miR-129-5p, and heterogeneous nuclear ribonucleoprotein K (HNRNPK) mRNA were measured by qRT-PCR. Cell proliferation, invasion and apoptosis were determined by MTT, colony formation, transwell and flow cytometry assays. The protein levels of Ki-67, HNRNPK and Wnt signaling-associated genes were examined by western blot assay. The possible action mechanism of FGD5-AS1 was detected by bioinformatic tools, luciferase reporter, RIP and TOP/FOP Flash reporter assays. A nude mouse xenograft model was built to analyze the function of FGD5-AS1 in vivo. KEY FINDINGS FGD5-AS1 expression was increased in GBM tumor tissues and cells. Knockdown of FGD5-AS1 inhibited cell proliferation and invasion in vitro, and slowed tumor growth in vivo. Mechanistically, FGD5-AS1 served as a sponge of miR-129-5p to relieve its suppression on HNRNPK. Moreover, down-regulation of HNRNPK repressed cell proliferation and invasion, while enhanced apoptosis. Additionally, si-FGD5-AS1-mediated suppression of cell proliferation and invasion was obviously reversed by the decrease of miR-129-5p or restoration of HNRNPK. Furthermore, FGD5-AS1 promoted cell growth and invasion by stimulating Wnt/β-catenin signaling via regulation of miR-129-5p/HNRNPK. SIGNIFICANCE FGD5-AS1 promoted GBM progression at least partly by regulating miR-129-5p/HNRNPK to activate Wnt/β-catenin signaling, suggesting the potential of FGD5-AS1 as a candidate target to improve GBM therapy.
Collapse
Affiliation(s)
- Lixin Wu
- Department of Neurosurgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xuqiang Zhu
- Department of Neurosurgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zhenyu Song
- Department of Neurosurgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Mengguo Guo
- Department of Neurosurgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Junxin Liang
- Department of Neurosurgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Dongming Yan
- Department of Neurosurgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
32
|
SOX15 exerts antitumor function in glioma by inhibiting cell proliferation and invasion via downregulation of Wnt/β-catenin signaling. Life Sci 2020; 255:117792. [PMID: 32416168 DOI: 10.1016/j.lfs.2020.117792] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 02/08/2023]
Abstract
AIMS Sex-determining region of Y chromosome-related high-mobility-group box 15 (SOX15) has recently emerged as a candidate tumor-inhibitor in multiple types of human tumors. To date, the involvement of SOX15 in glioma is undetermined. The purpose of this study was to investigate the expression, function and potential molecular mechanism of SOX15 in glioma. MAIN METHODS Relative mRNA expression was analyzed by real-time quantitative PCR. Protein expression was determined by Western blot. Cell proliferation was assessed by cell counting kit-8 and colony formation assay. Cell invasion was evaluated by Matrigel invasion assay. Wnt/β-catenin activation was monitored by luciferase reporter assay. KEY FINDINGS SOX15 expression was decreased in glioma tissues and cell lines compared with normal controls. Kaplan-Meier analysis revealed that patients with low expression of SOX15 had shorter survival than those who had high expression of SOX15. The upregulation of SOX15 markedly repressed the proliferation and invasion of glioma cells, whereas its depletion enhanced glioma cell proliferation and invasion. Research into the mechanism revealed that SOX15 exerted an inhibitory effect on Wnt/β-catenin signaling in glioma cells. Notably, overexpression of β-catenin partially reversed the SOX15 overexpression-mediated tumor-suppressive effect. In addition, SOX15 overexpression significantly impeded tumor formation by glioma cells in vivo in a mouse xenograft model associated with downregulation of active β-catenin expression. SIGNIFICANCE These data demonstrate that SOX15 functions as a potential tumor-suppressor in glioma by inhibiting cell proliferation and invasion via the downregulation of Wnt/β-catenin signaling.
Collapse
|
33
|
Han J, Shen X, Zhang Y, Wang S, Zhou L. Astragaloside IV suppresses transforming growth factor-β1-induced epithelial-mesenchymal transition through inhibition of Wnt/β-catenin pathway in glioma U251 cells. Biosci Biotechnol Biochem 2020; 84:1345-1352. [PMID: 32154763 DOI: 10.1080/09168451.2020.1737502] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Astragaloside IV (AS#IV) has previously demonstrated antitumoractivity. We investigated the effect and mechanisms of AS#IV in relation to epithelial-mesenchymal transition (EMT), viainterference with the Wnt/β-catenin signaling pathway in gliomaU251 cells. Induction of glioma U251 cells by transforming growthfactor (TGF)#β1 activated EMT, including switching E#cadherin toN-cadherin and altering the expression of Wnt/β-catenin signalingpathway components such as vimentin, β-catenin, and cyclin-D1.AS-IV inhibited the viability, invasion, and migration of TGF-β1-induced glioma U251 cells. AS-IV also interfered with the TGF#β1-induced Wnt/β-catenin signaling pathway in glioma U251 cells.These findings indicate that AS#IV prohibits TGF#β1-induced EMTby disrupting the Wnt/β-catenin pathway in glioma U251 cells. AS#IV may thus be a potential candidate agent for treating glioma andother central nervous system tumors.
