101
|
Liu PF, Farooqi AA, Peng SY, Yu TJ, Dahms HU, Lee CH, Tang JY, Wang SC, Shu CW, Chang HW. Regulatory effects of noncoding RNAs on the interplay of oxidative stress and autophagy in cancer malignancy and therapy. Semin Cancer Biol 2020; 83:269-282. [PMID: 33127466 DOI: 10.1016/j.semcancer.2020.10.009] [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] [Received: 09/15/2020] [Revised: 10/15/2020] [Accepted: 10/18/2020] [Indexed: 12/15/2022]
Abstract
Noncoding RNAs (ncRNAs) regulation of various diseases including cancer has been extensively studied. Reactive oxidative species (ROS) elevated by oxidative stress are associated with cancer progression and drug resistance, while autophagy serves as an ROS scavenger in cancer cells. However, the regulatory effects of ncRNAs on autophagy and ROS in various cancer cells remains complex. Here, we explore how currently investigated ncRNAs, mainly miRNAs and lncRNAs, are involved in ROS production through modulating antioxidant genes. The regulatory effects of miRNAs and lncRNAs on autophagy-related (ATG) proteins to control autophagy activity in cancer cells are discussed. Moreover, differential expression of ncRNAs in tumor and normal tissues of cancer patients are further analyzed using The Cancer Genome Atlas (TCGA) database. This review hypothesizes links between ATG genes- or antioxidant genes-modulated ncRNAs and ROS production, which might result in tumorigenesis, malignancy, and cancer recurrence. A better understanding of the regulation of ROS and autophagy by ncRNAs might advance the use of ncRNAs as diagnostic and prognostic markers as well as therapeutic targets in cancer therapy.
Collapse
Affiliation(s)
- Pei-Feng Liu
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Science, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan; Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan.
| | - Ammad Ahmad Farooqi
- Department of Molecular Oncology, Institute of Biomedical and Genetic Engineering (IBGE), Islamabad, Pakistan.
| | - Sheng-Yao Peng
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Science, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Tzu-Jung Yu
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Hans-Uwe Dahms
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Science, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan; Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan.
| | - Cheng-Hsin Lee
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Science, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Jen-Yang Tang
- Department of Radiation Oncology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.
| | - Sheng-Chieh Wang
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Science, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Chih-Wen Shu
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan; Institute of Biopharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan.
| | - Hsueh-Wei Chang
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Science, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan; Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.
| |
Collapse
|
102
|
Wu D, Li H, Wang J, Li H, Xiao Q, Zhao X, Huo Z. LncRNA NEAT1 promotes gastric cancer progression via miR-1294/AKT1 axis. Open Med (Wars) 2020; 15:1028-1038. [PMID: 33336058 PMCID: PMC7718639 DOI: 10.1515/med-2020-0218] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 06/17/2020] [Accepted: 07/08/2020] [Indexed: 12/22/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) were reported to promote the development of gastric cancer (GC). Nuclear-enriched abundant transcript 1 (NEAT1) played a great role in diverse cancers, but the mechanism of NEAT1 in GC remains indistinct. NEAT1 and AKT1 were distinctly up-regulated and miR-1294 was down-regulated in GC tissues and cells. Cell proliferation and metastasis were refrained but apoptosis was promoted in GC cells after knockdown of NEAT1. NEAT1 negatively regulated miR-1294 expression, and the miR-1294 inhibitor reverted the si-NEAT1-induced effect on GC cells. NEAT1 modulated AKT1 expression through miR-1294, and the si-NEAT1-induced effect was relieved by AKT1. NEAT1 affected phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) signaling pathway via regulating miR-1294 and AKT1. NEAT1 could modulate cell proliferation, apoptosis, and metastasis in GC cells by regulating the PI3K/AKT/mTOR signaling pathway via the miR-1294/AKT1 axis, showing the great potential for NEAT1 as a valid biomarker in the progression and treatment of GC.
Collapse
Affiliation(s)
- Dianchao Wu
- Department of Surgical Oncology, Xingtai People’s Hospital, No.16, Hongxing Street, Xingtai, 054031, Hebei, China
| | - Hui Li
- Department of Surgical Oncology, Xingtai People’s Hospital, No.16, Hongxing Street, Xingtai, 054031, Hebei, China
| | - Junfeng Wang
- Department of Colorectal Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Hua Li
- Department of Surgical Oncology, Xingtai People’s Hospital, No.16, Hongxing Street, Xingtai, 054031, Hebei, China
| | - Qihai Xiao
- Department of Surgical Oncology, Xingtai People’s Hospital, No.16, Hongxing Street, Xingtai, 054031, Hebei, China
| | - Xiaofeng Zhao
- Department of Surgical Oncology, Xingtai People’s Hospital, No.16, Hongxing Street, Xingtai, 054031, Hebei, China
| | - Zhibin Huo
- Department of Surgical Oncology, Xingtai People’s Hospital, No.16, Hongxing Street, Xingtai, 054031, Hebei, China
| |
Collapse
|
103
|
Wang Z, Wu H, Yan H, Cai T, Dai J, Liu Q. LINC00210 exerts oncogenic roles in glioma by sponging miR-328. Exp Ther Med 2020; 20:137. [PMID: 33110451 PMCID: PMC7581018 DOI: 10.3892/etm.2020.9266] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Accepted: 08/21/2020] [Indexed: 12/13/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) have been reported to serve key roles in human cancer types, including glioma. However, to the best of our knowledge, the expression and function of lncRNA LINC00210 in glioma have not previously been investigated. The present study was conducted to explore the regulatory role of LINC00210 in glioma cells. The present study demonstrated that LINC00210 was significantly upregulated in glioma tissues, and high expression of LINC00210 was significantly associated with advanced clinical stage and poor prognosis in patients with glioma. It was found that LINC00210 knockdown significantly inhibited the proliferation and migration of U251 and T98G cells. The results of luciferase reporter assays indicated that LINC00210 could directly target microRNA (miR)-328 in glioma cells, and miR-328 expression was negatively correlated with LINC00210 expression in glioma tissues. LINC00210 knockdown significantly promoted the expression of miR-328 in U251 and T98G cells. Moreover, silencing miR-328 impaired the inhibitory effects of LINC00210 knockdown on the proliferation and migration of U251 and T98G cells. Therefore, the present results suggested that LINC00210 may exert an oncogenic role in glioma via sponging miR-328.
Collapse
Affiliation(s)
- Zhifei Wang
- Department of Neurosurgery, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Hao Wu
- Department of Neurosurgery, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Hui Yan
- Department of Neurosurgery, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Tao Cai
- Department of Neurosurgery, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Jin Dai
- Department of Neurosurgery, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Qiang Liu
- Department of Neurosurgery, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
104
|
Hu W, Liu W, Liang H, Zhang C, Zou M, Zou B. Silencing of methyltransferase-like 3 inhibits oesophageal squamous cell carcinoma. Exp Ther Med 2020; 20:138. [PMID: 33082869 PMCID: PMC7557329 DOI: 10.3892/etm.2020.9267] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 10/22/2019] [Indexed: 12/13/2022] Open
Abstract
Methyltransferase-like 3 (METTL3) is a methyltransferase responsible for N6-methyladenosine mRNA modifications, which has been demonstrated to serve oncogenic roles in various types of cancer; however, the exact function of METTL3 in oesophageal squamous cell carcinoma (ESCC) has not been determined. The present study aimed to explore the regulatory role of METTL3 in ESCC. In the present study, reverse transcription-quantitative PCR and western blotting were used to examine mRNA and protein expression, CCK-8 assays and flow cytometry were used to determine cellular viability and apoptosis, and wound healing and Transwell assays were conducted to study cellular migration and invasion. The expression levels of METTL3 were significantly higher in ESCC tissues and cell lines compared with adjacent non-tumour tissues and the normal oesophageal epithelial cell line HET-1A, respectively. Increased METTL3 expression was associated with an advanced clinical stage of ESCC and poorer prognosis. Furthermore, the genetic knockdown of METTL3 using small interfering RNA significantly suppressed ESCC growth, invasion and migration in vitro, and induced cellular apoptosis, in addition to reducing the phosphorylation levels of PI3K and AKT. In conclusion, the present study demonstrated that the upregulation of METTL3 promoted ESCC progression, and that inhibition of METTL3 significantly suppressed the malignant phenotypes of ESCC cells, at least in part, by downregulating PI3K/AKT signalling activity. Thus, it is suggested that METTL3 may be a promising therapeutic target for ESCC.
Collapse
Affiliation(s)
- Wen Hu
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Wei Liu
- Department of Cardiac Major Vascular Surgery, Xiangya Hospital of Central South University, Changsha, Hunan 410008, P.R. China
| | - Hengxing Liang
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Chunmin Zhang
- Institute of Foreign Languages, Central South University, Changsha, Hunan 410075, P.R. China
| | - Min Zou
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Bibo Zou
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
105
|
Xie YH, Hu J. Suppression of long non-coding RNA PCAT19 inhibits glioma cell proliferation and invasion, and increases cell apoptosis through regulation of MELK targeted by miR-142-5p. Genes Genomics 2020; 42:1299-1310. [PMID: 32980991 DOI: 10.1007/s13258-020-01003-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 09/15/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Glioma has the chief type of primary brain tumors worldwide. The glioma may be controlled by regulators including some lncRNAs, miRNAs, and proteins. OBJECTIVE Our study aims to discover the underlying mechanism for lncPCAT19/miR-142-5p/MELK axis in glioma progression. METHODS The clinical samples were from patients with gliomas in our Hospital. Hematoxylin-eosin staining (H&E) was applied to determine the clinical pathological changes. Real time PCR was performed to measure the levels of lncPCAT19, miR-142-5p, MELK, and expression of other genes. Western blot was conducted to detect the protein level of MELK. RIP assay was performed to analyze the interaction between lncPCAT19 and miR-142-5p, and dual-luciferase reporter assay was used to determine the binding site between lncPCAT19 and miR-142-5p. CCK-8, colony formation assay, flow cytometry, and trans-well assay were carried out to confirm cell proliferation, colony formation, apoptosis, and invasion, respectively. RESULTS LncPCAT19 was increased in cancer tissues. Then, lncPCAT19 could interact with and down-regulate miR-142-5p. Knockdown of lncPCAT19 distinctly inhibited tumor growth in vivo. Interfering lncPCAT19/overexpression of miR-142-5p decreased glioma cell proliferation, colony formation and invasion, and promoted cell apoptosis by down-regulating expression of Cyclin B1, CDK2, N-cadherin, Bcl-2, and by up-regulating expression of Bax and E-cadherin. Moreover, overexpression of lncPCAT19 overturned tumor-suppressing role of miR-142-5p in cells. Additionally, lncPCAT19 and miR-142-5p synergistically regulated expression of MELK. In conclusion, lncPCAT19 enhanced glioma development via increasing MELK by performing as a sponge of miR-142-5p. CONCLUSIONS LncPCAT19 promotes glioma progression by sponging miR-142-5p to upregulate MELK levels. Thus, lncPCAT19/miR-142-5p/MELK signaling would be a potential target for glioma treatment.
Collapse
Affiliation(s)
- Yu-Hua Xie
- Department of Rehabilitation Medicine, Chenzhou No. 1 People's Hospital, Luo Jia Jin Street, Chenzhou, 423000, Hunan, China
| | - Jiao Hu
- Emergency Department, Chenzhou No. 1 People's Hospital, Luo Jia Jin Street, Chenzhou, 423000, Hunan, China.
| |
Collapse
|
106
|
Non-coding RNAs in Brain Tumors, the Contribution of lncRNAs, circRNAs, and snoRNAs to Cancer Development-Their Diagnostic and Therapeutic Potential. Int J Mol Sci 2020; 21:ijms21197001. [PMID: 32977537 PMCID: PMC7582339 DOI: 10.3390/ijms21197001] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/18/2020] [Accepted: 09/20/2020] [Indexed: 12/17/2022] Open
Abstract
Brain tumors are one of the most frightening ailments that afflict human beings worldwide. They are among the most lethal of all adult and pediatric solid tumors. The unique cell-intrinsic and microenvironmental properties of neural tissues are some of the most critical obstacles that researchers face in the diagnosis and treatment of brain tumors. Intensifying the search for potential new molecular markers in order to develop new effective treatments for patients might resolve this issue. Recently, the world of non-coding RNAs (ncRNAs) has become a field of intensive research since the discovery of their essential impact on carcinogenesis. Some of the most promising diagnostic and therapeutic regulatory RNAs are long non-coding RNAs (lncRNAs), circular RNAs (circRNAs), and small nucleolar RNAs (snoRNAs). Many recent reports indicate the important role of these molecules in brain tumor development, as well as their implications in metastasis. In the following review, we summarize the current state of knowledge about regulatory RNAs, namely lncRNA, circRNAs, and snoRNAs, and their impact on the development of brain tumors in children and adults with particular emphasis on malignant primary brain tumors-gliomas and medulloblastomas (MB). We also provide an overview of how these different ncRNAs may act as biomarkers in these tumors and we present their potential clinical implications.
Collapse
|
107
|
Du F, Hou Q. SNHG17 drives malignant behaviors in astrocytoma by targeting miR-876-5p/ERLIN2 axis. BMC Cancer 2020; 20:839. [PMID: 32883232 PMCID: PMC7469335 DOI: 10.1186/s12885-020-07280-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 08/09/2020] [Indexed: 12/20/2022] Open
Abstract
Background Astrocytoma is a common tumor type in primary central nervous system and has a high death rate around the world. Aberrant expression of long non-coding RNAs (lncRNAs) has been introduced by emerging studies to result in the development of diverse cancers. Methods RT-qPCR examined the expression of SNHG17, miR-876-5p and ERLIN2, and western blot evaluated ERLIN2 protein level. RNA pull down and luciferase reporter assays illustrated the relationships between SNHG17 and its downstream molecules. Results SNHG17 was up-regulated in astrocytoma cells. Moreover, SNHG17 silence could repress the proliferation, migration and invasion of astrocytoma cells. Besides, miR-876-5p was selected out as a downstream molecule of SNHG17 in astrocytoma. ERLIN2 was determined to be targeted by miR-876-5p. ERLIN2 mRNA and protein levels were lessened by miR-876-5p overexpression and SNHG17 silence. Additionally, miR-876-5p overexpression decelerated the biological processes of astrocytoma cells, so did ERLIN2 knockdown. More importantly, the impacts of SNHG17 down-regulation on the malignant behaviors of astrocytoma cells were counteracted by overexpressed ERLIN2 or inhibited miR-876-5p. Conclusions SNHG17 could induce the progression of astrocytoma by sponging miR-876-5p to elevate the expression of ERLIN2. This study indicated that SNHG17 has a high potential to be a therapeutic target for astrocytoma.
