1
|
Li Z, Zhang G. Glut-1 exerts anti-tumor activity in glioma by regulating AKT / mTOR signaling pathway. Technol Health Care 2025:9287329241304910. [PMID: 39973866 DOI: 10.1177/09287329241304910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
OBJECTIVE Glioma is a common tumor in neurosurgery. Glut-1 is the main carrier of glucose uptake by cells and can provide energy through the glycolytic pathway. However, the role and related mechanisms of Glut-1 in glioma have not yet been elucidated. METHODS Real time PCR and Western blot were done to assess Glut-1 level in glioma tumor tissues and adjacent tissues. Glioma U87 cells were separated into control group; Glut-1 negative control (NC group); and Glut-1 siRNA group followed by analysis of Glut-1 expression, cell proliferation by MTT assay, cell invasion, cell apoptosis and cycle by flow cytometry and AKT / mTOR signaling proteins level by Western blot. RESULTS Glut-1 expression was significantly increased in the tissues of glioma patients (P < 0.05) compared to adjacent tissues and its level was related to tumor size, pathological grade and survival. Down-regulating the expression of Glut-1 can significantly inhibit tumor cell proliferation and invasion, increase apoptosis and induce G0 phase of cell cycle arrest, and inhibit the expression of AKT / mTOR signaling proteins phosphorylation (P < 0.05). CONCLUSIONS Glut-1 level in glioma tissues is significantly increased, which is related to the pathological features. Down-regulating Glut-1 can inhibit glioma by regulating the AKT / mTOR signaling pathway.
Collapse
Affiliation(s)
- Zhimei Li
- Department of Neurosurgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Guofeng Zhang
- Department of Neurosurgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
2
|
Sun Y, Shen Y, Li X. Retracted article: Knockdown of long non-coding RNA AGAP2-AS1 suppresses the proliferation and metastasis of glioma by targeting microRNA-497-5p. Bioengineered 2024; 15:1995573. [PMID: 34709983 PMCID: PMC10802192 DOI: 10.1080/21655979.2021.1995573] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 12/15/2022] Open
Abstract
Yi Sun, Yulong Shen and Xing Li. Knockdown of long non-coding RNA AGAP2-AS1 suppresses the proliferation and metastasis of glioma by targeting microRNA-497-5p. Bioengineered. 2021 Oct. doi: 10.1080/21655979.2021.1995573.Since publication, significant concerns have been raised about the compliance with ethical policies for human research and the integrity of the data reported in the article.When approached for an explanation, the authors provided some original data but were not able to provide all the necessary supporting information. As verifying the validity of published work is core to the scholarly record's integrity, we are retracting the article. All authors listed in this publication have been informed.We have been informed in our decision-making by our editorial policies and the COPE guidelines. The retracted article will remain online to maintain the scholarly record, but it will be digitally watermarked on each page as 'Retracted.'
Collapse
Affiliation(s)
- Yi Sun
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing City, Jiangsu Province, China
| | - Yulong Shen
- Department of Neurosurgery, Huaihua First People’s Hospital, Huaihua City, Hunan Province, China
| | - Xing Li
- Department of Neurosurgery, Taizhou First People’s Hospital, Taizhou City, Zhejiang Province, China
| |
Collapse
|
3
|
Palaniappan A, Muthamilselvan S, Sarathi A. COADREADx: A comprehensive algorithmic dissection of colorectal cancer unravels salient biomarkers and actionable insights into its discrete progression. PeerJ 2024; 12:e18347. [PMID: 39484215 PMCID: PMC11526798 DOI: 10.7717/peerj.18347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 09/27/2024] [Indexed: 11/03/2024] Open
Abstract
Background Colorectal cancer is a common condition with an uncommon burden of disease, heterogeneity in manifestation, and no definitive treatment in the advanced stages. Renewed efforts to unravel the genetic drivers of colorectal cancer progression are paramount. Early-stage detection contributes to the success of cancer therapy and increases the likelihood of a favorable prognosis. Here, we have executed a comprehensive computational workflow aimed at uncovering the discrete stagewise genomic drivers of colorectal cancer progression. Methods Using the TCGA COADREAD expression data and clinical metadata, we constructed stage-specific linear models as well as contrast models to identify stage-salient differentially expressed genes. Stage-salient differentially expressed genes with a significant monotone trend of expression across the stages were identified as progression-significant biomarkers. The stage-salient genes were benchmarked using normals-augmented dataset, and cross-referenced with existing knowledge. The candidate biomarkers were used to construct the feature space for learning an optimal model for the digital screening of early-stage colorectal cancers. The candidate biomarkers were also examined for constructing a prognostic model based on survival analysis. Results Among the biomarkers identified are: CRLF1, CALB2, STAC2, UCHL1, KCNG1 (stage-I salient), KLHL34, LPHN3, GREM2, ADCY5, PLAC2, DMRT3 (stage-II salient), PIGR, HABP2, SLC26A9 (stage-III salient), GABRD, DKK1, DLX3, CST6, HOTAIR (stage-IV salient), and CDH3, KRT80, AADACL2, OTOP2, FAM135B, HSP90AB1 (top linear model genes). In particular the study yielded 31 genes that are progression-significant such as ESM1, DKK1, SPDYC, IGFBP1, BIRC7, NKD1, CXCL13, VGLL1, PLAC1, SPERT, UPK2, and interestingly three members of the LY6G6 family. Significant monotonic linear model genes included HIGD1A, ACADS, PEX26, and SPIB. A feature space of just seven biomarkers, namely ESM1, DHRS7C, OTOP3, AADACL2, LPHN3, GABRD, and LPAR1, was sufficient to optimize a RandomForest model that achieved > 98% balanced accuracy (and performant recall) of cancer vs. normal on external validation. Design of an optimal multivariate model based on survival analysis yielded a prognostic panel of three stage-IV salient genes, namely HOTAIR, GABRD, and DKK1. Based on the above sparse signatures, we have developed COADREADx, a web-server for potentially assisting colorectal cancer screening and patient risk stratification. COADREADx provides uncertainty measures for its predictions and needs clinical validation. It has been deployed for experimental non-commercial use at: https://apalanialab.shinyapps.io/coadreadx/.
Collapse
Affiliation(s)
- Ashok Palaniappan
- Systems Computational Biology Lab, Department of Bioinformatics, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | - Sangeetha Muthamilselvan
- Systems Computational Biology Lab, Department of Bioinformatics, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | - Arjun Sarathi
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
4
|
Xue Y, Diao M, Lyu J, Li K, He L, Chen J, Li X. Long Noncoding RNAs PTPRG Antisense RNA 1 Targets Cyclin D1 to Facilitate Cell Proliferation in Lung Adenocarcinoma. Cancer Biother Radiopharm 2024; 39:573-583. [PMID: 34767727 DOI: 10.1089/cbr.2021.0168] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background: Numerous studies have recorded the function of long noncoding RNAs (lncRNAs) in cancer development, including lung adenocarcinoma (LUAD). Previous studies have reported the crucial role of lncRNA PTPRG antisense RNA 1 (PTPRG-AS1) in various cancers. However, the role of PTPRG-AS1 in LUAD remains unknown. Materials and Methods: Real-time quantitative polymerase chain reaction (RT-qPCR) was applied for detecting PTPRG-AS1 expression in LUAD cell lines. Functional assays and in vivo experiments explored cell proliferation, whereas flow cytometry analysis was used to detect cell cycle. In addition, fluorescent in situ hybridization (FISH) and subcellular fractionation assay measured the localization of PTPRG-AS1 in LUAD cells. RNA pulldown, luciferase reporter, and RNA immunoprecipitation (RIP) assays were used to investigate the interaction of PTPRG-AS1/miR-124-3p/cyclin D1 (CCND1) axis. Results: PTPRG-AS1 expression was notably high in LUAD cell lines. PTPRG-AS1 knockdown suppressed cell proliferation and cycle as well as the level of CCND1. Moreover, miR-124-3p was the mutual target of PTPRG-AS1 and CCND1. In addition, PTPRG-AS1 sponged miR-124-3p to upregulate CCND1 in LUAD cells. Moreover, miR-124-3p depletion reversed the suppression of PTPRG-AS1 silence on LUAD cell behaviors, but then CCND1 knockdown countervailed the promoting influence of downregulated miR-124-3p. Conclusions: PTPRG-AS1 propels cell proliferation and cell cycle of LUAD by targeting miR-124-3p/CCND1 axis.