Collapse
Affiliation(s)
- Jinming Han
- Department of Spine Surgery, Ningbo No. 6 Hospital , Ningbo, Zhejiang, China
| | - Xiaohan Shen
- Ningbo Diagnostic Pathology Center, Shanghai Cancer Center Ningbo Pathology Center, Ningbo Medical Center Lihuili Hospital , Ningbo, Zhejiang, China
| | - Yong Zhang
- Department of Orthopedics, Ningbo No. 6 Hospital , Ningbo, Zhejiang, China
| | - Suying Wang
- Ningbo Diagnostic Pathology Center, Shanghai Cancer Center Ningbo Pathology Center, Ningbo, Zhejiang, China
| | - Leijie Zhou
- Department of Spine Surgery, Ningbo No. 6 Hospital , Ningbo, Zhejiang, China
| |
Collapse
|
34
|
Vengoji R, Ponnusamy MP, Rachagani S, Mahapatra S, Batra SK, Shonka N, Macha MA. Novel therapies hijack the blood-brain barrier to eradicate glioblastoma cancer stem cells. Carcinogenesis 2019; 40:2-14. [PMID: 30475990 DOI: 10.1093/carcin/bgy171] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 10/12/2018] [Accepted: 11/21/2018] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma (GBM) is amongst the most aggressive brain tumors with a dismal prognosis. Despite significant advances in the current multimodality therapy including surgery, postoperative radiotherapy (RT) and temozolomide (TMZ)-based concomitant and adjuvant chemotherapy (CT), tumor recurrence is nearly universal with poor patient outcomes. These limitations are in part due to poor drug penetration through the blood-brain barrier (BBB) and resistance to CT and RT by a small population of cancer cells recognized as tumor-initiating cells or cancer stem cells (CSCs). Though CT and RT kill the bulk of the tumor cells, they fail to affect CSCs, resulting in their enrichment and their development into more refractory tumors. Therefore, identifying the mechanisms of resistance and developing therapies that specifically target CSCs can improve response, prevent the development of refractory tumors and increase overall survival of GBM patients. Small molecule inhibitors that can breach the BBB and selectively target CSCs are emerging. In this review, we have summarized the recent advancements in understanding the GBM CSC-specific signaling pathways, the CSC-tumor microenvironment niche that contributes to CT and RT resistance and the use of novel combination therapies of small molecule inhibitors that may be used in conjunction with TMZ-based chemoradiation for effective management of GBM.
Collapse
Affiliation(s)
- Raghupathy Vengoji
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Satyanarayana Rachagani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sidharth Mahapatra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.,Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.,Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | - Nicole Shonka
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.,Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Muzafar A Macha
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,Department of Otolaryngology/Head and Neck Surgery, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
35
|
Zhuo C, Xun Z, Hou W, Ji F, Lin X, Tian H, Zheng W, Chen M, Liu C, Wang W, Chen C. Surprising Anticancer Activities of Psychiatric Medications: Old Drugs Offer New Hope for Patients With Brain Cancer. Front Pharmacol 2019; 10:1262. [PMID: 31695618 PMCID: PMC6817617 DOI: 10.3389/fphar.2019.01262] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 09/30/2019] [Indexed: 12/17/2022] Open
Abstract
Despite decades of research and major efforts, malignant brain tumors remain among the deadliest of all cancers. Recently, an increasing number of psychiatric drugs has been proven to possess suppressing activities against brain tumors, and rapid progress has been made in understanding the potential mechanisms of action of these drugs. In particular, the traditional mood stabilizer valproic acid, the widely used antidepressants fluoxetine and escitalopram oxalate, and the atypical psychiatric drug aripiprazole have demonstrated promise for application in brain tumor treatment strategies through multiple lines of laboratory, preclinical, and clinical evidence. The unexpected discovery of the anticancer properties of these drugs has ignited interest in the repurposing of other psychiatric drugs to combat brain cancer. In this review, we synthesize recent progress in understanding the potential molecular mechanisms underlying the brain cancer-killing activities of representative psychiatric drugs. We also identify key limitations in the repurposing of these medications that must be overcome to enhance our ability to successfully prevent and treat brain cancer, especially in the most vulnerable groups of patients, such as children and adolescents, pregnant women, and those with unfavorable genetic variants. Moreover, we propose perspectives that may guide future research and provide long-awaited new hope to patients with brain cancer and their families.
Collapse
Affiliation(s)
- Chuanjun Zhuo
- Department of Psychiatry, School of Mental Health, Psychiatric Genetics Laboratory (PSYG-Lab), Jining Medical University, Jining, China.,Department of Psychiatry, Wenzhou Seventh People's Hospital, Wenzhou, China.,Department of China-Canada Biological Psychiatry Lab, Xiamen Xianyue Hospital, Xiamen, China.,Department of Psychiatric-Neuroimaging-Genetics and Morbidity Laboratory (PNGC-Lab), Nankai University Affiliated Anding Hospital, Tianjin Mental Health Center, Mental Health Teaching Hospital, Tianjin Medical University, Tianjin, China
| | - Zhiyuan Xun
- Department of Psychiatric-Neuroimaging-Genetics and Morbidity Laboratory (PNGC-Lab), Nankai University Affiliated Anding Hospital, Tianjin Mental Health Center, Mental Health Teaching Hospital, Tianjin Medical University, Tianjin, China
| | - Weihong Hou
- Department of Biochemistry and Molecular Biology, Zhengzhou University, Zhengzhou, China.,Department of Biology, University of North Carolina at Charlotte, Charlotte, NC, United States
| | - Feng Ji
- Department of Psychiatry, School of Mental Health, Psychiatric Genetics Laboratory (PSYG-Lab), Jining Medical University, Jining, China