Collapse
Affiliation(s)
- Fengping Du
- Department of Neurology, the Second Hospital of Heibei Medical University, No. 215 West Heping Road, Shijiazhuang, 050000, Hebei, China
| | - Qian Hou
- Department of Neurology, the Second Hospital of Heibei Medical University, No. 215 West Heping Road, Shijiazhuang, 050000, Hebei, China.
| |
Collapse
|
108
|
Lin JZ, Lin N, Zhao WJ. Identification and validation of a six-lncRNA prognostic signature with its ceRNA networks and candidate drugs in lower-grade gliomas. Genomics 2020; 112:2990-3002. [PMID: 32447005 DOI: 10.1016/j.ygeno.2020.05.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/08/2020] [Accepted: 05/18/2020] [Indexed: 02/05/2023]
Abstract
Gliomas account for 75% of the primary malignant brain tumors and a majority of lower-grade gliomas (LGG) inevitably develop into glioblastoma. The dysregulation of lncRNAs play a crucial role in LGG. In the present study, we first screened out six differentially expressed lncRNAs (AC021739.2, AL031722.1, AL354740.1, FGD5-AS1, LINC00844, and NEAT1) based on TCGA and GTEx RNA-seq databases. LncRNA prognostic signature was then established by Kaplan-Meier and multivariate Cox proportional hazards regression, with its predictive value validated by time-dependent receiver operating characteristic (ROC) curves. After lncRNA-miRNA-mRNA regulatory networks were established by Cytoscape 3.7.2, Gene Oncology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed, with results enriched in various malignancy-related functions and pathways. Finally, six putative drugs (irinotecan, camptothecin, mitoxantrone, azacitidine, mestranol, and enilconazole) were predicted by Connectivity Map. In conclusion, we identified a 6-lncRNA prognostic signature with its ceRNA networks, and six candidate drugs against LGG.
Collapse
Affiliation(s)
- Jia-Zhe Lin
- Neurosurgical Department, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Nuan Lin
- Obstetrics & Gynecology Department, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Wei-Jiang Zhao
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
109
|
Zheng Y, Miao Y, Xie J, Lin Y, Yao Q, Cai J, Yang X. Long noncoding RNA lysophospholipase-like 1-2 as ceRNA modulates glioma metastasis by regulating miR-217/YWHAG. Am J Transl Res 2020; 12:4204-4215. [PMID: 32913498 PMCID: PMC7476125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 07/04/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND LncRNA-LYPLAL1-2 (lysophospholipase-like 1-2) is expressed at a very low level in gliomas, which are some of the most aggressive tumors. However, the function and mechanism of LYPLAL1-2 are not clear. The purpose of this study was to explore the role of LYPLAL1-2 in glioma. METHODS Reverse transcription and quantitative PCR (qRT-PCR) was used to quantify the levels of lncRNA-LYPLAL1-2, miR-127, and tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein gamma (YWHAG) in glioma tumor tissue and cells. Transwell assays were used to determine the migratory capacity and invasiveness of glioma cells. Hematoxylin and eosin staining was used to identify the metastatic capacity of glioma cells transfected with lncRNA-LYPLAL1-2 in vivo. Western blot analysis was used to identify the levels of YWHAG and related proteins. Luciferase reporter assay was used to identify whether miR-217 is the direct target of lncRNA-LYPLAL1-2. RESULTS LncRNA-LYPLAL1-2 was significantly downregulated in glioma tumor tissue. LncRNA-LYPLAL1-2 overexpression suppressed migration and invasion in vitro and in vivo. LncRNA-LYPLAL1-2 acted as a sponge molecule and targeted miR-217 in glioma cells. YWHAG was identified as the target gene of miR-217 and was indirectly regulated by lncRNA-LYPLAL1-2. CONCLUSIONS LncRNA-LYPLAL1-2 suppressed glioma metastasis via the miR-217/YWHAG axis and is expected to be a potential target for early diagnosis and treatment of gliomas.
Collapse
Affiliation(s)
- Yanyan Zheng
- Department of Neurology, Wenzhou No. 3 Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People’s HospitalWenzhou 325000, Zhejiang Province, China
| | - Yiting Miao
- Department of Neurology, Wenzhou No. 3 Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People’s HospitalWenzhou 325000, Zhejiang Province, China
| | - Jing Xie
- Department of Dermatology, Wenzhou No. 3 Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People’s HospitalWenzhou 325000, Zhejiang Province, China
| | - Yijun Lin
- Department of Urology, Wenzhou No. 3 Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People’s HospitalWenzhou 325000, Zhejiang Province, China
| | - Qiong Yao
- Department of Neurology, Wenzhou No. 3 Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People’s HospitalWenzhou 325000, Zhejiang Province, China
| | - Jianyong Cai
- Department of Neurology, Wenzhou Central HospitalWenzhou 325000, Zhejiang Province, China
| | - Xiaokai Yang
- Department of Neurology, Wenzhou No. 3 Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People’s HospitalWenzhou 325000, Zhejiang Province, China
| |
Collapse
|
110
|
Cheng Z, Wang G, Zhu W, Luo C, Guo Z. LEF1-AS1 accelerates tumorigenesis in glioma by sponging miR-489-3p to enhance HIGD1A. Cell Death Dis 2020; 11:690. [PMID: 32826866 PMCID: PMC7442828 DOI: 10.1038/s41419-020-02823-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 11/25/2022]
Abstract
Long non-coding (lncRNA) lymphoid enhancer-binding factor 1 antisense RNA 1 (LEF1-AS1) has been validated to be implicated in manifold cancers, whereas its function in glioma has not been understood thoroughly. Hence, in this study, we tested that LEF1-AS1 expression was significantly upregulated in glioma tissues and cell lines. Besides, knockdown of LEF1-AS1 repressed cell proliferation while activated apoptosis in glioma cells in vitro, and also suppressed tumor growth in vivo. RNA pull-down and luciferase reporter assays affirmed that LEF1-AS1 could bind with miR-489-3p. In addition, miR-489-3p expression was downregulated in glioma cells. Moreover, miR-489-3p depletion partly offset LEF1-AS1 knockdown-mediated function on proliferation and apoptosis. Further, HIGD1A identified as the target gene of miR-489-3p was upregulated in glioma cells. HIGD1A silence could restrict the process of glioma. In rescue assays, upregulation of HIGD1A remedied the inhibitory impacts of LEF1-AS1 silence on glioma cell growth. In summary, our studies corroborated the regulatory mechanism of LEF1-AS1/miR-489-3p/HIGD1A axis in glioma, suggesting that targeting LEF1-AS1 might be a promising method for glioma therapy in the future.
Collapse
Affiliation(s)
- Zhihua Cheng
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, No. 639 Zhizaoju Road, 200011, Shanghai, China
| | - Guangyu Wang
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, No. 639 Zhizaoju Road, 200011, Shanghai, China
| | - Weiyi Zhu
- Department of Neurosurgery, Shanghai Jing'an District Central Hospital, No. 259 Xikang Road, 200040, Shanghai, China
| | - Cong Luo
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, No. 639 Zhizaoju Road, 200011, Shanghai, China
| | - Zhilin Guo
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, No. 639 Zhizaoju Road, 200011, Shanghai, China.
| |
Collapse
|
111
|
Gao S, Duan H, An D, Yi X, Li J, Liao C. Knockdown of long non-coding RNA LINC00467 inhibits glioma cell progression via modulation of E2F3 targeted by miR-200a. Cell Cycle 2020; 19:2040-2053. [PMID: 32684096 PMCID: PMC7469466 DOI: 10.1080/15384101.2020.1792127] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 11/28/2019] [Indexed: 12/19/2022] Open
Abstract
Studies have found that LINC00467 is an important regulator of cancer. However, the function of LINC00467 in glioma cell is unclear. Therefore, this experimental design based on LINC00467 to explore its mechanism of action in glioma cell. RT-qPCR was used to detect the expression of LINC0046 and miR-200a in glioma cell lines. MTT assay, Edru assay and Transwell assay and flow cytometry were used to detect the effects of LINC0046 and miR-200a on PC cell proliferation, migration and apoptosis. Target gene prediction and screening, luciferase reporter assays were used to validate downstream target genes for LINC0046 and miR-200a. Western blotting was used to detect the protein expression of E2F3. The tumor changes in mice were detected by in vivo experiments in nude mice. LINC00467 was up-regulated in glioma cells. Knockdown of LINC00467 inhibited the viability, migration and invasion of glioma cells. In glioma cells, miR-200a was significantly reduced, while E2F3 was significantly rised. LINC00467 negatively regulated the expression of miR-200a in gliomas, while miR-200a negatively regulated the expression of E2F3 in gliomas. INC00467 promoted the development of glioma by inhibiting miR-200a and promoting E2F3 expression. LINC00467 may be a potential therapeutic target for gliomas.
Collapse
Affiliation(s)
- Shuzi Gao
- Department of Cerebrovascular Diseases, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai City, Guangdong Province, PR. China
| | - Haixia Duan
- Department of Ophthalmology, Zhuhai Hospital of Integrated Traditional Chinese and Western Medicine, Zhuhai City, Guangdong Province, PR. China
| | - Dezhu An
- Department of Neurosurgery, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai City, Guangdong Province, PR. China
| | - Xinfeng Yi
- Department of Neurosurgery, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai City, Guangdong Province, PR. China
| | - Jiayan Li
- Department of Neurosurgery, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai City, Guangdong Province, PR. China
| | - Changchun Liao
- Department of Neurosurgery, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai City, Guangdong Province, PR. China
| |
Collapse
|
112
|
Zhao JB, Xue JF, Zhang WZ, Ren YL, Yan DM. Long Noncoding RNA FGD5-AS1 Promotes Glioma Cell Proliferation, Migration and Invasion by Regulating wnt/β-Catenin Pathway. Cancer Manag Res 2020; 12:6187-6193. [PMID: 32801867 PMCID: PMC7398887 DOI: 10.2147/cmar.s250284] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 05/17/2020] [Indexed: 12/15/2022] Open
Abstract
Purpose To investigate the specific function of long noncoding RNA FGD5 antisense RNA 1 (lncRNA FGD5-AS1) in glioma. Materials and Methods The level of FGD5-AS1 was detected in clinical samples and cell lines by qRT-PCR. Small interfering RNA (siRNA) of FGD5-AS1 or scramble siRNA was transfected into U87 cell lines to examine the role of FGD5-AS1 on glioma development. The proliferation of glioma cells was tested by Cell Counting Kit-8 (CCK-8), the migration and invasion of glioma cells were tested by transwell assay without matrigel or with matrigel. Western blot was used to detect the protein expression, and XAV-939 was used to inhibit wnt/β-catenin pathway. The effect of FGD5-AS1 on tumorigenesis of glioma was confirmed by xenograft nude mice model. Results FGD5-AS1 was significantly increased in glioma tissues and cells. Loss of FGD5-AS1 inhibited the proliferation, migration and invasion of U87 cells. Furthermore, overexpression of FGD5-AS1 increased the mRNA and protein levels of β-catenin and cyclin D1. Blocking of wnt/β-catenin using XAV-939 reversed the promotion role of FGD3-AS1 on glioma cells’ migration and invasion. The in vivo tumor growth assay showed that FGD3-AS1 accelerated glioma tumorigenesis with activating wnt/β-catenin pathway. Conclusion Our research emphasized FGD5-AS1 acting as an oncogene by regulating wnt/β-catenin signaling pathway, thus providing some novel experimental basis for clinical treatment of glioma.
Collapse
Affiliation(s)
- Jun Bo Zhao
- Department of Neurosurgery, Jiaozuo People's Hospital, Jiaozuo 454000, Henan Province, People's Republic of China
| | - Jun Feng Xue
- Department of Neurosurgery, Jiaozuo People's Hospital, Jiaozuo 454000, Henan Province, People's Republic of China
| | - Wu Zhong Zhang
- Department of Neurosurgery, Jiaozuo People's Hospital, Jiaozuo 454000, Henan Province, People's Republic of China
| | - Yong Lu Ren
- Department of Neurosurgery, Jiaozuo People's Hospital, Jiaozuo 454000, Henan Province, People's Republic of China
| | - Dong Ming Yan
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, People's Republic of China
| |
Collapse
|
113
|
Han M, Wang S, Fritah S, Wang X, Zhou W, Yang N, Ni S, Huang B, Chen A, Li G, Miletic H, Thorsen F, Bjerkvig R, Li X, Wang J. Interfering with long non-coding RNA MIR22HG processing inhibits glioblastoma progression through suppression of Wnt/β-catenin signalling. Brain 2020; 143:512-530. [PMID: 31891366 PMCID: PMC7009478 DOI: 10.1093/brain/awz406] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 10/05/2019] [Accepted: 11/09/2019] [Indexed: 01/20/2023] Open
Abstract
Long non-coding RNAs play critical roles in tumour progression. Through analysis of publicly available genomic datasets, we found that MIR22HG, the host gene of microRNAs miR-22-3p and miR-22-5p, is ranked among the most dysregulated long non-coding RNAs in glioblastoma. The main purpose of this work was to determine the impact of MIR22HG on glioblastoma growth and invasion and to elucidate its mechanistic function. The MIR22HG/miR-22 axis was highly expressed in glioblastoma as well as in glioma stem-like cells compared to normal neural stem cells. In glioblastoma, increased expression of MIR22HG is associated with poor prognosis. Through a number of functional studies, we show that MIR22HG silencing inhibits the Wnt/β-catenin signalling pathway through loss of miR-22-3p and -5p. This leads to attenuated cell proliferation, invasion and in vivo tumour growth. We further show that two genes, SFRP2 and PCDH15, are direct targets of miR-22-3p and -5p and inhibit Wnt signalling in glioblastoma. Finally, based on the 3D structure of the pre-miR-22, we identified a specific small-molecule inhibitor, AC1L6JTK, that inhibits the enzyme Dicer to block processing of pre-miR-22 into mature miR-22. AC1L6JTK treatment caused an inhibition of tumour growth in vivo. Our findings show that MIR22HG is a critical inducer of the Wnt/β-catenin signalling pathway, and that its targeting may represent a novel therapeutic strategy in glioblastoma patients.
Collapse
Affiliation(s)
- Mingzhi Han
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University; Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012, China.,NorLux Neuro-Oncology, Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway
| | - Shuai Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University; Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012, China
| | - Sabrina Fritah
- NorLux Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg
| | - Xu Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University; Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012, China
| | - Wenjing Zhou
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University; Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012, China
| | - Ning Yang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University; Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012, China
| | - Shilei Ni
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University; Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012, China
| | - Bin Huang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University; Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012, China
| | - Anjing Chen
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University; Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012, China
| | - Gang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University; Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012, China
| | - Hrvoje Miletic
- NorLux Neuro-Oncology, Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway.,Department of Pathology, Haukeland University Hospital, 5021 Bergen, Norway
| | - Frits Thorsen
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University; Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012, China.,NorLux Neuro-Oncology, Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway.,The Molecular Imaging Center, Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway
| | - Rolf Bjerkvig
- NorLux Neuro-Oncology, Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway.,NorLux Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University; Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012, China
| | - Jian Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University; Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012, China.,NorLux Neuro-Oncology, Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway
| |
Collapse
|
114
|
Deng B, Zhang X, Liang Y, Jiang H, Huang W, Wu Y, Deng W. Nonadherent culture method promotes MSC-mediated vascularization in myocardial infarction via miR-519d/VEGFA pathway. Stem Cell Res Ther 2020; 11:266. [PMID: 32616068 PMCID: PMC7330937 DOI: 10.1186/s13287-020-01780-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/12/2020] [Accepted: 06/18/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) can provide therapeutic benefits for myocardial infarction (MI) recovery; however, the molecular mechanism by which MSCs improve the heart function is unclear. METHODS Microarray analysis was performed to examine the expression profiling of human MSCs (hMSCs) grown as adherent cultures (AC-hMSCs) or nonadherent cultures on ultra-low-adherent plates (nonAC-hMSCs). Real-time quantitative polymerase chain reaction (RT-qPCR), western blotting, and enzyme-linked immunosorbent assays (ELISA) were used to assess VEGFA expression and secretion in the AC-hMSCs and nonAC-hMSCs. The paracrine effect of VEGFA-overexpressing AC-MSCs (AC-VEGFA-hMSCs) or VEGFA-knockdown nonAC-hMSCs (nonAC-shVEGFA-hMSCs) on the angiogenic ability of human umbilical vein endothelial cells (HUVECs) was evaluated using tube formation assay. AC-VEGFA-hMSCs or nonAC-shVEGFA-hMSCs were transplanted into myocardial infarction rats to investigate the therapeutic effect of AC-VEGFA-hMSCs or nonAC-shVEGFA-hMSCs. Luciferase reporter assay was used to confirm the association of VEGFA with miR-519d. RESULTS Microarray analysis revealed that VEGFA is downregulated in AC-hMSCs compared to nonAC-hMSCs. Functional assays revealed that high levels of VEGFA produced from AC-VEGFA-hMSCs increased the tube formation capacity of HUVECs in vitro, improved angiogenesis and cardiac performance, and reduced infarct size in a rat MI model. Low levels of VEGFA secretion from nonAC-shVEGFA-hMSCs had the opposite effects. Mechanistically, we found that miR-519d directly targets VEGFA. High levels of VEGFA secreted from VEGFA-overexpressing nonAC-hMSCs abolished the repressive effect of miR-519d on HUVEC angiogenesis. CONCLUSION Our findings indicate that nonadherent culture-induced secretion of VEGFA plays an important role in MSCs via the miR-519d/VEGFA pathway and may provide a novel therapeutic strategy for MI treatment.