Collapse
Affiliation(s)
- Yang Xue
- Cardio-Thoracic Surgery, People's Hospital of Deyang, Deyang, China
| | - Mingqiang Diao
- Cardio-Thoracic Surgery, People's Hospital of Deyang, Deyang, China
| | - Jing Lyu
- Cardio-Thoracic Surgery, People's Hospital of Deyang, Deyang, China
| | - Kang Li
- Department of Respiratory Medicine, Wenling Hospital, Wenzhou Medical University, Wenling, China
| | - Long He
- Department of Laboratory, Wenling Hospital, Wenzhou Medical University, Wenling, China
| | - Junfeng Chen
- Department of Respiratory Medicine, Wenling Hospital, Wenzhou Medical University, Wenling, China
| | - Xiangguo Li
- Department of Respiratory Medicine, Wenling Hospital, Wenzhou Medical University, Wenling, China
| |
Collapse
|
5
|
Tahmasebi Dehkordi H, Khaledi F, Ghasemi S. Immunological processes of enhancers and suppressors of long non-coding RNAs associated with brain tumors and inflammation. Int Rev Immunol 2024; 43:178-196. [PMID: 37974420 DOI: 10.1080/08830185.2023.2280581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 03/18/2023] [Accepted: 11/02/2023] [Indexed: 11/19/2023]
Abstract
Immunological processes, such as inflammation, can both cause tumor suppression and cancer progression. Moreover, deregulated levels of long non-coding RNA (lncRNA) expression in the brain may cause inflammation and lead to the growth of tumors. Like other biological processes, the immune system's role in cancer is complicated, varies, and can help or hurt the cancer's maintenance. According to research, inflammation and brain cancer are correlated via several signaling pathways. A variety of lncRNAs have recently been revealed to influence cancer by modulating inflammatory pathways. As a result, lncRNAs have the potential to influence carcinogenesis, tumor formation, or tumor suppression via an increase or decrease in inflammation functions. Although the study and targeting of lncRNAs have made great progress in the treatment of cancer, there are definitely limitations and challenges. Using new technologies like nanocarriers and cell-penetrating peptides (CPPs) to target treatments without hurting healthy body tissues has shown to be very effective. In this review article, we have collected significantly related lncRNAs and their inhibitory or stimulating roles in inflammation and brain cancer for the first time. However, there are limitations, such as side effects and damage to normal tissues. With the advancement of new targeting technologies, these lncRNAs may be candidates for the specific targeting therapy of brain cancers by limiting inflammation or stimulating the immune system against them in the future.
Collapse
Affiliation(s)
- Hossein Tahmasebi Dehkordi
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Fatemeh Khaledi
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Sorayya Ghasemi
- Cancer Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
6
|
Wang M, Vulcano S, Xu C, Xie R, Peng W, Wang J, Liu Q, Jia L, Li Z, Li Y. Potentials of ribosomopathy gene as pharmaceutical targets for cancer treatment. J Pharm Anal 2024; 14:308-320. [PMID: 38618250 PMCID: PMC11010632 DOI: 10.1016/j.jpha.2023.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/29/2023] [Accepted: 10/07/2023] [Indexed: 04/16/2024] Open
Abstract
Ribosomopathies encompass a spectrum of disorders arising from impaired ribosome biogenesis and reduced functionality. Mutation or dysexpression of the genes that disturb any finely regulated steps of ribosome biogenesis can result in different types of ribosomopathies in clinic, collectively known as ribosomopathy genes. Emerging data suggest that ribosomopathy patients exhibit a significantly heightened susceptibility to cancer. Abnormal ribosome biogenesis and dysregulation of some ribosomopathy genes have also been found to be intimately associated with cancer development. The correlation between ribosome biogenesis or ribosomopathy and the development of malignancies has been well established. This work aims to review the recent advances in the research of ribosomopathy genes among human cancers and meanwhile, to excavate the potential role of these genes, which have not or rarely been reported in cancer, in the disease development across cancers. We plan to establish a theoretical framework between the ribosomopathy gene and cancer development, to further facilitate the potential of these genes as diagnostic biomarker as well as pharmaceutical targets for cancer treatment.
Collapse
Affiliation(s)
- Mengxin Wang
- Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Stephen Vulcano
- Autoimmunity and Inflammation Program, HSS Research Institute, Hospital for Special Surgery New York, New York, NY, 10021, USA
| | - Changlu Xu
- Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, CA, 90095, USA
| | - Renjian Xie
- Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
- School of Medical Information Engineering, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Weijie Peng
- Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Jie Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Qiaojun Liu
- Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Lee Jia
- Institute of Oceanography, Minjiang University, Fuzhou, 350108, China
| | - Zhi Li
- Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, CA, 90095, USA
| | - Yumei Li
- Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| |
Collapse
|
7
|
Wang J, Guo X, Chen Y, Zhang W, Ren J, Gao A. The m6A reader IGF2BP1 attenuates the stability of RPL36 and cell proliferation to mediate benzene hematotoxicity by recognizing m6A modification. Toxicology 2024; 503:153758. [PMID: 38367942 DOI: 10.1016/j.tox.2024.153758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 02/05/2024] [Accepted: 02/15/2024] [Indexed: 02/19/2024]
Abstract
Benzene exposure leads to hematotoxicity, and epigenetic modification is considered to be a potential mechanism of benzene pathogenesis. As a newly discovered post-transcriptional modification, the roles of N6-methyladenosine (m6A) in benzene hematotoxicity are still unclear. m6A can only exert its gene regulatory function after being recognized by m6A reading proteins. In this study, we found that the expression of m6A reader IGF2BP1 decreased in benzene poisoning workers and in 20 μM benzene metabolite 1,4-BQ-treated AHH-1 cells. Further overexpression of IGF2BP1 in mice alleviated 50 ppm benzene-induced hematopoietic damage, suggesting that IGF2BP1 plays a critical role in benzene hematotoxicity. Next, we examined transcriptome-wide m6A methylation in vitro to search for target genes of IGF2BP1. We found that benzene metabolite 1,4-BQ treatment altered the m6A methylation levels of various genes. The comprehensive analysis of mRNA expression and m6A methylation uncovered that the hypomethylated Ribosomal Protein L36 (RPL36) and its consequent reduced expression impaired cell proliferation. Mechanically, m6A modification reduced RNA stability to down-regulate RPL36 expression. Moreover, overexpression of IGF2BP1 relieved RPL36 reduction and cell proliferation inhibition caused by benzene in vitro and in vivo by directly binding with RPL36 mRNA. In conclusion, the m6A reader IGF2BP1 attenuates the stability of RPL36 and cell proliferation to mediate benzene hematotoxicity by recognizing m6A modification. IGF2BP1 and RPL36 may be key molecules and potential therapeutic targets for benzene hematotoxicity.
Collapse
Affiliation(s)
- Jingyu Wang
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Xiaoli Guo
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China; Department of Cancer Epidemiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, PR China
| | - Yujiao Chen
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Wei Zhang
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Jing Ren
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Ai Gao
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China.
| |
Collapse
|
8
|
Nakahara S, Male AG, Turner JA, Calhoun VD, Lim KO, Mueller BA, Bustillo JR, O'Leary DS, Voyvodic J, Belger A, Preda A, Mathalon DH, Ford JM, Guffanti G, Macciardi F, Potkin SG, Van Erp TGM. Auditory oddball hypoactivation in schizophrenia. Psychiatry Res Neuroimaging 2023; 335:111710. [PMID: 37690161 DOI: 10.1016/j.pscychresns.2023.111710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/30/2023] [Accepted: 08/26/2023] [Indexed: 09/12/2023]
Abstract
Individuals with schizophrenia (SZ) show aberrant activations, assessed via functional magnetic resonance imaging (fMRI), during auditory oddball tasks. However, associations with cognitive performance and genetic contributions remain unknown. This study compares individuals with SZ to healthy volunteers (HVs) using two cross-sectional data sets from multi-center brain imaging studies. It examines brain activation to auditory oddball targets, and their associations with cognitive domain performance, schizophrenia polygenic risk scores (PRS), and genetic variation (loci). Both sample 1 (137 SZ vs. 147 HV) and sample 2 (91 SZ vs. 98 HV), showed hypoactivation in SZ in the left-frontal pole, and right frontal orbital, frontal pole, paracingulate, intracalcarine, precuneus, supramarginal and hippocampal cortices, and right thalamus. In SZ, precuneus activity was positively related to cognitive performance. Schizophrenia PRS showed a negative correlation with brain activity in the right-supramarginal cortex. GWA analyses revealed significant single-nucleotide polymorphisms associated with right-supramarginal gyrus activity. RPL36 also predicted right-supramarginal gyrus activity. In addition to replicating hypoactivation for oddball targets in SZ, this study identifies novel relationships between regional activity, cognitive performance, and genetic loci that warrant replication, emphasizing the need for continued data sharing and collaborative efforts.
Collapse
Affiliation(s)
- Soichiro Nakahara
- Clinical Translational Neuroscience Laboratory, Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, CA, 92617, United States; Discovery Accelerator Venture Unit Direct Reprogramming, Astellas Pharma Inc, 21, Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - Alie G Male
- Clinical Translational Neuroscience Laboratory, Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, CA, 92617, United States
| | - Jessica A Turner
- Department of Psychiatry and Behavioral Health, Ohio State University, Columbus, OH, 43210, United States
| | - Vince D Calhoun
- Tri-institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State University, Georgia Institute of Technology, and Emory University 55 Park Pl NE, Atlanta, GA 30303, USA
| | - Kelvin O Lim
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, 55454, United States
| | - Bryon A Mueller
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, 55454, United States
| | - Juan R Bustillo
- Departments of Psychiatry & Neurosciences, University of New Mexico, Albuquerque, NM, 87131, United States
| | - Daniel S O'Leary
- Department of Psychiatry, University of Iowa, Iowa City, IA, 52242, United States
| | - James Voyvodic
- Brain Imaging and Analysis Center, Duke University Medical Center, Durham, NC, 27710, United States
| | - Aysenil Belger
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States
| | - Adrian Preda
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, CA, 92617, United States
| | - Daniel H Mathalon
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA, 94143, United States; Veterans Affairs San Francisco Healthcare System, San Francisco, CA, 94121, United States
| | - Judith M Ford
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA, 94143, United States; Veterans Affairs San Francisco Healthcare System, San Francisco, CA, 94121, United States; San Francisco Veterans Affairs Medical Center, San Francisco, CA 94121, United States
| | - Guia Guffanti
- Department of Psychiatry at McLean Hospital - Harvard Medical School, Boston, MA, 02478, United States
| | - Fabio Macciardi
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, CA, 92617, United States
| | - Steven G Potkin
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, CA, 92617, United States
| | - Theo G M Van Erp
- Clinical Translational Neuroscience Laboratory, Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, CA, 92617, United States; Center for the Neurobiology of Learning and Memory, University of California Irvine, 309 Qureshey Research Lab, Irvine, CA, 92697, United States.