| | - Xiaodong Lin
- Department of Psychiatry, Wenzhou Seventh People's Hospital, Wenzhou, China
| | - Hongjun Tian
- Department of Psychiatric-Neuroimaging-Genetics and Morbidity Laboratory (PNGC-Lab), Nankai University Affiliated Anding Hospital, Tianjin Mental Health Center, Mental Health Teaching Hospital, Tianjin Medical University, Tianjin, China
| | - Weifang Zheng
- Department of Psychiatry, Wenzhou Seventh People's Hospital, Wenzhou, China
| | - Min Chen
- Department of Psychiatry, School of Mental Health, Psychiatric Genetics Laboratory (PSYG-Lab), Jining Medical University, Jining, China
| | - Chuanxin Liu
- Department of Psychiatry, School of Mental Health, Psychiatric Genetics Laboratory (PSYG-Lab), Jining Medical University, Jining, China
| | - Wenqiang Wang
- Department of China-Canada Biological Psychiatry Lab, Xiamen Xianyue Hospital, Xiamen, China
| | - Ce Chen
- Department of Psychiatry, Wenzhou Seventh People's Hospital, Wenzhou, China
| |
Collapse
|
36
|
Sun QX, Wang RR, Liu N, Liu C. Dysregulation of miR-204-3p Driven by the Viability and Motility of Retinoblastoma via Wnt/β-catenin Pathway In Vitro and In Vivo. Pathol Oncol Res 2019; 26:1549-1558. [PMID: 31482398 DOI: 10.1007/s12253-019-00722-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 08/13/2019] [Indexed: 12/31/2022]
Abstract
Retinoblastoma (RB) is a malignant intraocular tumor that frequently occurs in infants and toddlers. Although the most of RB patients in the developed countries could survival from this cancer, the patients in undeveloped areas are still suffering. The human retinal pigment epithelial cell line ARPE-19 and human retinoblastoma (RB) cell lines HXO-RB44, Y79, and WERI-Rb1 were cultured. The mRNA levels of BANCR and miR-204-3p in these cell lines were measured by qRT-PCR. After transfection with sh-BANCR or treatment with miR-204-3p inhibitor in Y79 cells, the cell proliferation rate, growth, invasion, migration, apoptosis and Wnt/β-catenin signaling pathway activity were measured. The regular Y79 and Y79 cells stably expressed sh-BANCR were injected subcutaneously into nude mice, respectively. The volumes and pathohistological futures of tumors were compared. The biochemical features similar to the cell culture were detected and compered. The mRNA measurements showed that BANCR negatively modulate miR-204-3p expression via directly integration with it. Besides, miR-204-3p and Wnt/β-catenin signalling pathway were found to participate in the oncogenic effects of BANCR on RB cell line by Hoechst staining, cell Counting Kit-8 (CCK-8) assay, wound healing assay, transwell assay, and Western blot analysis in vitro. In addition, an in vivo tumorigenesis experiment in nude mice injected with Y79 cells stably expressed sh-BANCR conformed in the effects of BANCR on RB. Taken together, the knockdown of BANCR inhibited cell proliferation, apoptosis, invasion, and migration in RB via targeting miR-204-3p, the mechanism may involve inhibiting Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Qing-Xiu Sun
- Department of Ophthalmology, The Second Clinical Medical College of Qingdao University, Qingdao, China.,The Central Hospital of Qingdao, The Affiliated Central Hospital of Qingdao University, No. 127, Si-Liu South Road, Qingdao, 266000, Shandong Province, China
| | - Rong-Rong Wang
- Department of Ophthalmology, The Second Clinical Medical College of Qingdao University, Qingdao, China.,The Central Hospital of Qingdao, The Affiliated Central Hospital of Qingdao University, No. 127, Si-Liu South Road, Qingdao, 266000, Shandong Province, China
| | - Na Liu
- Department of Ophthalmology, The Second Clinical Medical College of Qingdao University, Qingdao, China.,The Central Hospital of Qingdao, The Affiliated Central Hospital of Qingdao University, No. 127, Si-Liu South Road, Qingdao, 266000, Shandong Province, China
| | - Chao Liu
- Department of Ophthalmology, The Second Clinical Medical College of Qingdao University, Qingdao, China. .,The Central Hospital of Qingdao, The Affiliated Central Hospital of Qingdao University, No. 127, Si-Liu South Road, Qingdao, 266000, Shandong Province, China.
| |
Collapse
|
37
|
Tompa M, Nagy A, Komoly S, Kalman B. Wnt pathway markers in molecular subgroups of glioblastoma. Brain Res 2019; 1718:114-125. [DOI: 10.1016/j.brainres.2019.05.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 05/03/2019] [Accepted: 05/06/2019] [Indexed: 12/20/2022]
|
38
|
Angelopoulou E, Paudel YN, Piperi C. Emerging Pathogenic and Prognostic Significance of Paired Box 3 (PAX3) Protein in Adult Gliomas. Transl Oncol 2019; 12:1357-1363. [PMID: 31352198 PMCID: PMC6664158 DOI: 10.1016/j.tranon.2019.07.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 07/08/2019] [Indexed: 01/19/2023] Open
Abstract
Gliomas present the most common type of brain tumors in adults, characterized by high morbidity and mortality. In search of potential molecular targets, members of paired box (PAX) family have been found expressed in neural crest cells, regulating their proliferation, apoptosis, migration and differentiation. Recently, PAX3 overexpression has been implicated in glioma tumorigenesis by enhancing proliferation, increasing invasiveness and inducing resistance to apoptosis of glioma cells, while maintaining brain glioma stem cells (BGSCs) stemness. Although the oncogenic potential of PAX3 in gliomas is still under investigation, experimental evidence suggests that PAX3 function is mainly mediated through the canonical and non-canonical Wnt signaling pathway as well as through its interaction with GFAP and p53 proteins. In addition, PAX3 may contribute to the chemoresistance of glioma cells and modulates the effectiveness of novel experimental therapies. Further evidence indicates that PAX3 may represent a novel diagnostic and prognostic biomarker for gliomas, facilitating personalized treatment. This review addresses the emerging role of PAX3 in glioma diagnosis, prognosis and treatment, aiming to shed more light on the underlying molecular mechanisms that could lead to more effective treatment approaches.