Collapse
Affiliation(s)
- Baoping Deng
- Department of Cardiothoracic Surgery, Zhongshan People's Hospital, 2 Sunwen East Road, Zhongshan, Guangdong, 528403, People's Republic of China
- Department of Vascular Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, People's Republic of China
| | - Xianlan Zhang
- Department of Vascular Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, People's Republic of China
| | - Yi Liang
- Department of Cardiothoracic Surgery, Zhongshan People's Hospital, 2 Sunwen East Road, Zhongshan, Guangdong, 528403, People's Republic of China
| | - Haiming Jiang
- Department of Cardiothoracic Surgery, Zhongshan People's Hospital, 2 Sunwen East Road, Zhongshan, Guangdong, 528403, People's Republic of China
| | - Weizhao Huang
- Department of Cardiothoracic Surgery, Zhongshan People's Hospital, 2 Sunwen East Road, Zhongshan, Guangdong, 528403, People's Republic of China
| | - Yinmeng Wu
- Department of Cardiothoracic Surgery, Zhongshan People's Hospital, 2 Sunwen East Road, Zhongshan, Guangdong, 528403, People's Republic of China
| | - Weiping Deng
- Department of Gastroenterology, Taihe Hospital, Hubei University of Medicine, 32 Ren Min South Road, Shiyan, 442000, Hubei, People's Republic of China.
| |
Collapse
|
115
|
Gu X, Jiang D, Yang Y, Zhang P, Wan G, Gu W, Shi J, Jiang L, Chen B, Zheng Y, Liu D, Guo S, Lu C. Construction and Comprehensive Analysis of Dysregulated Long Noncoding RNA-Associated Competing Endogenous RNA Network in Moyamoya Disease. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2020; 2020:2018214. [PMID: 32617116 PMCID: PMC7306867 DOI: 10.1155/2020/2018214] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 05/09/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Moyamoya disease (MMD) is a rare cerebrovascular disease characterized by chronic progressive stenosis or occlusion of the bilateral internal carotid artery (ICA), the anterior cerebral artery (ACA), and the middle cerebral artery (MCA). MMD is secondary to the formation of an abnormal vascular network at the base of the skull. However, the etiology and pathogenesis of MMD remain poorly understood. METHODS A competing endogenous RNA (ceRNA) network was constructed by analyzing sample-matched messenger RNA (mRNA), long non-coding RNA (lncRNA), and microRNA (miRNA) expression profiles from MMD patients and control samples. Then, a protein-protein interaction (PPI) network was constructed to identify crucial genes associated with MMD. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes pathway (KEGG) enrichment analyses were employed with the DAVID database to investigate the underlying functions of differentially expressed mRNAs (DEmRNAs) involved in the ceRNA network. CMap was used to identify potential small drug molecules. RESULTS A total of 94 miRNAs, 3649 lncRNAs, and 2294 mRNAs were differentially expressed between MMD patients and control samples. A synergistic ceRNA lncRNA-miRNA-mRNA regulatory network was constructed. Core regulatory miRNAs (miR-107 and miR-423-5p) and key mRNAs (STAT5B, FOSL2, CEBPB, and CXCL16) involved in the ceRNA network were identified. GO and KEGG analyses indicated that the DEmRNAs were involved in the regulation of the immune system and inflammation in MMD. Finally, two potential small molecule drugs, CAY-10415 and indirubin, were identified by CMap as candidate drugs for treating MMD. CONCLUSIONS The present study used bioinformatics analysis of candidate RNAs to identify a series of clearly altered miRNAs, lncRNAs, and mRNAs involved in MMD. Furthermore, a ceRNA lncRNA-miRNA-mRNA regulatory network was constructed, which provides insights into the novel molecular pathogenesis of MMD, thus giving promising clues for clinical therapy.
Collapse
Affiliation(s)
- Xuefeng Gu
- Research Department, Shanghai University of Medicine & Health Science Affiliated Zhoupu Hospital, Shanghai, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Dongyang Jiang
- Department of Cardiology, Pan-Vascular Medicine Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yue Yang
- Key Laboratory of Cancer Prevention and Treatment of Heilongjiang Province, Mudanjiang Medical University, Mudanjiang, China
- Department of Pathology, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, China
| | - Peng Zhang
- School of Clinical Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Guoqing Wan
- Research Department, Shanghai University of Medicine & Health Science Affiliated Zhoupu Hospital, Shanghai, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Wangxian Gu
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Junfeng Shi
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Liying Jiang
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Bing Chen
- Department of Neurosurgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yanjun Zheng
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Dingsheng Liu
- Department of Oncology and Hematology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, China
| | - Sufen Guo
- Key Laboratory of Cancer Prevention and Treatment of Heilongjiang Province, Mudanjiang Medical University, Mudanjiang, China
- Department of Pathology, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, China
| | - Changlian Lu
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China
| |
Collapse
|
116
|
Chi D, Zhang W, Jia Y, Cong D, Yu K, Hu S. LINC01207 Predicts Poor Prognosis and Suppresses Cell Growth and Metastasis via Regulating GSK-3β/β-Catenin Signaling Pathway in Malignant Glioma. Med Sci Monit 2020; 26:e923189. [PMID: 32533688 PMCID: PMC7309654 DOI: 10.12659/msm.923189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Background Recent literature has revealed that LINC01207 plays a vital part in tumorigenesis and malignancy progression. However, the potential mechanisms of LINC01207 in malignant glioma are still unknown. Material/Methods Quantitative real-time polymerase chain reaction (qRT-PCR) was applied to analyze LINC01207 mRNA levels in malignant glioma cell lines and tissue samples. The correlation between LINC01207 mRNA levels and clinical characteristics was explored, and the relative survival rate was observed using the Kaplan-Meier method. To examine the function of LINC01207, we performed cell viability, EdU assay, cell cycle assay, Transwell assay, and wound-healing assay to analyze relative cell proliferation, migration/invasion ability. Finally, qRT-PCR and western blot were used to investigate the potential mechanisms. Results LINC01207 mRNA was lowly expressed in malignant glioma cells and cancer tissue samples. Low expression of LINC01207 was associated with Karnofsky performance score (KPS), invasion condition, and tumor grade. Moreover, multivariate analysis confirmed LINC01207 expression and tumor grade were significant independent predictors of poor survival in malignant glioma. LINC01207 markedly inhibited cellar proliferation and viability via inducing G0/G1 phase cell cycle arrested and repressed cell metastasis through restraining epithelial-to-mesenchymal procession in vivo. In addition, we detected a reduction in the protein levels of β-catenin and p-GSK-3β, while GSK-3β expression was upregulated. Conclusions In summary, LINC01207 served as a tumor-related tumor suppress gene for malignant glioma through inhibiting of GSK-3β/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Dapeng Chi
- Department of Neurological Surgery, The Second Affiliated Hospital of the Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Wei Zhang
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China (mainland)
| | - Yulong Jia
- Department of Neurological Surgery, The Second Affiliated Hospital of the Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Damin Cong
- The Second Affiliated Hospital of the Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Kui Yu
- Hospital-Acquired Infection Control Department, Jingmen No. 1 Renmin Hospital, Jingmen, Hubei, China (mainland)
| | - Shaoshan Hu
- Department of Neurological Surgery, The Second Affiliated Hospital of the Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| |
Collapse
|
117
|
Peng X, Zhang K, Ma L, Xu J, Chang W. The Role of Long Non-Coding RNAs in Thyroid Cancer. Front Oncol 2020; 10:941. [PMID: 32596158 PMCID: PMC7300266 DOI: 10.3389/fonc.2020.00941] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 05/13/2020] [Indexed: 12/15/2022] Open
Abstract
Thyroid cancer, the most common endocrine malignancy, has become the most commonly diagnosed malignant solid tumor. Moreover, some cases have poor prognosis, and the survival period is only 3-5 months. Long noncoding RNAs (lncRNAs) are a group of functional RNA molecules more than 200 nucleotides in length that lack the ability to encode protein but participate in all aspects of gene regulation. Functionally, many lncRNAs play essential roles in epigenetic regulation at transcriptional and post-transcriptional levels via various molecular mechanisms. Recent studies have discovered important roles for lncRNAs during the complex process of carcinogenesis in thyroid cancer. In this review, we focus on lncRNAs dysregulated in thyroid cancer and summarize recently reported associations between lncRNAs and thyroid cancer in order to demonstrate the significant value of lncRNAs in diagnosis and treatment.
Collapse
Affiliation(s)
- Xuejiao Peng
- Department of Thyroid Surgery, Second Affiliated Hospital of Jilin University, Changchun, China
| | - Kun Zhang
- Medical Research Center, Second Affiliated Hospital of Jilin University, Changchun, China
| | - Li Ma
- Department of Thyroid Surgery, Second Affiliated Hospital of Jilin University, Changchun, China
| | - Junfeng Xu
- Department of Thyroid Surgery, Second Affiliated Hospital of Jilin University, Changchun, China
| | - Weiqin Chang
- Department of Thyroid Surgery, Second Affiliated Hospital of Jilin University, Changchun, China
| |
Collapse
|
118
|
Jiao M, Guo H, Chen Y, Li L, Zhang L. DARS-AS1 promotes clear cell renal cell carcinoma by sequestering miR-194-5p to up-regulate DARS. Biomed Pharmacother 2020; 128:110323. [PMID: 32526457 DOI: 10.1016/j.biopha.2020.110323] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 05/22/2020] [Accepted: 05/23/2020] [Indexed: 02/01/2023] Open
Abstract
Clear cell renal cell carcinoma (ccRCC), the most frequent subtype of renal cell carcinoma (RCC), is characterized by high relapse rate and mortality. Long non-coding RNAs (lncRNAs) are critical participants during cancer development. LncRNA DARS antisense RNA 1 (DARS-AS1), a newly-found lncRNA, is not specifically reported in ccRCC. However, Gene Expression Profiling Interactive Analysis (GEPIA) and starBase databases revealed the up-regulation of DARS-AS1 in ccRCC. Current study investigated the function and mechanism of DARS-AS1 in ccRCC. Functional assays including colony formation assay, EdU assay, caspase-3 activity detection, flow cytometry analysis and JC-1 assay were implemented to identify the role of DARS-AS1 in ccRCC. As a result, silencing of DARS-AS1 retarded proliferation and facilitated apoptosis in ccRCC cells. Moreover, mainly a cytoplasmic localization of lncRNA DARS-AS1 was verified in ccRCC cells. Then, we demonstrated that DARS-AS1 positively regulated its nearby gene, aspartyl-tRNA synthetase (DARS), by sequestering miR-194-5p. Moreover, DARS was testified as the oncogene in ccRCC and DARS-AS1 worked as a tumor-facilitator in ccRCC through miR-194-5p/DARS signaling. In a summary, this study firstly uncovered that DARS-AS1 boosted DARS expression via absorbing miR-194-5p, therefore contributing to malignancy in ccRCC. Our findings may be helpful for opening new strategies for ccRCC treatment.
Collapse
Affiliation(s)
- Min Jiao
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, PR China
| | - Hui Guo
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, PR China
| | - Yule Chen
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, PR China
| | - Lei Li
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, PR China
| | - Linlin Zhang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, PR China.
| |
Collapse
|
119
|
Chen L, Li X, Lu C, Zhao Y, Zhu J, Yang L. The long non‑coding RNA CASC7 inhibits growth and invasion of non‑small cell lung cancer cells through phosphatase and tensin homolog upregulation via sequestration of miR‑92a. Int J Oncol 2020; 57:466-477. [PMID: 32626930 PMCID: PMC7307594 DOI: 10.3892/ijo.2020.5076] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 04/16/2020] [Indexed: 02/07/2023] Open
Abstract
Accumulating evidence has demonstrated the crucial roles of long non-coding RNAs (lncRNAs) in various human cancers, including non-small cell lung cancer (NSCLC). However, to the best of our knowledge, the role of the lncRNA cancer susceptibility candidate 7 (CASC7) in NSCLC has not been clearly determined. The aim of the present study was to investigate the involvement of CASC7 in NSCLC. Marked downregulation of CASC7 was observed in NSCLC tissues and cell lines, and this downregulation of CASC7 was closely associated with distant metastasis, lymph node involvement and poor overall survival in NSCLC patients. Furthermore, overexpression of CASC7 significantly suppressed the proliferation, invasion and migration of the NSCLC cells A549 and H358, and promoted cell apoptosis in vitro. In addition, CASC7 was shown to act as a competing endogenous RNA by sponging miR-92a, which was proven to be an oncogenic miRNA in our previous study. The expression of miR-92a was upregulated in NSCLC tissues and cell lines, and was found to be inversely associated with CASC7 expression in NSCLC tissues. It was also demonstrated that CASC7 upregulated the expression of the tumor suppressor gene phosphatase and tensin homolog (a well-known target of miR-92a) by sequestration of miR-92a. Moreover, the tumor-suppressive effects of CASC7 were partly reversed by miR-92a overexpression in NSCLC cells. Collectively, the results of the present study indicated that CASC7 may act as a tumor-suppressive lncRNA that inhibits NSCLC progression by sponging miR-92a. These findings may improve our understanding of the potential mechanisms through which gain of CASC7 expression represses NSCLC progression.
Collapse
Affiliation(s)
- Ling Chen
- Department of Thoracic Surgery, Changhai Hospital, Shanghai 200433, P.R. China
| | - Xin Li
- Department of Thoracic Surgery, Changhai Hospital, Shanghai 200433, P.R. China
| | - Chaojing Lu
- Department of Thoracic Surgery, Changhai Hospital, Shanghai 200433, P.R. China
| | - Yue Zhao
- Department of Thoracic Surgery, Changhai Hospital, Shanghai 200433, P.R. China
| | - Ji Zhu
- Department of Thoracic Surgery, Changhai Hospital, Shanghai 200433, P.R. China
| | - Lixin Yang
- Department of Thoracic Surgery, Changhai Hospital, Shanghai 200433, P.R. China
| |
Collapse
|
120
|
Han W, Shi J, Cao J, Dong B, Guan W. Current advances of long non-coding RNAs mediated by wnt signaling in glioma. Pathol Res Pract 2020; 216:153008. [PMID: 32703485 DOI: 10.1016/j.prp.2020.153008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/14/2020] [Accepted: 05/10/2020] [Indexed: 12/21/2022]
Abstract
Glioma is the most common and aggressive brain tumor in the central nervous system (CNS), in which Wnt signaling pathway has been verified to play a pivotal role in regulating the initiation and progression. Currently, numerous studies have indicated that long non-coding RNAs (lncRNAs) have critical functions across biological processes including cell proliferation, colony formation, migration, invasion and apoptosis via Wnt signaling pathway in glioma. This review depicts canonical and non-canonical Wnt/β-catenin signaling pathway properties and relative processing mechanisms in gliomas, and summarizes the function and regulation of lncRNAs mediated by Wnt signaling pathway in the development and progression of glioma. Ultimately, we hope to seek out promising biomarkers and reliable therapeutic targets for glioma.