| |
Collapse
|
9
|
Hao C, Li R, Lu Z, He K, Shen J, Wang T, Qiu T. Predicting prognosis, immunotherapy and distinguishing cold and hot tumors in clear cell renal cell carcinoma based on anoikis-related lncRNAs. Front Immunol 2023; 14:1145450. [PMID: 37359524 PMCID: PMC10288194 DOI: 10.3389/fimmu.2023.1145450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 05/30/2023] [Indexed: 06/28/2023] Open
Abstract
Background Clear cell renal cell carcinoma (ccRCC) is the most frequently occurring malignant tumor within the kidney cancer subtype. It has low sensitivity to traditional radiotherapy and chemotherapy, the optimal treatment for localized ccRCC has been surgical resection, but even with complete resection the tumor will be eventually developed into metastatic disease in up to 40% of localized ccRCC. For this reason, it is crucial to find early diagnostic and treatment markers for ccRCC. Methods We obtained anoikis-related genes (ANRGs) integrated from Genecards and Harmonizome dataset. The anoikis-related risk model was constructed based on 12 anoikis-related lncRNAs (ARlncRNAs) and verified by principal component analysis (PCA), Receiver operating characteristic (ROC) curves, and T-distributed stochastic neighbor embedding (t-SNE), and the role of the risk score in ccRCC immune cell infiltration, immune checkpoint expression levels, and drug sensitivity was evaluated by various algorithms. Additionally, we divided patients based on ARlncRNAs into cold and hot tumor clusters using the ConsensusClusterPlus (CC) package. Results The AUC of risk score was the highest among various factors, including age, gender, and stage, indicating that the model we built to predict survival was more accurate than the other clinical features. There was greater sensitivity to targeted drugs like Axitinib, Pazopanib, and Sunitinib in the high-risk group, as well as immunotherapy drugs. This shows that the risk-scoring model can accurately identify candidates for ccRCC immunotherapy and targeted therapy. Furthermore, our results suggest that cluster 1 is equivalent to hot tumors with enhanced sensitivity to immunotherapy drugs. Conclusion Collectively, we developed a risk score model based on 12 prognostic lncRNAs, expected to become a new tool for evaluating the prognosis of patients with ccRCC, providing different immunotherapy strategies by screening for hot and cold tumors.
Collapse
Affiliation(s)
- Chao Hao
- Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang, China
| | - Rumeng Li
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zeguang Lu
- Department of Anesthesiology, Sun Yat-sen University Cancer Center/State Key Laboratory of Oncology in South China, Guangzhou, China
| | - Kuang He
- Department of Pathology, Dushu Lake Hospital Affiliated of Soochow University, Suzhou, China
| | - Jiayun Shen
- Afliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tengfei Wang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Tingting Qiu
- Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang, China
| |
Collapse
|
10
|
Li G, Li X, Li Z, Luo X, Jing L, Guo D, Guan K, Yuan F, Pan B. Sox2ot /miR-9 /Cthrc1 Promote Proliferation and Migration of Schwann Cells Following Nerve Injury. Neuroscience 2023; 519:47-59. [PMID: 36924985 DOI: 10.1016/j.neuroscience.2023.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 03/04/2023] [Accepted: 03/08/2023] [Indexed: 03/16/2023]
Abstract
The effects of traditional treatments for peripheral nerve injury (PNI) are not ideal, which has prompted the identification of new therapeutic strategies. As unique glial cells in the peripheral nervous system, Schwann cells (SCs) play an important role in the repair of PNI. Recent studies have demonstrated that long noncoding RNAs (lncRNAs) are involved in the regulation of nerve repair after PNI. In this study, we used microarray technology to detect mRNA and lncRNA expression profiles at different time points after PNI and identified lncRNA Sox2ot-miR-9-Cthrc1 as a competitive endogenous RNA (ceRNA) for further investigation. Expression of lncRNA Sox2ot was increased after PNI, and overexpression of Sox2ot promoted SCs migration and proliferation. Mechanistic analyses confirmed that Sox2ot can regulate the expression of Cthrc1 through competitive adsorption of miR-9 in SCs, ultimately affecting SCs migration and proliferation. Our findings reveal the key role of lncRNA Sox2ot in nerve regeneration and provide a new direction for PNI treatment.
Collapse
Affiliation(s)
- Gen Li
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Tissue Regeneration and Digital Medicine, Xuzhou Medical University, Xuzhou, China
| | - Xin Li
- Department of Orthopedics, The First People's Hospital of Lianyungang, Lianyungang, China
| | - Ziyang Li
- Department of Pediatrics, The First People's Hospital of Xuzhou, Xuzhou, China
| | - Xuanxiang Luo
- Department of Orthopedics, Nanjing Gaochun People's Hospital, Nanjing, China
| | - Li Jing
- Department of Orthopedics, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Di Guo
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Kai Guan
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Feng Yuan
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Tissue Regeneration and Digital Medicine, Xuzhou Medical University, Xuzhou, China.
| | - Bin Pan
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Tissue Regeneration and Digital Medicine, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
11
|
Silencing RPL8 inhibits the progression of hepatocellular carcinoma by down-regulating the mTORC1 signalling pathway. Hum Cell 2023; 36:725-737. [PMID: 36577883 DOI: 10.1007/s13577-022-00852-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 12/20/2022] [Indexed: 12/29/2022]
Abstract
This study aimed to explore the role of ribosomal protein L8 (RPL8) in controlling hepatocellular carcinoma (LIHC) development. We measured RPL8 expression, apoptosis, cell viability, proliferation, migration, invasion, glucose uptake, lactate production, and the ATP/ADP ratio of LIHC cells to investigate the effect of RPL8 on LIHC. Bioinformatic analysis was employed to analyse RPL8 expression and its potential mechanism in LIHC. RPL8 was upregulated in LIHC tissues and cells. RPL8 silencing accelerated apoptosis and suppressed viability, growth, and movement of LIHC cells. Additionally, RPL8 silencing inhibited glycolysis in LIHC cells. Bioinformatic analysis revealed that RPL8 is regulated by the upstream transcription factor upstream stimulating factor 1 (USF1) and activates the mTORC1 signalling pathway. USF1 overexpression eliminated the inhibitory effect of RPL8 silencing in LIHC cells. RPL8 overexpression increased cell growth, movement, and glycolysis in LIHC. However, inhibition of the mTORC1 signalling pathway eliminated the effect of RPL8 overexpression on LIHC cells. In conclusion, RPL8 may affect LIHC progression by regulating the mTORC1 signalling pathway.
Collapse
|
12
|
Zhang F, Lu J, Yang J, Dai Q, Du X, Xu Y, Zhang C. SNHG3 regulates NEIL3 via transcription factor E2F1 to mediate malignant proliferation of hepatocellular carcinoma. Immunogenetics 2023; 75:39-51. [PMID: 36114381 DOI: 10.1007/s00251-022-01277-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 08/30/2022] [Indexed: 02/06/2023]
Abstract
The involvement of small nucleolar RNA host gene 3 (SNHG3) in cancer regulation has been reported. This study attempted to deeply investigate the molecular regulatory mechanism of SNHG3 on malignant progression of hepatocellular carcinoma (HCC). According to TCGA analysis, high SNHG3 expression was a risk factor for poor prognosis of HCC patients. Therefore, we further detected the mRNA level of SNHG3 in HCC tissue and cells. It was found that SNHG3 was upregulated in HCC tissue and cells. Afterwards, CCK-8 and flow cytometry assays further proved that silencing SNHG3 inhibited HCC cell proliferation while inducing cell apoptosis and G0/G1 phase arrest. It was also attested in vivo experiments that silencing SNHG3 could reduce the volume and weight of tumors and downregulate the Ki-67 expression to suppress HCC tumor growth. Next, it was discovered that SNHG3 increased the binding of E2F1 and NEIL3 promoter region, thereby activating the transcription feature of NEIL3. Lastly, rescue assays indicated that NEIL3 participated in SNHG3-mediated HCC cell cycle, apoptosis and proliferation. All in all, this study revealed the specific regulatory mechanism of SNHG3 in HCC to enable SNHG3 a hopeful marker for HCC diagnosis and treatment.
Collapse
Affiliation(s)
- Fabiao Zhang
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Department of Hepatobiliary Surgery, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, 317000, China
| | - Jie Lu
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Department of Hepatobiliary Surgery, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, 317000, China
| | - Jian Yang
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Department of Hepatobiliary Surgery, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, 317000, China
| | - Qiqiang Dai
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Department of Hepatobiliary Surgery, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, 317000, China
| | - Xuefeng Du
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Department of Hepatobiliary Surgery, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, 317000, China
| | - Yongfu Xu
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Department of Hepatobiliary Surgery, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, 317000, China
| | - Caiming Zhang
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Department of Hepatobiliary Surgery, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, 317000, China.
| |
Collapse
|
13
|
Lu Q, Guo P, Li H, Liu Y, Yuan L, Zhang B, Wu Q, Wang X. Targeting the lncMST-EPRS/HSP90AB1 complex as novel therapeutic strategy for T-2 toxin-induced growth retardation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 247:114243. [PMID: 36332407 DOI: 10.1016/j.ecoenv.2022.114243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/11/2022] [Accepted: 10/21/2022] [Indexed: 06/16/2023]
Abstract
Growth retardation is a global public health problem that is highly prevalent especially in low-and middle-income countries, which is closely related to the consumption of grains contaminated with T-2 toxin, a risk for human and animal health. However, the possible targets that can relieve T-2 toxin-induced growth retardation still need to be explored. In the present study, T-2 toxin was used as an environmental exposure factor to induce growth retardation and further explore the regulatory role of lncRNA in growth retardation. The present study systematically characterised the expression profiles of lncRNAs and identified a lncRNA lncMST that is related to growth retardation in T-2 toxin-administered rats. Functionally, lncMST could alleviate cell cycle arrest and apoptosis in T-2 toxin-treated GH3 cells. Mechanistically, lncMST, serve as an inducible chaperone RNA, involved in the paradigm "Chemical-induced stress related growth retardation", through recruiting the EPRS/HSP90AB1 complex to increase HDAC6 expression, thus further alleviating T-2 toxin-induced growth retardation. These findings for the first time demonstrate that the probable therapeutic relationship between lncMST and growth retardation, providing an explanation and therapeutic targets for the pathogenesis of growth retardation.