Collapse
Affiliation(s)
- Efthalia Angelopoulou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Yam Nath Paudel
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
39
|
Hong X, Zhang Z, Pan L, Ma W, Zhai X, Gu C, Zhang Y, Bi X, Huang W, Pei H, Liu Z. MicroRNA-301b promotes the proliferation and invasion of glioma cells through enhancing activation of Wnt/β-catenin signaling via targeting Glypican-5. Eur J Pharmacol 2019; 854:39-47. [PMID: 30951720 DOI: 10.1016/j.ejphar.2019.03.057] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 03/14/2019] [Accepted: 03/28/2019] [Indexed: 02/08/2023]
Abstract
Accumulating evidence has suggested that Glypican-5 (GPC5) is a tumor suppressor gene in many types of cancers. However, whether GPC5 is involved in glioma remains unknown. This study was designed to explore the expression, biological function and regulatory mechanism of GPC5 in glioma. Our results demonstrated that GPC5 expression was significantly decreased in multiple glioma cell lines. Gain-of-function experiments showed that the ectopic expression of GPC5 markedly inhibited the proliferation, invasion and Wnt/β-catenin signaling of glioma cell lines. GPC5 was identified as a target gene of microRNA-301b (miR-301b). Further data showed that miR-301b expression was significantly up-regulated in glioma tissues and cell lines. In addition, miR-301b expression was inversely correlated with GPC5 expression in clinical glioma tissues. The overexpression of miR-301b promoted the proliferation, invasion and Wnt/β-catenin signaling of glioma cell lines, whereas the inhibition of miR-301b showed the opposite effect. However, the silencing of GPC5 significantly reversed the antitumor effect of miR-301b inhibition. Overall, our results revealed a tumor suppressive role of GPC5 in glioma and suggested that GPC5 expression was regulated by miR-301b. Our study indicates that the inhibition of miR-301b represses the proliferation and invasion of glioma cells by up-regulating GPC5 expression.
Collapse
Affiliation(s)
- Xin Hong
- Department of Emergency Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, PR China
| | - Zhengliang Zhang
- Department of Emergency Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, PR China
| | - Longfei Pan
- Department of Emergency Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, PR China
| | - Wei Ma
- Department of Emergency Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, PR China
| | - Xu Zhai
- Department of Emergency Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, PR China
| | - Changwei Gu
- Department of Emergency Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, PR China
| | - Yaru Zhang
- Department of Emergency Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, PR China
| | - Xiaoju Bi
- Department of Emergency Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, PR China
| | - Wan Huang
- Department of Emergency Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, PR China
| | - Honghong Pei
- Department of Emergency Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, PR China.
| | - Zhong Liu
- Department of Emergency Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, PR China.
| |
Collapse
|
40
|
Cao Y, Shi L, Wang M, Hou J, Wei Y, Du C. ATDC contributes to sustaining the growth and invasion of glioma cells through regulating Wnt/β-catenin signaling. Chem Biol Interact 2019; 305:148-155. [DOI: 10.1016/j.cbi.2019.03.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 03/07/2019] [Accepted: 03/26/2019] [Indexed: 02/09/2023]
|
41
|
Liang X, Dong Z, Bin W, Dekang N, Xuhang Z, Shuyuan Z, Liwen L, Kai J, Caixing S. PAX3 Promotes Proliferation of Human Glioma Cells by WNT/β-Catenin Signaling Pathways. J Mol Neurosci 2019; 68:66-77. [PMID: 30826985 DOI: 10.1007/s12031-019-01283-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 02/20/2019] [Indexed: 12/14/2022]
Abstract
The PAX3 (paired box 3) gene plays an important role in embryonic development, diseases, and cancer formation. Our preliminary studies have shown that PAX3 gene is upregulated in glioma cells, which is associated with a worse prognosis. Moreover, PAX3, by facilitating cell proliferation and invasion and inhibiting cell apoptosis, plays an oncogenic role in glioma. However, the specific molecular mechanism of PAX3 acting as an oncogene in glioma remains unclarified. In the present study, we have found that PAX3 overexpression was observed in high grade glioma and predicted a worse prognosis. PAX3 overexpression did not correlate significantly to IDH1 mutation and MGMT methylation. Moreover, the expression of PAX3 was positively correlated with that of β-catenin. In U87 glioma cells, PAX3 interacted with β-catenin, as was confirmed by CO-IP. Besides, PAX3 overexpression promoted cell proliferation and cell cycle progression, while it inhibited cell apoptosis by altering the expressions of important molecules associated with the Wnt signaling pathway, including β-catenin, Myc, VEGF, cyclinD1, MMP7, and Wnt1. In the meantime, it was also proved that PAX3 correlated to β-catenin through a negative regulatory mechanism with respect to the promotion of U87 glioma cell proliferation and cell cycle progression and inhibition of the cell apoptosis. Our experiment demonstrated the role of PAX3 in promoting glioma growth and development, possibly by interacting directly with β-catenin and regulating the Wnt signaling pathway.
Collapse
Affiliation(s)
- Xia Liang
- Department of Neurosurgery, Zhejiang Cancer Hospital, 1 Ban Shan east road, Hangzhou, 310022, Zhejiang Province, China
| | - Zhao Dong
- Department of Neurosurgery, Zhejiang Hospital, 12 Lingyin road, Hangzhou, 310013, Zhejiang Province, China
| | - Wu Bin
- Department of Neurosurgery, Zhejiang Cancer Hospital, 1 Ban Shan east road, Hangzhou, 310022, Zhejiang Province, China
| | - Nie Dekang
- Department of Neurosurgery, Yancheng First Peoples' Hospital, Yancheng, 224001, Jiangsu Province, China
| | - Zhu Xuhang
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital, 1 Ban Shan east road, Hangzhou, 310022, Zhejiang Province, China
| | - Zhang Shuyuan
- Department of Neurosurgery, Zhejiang Cancer Hospital, 1 Ban Shan east road, Hangzhou, 310022, Zhejiang Province, China
| | - Li Liwen
- Department of Neurosurgery, Zhejiang Cancer Hospital, 1 Ban Shan east road, Hangzhou, 310022, Zhejiang Province, China
| | - Jin Kai
- Department of Neurosurgery, Zhejiang Cancer Hospital, 1 Ban Shan east road, Hangzhou, 310022, Zhejiang Province, China
| | - Sun Caixing
- Department of Neurosurgery, Zhejiang Cancer Hospital, 1 Ban Shan east road, Hangzhou, 310022, Zhejiang Province, China.