Collapse
Affiliation(s)
- Wei Han
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jia Shi
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jiachao Cao
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Bo Dong
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Wei Guan
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China.
| |
Collapse
|
121
|
Li B, Lu X, Ma C, Sun S, Shu X, Wang Z, Sun W. Long non-coding RNA NEAT1 promotes human glioma tumor progression via miR-152-3p/CCT6A pathway. Neurosci Lett 2020; 732:135086. [PMID: 32454145 DOI: 10.1016/j.neulet.2020.135086] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 05/07/2020] [Accepted: 05/21/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Long non-coding RNA (lncRNA) nuclear enriched abundant transcript 1 (NEAT1) has been documented to implicate in diverse tumor progression. However, the mechanism of NEAT1 in glioma was rarely reported. METHODS The levels of NEAT1, microRNA-152-3p (miR-152-3p) and chaperonin containing TCP1 subunit 6A (CCT6A) in glioma tissues and cells were measured by quantitative real-time polymerase chain reaction (qRT-PCR). The cell viability, apoptotic rate, the migrated and invaded abilities of A172 and U251 cells were evaluated via cell counting kit-8 (CCK-8), flow cytometry and Transwell assay, respectively. The mice xenograft model was constructed to further verify the effect of NEAT1. The interactions between miR-152-3p and NEAT1 or CCT6A were predicted by starBase v2.0 or TargetScan, then luciferase reporter assay, RNA immunoprecipitation (RIP) and RNA pull-down assay were performed to validate the interaction. The protein level of CCT6A was detected by Western blot assay. RESULTS The levels of NEAT1, CCT6A were highly expressed, but miR-152-3p was decreased in glioma tissues and cells. NEAT1 depletion or miR-152-3p mimics suppressed cell viability, migrated and invaded abilities but induced apoptotic rate in A172 and U251 cells, while the introduction of CCT6A partly counteracted these impacts. In addition, NEAT1 silencing impeded xenograft tumor growth in vivo. MiR-152-3p was verified as a direct target of NEAT1 and directly targeted CCT6A. CCT6A expression was upregulated by NEAT1 and reversed by miR-152-3p. CONCLUSION NEAT1 enhanced glioma progression, partially through miR-152-3p/CCT6A pathway. The novel regulatory network might contribute to the diagnosis and treatment of glioma.
Collapse
Affiliation(s)
- Bin Li
- Department of Pathology, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai Clincal Center, CAS, Shanghai, PR China
| | - Xiangui Lu
- Department of Neurosurgery, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai Clincal Center, CAS, Shanghai, PR China
| | - Cong Ma
- Department of Endocrinology, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai Clincal Center, CAS, Shanghai, PR China
| | - Shujie Sun
- Department of Neurosurgery, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai Clincal Center, CAS, Shanghai, PR China
| | - Xiaoyan Shu
- Department of Neurosurgery, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai Clincal Center, CAS, Shanghai, PR China
| | - Zhiyu Wang
- Department of Neurosurgery, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai Clincal Center, CAS, Shanghai, PR China.
| | - Wanqun Sun
- Department of Science and Education, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai Clincal Center, CAS, Shanghai, PR China.
| |
Collapse
|
122
|
Shi J, Huang Y, Wen C, He S, Wu L, Zhou H. Genome-wide identification and characterization of long non-coding RNAs involved in acquired resistance to gefitinib in non-small-cell lung cancer. Comput Biol Chem 2020; 87:107288. [PMID: 32521497 DOI: 10.1016/j.compbiolchem.2020.107288] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 05/12/2020] [Accepted: 05/14/2020] [Indexed: 12/28/2022]
Abstract
Acquired resistance is a major obstacle to the therapeutic efficacy of gefitinib in non-small-cell lung cancer (NSCLC). Current knowledge about the role of long non-coding RNAs (lncRNAs) in this phenomenon is insufficient. In this study, we searched RNA sequencing data for lncRNAs associated with acquired resistance to gefitinib in NSCLC, and constructed a functional lncRNA-mRNA co-expression network and protein-protein interaction (PPI) network to analyze their putative target genes and biological functions. The expression levels of 14 outstanding dysregulated lncRNAs and mRNA were verified using real-time PCR. Changes in the expression levels of 39 lncRNAs and 121 mRNAs showed common patterns in our two pairs of gefitinib-sensitive and gefitinib-resistant NSCLC cell lines. The co-expression network included 1235 connections among these common differentially expressed lncRNAs and mRNAs. The significantly enriched signaling pathways based on dysregulated mRNAs were mainly involved in the Hippo signaling pathway; proteoglycans in cancer; and valine, leucine, and isoleucine biosynthesis. The results show that LncRNAs play an important part in acquired gefitinib resistance in NSCLC by regulating mRNA expression and function, and may represent potential new molecular biomarkers and therapeutic targets for gefitinib-resistant NSCLC.
Collapse
Affiliation(s)
- Jingjing Shi
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Yutang Huang
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Chunjie Wen
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Shuai He
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Lanxiang Wu
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China.
| | - Honghao Zhou
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China; Pharmacogenetics Research Institute, Institute of Clinical Pharmacology, Central South University, Changsha 410078, China
| |
Collapse
|
123
|
Shen J, Xiong J, Shao X, Cheng H, Fang X, Sun Y, Di G, Mao J, Jiang X. Knockdown of the long noncoding RNA XIST suppresses glioma progression by upregulating miR-204-5p. J Cancer 2020; 11:4550-4559. [PMID: 32489472 PMCID: PMC7255366 DOI: 10.7150/jca.45676] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 04/26/2020] [Indexed: 12/18/2022] Open
Abstract
Background: Gliomas are the most prevalent primary malignant tumors of the central nervous system. Our previous study showed that miR-204-5p is a tumor suppressor gene in glioma. Bioinformatic analyses suggest that long noncoding RNA (lncRNA) X-inactive specific transcript (XIST) is a potential target gene of miR-204-5p. Methods: We analyzed the expression of XIST and miR-204-5p in glioma tissues and the correlation with glioma grade. A series of in vitro experiments were carried out to elucidate the role of XIST in glioma progression. A mouse xenograft model was established to detect whether knockdown of XIST can inhibit glioma growth. A luciferase assay was performed to determine whether XIST can bind to miR-204-5p and the binding specificity. Cells stably expressing shXIST or shNC were transfected with anti-miR-204-5p or anti-miR-204-5p-NC to evaluate whether XIST mediates the tumor-suppressive effects of miR-204-5p. Results: XIST was upregulated in glioma tissues compared with normal brain tissues (NBTs), while miR-204-5p expression was significantly decreased in glioma tissues compared with NBTs. Both XIST and miR-204-5p expression levels were clearly related to glioma grade, and the expression of XIST was obviously negatively correlated with miR-204-5p expression. Knockdown of XIST inhibited glioma cell proliferation, migration, and invasion, promoted apoptosis of glioma cells, inhibited tumor growth and increased the survival time in nude mice. miR-204-5p could directly bind to XIST and negatively regulate XIST expression. XIST mediated glioma progression by targeting miR-204-5p in glioma cells. XIST crosstalk with miR-204-5p regulated Bcl-2 expression to promote apoptosis. Conclusion: Our results provide evidence that XIST, miR-204-5p and Bcl-2 form a regulatory axis that controls glioma progression and can serve as a potential therapeutic target for glioma.
Collapse
Affiliation(s)
- Jun Shen
- Department of Neurosurgery, The First Affiliated Hospital (Yijishan Hospital) of Wannan Medical College, Wuhu, P.R. China
| | - Jianhua Xiong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, P.R. China
| | - Xuefei Shao
- Department of Neurosurgery, The First Affiliated Hospital (Yijishan Hospital) of Wannan Medical College, Wuhu, P.R. China
| | - Hao Cheng
- Department of Neurosurgery, The First Affiliated Hospital (Yijishan Hospital) of Wannan Medical College, Wuhu, P.R. China
| | - Xinyun Fang
- Department of Neurosurgery, The First Affiliated Hospital (Yijishan Hospital) of Wannan Medical College, Wuhu, P.R. China
| | - Yongkang Sun
- Department of Neurosurgery, The First Affiliated Hospital (Yijishan Hospital) of Wannan Medical College, Wuhu, P.R. China
| | - Guangfu Di
- Department of Neurosurgery, The First Affiliated Hospital (Yijishan Hospital) of Wannan Medical College, Wuhu, P.R. China
| | - Jie Mao
- Department of Neurosurgery, Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, P.R. China
| | - Xiaochun Jiang
- Department of Neurosurgery, The First Affiliated Hospital (Yijishan Hospital) of Wannan Medical College, Wuhu, P.R. China
| |
Collapse
|
124
|
Madrer N, Soreq H. Cholino-ncRNAs modulate sex-specific- and age-related acetylcholine signals. FEBS Lett 2020; 594:2185-2198. [PMID: 32330292 PMCID: PMC7496432 DOI: 10.1002/1873-3468.13789] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/19/2020] [Accepted: 03/24/2020] [Indexed: 12/13/2022]
Abstract
Acetylcholine (ACh) signaling orchestrates mammalian movement, mental capacities, and inflammation. Dysregulated ACh signaling associates with many human mental disorders and neurodegeneration in an individual‐, sex‐, and tissue‐related manner. Moreover, aged patients under anticholinergic therapy show increased risk of dementia, but the underlying molecular mechanisms are incompletely understood. Here, we report that certain cholinergic‐targeting noncoding RNAs, named Cholino‐noncoding RNAs (ncRNAs), can modulate ACh signaling, agonistically or antagonistically, via distinct direct and indirect mechanisms and at different timescales. Cholino‐ncRNAs include both small microRNAs (miRNAs) and long noncoding RNAs (lncRNAs). The former may attenuate translation and/or induce destruction of target mRNAs that code for either ACh‐signaling proteins or transcription factors controlling the expression of cholinergic genes. lncRNAs may block miRNAs via ‘sponging’ events or by competitive binding to the cholinergic target mRNAs. Also, single nucleotide polymorphisms in either Cholino‐ncRNAs or in their recognition sites in the ACh‐signaling associated genes may modify ACh signaling‐regulated processes. Taken together, both inherited and acquired changes in the function of Cholino‐ncRNAs impact ACh‐related deficiencies, opening new venues for individual, sex‐related, and age‐specific oriented research, diagnosis, and therapeutics.
Collapse
Affiliation(s)
- Nimrod Madrer
- The Life Sciences Institute and the Edmond and Lily Safra Center of Brain Science, The Hebrew University of Jerusalem, Israel
| | - Hermona Soreq
- The Life Sciences Institute and the Edmond and Lily Safra Center of Brain Science, The Hebrew University of Jerusalem, Israel
| |
Collapse
|
125
|
Chang X, Zhang H, Yang Q, Pang L. LncRNA SOX2OT affects cervical cancer cell growth, migration and invasion by regulating SOX2. Cell Cycle 2020; 19:1391-1403. [PMID: 32286144 DOI: 10.1080/15384101.2020.1750812] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Long non-coding RNA (lncRNA) SOX2 overlapping transcript (SOX2OT) has been shown to play an oncogenic role in diverse cancers, generating eight transcript variants. SOX2 is located in the third intron of SOX2OT. However, the biological function of SOX2OT in cervical cancer and implication with SOX2 remain to be further explored. In this study, we screened the expression pattern of different SOX2OT transcript variants in cervical cancer cells. Interestingly, both high-expression levels of SOX2OT transcript 7 (SOX2OT-7) and SOX2 were detected in C-33A (HPV-) and SiHa (HPV16+) cells. Thus, C-33A and SiHa cells were conducted to investigate the effects of SOX2OT on cell growth, migration and invasion. Finally, rescue experiments were performed to confirm the role of SOX2 in SOX2OT-mediated regulation of cervical cancer progression. The results showed that knockdown of SOX2OT suppressed cell viability, arrested cell cycle and ameliorated migration and invasion ability of C-33A and SiHa cells. Ectopic expression of SOX2OT-7 exacerbated cervical cancer cell proliferation, migration and invasion. In addition, we found that the expression levels and protein stability of SOX2 were positively regulated by SOX2OT. Inhibition of SOX2 could block the malignant phenotypes of C-33A and SiHa cells by SOX2OT-7. In conclusion, these findings indicate that lncRNA SOX2OT contributes to the growth, migration and invasion of cervical cancer cells by modulating SOX2. Importantly, we demonstrate that the transcript SOX2OT-7 may be a novel and promising biomarker for both HPV- and HPV16+ cervical cancer.
Collapse
Affiliation(s)
- Xiaohan Chang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University , Shenyang, People's Republic of China
| | - Huijie Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University , Shenyang, People's Republic of China
| | - Qing Yang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University , Shenyang, People's Republic of China
| | - Li Pang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University , Shenyang, People's Republic of China
| |
Collapse
|
126
|
DeOcesano-Pereira C, Machado RAC, Chudzinski-Tavassi AM, Sogayar MC. Emerging Roles and Potential Applications of Non-Coding RNAs in Glioblastoma. Int J Mol Sci 2020; 21:E2611. [PMID: 32283739 PMCID: PMC7178171 DOI: 10.3390/ijms21072611] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 03/30/2020] [Accepted: 03/30/2020] [Indexed: 02/06/2023] Open
Abstract
Non-coding RNAs (ncRNAs) comprise a diversity of RNA species, which do not have the potential to encode proteins. Non-coding RNAs include two classes of RNAs, namely: short regulatory ncRNAs and long non-coding RNAs (lncRNAs). The short regulatory RNAs, containing up to 200 nucleotides, include small RNAs, such as microRNAs (miRNA), short interfering RNAs (siRNAs), piwi-interacting RNAs (piRNAs), and small nucleolar RNAs (snoRNAs). The lncRNAs include long antisense RNAs and long intergenic RNAs (lincRNAs). Non-coding RNAs have been implicated as master regulators of several biological processes, their expression being strictly regulated under physiological conditions. In recent years, particularly in the last decade, substantial effort has been made to investigate the function of ncRNAs in several human diseases, including cancer. Glioblastoma is the most common and aggressive type of brain cancer in adults, with deregulated expression of small and long ncRNAs having been implicated in onset, progression, invasiveness, and recurrence of this tumor. The aim of this review is to guide the reader through important aspects of miRNA and lncRNA biology, focusing on the molecular mechanism associated with the progression of this highly malignant cancer type.