Collapse
Affiliation(s)
- Qirong Lu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China; National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Pu Guo
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China; National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Houpeng Li
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China; MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Hubei 430070, China
| | - Yanan Liu
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China; MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Hubei 430070, China
| | - Ling Yuan
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China; MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Hubei 430070, China
| | - Boyue Zhang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China; MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Hubei 430070, China
| | - Qinghua Wu
- College of Life Science, Yangtze University, Jingzhou 434025, China.
| | - Xu Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China; MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Hubei 430070, China.
| |
Collapse
|
14
|
Mukherjee S, Kundu U, Desai D, Pillai PP. Particulate Matters Affecting lncRNA Dysregulation and Glioblastoma Invasiveness: In Silico Applications and Current Insights. J Mol Neurosci 2022; 72:2188-2206. [PMID: 36370303 DOI: 10.1007/s12031-022-02069-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/14/2022] [Indexed: 11/15/2022]
Abstract
With a reported rise in global air pollution, more than 50% of the population remains exposed to toxic air pollutants in the form of particulate matters (PMs). PMs, from various sources and of varying sizes, have a significant impact on health as long-time exposure to them has seen a correlation with various health hazards and have also been determined to be carcinogenic. In addition to disrupting known cellular pathways, PMs have also been associated with lncRNA dysregulation-a factor that increases predisposition towards the onset or progression of cancer. lncRNA dysregulation is further seen to mediate glioblastoma multiforme (GBM) progression. The vast array of information regarding cancer types including GBM and its various precursors can easily be obtained via innovative in silico approaches in the form of databases such as GEO and TCGA; however, a need to obtain selective and specific information correlating anthropogenic factors and disease progression-in the case of GBM-can serve as a critical tool to filter down and target specific PMs and lncRNAs responsible for regulating key cancer hallmarks in glioblastoma. The current review article proposes an in silico approach in the form of a database that reviews current updates on correlation of PMs with lncRNA dysregulation leading to GBM progression.
Collapse
Affiliation(s)
- Swagatama Mukherjee
- Division of Neurobiology, Department of Zoology, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Uma Kundu
- Division of Neurobiology, Department of Zoology, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Dhwani Desai
- Integrated Microbiome Resource, Department of Pharmacology and Marine Microbial Genomics and Biogeochemistry lab, Department of Biology, Dalhousie University, Halifix, Canada
| | - Prakash P Pillai
- Division of Neurobiology, Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, 390 002, Gujarat, India.
| |
Collapse
|
15
|
Qian G, Yu Y, Dong Y, Hong Y, Wang M. LncRNA AWPPH is downregulated in osteoporosis and regulates type I collagen α1 and α2 ratio. Arch Physiol Biochem 2022; 128:1297-1301. [PMID: 32552067 DOI: 10.1080/13813455.2020.1767150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Normal ratio of type I collagen α1 to α2 (2:1) maintains normal bone microarchitecture. Altered ratios lead to formation of collagen homotrimers and deteriorated bone microarchitecture. In this study, we aimed to investigate the role of lncRNA AWPPH in osteoporosis. We observed that the expression of lncRNA AWPPH was downregulated in osteoporosis patients than that in healthy controls. Downregulated expression of lncRNA AWPPH distinguished osteoporosis patients from healthy controls. In vitro cell experiments showed that knockdown of lncRNA AWPPH led to upregulated α1 but downregulated expression of α2 in osteoblasts, which made the α1 to α2 ratio higher than 2:1. In contrast, overexpression of lncRNA AWPPH led to downregulated α1 but upregulated α2 in osteoblasts, which made the α1 to α2 ratio lower than 2:1. Therefore, lncRNA AWPPH is downregulated in osteoporosis and altered the expression of lncRNA AWPPH regulates type I collagen α1 and α2 ratio in osteoblasts.
Collapse
Affiliation(s)
- Guang Qian
- Department of Orthopedics, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, P. R. China
| | - Yueming Yu
- Department of Orthopedics, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, P. R. China
| | - Youhai Dong
- Department of Orthopedics, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, P. R. China
| | - Yang Hong
- Department of Orthopedics, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, P. R. China
| | - Minghai Wang
- Department of Orthopedics, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, P. R. China
| |
Collapse
|
16
|
Xie G, Zhu Y, Lin Z, Sun Y, Gu G, Li J, Wang W. HBRWRLDA: predicting potential lncRNA-disease associations based on hypergraph bi-random walk with restart. Mol Genet Genomics 2022; 297:1215-1228. [PMID: 35752742 DOI: 10.1007/s00438-022-01909-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 05/20/2022] [Indexed: 10/17/2022]
Abstract
Accumulating evidence indicates that the regulation of long non-coding RNAs (lncRNAs) is closely related to a variety of diseases. Identifying meaningful lncRNA-disease associations will help to contribute to the understanding of the molecular mechanisms underlying these diseases. However, only a limited number of associations between lncRNAs and diseases have been inferred from traditional biological experiments due to the high cost and highly specialized. Therefore, computational methods are increasingly used to reduce time of biological experiments and complement biological research. In this paper, a computational method called HBRWRLDA is proposed to predict lncRNA-disease associations. First, HBRWRLDA models the relationships between multiple nodes using hypergraphs, which allows HBRWRLDA to integrate the expression similarity of lncRNAs and the semantic similarity of diseases to construct hypergraphs. Then, a bi-random walk on hypergraphs is used to predict potential lncRNA-disease associations. HBRWRLDA achieves a higher area under the curve value of 0.9551 and [Formula: see text], respectively, compared with the other five advanced methods under the framework of one-leave cross validation (LOOCV) and five-fold cross-validation (5-fold CV). In addition, the prediction effect of HBRWRLDA was confirmed case studies of three diseases: renal cell carcinoma, gastric cancer, and hepatocellular carcinoma. Case studies demonstrates the capacity of HBRWRLDA to identify potentially disease-associated lncRNAs. Overall, HBRWRLDA is excellent at predicting potential lncRNA-disease associations and could be useful in conducting further biological experiments by helping researchers identify candidates of lncRNA-disease association.
Collapse
Affiliation(s)
- Guobo Xie
- School of Computing, Guangdong University of Technology, Guangzhou, 510000, China
| | - Yinting Zhu
- School of Computing, Guangdong University of Technology, Guangzhou, 510000, China
| | - Zhiyi Lin
- School of Computing, Guangdong University of Technology, Guangzhou, 510000, China.
| | - Yuping Sun
- School of Computing, Guangdong University of Technology, Guangzhou, 510000, China
| | - Guosheng Gu
- School of Computing, Guangdong University of Technology, Guangzhou, 510000, China
| | - Jianming Li
- School of Computing, Guangdong University of Technology, Guangzhou, 510000, China
| | - Weiming Wang
- School of Computing, Guangdong University of Technology, Guangzhou, 510000, China
| |
Collapse
|
17
|
Chen Z, Zhang W, Yan Z, Zhang M. Comprehensive analyses indicated the association between m6A related long non-coding RNAs and various pathways in glioma. Cancer Med 2022; 12:760-788. [PMID: 35668574 PMCID: PMC9844638 DOI: 10.1002/cam4.4913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 04/23/2022] [Accepted: 05/25/2022] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Glioma is one of the most malignant brain tumors and diseases. N6-methyladenosine modification (m6A) is the most abundant and prevalent internal chemical modification of mRNA and long non-coding RNAs (lncRNAs) in eukaryotes. Nevertheless, the correlated pathways and clinical utilization of m6A-related lncRNAs have not been fully evaluated in glioma. METHODS Public RNA-sequencing and clinical annotation data were retrieved from TCGA, CGGA and GEO database. Differential expression analysis and univariate Cox regression analysis were performed to identify the m6A-related and differentially expressed lncRNAs with prognostic function (m6A-DELPF). The consensus clustering was performed to identify the expression pattern of m6A-DELPF. LASSO Cox regression analysis was performed to construct the lncRNA-based signature. The CIBERSORT and ESTIMATE algorithms were performed to analyze immune infiltration and tumor microenvironment, respectively. Immunotherapy sensitivity analysis was performed using data from TCIA. The small molecule drugs prediction analysis was performed using The Connectivity Map (CMap) database and STITCH database. A competing endogenous RNAs (ceRNA) network was constructed based on miRcode, miRDB, miRTarBase, TargetScan database. RESULTS Two clusters (cluster1 and cluster2) were identified after unsupervised cluster analysis based on m6A-DELPF. Additionally, a 15-gene prognostic signature namely m6A-DELPFS was constructed. Analyses of epithelial-mesenchymal-transition score, tumor microenvironment, immune infiltration, clinical characterization analysis, and putative drug prediction were performed to confirm the clinical utility and efficacy of m6A-DELPFS. The potential mechanisms including tumor immune microenvironment of m6A-DELPF influence the initiation and progression of glioma. A clinically accessible nomogram was also constructed based on the m6A-DELPF and other survival-relevant clinical parameters. Two miRNAs and 114 mRNAs were identified as the downstream of seven m6A-related lncRNAs in a ceRNA network. CONCLUSION Our present research confirmed the clinical value of m6A related lncRNAs and their high correlation with tumor immunity, tumor microenvironment, tumor mutation burden and drug sensitivity in glioma.