| |
Collapse
|
42
|
Kafka A, Bačić M, Tomas D, Žarković K, Bukovac A, Njirić N, Mrak G, Krsnik Ž, Pećina‐Šlaus N. Different behaviour of DVL1, DVL2, DVL3 in astrocytoma malignancy grades and their association to TCF1 and LEF1 upregulation. J Cell Mol Med 2019; 23:641-655. [PMID: 30468298 PMCID: PMC6307814 DOI: 10.1111/jcmm.13969] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 09/04/2018] [Accepted: 09/27/2018] [Indexed: 01/21/2023] Open
Abstract
Key regulators of the Wnt signalling, DVL1, DVL2 and DVL3, in astrocytomas of different malignancy grades were investigated. Markers for DVL1, DVL2 and DVL3 were used to detect microsatellite instability (MSI) and gross deletions (LOH), while immunohistochemistry and immunoreactivity score were used to determine the signal strengths of the three DVL proteins and transcription factors of the pathway, TCF1 and LEF1. Our findings demonstrated that MSI at all three DVL loci was constantly found across tumour grades with the highest number in grade II (P = 0.008). Collectively, LOHs were more frequent in high-grade tumours than in low grade ones. LOHs of DVL3 gene were significantly associated with grade IV tumours (P = 0.007). The results on protein expressions indicated that high-grade tumours expressed less DVL1 protein as compared with low grade ones. A significant negative correlation was established between DVL1 expression and malignancy grades (P < 0.001). The expression of DVL2 protein was found similar across grades, while DVL3 expression significantly increased with malignancy grades (P < 0.001). The signal strengths of expressed DVL1 and DVL3 were negatively correlated (P = 0.002). However, TCF1 and LEF1 were both significantly upregulated and increasing with astrocytoma grades (P = 0.001). A positive correlation was established between DVL3 and both TCF1 (P = 0.020) and LEF1 (P = 0.006) suggesting their joint involvement in malignant progression. Our findings suggest that DVL1 and DVL2 may be involved during early stages of the disease, while DVL3 may have a role in later phases and together with TCF1 and LEF1 promotes the activation of Wnt signalling.
Collapse
Affiliation(s)
- Anja Kafka
- Laboratory of Neuro‐oncologyCroatian Institute for Brain ResearchSchool of MedicineUniversity of ZagrebZagrebCroatia
- Department of BiologySchool of MedicineUniversity of ZagrebZagrebCroatia
| | | | - Davor Tomas
- Department of PathologySchool of MedicineUniversity of ZagrebZagrebCroatia
- Department of PathologyUniversity Hospital Center “Sisters of Charity”ZagrebCroatia
| | - Kamelija Žarković
- Department of PathologySchool of MedicineUniversity of ZagrebZagrebCroatia
- Division of PathologyUniversity Hospital Center “Zagreb”ZagrebCroatia
| | - Anja Bukovac
- Laboratory of Neuro‐oncologyCroatian Institute for Brain ResearchSchool of MedicineUniversity of ZagrebZagrebCroatia
- Department of BiologySchool of MedicineUniversity of ZagrebZagrebCroatia
| | - Niko Njirić
- Laboratory of Neuro‐oncologyCroatian Institute for Brain ResearchSchool of MedicineUniversity of ZagrebZagrebCroatia
- Department of NeurosurgeryUniversity Hospital Center “Zagreb”School of MedicineUniversity of ZagrebZagrebCroatia
| | - Goran Mrak
- Department of NeurosurgeryUniversity Hospital Center “Zagreb”School of MedicineUniversity of ZagrebZagrebCroatia
| | - Željka Krsnik
- Department of NeuroscienceCroatian Institute for Brain ResearchSchool of MedicineUniversity of ZagrebZagrebCroatia
| | - Nives Pećina‐Šlaus
- Laboratory of Neuro‐oncologyCroatian Institute for Brain ResearchSchool of MedicineUniversity of ZagrebZagrebCroatia
- Department of BiologySchool of MedicineUniversity of ZagrebZagrebCroatia
| |
Collapse
|
43
|
Chen X, Dong D, Pan C, Xu C, Sun Y, Geng Y, Kong L, Xiao X, Zhao Z, Zhou W, Huang L, Song Y, Zhang L. Identification of Grade-associated MicroRNAs in Brainstem Gliomas Based on Microarray Data. J Cancer 2018; 9:4463-4476. [PMID: 30519352 PMCID: PMC6277643 DOI: 10.7150/jca.26417] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 08/01/2018] [Indexed: 12/19/2022] Open
Abstract
Gliomas arising in the brainstem are rare tumours that are difficult to surgically resect, and the microRNAs (miRNAs) and signalling pathways associated with brainstem gliomas (BSGs) are largely unknown. To identify grade-associated miRNAs in BSGs, a microarray analysis of 10 low-grade and 15 high-grade BSGs was performed in this study. Differentially expressed miRNAs (DE-miRNAs) were identified, and the functional DE-miRNAs were selected. The potential target genes and enriched pathways were analysed, and a target gene-associated protein-protein interaction (PPI) network was generated. Grade-associated functional DE-miRNAs were confirmed by real-time quantitative PCR. First, 28 functional DE-miRNAs, including 13 upregulated miRNAs and 15 downregulated miRNAs, were identified. Second, 2546 target genes that were involved in BSG-related pathways, such as signalling pathways regulating the pluripotency of stem cells, the AMPK signalling pathway, the HIF-1 signalling pathway, the PI3K-Akt signalling pathway, the Wnt signalling pathway and the Hippo signalling pathway, were screened. Third, PHLPP2 and VEGFA were identified as hub genes in the PPI network. Last, we found that hsa-miR-34a-5p inhibits BSG cell invasion in vitro. In summary, using integrated bioinformatics analysis, we have identified the potential target genes and pathways of grade-associated functional DE-miRNAs in BSGs, which could improve the accuracy of prognostic evaluation. Furthermore, these hub genes and pathways could be therapeutic targets for the treatment of BSGs.