Collapse
Affiliation(s)
- Carlos DeOcesano-Pereira
- Center of Excellence in New Target Discovery (CENTD), Butantan Institute, 1500 Vital Brazil Avenue, São Paulo 05503-900 SP, Brazil; (C.D.-P.); (A.M.C.-T.)
| | - Raquel A. C. Machado
- Department of Life Science and Medicine, University of Luxembourg, Campus Belval, Avenue des Hauts-Fourneaux, L-4362 Esch-sur-Alzette, Luxembourg;
| | - Ana Marisa Chudzinski-Tavassi
- Center of Excellence in New Target Discovery (CENTD), Butantan Institute, 1500 Vital Brazil Avenue, São Paulo 05503-900 SP, Brazil; (C.D.-P.); (A.M.C.-T.)
| | - Mari Cleide Sogayar
- Biochemistry Department, Chemistry Institute, University of São Paulo, São Paulo 05508-000, Brazil
- Cell and Molecular Therapy Center (NUCEL), School of Medicine, University of São Paulo, São Paulo 05360-130 SP, Brazil
| |
Collapse
|
127
|
Lang C, Dai Y, Wu Z, Yang Q, He S, Zhang X, Guo W, Lai Y, Du H, Wang H, Ren D, Peng X. SMAD3/SP1 complex-mediated constitutive active loop between lncRNA PCAT7 and TGF-β signaling promotes prostate cancer bone metastasis. Mol Oncol 2020; 14:808-828. [PMID: 31925912 PMCID: PMC7138406 DOI: 10.1002/1878-0261.12634] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 12/16/2019] [Accepted: 01/09/2020] [Indexed: 01/05/2023] Open
Abstract
Bone metastasis is associated with cancer-related death in patients with prostate cancer (PCa). Long noncoding RNAs (lncRNAs) play critical roles in tumor progression of PCa. Nevertheless, the biological function of lncRNAs in PCa bone metastasis remains unclear. PCAT7 was identified as a bone metastasis-related lncRNA via analyzing TCGA dataset. Meanwhile, PCAT7 was found to be elevated in primary PCa tissues with bone metastasis and associated with bone metastasis status and poor prognosis of patients with PCa. Functionally, our results reveal that PCAT7 overexpression promotes PCa bone metastasis in vivo, as well as migration, invasion, and EMT of PCa cells in vitro; on the contrary, PCAT7 knockdown has an inverse effect. Mechanistically, PCAT7 activates TGF-β/SMAD signaling by upregulating TGFBR1 expression via sponging miR-324-5p. In turn, TGF-β signaling forms a positive feedback loop with PCAT7 via SMAD3/SP1 complex-induced PCAT7 upregulation. Finally, the clinical positive correlation between PCAT7 and TGFBR1 and TGF-β signaling activity, and the negative association with miR-324-5p are further demonstrated in PCa tissues and clinical primary PCa cells. This study reveals a novel mechanism that is responsible for the constitutive activation of TGF-β signaling in PCa bone metastasis, implying that PCAT7 can act as a potential therapeutic target against bone metastasis of PCa via disrupting the constitutive active loop between PCAT7 and TGF-β signaling.
Collapse
Affiliation(s)
- Chuandong Lang
- Department of Orthopaedic SurgeryThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Orthopedics and TraumatologyGuangzhouChina
| | - Yuhu Dai
- Department of Orthopaedic SurgeryThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Orthopedics and TraumatologyGuangzhouChina
| | - Zhengquan Wu
- Department of Orthopaedic SurgeryThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Orthopedics and TraumatologyGuangzhouChina
| | - Qing Yang
- Department of Orthopaedic SurgeryThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Orthopedics and TraumatologyGuangzhouChina
| | - Shaofu He
- Department of RadiologyThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Xin Zhang
- Clinical Experimental CenterJiangmen Central HospitalAffiliated Jiangmen HospitalSun Yat‐sen UniversityJiangmenChina
| | - Wei Guo
- Department of Orthopaedic SurgeryThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Orthopedics and TraumatologyGuangzhouChina
| | - Yingrong Lai
- Department of PathologyThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Hong Du
- Department of PathologyThe First People’s Hospital of Guangzhou CityGuangzhouChina
| | - Hehe Wang
- Department of Medical LaboratoryWeifang Medical UniversityWeifangChina
| | - Dong Ren
- Department of Orthopaedic SurgeryThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Orthopedics and TraumatologyGuangzhouChina
| | - Xinsheng Peng
- Department of Orthopaedic SurgeryThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Orthopedics and TraumatologyGuangzhouChina
| |
Collapse
|
128
|
Jia L, Song Y, Mu L, Li Q, Tang J, Yang Z, Meng W. Long noncoding RNA TPT1-AS1 downregulates the microRNA-770-5p expression to inhibit glioma cell autophagy and promote proliferation through STMN1 upregulation. J Cell Physiol 2020; 235:3679-3689. [PMID: 31637705 DOI: 10.1002/jcp.29262] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 08/26/2019] [Indexed: 12/17/2022]
Abstract
Through the microarray analysis, long noncoding RNA TPT1-AS1 (TPT1-AS1) was identified in the development of glioma. However, the specific effect of TPT1-AS1 on glioma autophagy in the recent years has not fully been investigated. Therefore, the purpose of our present study is to investigate the function of TPT1-AS1 on affecting autophagy of glioma cells through regulation of microRNA-770-5p (miR-770-5p)-mediated stathmin 1 (STMN1). Initially, the expression of TPT1-AS1, miR-770-5p, and STMN1 were determined in glioma cell lines, followed by the prediction and validation of their interaction. After that, the effects of TPT1-AS1, miR-770-5p, and STMN1 on the in vitro glioma cell proliferation and autophagy were assessed using EdU assay and macrophage-derived chemokine (MDC) and on the in vivo tumor development and autophagy were evaluated using a nude mouse xenograft tumor assay and immunofluorescence assay. In comparison with the normal cells, the glioma cells displayed upregulated expression of TPT1-AS1 and STMN1, but a downregulated miR-770-5p expression. miR-770-5p, which directly targeted STMN1, could be downregulated by TPT1-AS1. Subsequently, in glioma cells, TPT1-AS1 can function to competitively bind to miR-770-5p, thus regulatEing STMN1 expression. Moreover, glioma cell proliferation and autophagy could be mediated through the TPT1-AS1/miR-770-5p/STMN1 axis. From our data we conclude an inhibitory function of TPT1-AS1 in glioma cell autophagy by downregulating miR-770-5p and upregulating STMN1, which may be instrumental for the therapeutic targeting and clinical management of glioma.
Collapse
Affiliation(s)
- Lei Jia
- Department of Neurosurgery, The 4th Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yuwen Song
- Department of Minimally Invasive Neurosurgery, The 4th Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Luyan Mu
- Department of Minimally Invasive Neurosurgery, The 4th Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qingla Li
- Department of Minimally Invasive Neurosurgery, The 4th Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiabin Tang
- Department of Minimally Invasive Neurosurgery, The 4th Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhao Yang
- Department of Minimally Invasive Neurosurgery, The 4th Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenjuan Meng
- Department of Comprehensive Archives, The 1st Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
129
|
Ye F, Xu R, Ge Y, Zheng Y, Liu X, Deng P, Xu X. LINC00963 Confers Oncogenic Properties in Glioma by Regulating the miR-506/BCAT1 Axis. Cancer Manag Res 2020; 12:2339-2351. [PMID: 32273770 PMCID: PMC7108718 DOI: 10.2147/cmar.s246332] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 03/16/2020] [Indexed: 12/21/2022] Open
Abstract
Background Glioma is a prevalent disease of the central nervous system with a high incidence and mortality rate. Many long noncoding RNAs (lncRNAs) have been determined to be critical regulators of glioma oncogenesis. However, the function and mechanism of LINC00963 in glioma have not been fully elucidated. Methods The expression level of RNA was determined by qRT-PCR, and the protein level was determined by Western blot analysis. A luciferase activity assay was conducted to verify the interaction between miRNA and lncRNA or the target gene. The proliferation, cell cycle distribution, invasion, and migration were evaluated by MTT, EdU, flow cytometry, wound-healing and Transwell invasion assays, respectively. In vivo tumor growth was evaluated in a xenograft nude mouse model. Results We found that LINC00963 was upregulated in glioma cells and tissues and associated with the poor prognosis of patients with glioma. Ectopic expression of LINC00963 promoted cell proliferation, cell cycle progression, migration, and invasion in vitro and tumorigenesis in vivo. Mechanistically, the results of luciferase activity and RNA pulldown assays validated that LINC00963 could act as a molecular sponge of miR-506. Reciprocal repression was found between LINC00963 and miR-506. In addition, BCAT1 was identified as a target of miR-506, and both the mRNA and protein levels of BCAT1 were reduced by miR-506. In tumor tissues, the expression of BCAT1 was negatively and positively correlated with miR-506 and LINC00963 expression, respectively. The reintroduction of BCAT1 in glioma cells abolished the tumor suppressive function of miR-506 by promoting cell viability and motility. The upregulated LINC00963 and BCAT1 were associated with the aggressive phenotypes of tumors. Conclusion Our data revealed that LINC00963 confers oncogenic function in the progression of glioma and that the LINC00963/miR-506/BCAT1 axis may be a novel mechanism and therapeutic strategy for this disease.
Collapse
Affiliation(s)
- Feng Ye
- Department of Neurosurgery, The Second People's Hospital of Chengdu, Chengdu, Sichuan 610021, People's Republic of China
| | - Ronghua Xu
- Department of Neurosurgery, The Second People's Hospital of Chengdu, Chengdu, Sichuan 610021, People's Republic of China
| | - Yuanhong Ge
- Department of Neurosurgery, The Second People's Hospital of Chengdu, Chengdu, Sichuan 610021, People's Republic of China
| | - Yi Zheng
- Department of Neurosurgery, The Second People's Hospital of Chengdu, Chengdu, Sichuan 610021, People's Republic of China
| | - Xiaowei Liu
- Department of Neurosurgery, The Second People's Hospital of Chengdu, Chengdu, Sichuan 610021, People's Republic of China
| | - Pingfu Deng
- Department of Neurosurgery, The Second People's Hospital of Chengdu, Chengdu, Sichuan 610021, People's Republic of China
| | - Xuejun Xu
- Department of Neurosurgery, The Second People's Hospital of Chengdu, Chengdu, Sichuan 610021, People's Republic of China
| |
Collapse
|
130
|
Ding C, Yi X, Wu X, Bu X, Wang D, Wu Z, Zhang G, Gu J, Kang D. Exosome-mediated transfer of circRNA CircNFIX enhances temozolomide resistance in glioma. Cancer Lett 2020; 479:1-12. [PMID: 32194140 DOI: 10.1016/j.canlet.2020.03.002] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/02/2020] [Accepted: 03/03/2020] [Indexed: 12/14/2022]
Abstract
Development of chemotherapy resistance remains a major obstacle for glioma management. Exosome-mediated transfer of circular RNAs (circRNAs) are being found to have relevance to many human cancers, including glioma. The purpose of this study is to explore the effect and underlying mechanism of exosomal circRNA nuclear factor I X (CircNFIX) on temozolomide (TMZ) chemoresistance in glioma. Our results indicated that exosomal CircNFIX was up-regulated in the serum of TMZ-resistant patients and predicted poor prognosis. Exosomal CircNFIX from TMZ-resistant cells conferred TMZ resistance to recipient sensitive cells through the enhancement of cell migration and invasion and the repression of cell apoptosis under TMZ exposure. CircNFIX directly interacted with miR-132 by binding to miR-132. CircNFIX knockdown enhanced TMZ sensitivity in resistant glioma cells by up-regulating miR-132. Additionally, exosomal CircNFIX promoted tumor growth and its depletion enhanced TMZ sensitivity in glioma cells in vivo. Taken together, our study suggests that exosome-mediated transfer of CircNFIX enhances TMZ resistance in glioma at least partially through sponging miR-132, highlighting a potentially prognostic biomarker and therapeutic target for improving the clinical benefits of TMZ treatment in patients with glioma.
Collapse
Affiliation(s)
- Chenyu Ding
- Department of Neurosurgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350001, Fujian, People's Republic of China
| | - Xuehan Yi
- Department of Otolaryngology Head and Neck Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, People's Republic of China
| | - Xiyue Wu
- Department of Neurosurgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350001, Fujian, People's Republic of China
| | - Xingyao Bu
- Department of Neurosurgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450000, Henan, People's Republic of China
| | - Desheng Wang
- Department of Otolaryngology Head and Neck Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, People's Republic of China
| | - Zanyi Wu
- Department of Neurosurgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350001, Fujian, People's Republic of China
| | - Gaoqi Zhang
- Department of Neurosurgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450000, Henan, People's Republic of China
| | - Jianjun Gu
- Department of Neurosurgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450000, Henan, People's Republic of China.
| | - Dezhi Kang
- Department of Neurosurgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350001, Fujian, People's Republic of China.
| |
Collapse
|
131
|
Huang J, Lu D, Xiang T, Wu X, Ge S, Wang Y, Wang J, Cheng N. MicroRNA-132-3p regulates cell proliferation, apoptosis, migration and invasion of liver cancer by targeting Sox4. Oncol Lett 2020; 19:3173-3180. [PMID: 32256813 PMCID: PMC7074496 DOI: 10.3892/ol.2020.11431] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 12/13/2019] [Indexed: 01/08/2023] Open
Abstract
The present study investigated whether microRNA (miR)-132-3p targeted transcription factor SOX-4 (Sox4) for the inhibition of proliferation, migration, invasion and promotion of apoptosis in liver cancer (LC) cells. The expression of miR132-3p and Sox4 mRNA was evaluated by quantitative PCR and protein expression was determined by western blot analysis. Cell proliferation, apoptosis, migration, and invasion were assessed at different time points by the MTT assay, flow cytometry analysis, wound healing assay and Transwell migration assay, respectively. Bioinformatics prediction and luciferase assays were performed to validate and confirm Sox4as a potential target of miR-132p. There was a reduced expression of miR-132-3p in HepG2 and Huh7 cell lines compared with HccLM3 cells. Overexpression of miR-132-3p resulted in significant inhibition of proliferation and induction of apoptosis in LC cells. Moreover, migration and invasion of HepG2 cells were suppressed by over expressing miR-132-3p. However, downregulation of miR-132-3p in Hep-G2 cells promoted cell growth, invasion and migration and inhibited apoptosis. Bioinformatics analysis predicted Sox4 as a potential target of miR-132-3p, which was further confirmed by the luciferase reporter assay. In addition, an inverse association was observed between miR-132-3p and Sox4 expression. miR-132-3p may regulate the proliferation, apoptosis, migration and invasion of HepG2 cells by targeting Sox4.
Collapse
Affiliation(s)
- Jiansheng Huang
- Department of Infectious Diseases, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Dudan Lu
- Department of Infectious Diseases, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Tianxin Xiang
- Department of Infectious Diseases, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xiaoping Wu
- Department of Infectious Diseases, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Shanfei Ge
- Department of Infectious Diseases, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yue Wang
- Department of Infectious Diseases, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jiaxin Wang
- Department of Infectious Diseases, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Na Cheng
- Department of Infectious Diseases, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
132
|
Cheng M, Zhang ZW, Ji XH, Xu Y, Bian E, Zhao B. Super-enhancers: A new frontier for glioma treatment. Biochim Biophys Acta Rev Cancer 2020; 1873:188353. [PMID: 32112817 DOI: 10.1016/j.bbcan.2020.188353] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/21/2020] [Accepted: 02/21/2020] [Indexed: 01/17/2023]
Abstract
Glioma is the most common primary malignant tumor in the human brain. Although there are a variety of treatments, such as surgery, radiation and chemotherapy, glioma is still an incurable disease. Super-enhancers (SEs) are implicated in the control of tumor cell identity, and they promote oncogenic transcription, which supports tumor cells. Inhibition of the SE complex, which is required for the assembly and maintenance of SEs, may repress oncogenic transcription and impede tumor growth. In this review, we discuss the unique characteristics of SEs compared to typical enhancers, and we summarize the recent advances in the understanding of their properties and biological role in gene regulation. Additionally, we highlight that SE-driven lncRNAs, miRNAs and genes are involved in the malignant phenotype of glioma. Most importantly, the application of SE inhibitors in different cancer subtypes has introduced new directions in glioma treatment.