Collapse
Affiliation(s)
- Zhuohui Chen
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaChina
| | - Wei Zhang
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina
| | - Zhouyi Yan
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaChina
| | - Mengqi Zhang
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaChina,National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaChina
| |
Collapse
|
18
|
Guo A, Fang G, Lin Z, Zheng S, Zhuang Z, Lin R, Lin Y. Overexpression of lncRNA IRAIN restrains the progression and Temozolomide resistance of glioma via repressing IGF-1R-PI3K-NF-κB signaling pathway. Histol Histopathol 2022; 37:543-554. [PMID: 35102541 DOI: 10.14670/hh-18-425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
BACKGROUND Increasing studies have found that long noncoding RNAs (lncRNAs) contribute to regulating tumor progression. This study explores the expression characteristics, effects, and related mechanisms of lncRNA IGF1R antisense imprinted non-protein coding RNA (IRAIN) in glioma. METHODS Quantitative real-time PCR (qRT-PCR) was implemented to testify the IRAIN profile in glioma tissues and paracancerous tissues, and the link between the IRAIN level and the clinicopathological indicators of glioma was analyzed. IRAIN overexpression and knockdown cell models were constructed in glioma cells. Cell proliferation was verified by the colony formation experiment, while flow cytometry was implemented to monitor apoptosis. Transwell assay was performed to examine cell invasion and migration. Western blot (WB) was adopted to compare the profiles of the apoptosis-related proteins (Bax, Bcl2, and Caspase3) and IGF-1R-PI3K-NF-κB pathway. RESULTS IRAIN was down-regulated in glioma tissues (compared with adjacent normal tissues), and the low IRAIN expression was significantly linked with the larger tumor volume and higher pathological stages. Functionally, overexpressing IRAIN abated glioma cell proliferation, invasion, and migration, promoted apoptosis, and attenuated IGF-1R-PI3K-NF-κB expression and temozolomide (TMZ) resistance, which was also confirmed in the xenograft tumor experiment. The WB result showed that overexpressing IRAIN inactivated the IGF-1R-PI3K-NF-κB pathway. Additionally, the IGF-1R knockdown model was established in U251 cells. Si-IGF-1R induced cell proliferation inhibition, promoted cell death, and reduced cell migration and TMZ resistance, whereas Si-IGF-1R+IRAIN group showed no additional effects on glioma cells compared with the Si-IGF-1R group. CONCLUSION IRAIN repressed glioma development and TMZ resistance by inactivating the IGF-1R-PI3K-NF-κB axis.
Collapse
Affiliation(s)
- Aishun Guo
- Department of Neurosurgery, Zhangzhou Municipal Hospital of Fujian Province and Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian Province, China
| | - Guixia Fang
- Department of Nephrology, Zhangzhou Municipal Hospital of Fujian Province and Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian Province, China
| | - Zhenrong Lin
- Department of Neurosurgery, Zhangzhou Municipal Hospital of Fujian Province and Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian Province, China
| | - Shuishun Zheng
- Department of Neurosurgery, Zhangzhou Municipal Hospital of Fujian Province and Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian Province, China
| | - Zhijun Zhuang
- Department of Neurosurgery, Zhangzhou Municipal Hospital of Fujian Province and Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian Province, China
| | - Ruisheng Lin
- Department of Neurosurgery, Zhangzhou Municipal Hospital of Fujian Province and Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian Province, China
| | - Yanling Lin
- Department of Pathology, Zhangzhou Municipal Hospital of Fujian Province and Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian Province, China.
| |
Collapse
|
19
|
Identification of an Inflammatory Response-Related Gene Signature to Predict Survival and Immune Status in Glioma Patients. J Immunol Res 2022; 2022:8972730. [PMID: 35647198 PMCID: PMC9132661 DOI: 10.1155/2022/8972730] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/14/2022] [Accepted: 04/21/2022] [Indexed: 12/26/2022] Open
Abstract
Background Glioma is the most common primary brain tumor with high mortality and poor outcomes. As a hallmark of cancers, inflammatory responses are crucial for their progression. The present study is aimed at exploring the prognostic value of inflammatory response-related genes (IRRGs) and constructing a prognostic IRRG signature for gliomas. Materials and Methods We investigated the relationship between IRRGs and gliomas by integrating the transcriptomic data for gliomas from public databases. Differentially expressed IRRGs (DE-IRRGs) were identified in the GSE4290 cohort. Further, univariate, least absolute shrinkage and selection operator, and multivariate Cox regression analyses were conducted to construct an IRRG signature using The Cancer Genome Atlas (TCGA) cohort. Gliomas from the Chinese Glioma Genome Atlas (CGGA) cohort were employed for independent validation. The performance of gene signature was assessed by survival and receiver operating characteristic curve analyses. The differences in clinical correlations, immune infiltrate types, immunotherapeutic response predictions, and pathway enrichment among subgroups were investigated via bioinformatic algorithms. Results In total, 37 DE-IRRGs were determined, of which 31 were found to be associated with survival. Ultimately, eight genes were retained to construct an IRRG signature that further classified glioma patients into two groups; the high-risk group suffered a poorer outcome as compared to the low-risk group. Furthermore, the high-risk group was significantly correlated with several risk factors, including older age, higher tumor grade, IDH wild type, 1p19q noncodel, and MGMT unmethylation. The nomogram was constructed by integrating the risk scores and other independent clinical characteristics. Moreover, the high-risk group had a greater immune infiltration and was most likely to benefit from immunotherapy. Gene set enrichment analysis suggested that immune and oncogenic pathways were enriched in high-risk glioma patients. Conclusion We constructed a signature composed of eight IRRGs for gliomas, which could effectively predict survival and guide decision-making for treatment.
Collapse
|
20
|
Zhu R, Wang Z. Inhibitory Effect of MicroRNA-376b-Overexpressing Bone Marrow Mesenchymal Stem Cells (BMSCs) on Malignant Characteristics of Glioma Cells Through Targeting Forkhead Box Protein P2 (FOXP2). J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.2999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
This study investigated the impact of microRNA (miR)-376b derived from BMSCs on glioma progression. BMSCs were transfected with miR-376b mimic, miR-376b inhibitor or NC and then cocultured with glioma cells followed by measuring cell behaviors by MTT assay, Transwell assay and flow
cytometry, FOXP2 and miR-376b expression by Western blot and RT-qPCR. After confirming the inhibitory and mimicking activity of transfection, we found that overexpression of miR-376b in BMSCs decreased glioma cell invasion, migration and proliferation but promoted cell apoptosis within 24
h and 48 h after transfection along with reduced number of cells in S-phase. Mechanically, miR-376b targeted miR-376b and up-regulation of miR-376b caused down-regulation of FOXP2 (p < 0.05). Overexpression of miR-376b in BMSCs decelerated glioma cell cycle and inhibitedmalignant
behaviors of glioma cells by targeting FOXP2 expression. These evidence unveils the potential role of FOXP2 as a biomarker for the treatment of gliomas.
Collapse
Affiliation(s)
- Ruoyu Zhu
- Faculty of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jianan, Shandong, 250014, China
| | - Zhonglin Wang
- Department of Neurology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250014, China
| |
Collapse
|
21
|
Wang H, Zhang X, Chen X, Zhang S, Yun Z, Gao Q, Sheng H, Wang J. Long non-coding RNA placenta‑specific protein 2 regulates the chemosensitivity of cancer cells to cisplatin in hepatocellular carcinoma (HCC) by sponging microRNA-96 to upregulate X-linked inhibitor of apoptosis protein. Bioengineered 2022; 13:10765-10773. [PMID: 35475470 PMCID: PMC9208526 DOI: 10.1080/21655979.2022.2056815] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
This study was conducted to investigate the roles of lncRNA PLAC2 and XiaP in hepatocellular carcinoma (HCC). HCC and paired non-tumor tissues were collected from 62 HCC patients who received cisplatin-based treatment. At 0, 2, and 4 months of post-cisplatin-based therapy, blood samples (5 ml) were collected from all patients and prepared plasma samples. LncRNA PLAC2 expression in tissue and plasma samples was determined by RT-qPCR. The interactions between lncRNA PLAC2 and XiaP in HCC cell lines were assessed by overexpression experiments. Cell viability and apoptosis under cisplatin treatment were analyzed by MTT assay and cell apoptosis assay, respectively. The direct interaction between lncRNA PLAC2 and miR-96, which can target XiaP, was analyzed by performing RNA-RNA pulldown assay. It was observed that lncRNA PLAC2 was upregulated in HCC tissues than in non-tumor tissues. LncRNA PLAC2 expression in HCC tissues was not affected by HBV and HCV but upregulated after cisplatin-based treatment. Similarly, cisplatin treatment of HCC cells increased PLAC2 expression. LncRNA PLAC2 and XiaP overexpression increased viability and decreased apoptosis of cisplatin-treated HCC cells, while lncRNA PLAC2 knockdown decreased viability and increased apoptosis of cisplatin-treated HCC cells. Western blot analysis showed that lncRNA PLAC2 increased XiaP protein accumulation, while lncRNA PLAC2 siRNA silencing decreased XiaP expression in HCC cells. LncRNA PLAC2 and miR-96 directly interacted with each other, while they failed to regulate the expression of each other. In conclusion, lncRNA PLAC2 negatively regulates the chemosensitivity of HCC cells to cisplatin, possibly by sponging miR-96 to upregulate miR-96.