Collapse
Affiliation(s)
- Xin Chen
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Tiantanxili 6, Dongcheng District, Beijing, 100050, China
| | - Dezuo Dong
- Department of Radiation Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Changcun Pan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Tiantanxili 6, Dongcheng District, Beijing, 100050, China
| | - Cheng Xu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Tiantanxili 6, Dongcheng District, Beijing, 100050, China
| | - Yu Sun
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Tiantanxili 6, Dongcheng District, Beijing, 100050, China
| | - Yibo Geng
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Tiantanxili 6, Dongcheng District, Beijing, 100050, China
| | - Lu Kong
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Tiantanxili 6, Dongcheng District, Beijing, 100050, China
| | - Xiong Xiao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Tiantanxili 6, Dongcheng District, Beijing, 100050, 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, China
| | - Wei Zhou
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lijie Huang
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, 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, China
| | - Liwei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Tiantanxili 6, Dongcheng District, Beijing, 100050, China
| |
Collapse
|
44
|
Xu S, Fan Y, Li D, Liu Y, Chen X. Glycoprotein nonmetastatic melanoma protein B accelerates tumorigenesis of cervical cancer in vitro by regulating the Wnt/β-catenin pathway. ACTA ACUST UNITED AC 2018; 52:e7567. [PMID: 30484490 PMCID: PMC6262743 DOI: 10.1590/1414-431x20187567] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 10/01/2018] [Indexed: 11/28/2022]
Abstract
Cervical cancer is one of the most common cancers among women around the world. However, the underlying mechanism involved in cervical cancer progression is incompletely known. In the present study, we determined the role of glycoprotein nonmetastatic melanoma protein B (GPNMB) in tumorigenesis of cervical cancer. According to the GEO database, we found that GPNMB expression was significantly higher in cervical cancer than in normal cervix epithelium. A similar pattern was observed in GPNMB expression in cultured cervical cancer cells and normal cervical epithelial cells. Compared with the control, GPNMB knockdown significantly decreased the proliferation and migration capacity, but enhanced the apoptosis capacity of SiHa and HeLa cells. Additionally, the activity of MMP-2 and MMP-9 were aberrantly increased in SiHa and HeLa cells compared with normal cervical epithelial cells, whereas their activities were strongly inhibited by GPNMB siRNA. Furthermore, Wnt/β-catenin signaling was activated by GPNMB in SiHa and HeLa cells. Increased MMP-2/MMP-9 expression was suppressed by Dkk-1, inhibitor of Wnt/β-catenin signaling, while it was enhanced by stimulator BIO. The proliferation, migration, and apoptosis capacity of HeLa cells were found to be affected by Dkk-1 and BIO to different extents. In conclusion, we demonstrated that GPNMB contributed to the tumorigenesis of cervical cancer, at least in part, by regulating MMP-2/MMP-9 activity in tumor cells via activation of canonical Wnt/β-catenin signaling. This might be a potential therapeutic target for treating human cervical cancer.
Collapse
Affiliation(s)
- Shuxiang Xu
- Department of Obstetrics and Gynecology, Huashan Hospital North, Fudan University, Shanghai, China
| | - Yingying Fan
- Department of Obstetrics and Gynecology, Huashan Hospital North, Fudan University, Shanghai, China
| | - Dongping Li
- Department of Obstetrics and Gynecology, Huashan Hospital North, Fudan University, Shanghai, China
| | - Yan Liu
- Department of Obstetrics and Gynecology, Huashan Hospital North, Fudan University, Shanghai, China
| | - Xu Chen
- Department of Obstetrics and Gynecology, Huashan Hospital North, Fudan University, Shanghai, China
| |
Collapse
|
45
|
Cao Q, Wang X, Shi Y, Zhang M, Yang J, Dong M, Mi Y, Zhang Z, Liu K, Jiang L, Wang N, Wang P. FOXC1 silencing inhibits the epithelial‑to‑mesenchymal transition of glioma cells: Involvement of β‑catenin signaling. Mol Med Rep 2018; 19:251-261. [PMID: 30431099 PMCID: PMC6297783 DOI: 10.3892/mmr.2018.9650] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 08/31/2018] [Indexed: 12/17/2022] Open
Abstract
Glioma is a type of malignant brain tumor. Forkhead box C1 (FOXC1) is a conserved transcription factor that is involved in tumorigenesis; however, the function of FOXC1 in glioma remains unclear. The present study aimed to investigate the effects of FOXC1 silencing on the epithelial-to-mesenchymal transition (EMT) of glioma cells. FOXC1-specific small interfering RNAs were employed to downregulate the expression levels of FOXC1 in glioma cells. The proliferation, migration and invasion of glioma cells were assessed by MTT assay, wound healing assay and Transwell assay. Western blot analysis was performed to reveal the effects of FOXC1 on EMT-associated proteins and β-catenin signaling. The results revealed that, following FOXC1 silencing, the proliferation, migration and invasion of glioma cells were decreased. The expression levels of EMT-associated proteins were also affected. Further examination demonstrated that β-catenin signaling was involved in the effects of FOXC1 on glioma cells. Previous results suggested that overexpression of β-catenin reversed the effects of FOXC1 silencing on glioma cells. The present study demonstrated that FOXC1 may regulate the EMT of glioma cells, potentially via β-catenin signaling. Therefore, FOXC1 may be a potential therapeutic target for the treatment of glioma.