Collapse
Affiliation(s)
- Meng Cheng
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei 230601, China
| | - Zheng Wei Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei 230601, China
| | - Xing Hu Ji
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei 230601, China
| | - Yadi Xu
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei 230601, China
| | - Erbao Bian
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei 230601, China.
| | - Bing Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei 230601, China.
| |
Collapse
|
133
|
Ding J, Zhang L, Chen S, Cao H, Xu C, Wang X. lncRNA CCAT2 Enhanced Resistance of Glioma Cells Against Chemodrugs by Disturbing the Normal Function of miR-424. Onco Targets Ther 2020; 13:1431-1445. [PMID: 32110042 PMCID: PMC7034969 DOI: 10.2147/ott.s227831] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 11/28/2019] [Indexed: 12/14/2022] Open
Abstract
Background Aggressive metastasis of tumor cells assumed a constructive role in strengthening chemoresistance of tumors, so this investigation was intended to elucidate if lncRNA CCAT2 sponging downstream miR-424 regulated chemotolerance of glioma cells by boosting metastasis of glioma cells. Methods One hundred and twenty-eight pairs of glioma tissues and corresponding adjacent tissues were resected from glioma patients during their operation, and we also purchased a series of glioma cell lines, including U251, U87, A172 and SHG44. Furthermore, pcDNA3.1-CCAT2, si-CCAT2, miR-424 mimic and miR-424 inhibitor were transfected into SHG44 and U251 cell lines, so as to evaluate impacts of CCAT2 and miR-424 on chemosensitivity of the glioma cells. Besides, proliferation, invasion and metastasis of the cells were determined through the implementation of colony formation assay, transwell assay and scratch assay. Results Glioma tissues and cells were monitored with higher CCAT2 expression and lower miR-424 expression than adjacent normal tissues and NHA cell line (P<0.05). Among the glioma cell lines, the SHG44 cell line showed the strongest resistance against teniposide, temozolomide and cisplatin (P<0.05), whereas the U251 cell line was more sensitive to teniposide, temozolomide, vincristine and cisplatin than any other cell line (P<0.05). Besides, pcDNA3.1-CCAT2 and miR-424 inhibitor could enhance tolerance of glioma cell lines against drugs (P<0.05). Moreover, in-vitro transfection of si-CCAT2 and miR-424 mimic could significantly retard proliferation, invasion and migration of SHG44 and U251 cells (P<0.05), and CCAT2 was found to negatively regulate miR-424 expression by sponging it (P<0.05). In addition, CHK1 was deemed as the molecule targeted by upstream miR-424, and its overexpression can changeover the effects of miR-424 mimic on proliferation and metastasis of SHG44 and U251 cells. Conclusion lncRNA CCAT2/miR-424/Chk1 axis might serve as a promising target for improving chemotherapeutic efficacies in glioma treatment.
Collapse
Affiliation(s)
- Jun Ding
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Lin Zhang
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Shiwen Chen
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Heli Cao
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Chen Xu
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Xuyang Wang
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| |
Collapse
|
134
|
Yao P, Li YL, Chen Y, Shen W, Wu KY, Xu WH. Overexpression of long non-coding RNA Rian attenuates cell apoptosis from cerebral ischemia-reperfusion injury via Rian/miR-144-3p/GATA3 signaling. Gene 2020; 737:144411. [PMID: 32006596 DOI: 10.1016/j.gene.2020.144411] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/14/2020] [Accepted: 01/27/2020] [Indexed: 12/31/2022]
Abstract
Long non-coding RNAs (lncRNAs) have been identified in cerebral ischemia-reperfusion (I/R) injury nowadays. Herein, we uncovered the function and underlying mechanism of the lncRNA Rian in cerebral I/R injury. The oxygen-glucose deprivation model in N2a cells was offered to mimic cerebral I/R injury in vitro. Trypan blue staining, reactive oxygen species (ROS) production, and caspase-3 activity were used to evaluate cell apoptosis. Then, middle cerebral artery occlusion was conducted to evaluate the function of lncRNA Rian in mice. Real-time PCR and western blotting were performed to determine the expression of lncRNA Rian, miR-144-3p, GATA binding protein 3 (GATA3), caspase-3, Bax, and Bcl-2. The results showed that both Rian and GATA3 were downregulated, and miR-144-3p was upregulated in cerebral I/R injury in vitro and in vivo. Overexpression of Rian could inhibit the cell apoptosis induced by oxygen-glucose deprivation. Furthermore, overexpression of Rian distinctly reduced the infarct size, and it also improved the neurological score. Overexpression of Rian could abolish miR-144-3p-mediated I/R injury in vitro and in vivo. Besides, GATA3 was the target of miR-144-3p and GATA3 could be regulated co-operatively by miR-144-3p and Rian. Consequently, these findings showed that the Rian/miR-144-3p/GATA3 axis is an essential signaling in cerebral I/R injury. The lncRNA Rian may serve as a potential target for novel treatment in patients with ischemic stroke.
Collapse
Affiliation(s)
- Peng Yao
- Department of Intensive Care Unit (ICU), Xiaogan Central Hospital, Wuhan University of Science and Technology, No. 6 Square Road, South District, Xiaogan, Hubei Province 432000, China
| | - Yi-Ling Li
- Department of Anesthesiology, Xiaogan Central Hospital, Wuhan University of Science and Technology, No. 6 Square Road, South District, Xiaogan, Hubei Province 432000, China
| | - Yong Chen
- Department of Anesthesiology, The Second Affiliated Medicine of Nanchang University, No. 1 Mingde Road, Nanchang, Jiangxi Province 330000, China
| | - Wei Shen
- Department of Intensive Care Unit (ICU), Xiaogan Central Hospital, Wuhan University of Science and Technology, No. 6 Square Road, South District, Xiaogan, Hubei Province 432000, China
| | - Ke-Yan Wu
- Department of Intensive Care Unit (ICU), Xiaogan Central Hospital, Wuhan University of Science and Technology, No. 6 Square Road, South District, Xiaogan, Hubei Province 432000, China
| | - Wen-Hao Xu
- Department of Intensive Care Unit (ICU), Xiaogan Central Hospital, Wuhan University of Science and Technology, No. 6 Square Road, South District, Xiaogan, Hubei Province 432000, China.
| |
Collapse
|
135
|
Long noncoding RNA SNHG17 induced by YY1 facilitates the glioma progression through targeting miR-506-3p/CTNNB1 axis to activate Wnt/β-catenin signaling pathway. Cancer Cell Int 2020; 20:29. [PMID: 32009853 PMCID: PMC6988207 DOI: 10.1186/s12935-019-1088-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 12/23/2019] [Indexed: 12/19/2022] Open
Abstract
Background Glioma is one of the most widely diagnosed malignancies worldwide. It has been reported that long noncoding RNAs (lncRNAs) are participators in the tumorgenesis of cancers. Nevertheless, the role and function of lncRNA SNHG17 among glioma is unclear. Methods RT-qPCR revealed SNHG17, YY1, miR-506-3p, CTNNB1 expression among glioma cells. CCK-8, colony formation, EdU, flow cytometry, TUNEL and western blot assays revealed the function of SNHG17 in glioma. RIP uncovered SNHG17, miR-506-3p and CTNNB1 enrichment in RISC complex. Luciferase reporter assays and RNA pull down revealed interaction of miR-506-3p with SNHG17 and CTNNB1. Results SNHG17 expression was up-regulated in glioma tissues and cells. SNHG17 silence attenuated cell proliferation and promoted apoptosis and repressed tumor growth. Moreover, SNHG17 was up-regulated by transcription factor YY1. Mechanistically, SNHG17 activated Wnt/β-catenin signaling pathway in glioma. CTNNB1 was referred to as the mRNA of β-catenin, we validated that SNHG17 bound to miR-506-3p to induce CTNNB1 and activate Wnt/β-catenin signaling pathway. Rescue experiments indicated that CTNNB1 overexpression abolished the inhibitory effects of SNHG7 inhibition on glioma progression. Conclusions The findings that YY1-induced SNHG17 facilitated the glioma progression through targeting miR-506-3p/CTNNB1 axis to activate Wnt/β-catenin signaling pathway offered a brand-new prospects to molecular-targeted treatment for glioma.
Collapse
|
136
|
Li F, Li F, Chen W. Propofol Inhibits Cell Proliferation, Migration, and Invasion via mir-410-3p/Transforming Growth Factor-β Receptor Type 2 (TGFBR2) Axis in Glioma. Med Sci Monit 2020; 26:e919523. [PMID: 31960827 PMCID: PMC6993559 DOI: 10.12659/msm.919523] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Propofol is a common intravenous anesthetic used to induce and maintain anesthesia. Numerous studies have reported that propofol plays an anti-tumor role in diverse human cancers, including glioma. In this research, we explored the roles of propofol and its related molecular mechanisms in glioma. MATERIAL AND METHODS U251 and A172 cells were exposed to different doses of propofol for 24 h. Cell proliferation, migration, and invasion in glioma were evaluated using MTT assay and Transwell assay, respectively. The levels of microRNA-410-3p (miR-410-3p) and transforming growth factor-ß receptor type 2 (TGFBR2) were detected by quantitative real-time polymerase chain reaction (qRT-PCR) assay and Western blot assay, respectively. The association between miR-410-3p and TGFBR2 was predicted by TargetScan and confirmed by dual-luciferase reporter assay. RESULTS Propofol inhibited the proliferation, migration, and invasion of glioma cells in a concentration-dependent way. miR-410-3p was induced and TGFBR2 was inhibited by different concentrations of propofol treatment. Moreover, TGFBR2 was confirmed to be a target gene of miR-410-3p and TGFBR2 was inversely modulated by miR-410-3p in glioma cells. Depletion of miR-410-3p reversed the inhibition of propofol treatment on U251 and A172 cell growth and metastasis, but the effects were further abolished by knocking down the expression of TGFBR2. CONCLUSIONS Propofol can suppress cell growth and metastasis by regulating the miR-410-3p/TGFBR2 axis in glioma.
Collapse
Affiliation(s)
- Fengli Li
- Department of Anesthesiology, Linyi Central Hospital, Linyi, Shandong, China (mainland)
| | - Fengliang Li
- Department of Psychiatry, Third Hospital of Weifang, Weifang, Shandong, China (mainland)
| | - Wei Chen
- Department of Neurosurgery, Linyi Central Hospital, Linyi, Shandong, China (mainland)
| |
Collapse
|
137
|
Song LR, Li D, Weng JC, Li CB, Wang L, Wu Z, Zhang JT. MicroRNA-195 Functions as a Tumor Suppressor by Directly Targeting Fatty Acid Synthase in Malignant Meningioma. World Neurosurg 2020; 136:e355-e364. [PMID: 31927122 DOI: 10.1016/j.wneu.2019.12.182] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/30/2019] [Accepted: 12/31/2019] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Meningiomas are among the most common primary intracranial tumors. Up to 20% of cases will show increased malignancy at histological examination (World Health Organization grade II or III). Effective pharmacotherapy, except for radiotherapy, is lacking. Therefore, it is necessary to study the pathogenesis of malignant meningioma to provide more treatment strategies. METHODS RNA sequencing and micro-RNA (miRNA) microarray detection were applied to identify differentially expressed messenger RNAs (mRNAs) and miRNAs in benign and malignant meningioma. The miRDB and TargetScan databases were used to predict the potential interaction between miRNAs and mRNAs. A proliferation assay was used to evaluate the cell growth. A wound healing assay and Transwell assay were performed to assess the cell migration and invasion abilities, respectively. The interaction between miRNA and mRNA was identified using a luciferase reporter assay. RESULTS We found fatty acid synthase (FASN) was significantly upregulated in malignant meningioma compared with benign meningioma. Knockdown of FASN significantly inhibited proliferation, migration, and invasion of IOMM-Lee cells. Moreover, miR-195 was verified to directly target FASN using a luciferase reporter assay. Upregulation of miR-195 also significantly inhibited proliferation, migration, and invasion of IOMM-Lee cells. Furthermore, we performed bioinformatics analysis to predict the competing endogenous RNAs (ceRNAs) and found that NUP210, SPIRE2, SLC7A1, and DMTN might function as ceRNAs of FASN by sponging miR-195 in meningioma. CONCLUSIONS Our results have suggested a tumor suppressive role for miR-195 in the tumorigenesis and progression of malignant meningioma by targeting FASN. In addition, NUP210, SPIRE2, SLC7A1, and DMTN might act as ceRNAs to regulate FASN expression by sponging miR-195.
Collapse
Affiliation(s)
- Lai-Rong Song
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Department of Neuro-Oncology, China National Clinical Research Center for Neurological Diseases, Beijing, China; Department of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China; Department of Cancer Biology, Beijing Key Laboratory of Brain Tumor, Beijing, China
| | - Da Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Department of Neuro-Oncology, China National Clinical Research Center for Neurological Diseases, Beijing, China; Department of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China; Department of Cancer Biology, Beijing Key Laboratory of Brain Tumor, Beijing, China
| | - Jian-Cong Weng
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Department of Neuro-Oncology, China National Clinical Research Center for Neurological Diseases, Beijing, China; Department of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China; Department of Cancer Biology, Beijing Key Laboratory of Brain Tumor, Beijing, China
| | - Cheng-Bei Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Department of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China; Department of Cancer Biology, Beijing Key Laboratory of Brain Tumor, Beijing, China
| | - Liang Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Department of Neuro-Oncology, China National Clinical Research Center for Neurological Diseases, Beijing, China; Department of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China; Department of Cancer Biology, Beijing Key Laboratory of Brain Tumor, Beijing, China
| | - Zhen Wu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Department of Neuro-Oncology, China National Clinical Research Center for Neurological Diseases, Beijing, China; Department of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China; Department of Cancer Biology, Beijing Key Laboratory of Brain Tumor, Beijing, China
| | - Jun-Ting Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Department of Neuro-Oncology, China National Clinical Research Center for Neurological Diseases, Beijing, China; Department of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China; Department of Cancer Biology, Beijing Key Laboratory of Brain Tumor, Beijing, China.
| |
Collapse
|
138
|
Lv T, Miao Y, Xu T, Sun W, Sang Y, Jia F, Zhang X. Circ-EPB41L5 regulates the host gene EPB41L5 via sponging miR-19a to repress glioblastoma tumorigenesis. Aging (Albany NY) 2020; 12:318-339. [PMID: 31905344 PMCID: PMC6977680 DOI: 10.18632/aging.102617] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 12/05/2019] [Indexed: 12/14/2022]
Abstract
Background: Circular RNAs (circRNAs) are widely expressed non-coding RNAs in eukaryotic cells, involved in regulating tumorigenesis of several types of cancers. However, the expression profiles and the precise functional role in glioblastoma remain unclear. Results: Circ-EPB41L5 was downregulated in glioblastoma tissues and cell lines compared to the normal brain tissues and cell lines. Low circ-EPB41L5 expression was correlated to the poor prognosis of glioblastoma patients, while the overexpression inhibited proliferation, clone formation, migration, and invasion abilities of glioma cells, and the suppression had counter effects. Furthermore, RNA-seq results determined that the host gene was the target gene of circ-EPB41L5, which served as a sponge against miR-19a and inhibited miR-19a activity from upregulating the expression of EPB41L5. Finally, we found that circ-EPB41L5 regulated the RhoC expression and phosphorylation of AKT through EPB41L5. Conclusion: The current study highlights a novel suppressive function of circ-EPB41L5 and reveals that circ-EPB41L5/miR-19a/EPB41L5/p-AKT regulatory axis plays a striking role in the progression of glioblastoma, which provides a novel insight into the mechanisms underlying glioblastoma. Methods: The expression profiles of circRNAs in glioblastoma were determined by Illumina HiSeq from six glioblastoma tissues and six normal brain tissues. Then, the correlation between circ-EPB41L5 expression and clinical features and the survival time of 45 glioblastoma patients was detected. The interaction between circ-EPB41L5, miR-19a, and EPB41L5 was assessed by luciferase reporter and RNA pull-down assays. The effects of expression of the ectopic intervention of circ-EPB41L5 or EPB41L5 on proliferation, clone formation, migration, and invasion in vitro and tumorigenesis in vivo were observed to evaluate the function of circ-EPB41L5 or EPB41L5.