Collapse
Affiliation(s)
- Huixiong Wang
- Department of Hepatobiliary Surgery, Hospital of Inner Mongolia Baotou Steel, Baotou, Inner Mongolia, China
| | - Xin Zhang
- Department of Pathology, The Second People's Hospital of Foshan Affiliated Foshan Hospital of Southern Medical University, Foshan, Guangdong, China
| | - Xiaoting Chen
- Department of Plastic surgery, Hospital of Inner Mongolia Baotou Steel, Baotou City, Inner Mongolia, China
| | - Shengbin Zhang
- Department of Hepatobiliary Surgery, Hospital of Inner Mongolia Baotou Steel, Baotou, Inner Mongolia, China
| | - Zhelin Yun
- Department of Hepatobiliary Surgery, Hospital of Inner Mongolia Baotou Steel, Baotou, Inner Mongolia, China
| | - Qiang Gao
- Department of Hepatobiliary Surgery, Hospital of Inner Mongolia Baotou Steel, Baotou, Inner Mongolia, China
| | - Haitao Sheng
- Department of Hepatobiliary Surgery, Hospital of Inner Mongolia Baotou Steel, Baotou, Inner Mongolia, China
| | - Junjie Wang
- Department of Hepatobiliary Surgery, Hospital of Inner Mongolia Baotou Steel, Baotou, Inner Mongolia, China
| |
Collapse
|
22
|
Liu K, Chen H, Wang Y, Jiang L, Li Y. Evolving Insights Into the Biological Function and Clinical Significance of Long Noncoding RNA in Glioblastoma. Front Cell Dev Biol 2022; 10:846864. [PMID: 35531099 PMCID: PMC9068894 DOI: 10.3389/fcell.2022.846864] [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: 12/31/2021] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
Glioblastoma (GBM) is one of the most prevalent and aggressive cancers worldwide. The overall survival period of GBM patients is only 15 months even with standard combination therapy. The absence of validated biomarkers for early diagnosis mainly accounts for worse clinical outcomes of GBM patients. Thus, there is an urgent requirement to characterize more biomarkers for the early diagnosis of GBM patients. In addition, the detailed molecular basis during GBM pathogenesis and oncogenesis is not fully understood, highlighting that it is of great significance to elucidate the molecular mechanisms of GBM initiation and development. Recently, accumulated pieces of evidence have revealed the central roles of long noncoding RNAs (lncRNAs) in the tumorigenesis and progression of GBM by binding with DNA, RNA, or protein. Targeting those oncogenic lncRNAs in GBM may be promising to develop more effective therapeutics. Furthermore, a better understanding of the biological function and underlying molecular basis of dysregulated lncRNAs in GBM initiation and development will offer new insights into GBM early diagnosis and develop novel treatments for GBM patients. Herein, this review builds on previous studies to summarize the dysregulated lncRNAs in GBM and their unique biological functions during GBM tumorigenesis and progression. In addition, new insights and challenges of lncRNA-based diagnostic and therapeutic potentials for GBM patients were also introduced.
Collapse
Affiliation(s)
- Kun Liu
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Hong Chen
- Department of Oncology, 920th Hospital of Joint Logistics Support Force, Teaching Hospital of Kunming Medical University, Kunming, China
| | - Yuanyuan Wang
- Department of Pathology, 920th Hospital of Joint Logistics Support Force, Teaching Hospital of Kunming Medical University, Kunming, China
| | - Liping Jiang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, United States
- *Correspondence: Yi Li, ; Liping Jiang,
| | - Yi Li
- Department of Oncology, 920th Hospital of Joint Logistics Support Force, Teaching Hospital of Kunming Medical University, Kunming, China
- *Correspondence: Yi Li, ; Liping Jiang,
| |
Collapse
|
23
|
Yang Z, Li X, Luo W, Wu Y, Tang T, Wang Y. The Involvement of Long Non-coding RNA and Messenger RNA Based Molecular Networks and Pathways in the Subacute Phase of Traumatic Brain Injury in Adult Mice. Front Neuroinform 2022; 16:794342. [PMID: 35311004 PMCID: PMC8931714 DOI: 10.3389/fninf.2022.794342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 01/26/2022] [Indexed: 12/24/2022] Open
Abstract
Traumatic brain injury (TBI) is a complex injury with a multi-faceted recovery process. Long non-coding RNAs (lncRNAs) are demonstrated to be involved in central nervous system (CNS) disorders. However, the roles of lncRNAs in long-term neurological deficits post-TBI are poorly understood. The present study depicted the microarray’s lncRNA and messenger RNA (mRNA) profiles at 14 days in TBI mice hippocampi. LncRNA and mRNA microarray was used to identify differentially expressed genes. Quantitative real-time polymerase chain reaction (qRT-PCR) was employed to validate the microarray results. Bioinformatics analysis [including Gene Ontology (GO), the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway, lncRNA-mRNA co-expression network, and lncRNA-miRNA-mRNA network] were applied to explore the underlying mechanism. A total of 264 differentially expressed lncRNAs and 232 expressed mRNAs were identified (fold change > 1.5 and P-value < 0.05). Altered genes were enriched in inflammation, immune response, blood–brain barrier, glutamatergic neurological effects, and neuroactive ligand-receptor, which may be associated with TBI-induced pathophysiologic changes in the long-term neurological deficits. The lncRNAs-mRNAs co-expression network was generated for 74 lncRNA-mRNA pairs, most of which are positive correlations. The lncRNA-miRNA-mRNA interaction network included 12 lncRNAs, 59 miRNAs, and 25 mRNAs. Numerous significantly altered lncRNAs and mRNAs in mice hippocampi were enriched in inflammation and immune response. Furthermore, these dysregulated lncRNAs and mRNAs may be promising therapeutic targets to overcome obstacles in long-term recovery following TBI.
Collapse
Affiliation(s)
- Zhaoyu Yang
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xuexuan Li
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Weikang Luo
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yao Wu
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Tao Tang
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Tao Tang,
| | - Yang Wang
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Yang Wang,
| |
Collapse
|
24
|
Zhang Z, Wang Y, Wang X, Zhang H, Wang S. The up-regulation of LRIG1 expression inhibits the proliferation, apoptosis and invasion of glioma cells. Am J Transl Res 2022; 14:788-797. [PMID: 35273685 PMCID: PMC8902577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 11/22/2021] [Indexed: 06/14/2023]
Abstract
OBJECTIVE To illustrate the role of LRIG1 in regulating the Notch signaling pathway and glioma cell proliferation, apoptosis and invasion. METHODS The glioma cells (U373) were divided into control group, NC group and LRIG1 group. After transfection, the CCK-8 assay, Transwell assay, and Flow cytometry were used to explore the biological function of LRIG1 in glioma cells. At the end, Western blot was used to detect the expression of LRIG1, Notch1, Hes1, Bcl-2, and Bax. RESULTS The LRIG1 expression in U373 cells was remarkably lower than that in normal glial cells (P=0.019). The LRIG1 expression in the LRIG1 group was successfully increased when compared with that in the control group (P=0.004). The cell viability of the LRIG1 group was significantly lower than that of the NC group and control group at 24 h, 48 h, and 72 h (P=0.040, 0.025; P=0.041, 0.041; P=0.035, 0.035) respectively. Increased LRIG1 expression level in glioma cells strongly inhibits cell migration in transwell experiment. Flow cytometry results indicated that the apoptosis rate of the LRIG1 group was critically higher than that of the NC group and control group (P=0.003; P=0.003). According to results of Western blot, the expression levels of Notch1, Hes1, Hes5, and Jagged1 in LRIG1 group were dramatically higher than that in NC group and control group (P=0.006, 0.013; P=0.025, 0.026; P=0.001, 0.004; P=0.025, 0.027; P=0.029, 0.021) reespectively. While Bax expression in LRIG1 group was lower than that of NC group and control group (P=0.018, 0.021). CONCLUSION The up-regulation of LRIG1 can inhibit the proliferation and migration of glioma cells and promote apoptosis by regulating the Notch signaling pathway.