Collapse
Affiliation(s)
- Qinchen Cao
- Department of Radiation Therapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Xinxin Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yonggang Shi
- Department of Radiation Therapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Jing Yang
- Department of Radiation Therapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Meilian Dong
- Department of Radiation Therapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yin Mi
- Department of Radiation Therapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Zhigang Zhang
- Department of Radiation Therapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Ke Liu
- Department of Radiation Therapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Li Jiang
- Department of Radiation Therapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Na Wang
- Department of Radiation Therapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Ping Wang
- Department of Radiation Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China
| |
Collapse
|
46
|
Valproic Acid Inhibits Proliferation and Reduces Invasiveness in Glioma Stem Cells Through Wnt/β Catenin Signalling Activation. Genes (Basel) 2018; 9:genes9110522. [PMID: 30373123 PMCID: PMC6267016 DOI: 10.3390/genes9110522] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/17/2018] [Accepted: 10/22/2018] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma is the most common malignant brain tumour in adults. The failure of current therapies can be ascribed to glioma stem cells (GSCs), which can rapidly repopulate the tumour following the initial treatment. The study of histone deacetylase inhibitors, such as valproic acid (VPA), is becoming an attractive field in cancer research. However, the exact mechanisms underlying its anti-cancer effect remain to be elucidated due to its pleiotropic effects on several cell-signalling pathways. Ingenuity Pathway Analysis (IPA) bioinformatics analysis was performed on genome-wide data regarding GSCs methylome to identify the signalling pathways mainly affected by methylation changes induced by VPA. Real time PCR and luciferase reporter assay were used to better investigate VPA effects on Wnt/β-catenin signalling pathway. VPA effect on GSC proliferation was evaluated by 3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide (MTT) and Trypan blue assays. Finally, VPA impact on GSC motility was demonstrated by Boyden chamber assay and further confirmed evaluating the expression levels or localisation, through western blot or immunofluorescence, of Twist1, Snail1, E-Cadherin and N-Cadherin. The bioinformatics analyses performed on GSCs methylome highlighted that Wnt/β-catenin signalling was affected by the methylation changes induced by VPA, which could influence its activation status. In particular, we pointed out a general activation of this pathway after VPA exposure, which was accompanied by an inhibitory potential on GSCs proliferation. Finally, we also proved VPA's ability to inhibit GSCs invasion through Snail1 and Twist1 downregulation and E-Cadherin relocalisation. VPA treatment may represent a new, interesting therapeutic approach to affect GSC proliferation and motility, but further investigations are certainly needed.
Collapse
|
47
|
Tang SL, Gao YL, Hu WZ. Retracted Article: TRIM22 functions as an oncogene in gliomas through regulating the Wnt/β-catenin signaling pathway. RSC Adv 2018; 8:30894-30901. [PMID: 35548737 PMCID: PMC9085489 DOI: 10.1039/c8ra05684f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 08/20/2018] [Indexed: 12/20/2022] Open
Abstract
The tripartite motif-containing (TRIM) family is a group of proteins that are implicated in a plethora of pathological conditions. TRIM22 has been found to be involved in various cancers; however, the role of TRIM22 in gliomas has not been reported. The present study aimed to evaluate the expression pattern of TRIM22 and its function in gliomas. TRIM22 expressions in glioma tissues and cell lines were measured by RT-PCR and western blot analysis. To knockdown TRIM22 by small hairpin RNAs (shTRIM22), the U118 cells were transfected with pLKO.1-shTRIM22 plasmid or pLKO.1 plasmid. Cell proliferation was measured using CCK-8 assay. Transwell assays were performed to evaluate the migration and invasion. The epithelial-mesenchymal transition (EMT) was assessed by detecting the expressions of E-cadherin, N-cadherin and vimentin with western blot analysis. A xenograft mouse model was established to evaluate the effect of TRIM22 silencing on tumor growth in vivo. The expressions of β-catenin, cyclin D1, and c-Myc were analyzed by western blot analysis. TRIM22 was significantly overexpressed in glioma tissues and cell lines. In vitro studies demonstrated that TRIM22 knockdown inhibited cell proliferation, migration, and invasion. Additionally, TRIM22 silencing increased the expressions of E-cadherin, and decreased the expressions of N-cadherin and vimentin. Nude mouse xenograft assay showed that TRIM22 silencing inhibited tumor growth in vivo. Furthermore, silencing of TRIM22 inhibited the activation of the Wnt/β-catenin pathway. Treatment with LiCl, an activator of the Wnt/β-catenin pathway, attenuated the effects of shTRIM22 on U118 cells. Silencing of TRIM22 inhibited proliferation, migration and invasion, as well as repressing the EMT process in glioma cells. The Wnt/β-catenin pathway was involved in the effect of TRIM22.
Collapse
Affiliation(s)
- Shi-Lei Tang
- Department of Neurosurgery, Huaihe Hospital of Henan University No. 8 Baobei Road Kaifeng 475000 Henan Province China +86-0371-23906516 +86-0371-23906516
| | - Yuan-Lin Gao
- Department of Neurology, Kaifeng Central Hospital Kaifeng 475000 Henan Province China
| | - Wen-Zhong Hu
- Department of Neurosurgery, Huaihe Hospital of Henan University No. 8 Baobei Road Kaifeng 475000 Henan Province China +86-0371-23906516 +86-0371-23906516
| |
Collapse
|
48
|
RUNX3 inhibits glioma survival and invasion via suppression of the β-catenin/TCF-4 signaling pathway. J Neurooncol 2018; 140:15-26. [PMID: 29916101 DOI: 10.1007/s11060-018-2927-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 06/09/2018] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Runt-related transcription factor 3 (RUNX3) exerts a tumor suppressor gene associated with gastric and other cancers, including glioma. However, how its anti-tumor mechanism in functions glioma is unclear. METHODS We assayed expression of RUNX3 with a tissue microarray (TMA), frozen cancer tissues and malignant glioma cell lines using immunohistochemistry, qRT-PCR and Western bolt analysis. Cell proliferation, invasion, cell cycle distribution and apoptosis were also examined to confirm the effect of RUNX3 medicated malignant phenotype. TOP/FOP experiment was used to detect the β-catenin/Tcf-4 transcription activity by RUNX3. RESULTS Enforced RUNX3 expression inhibited proliferation and invasion, induced cell cycle arrest and promoted apoptosis in vitro and in vivo, Bim siRNA partically reversed the effect of RUNX3-induced apoptosis in LN229 and U87 cells, suggesting a dependent role of Bim-caspase pathway. Moreover, Mechanism investigations revealed that restoration of RUNX3 suppressed β-catenin/Tcf-4 transcription activity. CONCLUSIONS RUNX3 plays a pivotal role in glioma initiation and progression as a tumor suppressor via attenuation of Wnt signaling, highlighting it as a potential therapeutic target for glioma.