Collapse
Affiliation(s)
- Tao Lv
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yifeng Miao
- Department of Neurosurgery, Ren Ji Hospital South Campus, School of Medicine, Shanghai Jiao Tong University, Shanghai 201112, China
| | - Tianqi Xu
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Wenhua Sun
- Department of Neurosurgery, Ren Ji Hospital South Campus, School of Medicine, Shanghai Jiao Tong University, Shanghai 201112, China
| | - Youzhou Sang
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Feng Jia
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xiaohua Zhang
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
139
|
Kong B, Li M, Gao B, Han B, Zhao W, Wang F. Retracted Article: Overexpression of circ_0034642 contributes to hypoxia-induced glycolysis, cell proliferation, migration and invasion in gliomas by facilitating TAGLN2 expression via sponging miR-625-5p. RSC Adv 2020; 10:897-908. [PMID: 35494452 PMCID: PMC9048227 DOI: 10.1039/c9ra08600e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 12/17/2019] [Indexed: 11/21/2022] Open
Abstract
Glioma is an aggressive brain cancer with poor prognosis and high invasiveness. Dysregulation of circular RNAs (circRNAs) has been widely discovered in various cancers, including glioma. However, the molecular mechanism of circ_0034642 in glioma is still unclear. The expression of circ_0034642, microRNA (miR)-625-5p and transgelin-2 (TAGLN2) in glioma tumors and cells was detected by performing a quantitative real-time polymerase chain reaction (qRT-PCR). The stability of circ_0034642 was determined by carrying out RNase R treatment. Cell proliferation was evaluated by performing the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay. Glycolysis was analyzed by measuring the extracellular acidification rate (ECAR) using glucose detection and lactic acid detection kits. Cell migration and invasion were determined by performing the transwell assay. Protein expression levels of the proteins hexokinase 2 (HK2), matrix metalloproteinase-2 (MMP2), matrix metalloproteinase-9 (MMP9) and TAGLN2 were analyzed using western blots. The interaction between miR-625-5p and circ_0034642 or TAGLN2 was proved using a dual-luciferase reporter system. Animal models were established by subcutaneously injecting glioma cells stably transfected with sh-NC or sh-circ_0034642. Circ_0034642 and TAGLN2 were overexpressed whereas miR-625-5p was expressed at low levels in glioma tumors and cells. Moreover, circ_0034642 and TAGLN2 were upregulated while miR-625-5p was downregulated under hypoxic conditions in a time-dependent manner. Next, elimination of circ_0034642 was shown to inhibit cell glycolysis, proliferation, migration and invasion under hypoxic conditions in gliomas. Then, we found that circ_0034642 acted as a “sponge” of miR-625-5p while TAGLN2 acted as a target of miR-625-5p. In addition, recovery of circ_0034642 attenuated the repression mediated by miR-625-5p on glioma cell glycolysis and progression under hypoxic conditions. Meanwhile, an inhibitor of miR-625-5p alleviated TAGLN2 deficiency-induced inhibition of glioma cell development under hypoxic conditions. We also discovered that circ_0034642 could interact with miR-625-5p and further alter the expression of TAGLN2. Lastly, a circ_0034642 knockdown hindered tumor growth in vivo by regulating the miR-625-5p/TAGLN2 axis. Enhanced expression of circ_0034642 was found to promote cell glycolysis, proliferation, migration and invasion under hypoxic conditions in gliomas by sponging miR-625-5p to improve TAGLN2 expression, providing prospective biomarkers for the diagnosis of glioma. Circ_0034642 was upregulated under hypoxic conditions in gliomas.![]()
Collapse
Affiliation(s)
- Bo Kong
- Department of Neurosurgery
- Affiliated Hospital of Jining Medical University
- Jining
- China
| | - Mingxuan Li
- Department of Neurosurgery
- Affiliated Hospital of Jining Medical University
- Jining
- China
| | - Bo Gao
- Department of Neurosurgery
- Affiliated Hospital of Jining Medical University
- Jining
- China
| | - Bin Han
- Department of Neurosurgery
- Affiliated Hospital of Jining Medical University
- Jining
- China
| | - Wanju Zhao
- Department of Neurosurgery
- Affiliated Hospital of Jining Medical University
- Jining
- China
| | - Fujun Wang
- Department of Neurosurgery
- Affiliated Hospital of Jining Medical University
- Jining
- China
| |
Collapse
|
140
|
Li X, Liu Q, Wang K, Luo W, Liang T, Yuan S, Zhen Y, Yan D. Retracted Article: LncRNA SNHG5 regulates the cell viability and apoptosis of glioma cells by the miR-1297/KPNA2 axis. RSC Adv 2020; 10:1498-1506. [PMID: 35494689 PMCID: PMC9048252 DOI: 10.1039/c9ra08693e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 12/28/2019] [Indexed: 12/27/2022] Open
Abstract
Long non-coding RNA small nucleolar RNA host gene 5 (lncRNA SNHG5) has been reported to participate in the occurrence and development of glioma. However, the function and underlying molecular mechanisms of SNHG5 in glioma remain largely unknown. The expressions of SNHG5, microRNA-1297 (miR-1297) and karyopherin subunit alpha 2 (KPNA2) in glioma tissues and cells were evaluated by quantitative reverse transcription-polymerase chain reaction (qRT-PCR) or western blot. 3-(4,5-Dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) assay and flow cytometry were used to detect cell viability and apoptosis, respectively. Western blot was also performed to detect the expressions of autophagy-associated proteins. The relationship among lncRNA SNHG5, miR-1297 and KPNA2 was verified by luciferase reporter assay and RNA immunoprecipitation (RIP) assay. SNHG5 and KPNA2 were over expressed, and the level of miR-1297 was down-regulated in glioma tissues and cell lines. Knockdown of SHNG5 promoted apoptosis, while suppressing cell viability and autophagy of A172 and LN340 cells. Meanwhile, SHNG5 harbored the binding sites with miR-1297, and a negative correlation between the expression of SNHG5 and miR-1297 in glioma tissues was also observed. Interestingly, silencing of miR-1297 undermined the SHNG5 depletion-mediated effect on cell viability, apoptosis, and autophagy. KPNA2 was a direct target of miR-1297, and negatively regulated by miR-1297. More importantly, gain of KPNA2 mitigated the effect of SHNG5l knockdown on glioma cells. Silencing of SNHG5 had an implication in inhibiting apoptosis and stimulating cell viability and autophagy by the miR-1297/KPNA2 axis in glioma. Long non-coding RNA small nucleolar RNA host gene 5 (lncRNA SNHG5) has been reported to participate in the occurrence and development of glioma.![]()
Collapse
Affiliation(s)
- Xueyuan Li
- Department of Neurosurgery
- The First Affiliated Hospital of Zhengzhou University
- Zhengzhou City 450000
- China
| | - Qiankun Liu
- Department of Neurosurgery
- The First Affiliated Hospital of Zhengzhou University
- Zhengzhou City 450000
- China
| | - Kang Wang
- Department of Neurosurgery
- The First Affiliated Hospital of Zhengzhou University
- Zhengzhou City 450000
- China
| | - Wenzheng Luo
- Department of Neurosurgery
- The First Affiliated Hospital of Zhengzhou University
- Zhengzhou City 450000
- China
| | - Tiansong Liang
- Department of Radiotherapy
- The First Affiliated Hospital of Zhengzhou University
- Zhengzhou
- China
| | - Shanpeng Yuan
- Department of Neurosurgery
- The First Affiliated Hospital of Zhengzhou University
- Zhengzhou City 450000
- China
| | - Yingwei Zhen
- Department of Neurosurgery
- The First Affiliated Hospital of Zhengzhou University
- Zhengzhou City 450000
- China
| | - Dongming Yan
- Department of Neurosurgery
- The First Affiliated Hospital of Zhengzhou University
- Zhengzhou City 450000
- China
| |
Collapse
|
141
|
Meng X, Deng Y, Lv Z, Liu C, Guo Z, Li Y, Liu H, Xie B, Jin Z, Lin F, Zhu H. LncRNA SNHG5 Promotes Proliferation of Glioma by Regulating miR-205-5p/ZEB2 Axis. Onco Targets Ther 2019; 12:11487-11496. [PMID: 31920337 PMCID: PMC6939796 DOI: 10.2147/ott.s228439] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 12/03/2019] [Indexed: 12/21/2022] Open
Abstract
Background Glioma is a common primary brain tumor with extremely poor prognosis outcomes. Increasing evidences have proved the relation between lncRNAs and glioma onset and progression. LncRNA SNHG5 involves in the biological activities of tumor cells, such as proliferation, migration and metastasis. Nevertheless, it is still necessary to explain the molecular mechanism and biofunction of SNHG5 in glioma. Materials and methods Quantitative real-time PCR (qRT-PCR) was performed to analyze expressions of SNHG5, miR-205-5p and ZEB2 in tumor tissues and cell lines. The cell counting kit-8 (CCK-8) assay, plate and soft agar colony formation assays were performed to evaluate cell proliferation ability. RNA immunoprecipitation assay and dual-luciferase reporter assay were used to confirm the interaction among SNHG5, miR-205-5p and ZEB2. The protein level of ZEB2 was measured by Western blot. Results Based on our findings, compared with normal tissues, the elevated expression of SNHG5 and decreased expression of miR-205-5p were observed in glioma tissues. The downregulation of SNHG5 exerted an obvious inhibitory effect on glioma cells in terms of their proliferation. With regard to the underlying mechanism, SNHG5 presented a direct inhibitory influence on miR-205-5p which targeted to the 3'-UTR region of zinc finger E-box binding homeobox 2 (ZEB2) mRNA. As a competing endogenous RNA (ceRNA), SNHG5 sponged miR-205-5p, regulating the expression of ZEB2 thereby. Conclusion These discoveries indicate that SNHG5 promotes proliferation of glioma by regulating miR-205-5p/ZEB2 axis.
Collapse
Affiliation(s)
- Xiangrui Meng
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Yanyao Deng
- Department of Neurology, The First Hospital of Changsha, Changsha, Hunan, People's Republic of China
| | - Zhicheng Lv
- Department of Neurosurgery, Chenzhou First People's Hospital, Chenzhou, Hunan, People's Republic of China
| | - Chao Liu
- Department of Neurology, The First Hospital of Changsha, Changsha, Hunan, People's Republic of China
| | - Ziqing Guo
- Department of Neurology, The First Hospital of Changsha, Changsha, Hunan, People's Republic of China
| | - Yuan Li
- Department of Neurology, The First Hospital of Changsha, Changsha, Hunan, People's Republic of China
| | - Hua Liu
- Department of Neurology, The First Hospital of Changsha, Changsha, Hunan, People's Republic of China
| | - Bing Xie
- Department of Neurology, The First Hospital of Changsha, Changsha, Hunan, People's Republic of China
| | - Ziqi Jin
- Department of Neurology, The First Hospital of Changsha, Changsha, Hunan, People's Republic of China
| | - Fangbo Lin
- Department of Neurology, The First Hospital of Changsha, Changsha, Hunan, People's Republic of China
| | - Hongwei Zhu
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| |
Collapse
|
142
|
Zhang M, Wang X, Yao J, Qiu Z. Long non-coding RNA NEAT1 inhibits oxidative stress-induced vascular endothelial cell injury by activating the miR-181d-5p/CDKN3 axis. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:3129-3137. [PMID: 31352814 DOI: 10.1080/21691401.2019.1646264] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Endothelial cell (EC) dysfunction induces atherosclerotic coronary heart disease (CHD) development. Recent studies demonstrated that lncRNA NEAT1 mediates multiple biological functions of cells. How NEAT1 regulates EC function is still unclear, so this study explored the role and mechanism of NEAT1 in oxidative stress-induced ECs. The levels of NEAT1 and miR-181d-5p were measured in serum samples from ApoE-/- mice and t-BHP-treated human umbilical vein endothelial cells (HUVECs) by qRT-PCR. The potential role of NEAT1 in viability, migration and apoptosis was analyzed by CCK-8, cell metastasis, flow cytometry, dual-luciferase reporter, RNA immunoprecipitation and Western blot assays using HUVECs overexpressing NEAT1. The expression of NEAT1 was increased, but miR-181d-5p expression was decreased in serum samples from both ApoE-/- mice and t-BHP-treated HUVECs. Overexpression of NEAT1 increased viability, migration and CDKN3 expression but decreased apoptotic rates, caspase-3 activity and miR-181d-5p expression in HUVECs. In addition, NEAT1 acted as a promoter of the proangiogenic capacity of HUVECs by targeting miR-181d-5p/CDKN3. Altogether, these findings indicate that NEAT1 may exert a protective effect on HUVECs by regulating the miR-181d-5p/CDKN3A axis.
Collapse
Affiliation(s)
- Min Zhang
- a Division of Cardiology, TongRen Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Xueting Wang
- a Division of Cardiology, TongRen Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Jing Yao
- b Division of Cardiology, Huadong Hospital affiliated with Fudan University , Shanghai , China
| | - Zhaohui Qiu
- a Division of Cardiology, TongRen Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , China
| |
Collapse
|
143
|
Li H, Shen S, Ruan X, Liu X, Zheng J, Liu Y, Yang C, Wang D, Liu L, Ma J, Ma T, Wang P, Cai H, Li Z, Zhao L, Xue Y. Biosynthetic CircRNA_001160 induced by PTBP1 regulates the permeability of BTB via the CircRNA_001160/miR-195-5p/ETV1 axis. Cell Death Dis 2019; 10:960. [PMID: 31862871 PMCID: PMC6925104 DOI: 10.1038/s41419-019-2191-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 11/30/2019] [Accepted: 12/02/2019] [Indexed: 02/07/2023]
Abstract
The presence of the blood-tumor barrier (BTB) severely impedes the transport of anti-neoplasm drugs to the central nervous system, affecting the therapeutic effects of glioma. Glioma endothelial cells (GECs) are the main structural basis of the BTB. Circular RNA is considered to be an important regulator of endothelial cell growth. In this study, we found that polypyrimidine tract binding protein 1 (PTBP1) and circRNA_001160 were remarkably upregulated in GECs. Knockdown of PTBP1 or circRNA_001160 significantly increased BTB permeability, respectively. As a molecular sponge of miR-195-5p, circRNA_001160 attenuated its negative regulation of the target gene ETV1 by adsorbing miR-195-5p. In addition, ETV1 was overexpression in GECs. ETV1 bounded to the promoter regions of tight junction-related proteins and increased the promoter activities, which significantly promoted the expression levels of tight junction-related proteins. The present study showed that the combined application of PTBP1, circRNA_001160, and miR-195-5p with the anti-tumor drug Dox effectively promoted Dox through BTB and extremely induced the apoptosis of glioma cells. Our results demonstrated that the PTBP1/circRNA_001160/miR-195-5p/ETV1 axis was critical in the regulation of BTB permeability and provided new targets for the treatment of glioma.