Collapse
Affiliation(s)
- Zhiyong Zhang
- Department of Neurosurgery, Caoxian People’s HospitalHeze 274400, Shandong, China
| | - Yuanyuan Wang
- Department of Oncology, Linyi People’s HospitalLinyi 251500, Shandong, China
| | - Xianxing Wang
- Department of Neurosurgery, Yuncheng County People’s HospitalHeze 274700, Shandong, China
| | - Hongguang Zhang
- Department of Neurosurgery, Gaotang People’s HospitalLiaocheng 252800, Shandong, China
| | - Shuai Wang
- Department of Neurosurgery, First Hospital of ZiboZibo 255200, Shandong, China
| |
Collapse
|
25
|
Valsartan Regulates PI3K/AKT Pathways through lncRNA GASL1 to Improve Isoproterenol-Induced Heart Failure. DISEASE MARKERS 2022; 2022:1447399. [PMID: 35035607 PMCID: PMC8758289 DOI: 10.1155/2022/1447399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/08/2021] [Accepted: 12/14/2021] [Indexed: 12/04/2022]
Abstract
Objective This study is aimed at determining the expression and function of the GASL1 and PI3K/AKT pathways in isoproterenol- (ISO-) induced heart failure (HF). To determine the moderating effect of valsartan (VAL) on the progression of ISO-induced HF and to elucidate the related mechanism. Materials and Methods First, in in vivo experiment, we examined the effect of VAL on cardiac function in rats with ISO-induced HF. Similarly, quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot were used to detect the effect of VAL on ISO-treated rat primary cardiomyocytes. Then, si-GASL1-transfected primary cardiomyocytes were constructed and Ad-si-GASL1 was injected through rat tail vein to achieve the effect of lowering GASL1 expression, so as to investigate the role of GASL1 in VAL's treatment of ISO-induced HF. Results In ISO-induced HF rat models, the GASL1 decreased while PI3K and p-AKT expressions were abnormally elevated and cardiac function deteriorated, and VAL was able to reverse these changes. In primary cardiomyocytes, ISO induces apoptosis of cardiomyocytes, and expression of GASL1 decreased while PI3K and p-AKT were abnormally elevated, which can be reversed by VAL. The transfection of primary cardiomyocytes with si-GASL1 confirmed that GASL1 affected the expression of PI3K, p-AKT, and the apoptosis of primary cardiomyocytes. Rat myocardium injected with Ad-si-GASL1 was found to aggravate the cardiac function improved by VAL. Conclusions This study was the first to confirm that VAL improves ISO-induced HF by regulating the PI3K/AKT pathway through GASL1. And this study demonstrated a significant correlation between HF, VAL, GASL1, and the PI3K/AKT pathway.
Collapse
|
26
|
Zhao Y, Li F, Li S, Ji J, Qiao W, Fang J. Aluminum chloride induces G0/G1 phase arrest via regulating the reactive oxygen species-depended non-canonical STAT1 pathway in hFOB1.19 cells. Hum Exp Toxicol 2022; 41:9603271221129846. [PMID: 36154299 DOI: 10.1177/09603271221129846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Treatment with aluminum chloride (AlCl3) suppresses the growth of osteoblastic cells; however, the molecular mechanisms underlying the impact of AlCl3 on cell growth have not been fully characterized. In this study, we observed that exposure of hFOB1.19 cells to AlCl3 arrested cells at G0/G1 phase by inducing p21 expression. Further studies indicated that AlCl3 upregulated the phosphorylation level of signal transducer and activator of transcription 1 (STAT1) at serine 727 site (Ser727). By chromatin immunoprecipitation and electrophoretic mobility shift assay, we found that AlCl3 promoted STAT1/DNA binding activity to p21 promoter, thus resulting in the upregulation of p21. Moreover, siRNA-mediated knockdown of STAT1 attenuated p21 level induced by AlCl3. Notably, using hFOB1.19 cells stably expressing dominant-negative STAT1 (Ser727Ala), we demonstrated that phosphorylation of STAT1 at Ser727 site is required for p21-mediated cycle arrest induced by AlCl3. Mechanism investigation indicated that AlCl3 stimulated the phosphorylation of JNK, and administration of JNK inhibitor SP600125 prevented AlCl3-induced G0/G1 arrest through suppressing the phosphorylation of STAT1. Notably, pretreatment with N-acetyl-cysteine, a reactive oxygen species scavenger, conferred a significantly inhibitory effect on AlCl3-mediated activation of JNK/STAT1 signaling pathway. Taken together, our findings provide the molecular mechanism for G0/G1 arrest induced by AlCl3 in osteoblastic cells.
Collapse
Affiliation(s)
- Y Zhao
- Department of Orthopaedics, 12561The First Affiliated Hospital of Henan Polytechnic University. The Second People's Hospital of Jiaozuo City, Jiaozuo, P.R. China
| | - F Li
- Otolaryngology Head and Neck Surgery, 12561The First Affiliated Hospital of Henan Polytechnic University. The Second People's Hospital of Jiaozuo City, Jiaozuo, P.R. China
| | - S Li
- Department of Orthopaedics, 12561The First Affiliated Hospital of Henan Polytechnic University. The Second People's Hospital of Jiaozuo City, Jiaozuo, P.R. China
| | - J Ji
- Department of Orthopaedics, 618971The Central Hospital of Kaifeng City, Kaifeng, P.R. China
| | - W Qiao
- Department of Orthopaedics, 12561The First Affiliated Hospital of Henan Polytechnic University. The Second People's Hospital of Jiaozuo City, Jiaozuo, P.R. China
| | - J Fang
- Department of Orthopaedics, 12561The First Affiliated Hospital of Henan Polytechnic University. The Second People's Hospital of Jiaozuo City, Jiaozuo, P.R. China
| |
Collapse
|
27
|
Li G, Wang Y, Wang J, Chen G, Wang H. Long non-coding RNA placenta‑specific protein 2 regulates micorRNA-19a/tumor necrosis factor α to participate in polycystic ovary syndrome. Bioengineered 2022; 13:856-862. [PMID: 34967266 PMCID: PMC8805902 DOI: 10.1080/21655979.2021.2013722] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 11/25/2021] [Indexed: 01/21/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is a type of hormonal disorder that affects about 5-20% of females at their reproductive age worldwide. MicorRNA-19a (miR-19a) is a well-characterized miRNA in cancer biology and its function is mainly mediated by targeting tumor necrosis factor α (TNF-α), which plays critical roles in PCOS. Our preliminary analysis predicted the potential interaction between miR-19a and long non-coding RNA (lncRNA) placenta‑specific protein 2 (PLAC2). Therefore, this study aimed to explore the role of PLAC2 in PCOS. Ovarian tissues were collected from 62 PCOS patients and 62 healthy females. Granulosa-like tumor cells (KGN) was prepared, and transient transfections was conducted. Dual-luciferase activity assay was used to investigate the interaction between PLAC2 and miR-19a. qPCR assays were performed for the expression analysis of miR-19a/TNF-α. In addition, Western blot analysis and cell apoptosis assay were conducted. The results showed that PLAC2 was upregulated in PCOS. PLAC2 and miR-19a showed a direct interaction, while overexpression of PLAC2 and miR-19a did not affect the expression of each other in KGN cells. Instead, overexpression of PLAC2 led to upregulated TNF-α, which is a target of miR-19a. Cell apoptosis analysis showed that PLAC2 and TNF-α promoted the apoptosis of KGN cells. Overexpression of miR-19a played an opposite role. In addition, the overexpression of PLAC2 reduced the effects of overexpression of miR-19a. Therefore, PLAC2 may regulate miR-19a/TNF-α to participate in PCOS.
Collapse
Affiliation(s)
- Gang Li
- Department of Obstetrics and Gynecology, Huai ‘An Maternal and Child Health Hospital, Huai ‘An City, PR. China
| | - Yongli Wang
- Global Health Institute, Xi’an Jiaotong University, Xian City, PR. China
| | - Jingyuan Wang
- Department of Clinical Laboratory, First Affiliated Hospital of Xi’an Jiaotong University, Xian City, PR. China
| | - Gong Chen
- Department of Obstetrics and Gynecology, Huai ‘An Maternal and Child Health Hospital, Huai ‘An City, PR. China
| | - Haiyan Wang
- Department of Reproductive Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xian City, PR. China
| |
Collapse
|
28
|
Hu KQ, Ao XS. Long non-coding RNA DLGAP1 antisense RNA 1 accelerates glioma progression via the microRNA-628-5p/DEAD-box helicase 59 pathway. Clinics (Sao Paulo) 2022; 77:100002. [PMID: 35113786 PMCID: PMC8903805 DOI: 10.1016/j.clinsp.2021.100002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 10/15/2021] [Indexed: 12/03/2022] Open
Abstract
OBJECTIVES Abnormal expression of long non-coding RNAs (lncRNAs) plays a prominent role in glioma progression. However, the biological function and mechanism of lncRNA DLGAP1 antisense RNA 1 (DLGAP1-AS1) in gliomas are still unknown. METHODS The authors assessed DLGAP1-AS1 and miR-628-5p expression in glioma tissues and cell lines using quantitative real-time polymerase chain reaction (qRT-PCR) and evaluated their effects on glioma cell proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) using the cell counting kit-8 (CCK-8) assay, 5-Ethynyl-2'-deoxyuridine (EdU) assay, Transwell assay, and western blot, respectively. The expression of DEAD-box helicase 59 (DDX59) was quantified using western blotting, and a dual-luciferase reporter gene assay was performed to detect the interaction between DLGAP1-AS1 and miR-628-5p. RESULTS The authors observed increased DLGAP1-AS1 expression in glioma tissues and cell lines with higher WHO grades and shorter survival time. DLGAP1-AS1 promoted the proliferation, migration, invasion, and EMT of glioma cells, while miR-628-5p counteracted these effects. The authors identified DLGAP1-AS1 as a molecular sponge of miR-628-5p in glioma cells as the biological functions of DLGAP1-AS1 are partially mediated via miR-628-5p. In addition, DLGAP1-AS1 upregulated DDX59 expression by inhibiting miR-628-5p expression. CONCLUSION The DLGAP1-AS1/miR-628-5p/DDX59 axis regulates glioma progression.