Collapse
|
49
|
Vallée A, Lecarpentier Y, Guillevin R, Vallée JN. Opposite Interplay Between the Canonical WNT/β-Catenin Pathway and PPAR Gamma: A Potential Therapeutic Target in Gliomas. Neurosci Bull 2018; 34:573-588. [PMID: 29582250 PMCID: PMC5960455 DOI: 10.1007/s12264-018-0219-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 01/18/2018] [Indexed: 12/19/2022] Open
Abstract
In gliomas, the canonical Wingless/Int (WNT)/β-catenin pathway is increased while peroxisome proliferator-activated receptor gamma (PPAR-γ) is downregulated. The two systems act in an opposite manner. This review focuses on the interplay between WNT/β-catenin signaling and PPAR-γ and their metabolic implications as potential therapeutic target in gliomas. Activation of the WNT/β-catenin pathway stimulates the transcription of genes involved in proliferation, invasion, nucleotide synthesis, tumor growth, and angiogenesis. Activation of PPAR-γ agonists inhibits various signaling pathways such as the JAK/STAT, WNT/β-catenin, and PI3K/Akt pathways, which reduces tumor growth, cell proliferation, cell invasiveness, and angiogenesis. Nonsteroidal anti-inflammatory drugs, curcumin, antipsychotic drugs, adiponectin, and sulforaphane downregulate the WNT/β-catenin pathway through the upregulation of PPAR-γ and thus appear to provide an interesting therapeutic approach for gliomas. Temozolomide (TMZ) is an antiangiogenic agent. The downstream action of this opposite interplay may explain the TMZ-resistance often reported in gliomas.
Collapse
Affiliation(s)
- Alexandre Vallée
- Laboratory of Mathematics and Applications, Unités Mixtes de Recherche (UMR), Centre National de la Recherche Scientifique (CNRS) 7348, University of Poitiers, Poitiers, France.
- Délégation à la Recherche Clinique et à l'Innovation (DRCI), Hôpital Foch, Suresnes, France.
| | - Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien, Meaux, France
| | - Rémy Guillevin
- DACTIM, UMR CNRS 7348, University of Poitiers et CHU de Poitiers, Poitiers, France
| | - Jean-Noël Vallée
- Laboratory of Mathematics and Applications, Unités Mixtes de Recherche (UMR), Centre National de la Recherche Scientifique (CNRS) 7348, University of Poitiers, Poitiers, France
- CHU Amiens Picardie, University of Picardie Jules Verne, Amiens, France
| |
Collapse
|
50
|
Xu W, Zheng J, Bie S, Kang L, Mao Q, Liu W, Guo J, Lu J, Xia R. Propofol inhibits Wnt signaling and exerts anticancer activity in glioma cells. Oncol Lett 2018; 16:402-408. [PMID: 29928428 DOI: 10.3892/ol.2018.8606] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 12/11/2017] [Indexed: 11/06/2022] Open
Abstract
Aberrant activation of Wnt signaling is implicated in gliomagenesis. Propofol, the most commonly used intravenous anesthetic agent in clinics, exhibits potent antitumor activity in a variety of cancer cells through different mechanisms. However, the role of propofol on Wnt signaling and glioma cell growth remains to be fully elucidated. In the present study, propofol was identified as a potent inhibitor of Wnt signaling. In 293T cells transfected with Wnt1 or Wnt3 expression plasmids or treated with Wnt3A-conditioned medium, propofol significantly inhibited the transcriptional activity of the SuperTopFlash reporter and the expression of Wnt target genes. The inhibitory effect of propofol on Wnt signaling was also observed in glioma cells. Further experiments demonstrated that propofol suppressed glioma cell growth by decreasing cell proliferation and enhancing cell apoptosis. Finally, the potential antitumor efficiency of propofol was confirmed using xenograft experiments in vivo. Taken together, the results indicated a novel mechanism for the anticancer activity of propofol and provide supporting evidence for its use as a prospective anticancer drug to treat glioma in patients with deregulated Wnt signaling.
Collapse
Affiliation(s)
- Wei Xu
- Department of Anesthesiology, The First People's Hospital of Jingzhou, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Jiwei Zheng
- Department of Anesthesiology, The First People's Hospital of Jingzhou, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Shijie Bie
- Department of Anesthesiology, The First People's Hospital of Jingzhou, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Liuyu Kang
- Department of Anesthesiology, The First People's Hospital of Jingzhou, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Qingjun Mao
- Department of Anesthesiology, The First People's Hospital of Jingzhou, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Weiwei Liu
- Department of Anesthesiology, The First People's Hospital of Jingzhou, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Jinxin Guo
- Department of Anesthesiology, The First People's Hospital of Jingzhou, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Juan Lu
- Operating Room, The First People's Hospital of Jingzhou, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Rui Xia
- Department of Anesthesiology, The First People's Hospital of Jingzhou, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| |
Collapse
|