Collapse
Affiliation(s)
- Hua Li
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, People's Republic of China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, People's Republic of China
| | - Shuyuan Shen
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, People's Republic of China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, People's Republic of China
| | - Xuelei Ruan
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, People's Republic of China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, People's Republic of China
| | - Xiaobai Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, 110004, People's Republic of China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, People's Republic of China
| | - Jian Zheng
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, 110004, People's Republic of China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, People's Republic of China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, 110004, People's Republic of China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, People's Republic of China
| | - Chunqing Yang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, 110004, People's Republic of China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, People's Republic of China
| | - Di Wang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, 110004, People's Republic of China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, People's Republic of China
| | - Libo Liu
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, People's Republic of China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, People's Republic of China
| | - Jun Ma
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, People's Republic of China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, People's Republic of China
| | - Teng Ma
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, People's Republic of China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, People's Republic of China
| | - Ping Wang
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, People's Republic of China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, People's Republic of China
| | - Heng Cai
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, 110004, People's Republic of China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, People's Republic of China
| | - Zhen Li
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, 110004, People's Republic of China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, People's Republic of China
| | - Lini Zhao
- Department of pharmacology, Shenyang Medical College, Shenyang, 110034, People's Republic of China
| | - Yixue Xue
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, 110122, People's Republic of China. .,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, People's Republic of China. .,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, People's Republic of China.
| |
Collapse
|
144
|
Wang S, Cui Z, Li H, Li J, Lv X, Yang Z, Gao M, Bi Y, Zhang Z, Zhou B, Yin Z. LncRNA NEAT1 polymorphisms and lung cancer susceptibility in a Chinese Northeast Han Population: A case-control study. Pathol Res Pract 2019; 215:152723. [DOI: 10.1016/j.prp.2019.152723] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 10/14/2019] [Accepted: 10/26/2019] [Indexed: 01/23/2023]
|
145
|
LncRNA NEAT1 promotes endometrial cancer cell proliferation, migration and invasion by regulating the miR-144-3p/EZH2 axis. Radiol Oncol 2019; 53:434-442. [PMID: 31747378 PMCID: PMC6884930 DOI: 10.2478/raon-2019-0051] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 09/21/2019] [Indexed: 12/21/2022] Open
Abstract
Background Endometrial cancer (EC) is one of the most common gynaecological tumours in the worldwide. Long non-coding RNA (lncRNA) nuclear enriched abundant transcript 1 (NEAT1) promotes cell proliferation, migration and invasion in EC cells. However, the molecular mechanisms of NEAT1 in EC have not been fully clarified. We conducted this study to reveal the function of NEAT1 in EC tissues and cell lines. Materials and methods Cancer and adjacent tissues were collected from EC patients. HEC-1A and Ishikawa cells were cultured in vitro. NEAT1 expression was downregulated by transfecting small hairpin RNA (shRNA) and miR-144-3p was overexpressed by transfecting miR-144-3p mimics. Cell proliferation was detected by MTT assay and colony formation assay. Cell migration and invasion abilities were assessed by transwell assay. A dual-luciferase reporter assay was used to verify the relationship among NEAT1, EZH2, and miR-144-3p. The expression level of EZH2 was measured by Western blot and qPCR. Results NEAT1 was highly expressed in EC tissues and cells. Knockdown of NEAT1 inhibited the proliferation, migration and invasion of EC cells. Additionally, NEAT1 acted as a ceRNA of miR-144-3p, leading to EZH2 upregulation. Overexpression of miR-144-3p suppressed the proliferation and invasion of EC cells. Conclusions NEAT1 promotes EC cells proliferation and invasion by regulating the miR-144-3p/EZH2 axis.
Collapse
|
146
|
Ge X, Xu B, Xu W, Xia L, Xu Z, Shen L, Peng W, Huang S. Long noncoding RNA GAS5 inhibits cell proliferation and fibrosis in diabetic nephropathy by sponging miR-221 and modulating SIRT1 expression. Aging (Albany NY) 2019; 11:8745-8759. [PMID: 31631065 PMCID: PMC6834398 DOI: 10.18632/aging.102249] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 09/02/2019] [Indexed: 12/25/2022]
Abstract
Diabetic nephropathy (DN) is one of the leading causes of end-stage renal diseases worldwide. This study is designed to investigate the underlying function and mechanism of a novel lncRNA GAS5 in the progression of DN. We found that lncRNA GAS5 expression level was decreased in type 2 diabetes (T2D) with DN compared with that in patients without DN. Moreover, lncRNA GAS5 expression level was negatively associated with the severity of DN-related complications. lncRNA GAS5 inhibited MCs proliferation and caused G0/1 phase arrest. lncRNA GAS5 overexpression alleviated the expression of fibrosis-related protein in mesangial cells (MCs). The dual-luciferase reporter assay and RNA binding protein immunoprecipitation (RIP) assay results revealed that lncRNA GAS5 functions as an endogenous sponge for miR-221 via both the directly targeting way and Ago2-dependent manner. Furthermore, SIRT1 was confirmed as a target gene of miR-221. lncRNA GAS5 upregulated SIRT1 expression and inhibited MCs proliferation and fibrosis by acting as an miR-221 sponge. Finally, we found that lncRNA GSA5 suppressed the development of DN in vivo. Thus, lncRNA GAS5 was involved in the progression of DN by sponging miR-221 and contributed to lncRNA-directed diagnostics and therapeutics in DN.
Collapse
Affiliation(s)
- Xiaoxu Ge
- Department of Endocrinology, Tongren Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Bojin Xu
- Department of Endocrinology, Tongren Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Wenwei Xu
- Department of Geriatrics, Tongren Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Lili Xia
- Department of Endocrinology, Tongren Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Zhongqin Xu
- Department of Family Medicine, Tongren Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Lisha Shen
- Department of Endocrinology, Tongren Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Wenfang Peng
- Department of Endocrinology, Tongren Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Shan Huang
- Department of Endocrinology, Tongren Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
147
|
Li Z, Tan H, Zhao W, Xu Y, Zhang Z, Wang M, Zhou X. Integrative analysis of DNA methylation and gene expression profiles identifies MIR4435-2HG as an oncogenic lncRNA for glioma progression. Gene 2019; 715:144012. [PMID: 31357021 DOI: 10.1016/j.gene.2019.144012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 07/15/2019] [Accepted: 07/25/2019] [Indexed: 02/09/2023]
Abstract
Long noncoding RNAs (lncRNAs) have been shown to play an important role in tumor biogenesis and prognosis. The glioma is a grade classified cancer, however, we still lack the knowledge on their function during glioma progression. While previous studies have shown how lncRNAs regulate protein-coding gene epigenetically, it is still unclear how lncRNAs are regulated epigenetically. In this study, we firstly analyzed the RNA-seq data systematically across grades II, IV, and IV of glioma samples. We identified 60 lncRNAs that are significantly differentially expressed over disease progression (DElncRNA), including well-known PVT1, HOTAIR, H19 and rarely studied CARD8-AS, MIR4435-2HG. Secondly, by integrating HM450K methylation microarray data, we demonstrated that some of the lncRNAs are epigenetically regulated by methylation. Thirdly, we developed a DESeq2-GSEA-ceRNA-survival analysis strategy to investigate their functions. Particularly, MIR4435-2HG is highly expressed in high-grade glioma and may have an impact on EMT and TNFα signaling pathway by functioning as a miRNA sponge of miR-125a-5p and miR-125b-5p to increase the expression of CD44. Our results revealed the dynamic expression of lncRNAs in glioma progression and their epigenetic regulation mechanism.
Collapse
Affiliation(s)
- Zhijin Li
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China; Center for Computational Systems Medicine, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Hua Tan
- Center for Computational Systems Medicine, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Weiling Zhao
- Center for Computational Systems Medicine, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yungang Xu
- Center for Computational Systems Medicine, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Zhigang Zhang
- School of Information Management and Statistics, Hubei University of Economics, Wuhan, China
| | - Maode Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Xiaobo Zhou
- Center for Computational Systems Medicine, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA; McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA; School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
148
|
Wang J, Li X, Xiao Z, Wang Y, Han Y, Li J, Zhu W, Leng Q, Wen Y, Wen X. MicroRNA-488 inhibits proliferation and glycolysis in human prostate cancer cells by regulating PFKFB3. FEBS Open Bio 2019; 9:1798-1807. [PMID: 31410981 PMCID: PMC6768114 DOI: 10.1002/2211-5463.12718] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/12/2019] [Accepted: 08/12/2019] [Indexed: 12/21/2022] Open
Abstract
Prostate cancer (PCa) remains the second leading cause of cancer-related death among men in the United States, and its molecular mechanism remains to be elucidated. Recent studies have suggested that microRNAs may play an important role in cancer development and progression. By analyzing the Gene Expression Omnibus dataset, we found lower expression for miR-488 in PCa than in normal tissues. Moreover, CCK-8, EdU, glucose uptake, and lactate secrete assays revealed that overexpression of miR-488 in PCa cell lines PC3 and DU145 resulted in inhibition of proliferation and glycolysis. In contrast, downregulation of miR-488 expression promoted proliferation and glycolysis in PCa cells. Using a bioinformatic approach and dual-luciferase reporter assays, we identified 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase, isoform3 (PFKFB3), as a direct target of miR-488. Inhibition of PFKFB3 also suppressed PCa cell glycolysis and proliferation. Our study suggests that miR-488 inhibits PCa cell proliferation and glycolysis by targeting PFKFB3, and thus, miR-488 may be a novel therapeutic candidate for PCa.
Collapse
Affiliation(s)
- Jun Wang
- Department of UrologyThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Department of UrologyFudan University Shanghai Cancer CenterChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Xiaojuan Li
- Center of Health ManagementShenzhen HospitalSouthern Medical UniversityShenzhenChina
| | - Zhaoming Xiao
- Department of UrologyNanfang Hospital of Southern Medical UniversityGuangzhouChina
| | - Yu Wang
- Department of UrologyThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Yuefu Han
- Department of UrologyYue Bei People's HospitalShaoguanChina
| | - Jun Li
- Department of UrologyShenzhen HospitalSouthern Medical UniversityShenzhenChina
| | - Weian Zhu
- Department of UrologyShenzhen HospitalSouthern Medical UniversityShenzhenChina
| | - Qu Leng
- Department of UrologyShenzhen HospitalSouthern Medical UniversityShenzhenChina
| | - Yuehui Wen
- Department of UrologyShenzhen HospitalSouthern Medical UniversityShenzhenChina
| | - Xinqiao Wen
- Department of UrologyThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
149
|
Dong N, Guo J, Han S, Bao L, Diao Y, Lin Z. Positive feedback loop of lncRNA HOXC-AS2/miR-876-5p/ZEB1 to regulate EMT in glioma. Onco Targets Ther 2019; 12:7601-7609. [PMID: 31571911 PMCID: PMC6754333 DOI: 10.2147/ott.s216134] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 08/28/2019] [Indexed: 12/29/2022] Open
Abstract
Purpose Growing evidence has valued the diagnostic and therapeutic ability of long non-coding RNAs (lncRNAs) in various human tumors including glioma. Here, we investigated the biological function and potential mechanism of a novel cancer-related lncRNA, HOXC-AS2, in glioma. Materials and methods The expression of lncHOXC-AS2 was detected using qRT-PCR in glioma cells and tissues. A series of in vitro studies were performed to analyze the biological function of lncHOXC-AS2. Dual-luciferase reporter, RIP was used to determine the relation between lncHOXC-AS2, miR-876-5p and ZEB1. CHIP assay was performed to investigate the transcriptional regulation of HOXC-AS2. Results We found HOXC-AS2 was upregulated in glioma cells and tissues. Depletion of HOXC-AS2 was associated with the inhibition of migration, invasion and EMT process in glioma cells. Mechanism, HOXC-AS2 can sponge miR-876-5p to affect ZEB1 expression. Meanwhile, ZEB1 can bind promoter region of HOXC-AS2 and regulate HOXC-AS2 at transcriptional level. Conclusion Our results conclude that HOXC-AS2/miR-876-5p/ZEB1 constitutes a positive feedback loop to regulate EMT in GBM, providing a potential therapeutic target for glioma.
Collapse
Affiliation(s)
- Nan Dong
- Department of Neurosurgery, Beijing Sanbo Brain Hospital, Capital Medical University, Beijing 100093, People's Republic of China
| | - Junxiu Guo
- Department of Neurosurgery, Shanxi Children's Hospital, Taiyuan 030013, People's Republic of China
| | - Song Han
- Department of Neurosurgery, Beijing Sanbo Brain Hospital, Capital Medical University, Beijing 100093, People's Republic of China
| | - Long Bao
- Department of Neurosurgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, People's Republic of China
| | - Yi Diao
- Department of Neurosurgery, Xuzhou Central Hospital, Xuzhou, Jiangsu 221009, People's Republic of China
| | - Zhixiong Lin
- Department of Neurosurgery, Beijing Sanbo Brain Hospital, Capital Medical University, Beijing 100093, People's Republic of China
| |
Collapse
|
150
|
Liu N, Wang Z, Liu D, Xie P. HOXC13-AS-miR-122-5p-SATB1-C-Myc feedback loop promotes migration, invasion and EMT process in glioma. Onco Targets Ther 2019; 12:7165-7173. [PMID: 31564901 PMCID: PMC6731462 DOI: 10.2147/ott.s220027] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 08/16/2019] [Indexed: 01/15/2023] Open
Abstract
Purpose Differentially expressed long non-coding ribonucleic acids (lncRNAs) have been reported as a key factor of glioma carcinogenesis, but the underlying mechanism involved is still unknown. Materials and methods In the present study, lncRNA HOXC13 antisense RNA (HOXC13-AS) was identified as a potential oncogene in glioma, and Western blotting, wound healing and Transwell assays were carried out to explore the effects of HOXC13-AS on the epithelial-mesenchymal transition (EMT) process as well as the migration and invasion of glioma cells. Results A further mechanistic study showed that HOXC13-AS sponged miR-122-5p to indirectly regulate SATB1 expression and affect the EMT process via the Wnt/β-catenin pathway. Meanwhile, the promoter activity was significantly increased via c-Myc, a key factor of the Wnt/β-catenin pathway, thus forming a positive HOXC13-AS-miR-122-5p-SATB1-c-Myc feedback loop to drive the malignant behavior in glioma. Discussion This study evidences the constitutive HOXC13-AS-miR-122-5p-SATB1-c-Myc feedback loop and provides a potential therapeutic target for glioma treatment.
Collapse
Affiliation(s)
- Ning Liu
- Department of Neurosurgery, Huzhou Central Hospital, Huzhou, Zhejiang 313000, People's Republic of China
| | - Ziyu Wang
- Department of Emergency Intensive Care Unit, The Affiliated Huai'an Hospital of Xuzhou Medical University, The Second People's Hospital of Huai'an, Huai'an 223002, People's Republic of China
| | - Dachao Liu
- Department of Imaging, The Affiliated Huai'an Hospital of Xuzhou Medical University, The Second People's Hospital of Huai'an, Huai'an 223002, People's Republic of China
| | - Peng Xie
- Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, The Second People's Hospital of Huai'an, Huai'an 223002, People's Republic of China
| |
Collapse
|