Collapse
Affiliation(s)
- Ke-Qi Hu
- Department of Neurosurgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, China
| | - Xiang-Sheng Ao
- Department of Neurosurgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, China.
| |
Collapse
|
29
|
Liu L, Xu Q, Xiong Y, Deng H, Zhou J. LncRNA LINC01094 contributes to glioma progression by modulating miR-224-5p/CHSY1 axis. Hum Cell 2022; 35:214-225. [PMID: 34716872 DOI: 10.1007/s13577-021-00637-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 05/07/2021] [Indexed: 01/20/2023]
Abstract
Glioma serves as the most common malignancy influencing modern people and is associated with severe morbidity and high mortality. Long non-coding RNAs (lncRNAs) as crucial regulators participate in multiple cancer progression. However, the role of lncRNA LINC01094 in the development of glioma remains unclear. Here, we aimed to explore the effect of lncRNA LINC01094 on the glioma progression and the underlying mechanism. Significantly, we revealed that the expression levels of LINC01094 were elevated in the glioma patient tissues compared to adjacent normal tissues. The LINC01094 expression was enhanced in the glioma cell lines. The depletion of LINC01094 inhibited cell viability and colony formation in the glioma cells. Meanwhile, the migration and invasion of glioma cells were impaired by the depletion of LINC01094. Mechanically, we identified that LINC01094 was able to sponge the miR-224-5p in the glioma cells and miR-224-5p inhibitor could reverse the effect of LINC01094 on glioma progression. In addition, miR-224-5p targeted CHSY1 and LINC01094 up-regulated CHSY1 by targeting miR-224-5p in the glioma cells. LINC01094 promoted glioma progression by the positive regulation of CHSY1. Moreover, tumorigenicity analysis showed that LINC01094 enhanced tumor growth of glioma in vivo. Thus, we conclude that lncRNA LINC01094 promotes glioma progression by modulating miR-224-5p/CHSY1 axis. Our finding provides new insights into the mechanism by which lncRNA LINC01094 contributes to the development of glioma, improving the understanding of lncRNA LINC01094 and glioma. LncRNA LINC01094, miR-224-5p, and CHSY1 may serve as potential targets for glioma.
Collapse
Affiliation(s)
- Luotong Liu
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Qian Xu
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yu Xiong
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Huajiang Deng
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jie Zhou
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China.
| |
Collapse
|
30
|
Zhao X, He X, Wang M, Zhang H, Cheng J, Wang H. LncRNA PLAC2 upregulates CDK6 by directly targeting miR-29C to promote cell proliferation in lung squamous cell carcinoma. Crit Rev Eukaryot Gene Expr 2022; 32:55-67. [DOI: 10.1615/critreveukaryotgeneexpr.2022044134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
31
|
Fu T, Yang Y, Mu Z, Sun R, Li X, Dong J. Silencing lncRNA LINC01410 suppresses cell viability yet promotes apoptosis and sensitivity to temozolomide in glioblastoma cells by inactivating PTEN/AKT pathway via targeting miR-370-3p. Immunopharmacol Immunotoxicol 2021; 43:680-692. [PMID: 34435542 DOI: 10.1080/08923973.2021.1966031] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 07/31/2021] [Indexed: 02/04/2023]
Abstract
BACKGROUND Long non-coding RNAs (LncRNAs) are involved in glioblastoma (GBM), but the role of long intergenic non-protein coding RNA 01410 (lncRNA LINC01410) is poorly understood. METHODS The expression of LINC01410 in GBM tissues and cells was analyzed. After transfection or temozolomide (TMZ) treatment, the cell viability and apoptosis were detected using cell counting kit-8 assay and flow cytometry. The targeting relationship between LINC01410 and microRNA (miR)-370-3p was confirmed by dual-luciferase reporter assay. Expressions of LINC01410, miR-370-3p and drug resistance- and Phosphatase and Tensin Homolog (PTEN)/AKT pathway-related factors were evaluated by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot. RESULTS LINC01410 expression was upregulated in GBM, and silencing of LINC01410 decreased cell viability. A slowed decreased trend in cell viability yet an increased half maximal inhibitory concentration (IC50 for TMZ) value and increased expressions of drug resistance-related factors as well as LINC01410 were found in TMZ-resistant GBM cells. Silencing of LINC01410 also decreased the IC50 value yet promoted the sensitivity and apoptosis in TMZ-resistant cells, while upregulating the expression of PTEN and downregulating the phosphorylation of AKT. MiR-370-3p could competitively bind to LINC01410 and its expression was decreased in both parental and TMZ-resistant GBM cells. Downregulation of miR-370-3p reversed the effects of LINC01410 silencing on cell viability, apoptosis and the expressions of miR-370-3p and PTEN/AKT pathway-related factors. CONCLUSION Silencing of LINC01410 inhibits cell viability yet enhances apoptosis and sensitivity to TMZ in GBM cells by inactivating PTEN/AKT pathway via targeting miR-370-3p.
Collapse
Affiliation(s)
- Tingkai Fu
- Department of Neurosurgery, People's Hospital of Rizhao, Rizhao City, China
| | - Yunxue Yang
- Department of Neurosurgery, People's Hospital of Rizhao, Rizhao City, China
| | - Zhenxin Mu
- Department of Neurosurgery, People's Hospital of Rizhao, Rizhao City, China
| | - Rongwei Sun
- Department of Neurosurgery, People's Hospital of Rizhao, Rizhao City, China
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China
| | - Jun Dong
- Department of Neurosurgery, People's Hospital of Rizhao, Rizhao City, China
| |
Collapse
|
32
|
Tan JK, Ma XF, Wang GN, Jiang CR, Gong HQ, Liu H. LncRNA MIAT knockdown alleviates oxygen-glucose deprivation‑induced cardiomyocyte injury by regulating JAK2/STAT3 pathway via miR-181a-5p. J Cardiol 2021; 78:586-597. [PMID: 34489160 DOI: 10.1016/j.jjcc.2021.08.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 07/25/2021] [Accepted: 07/29/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND Coronary artery disease (CAD) is a common heart disease with high incidence and mortality. Myocardial ischemia is the main type of CAD, which negatively affects health worldwide. The aim of the present study was to investigate the function and mechanism of myocardial infarction-associated transcript (MIAT) in myocardial ischemia. METHODS Human cardiomyocytes (HCM) were treated with oxygen-glucose deprivation (OGD) to set the in vitro model and mouse myocardial ischemia/reperfusion (I/R) was set for in vivo model. Cell viability and apoptosis were detected by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide assay, flow cytometry, and immunofluorescence analysis. Inflammatory cytokines levels were detected by enzyme-linked immunosorbent assay. Gene and protein expressions were identified by quantitative real time-polymerase chain reaction or Western blotting. The interaction of MIAT, miR-181a-5p, and janus kinase 2 (JAK2) was identified by dual-luciferase report assay. Mouse heart tissues histopathological condition were observed by hematoxylin and eosin assays. RESULTS Expression of MIAT and JAK2 were increased in OGD-treated HCM and mice of I/R model group, and miR-181a-5p was decreased. MIAT silencing could reverse the OGD treatment induced cell proliferation inhibition, cleaved caspase-3 and Bcl2-associated X (Bax) levels increased, while those of B-cell lymphoma-2 (Bcl-2) and mitochondria's cyt-C decreased. Besides, MIAT knockdown attenuated the OGD-induced increase of tumor necrosis factor-α, interleukin (IL)-1β, and IL-6 levels. Moreover, MIAT targeted miR-181a-5p to enhance the expression of JAK2 and signal Transducer and Activator of Transcription 3 (STAT3), and miR-181a-5p overexpression promoted proliferation, whereas it inhibited apoptosis in OGD-induced cardiomyocytes. Furthermore, the regulatory effects of MIAT knockdown in cell proliferation, apoptosis, and inflammatory injury was reversed by inhibition of miR-181a-5p or overexpression of JAK2 in OGD-treated HCM. Knockdown of MIAT reduced myocardial injury caused by I/R treatment in vivo. CONCLUSION MIAT knockdown inhibited apoptosis and inflammation by regulating JAK2/STAT3 signaling pathway via targeting miR-181a-5p in myocardial ischemia model. MIAT can be a possible therapeutic target for controlling the progression of myocardial ischemia.
Collapse
Affiliation(s)
- Jian-Kai Tan
- The Affiliated Nanhua Hospital, Department of cardiology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Xiao-Feng Ma
- The Affiliated Nanhua Hospital, Department of cardiology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Guang-Neng Wang
- The Affiliated Nanhua Hospital, Department of cardiology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Chang-Rong Jiang
- The Affiliated Nanhua Hospital, Department of cardiology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Hui-Qin Gong
- The Affiliated Nanhua Hospital, Department of cardiology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Huan Liu
- The Affiliated Nanhua Hospital, Department of cardiology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
33
|
Kang J, Brajanovski N, Chan KT, Xuan J, Pearson RB, Sanij E. Ribosomal proteins and human diseases: molecular mechanisms and targeted therapy. Signal Transduct Target Ther 2021; 6:323. [PMID: 34462428 PMCID: PMC8405630 DOI: 10.1038/s41392-021-00728-8] [Citation(s) in RCA: 204] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 07/12/2021] [Accepted: 07/30/2021] [Indexed: 02/07/2023] Open
Abstract
Ribosome biogenesis and protein synthesis are fundamental rate-limiting steps for cell growth and proliferation. The ribosomal proteins (RPs), comprising the structural parts of the ribosome, are essential for ribosome assembly and function. In addition to their canonical ribosomal functions, multiple RPs have extra-ribosomal functions including activation of p53-dependent or p53-independent pathways in response to stress, resulting in cell cycle arrest and apoptosis. Defects in ribosome biogenesis, translation, and the functions of individual RPs, including mutations in RPs have been linked to a diverse range of human congenital disorders termed ribosomopathies. Ribosomopathies are characterized by tissue-specific phenotypic abnormalities and higher cancer risk later in life. Recent discoveries of somatic mutations in RPs in multiple tumor types reinforce the connections between ribosomal defects and cancer. In this article, we review the most recent advances in understanding the molecular consequences of RP mutations and ribosomal defects in ribosomopathies and cancer. We particularly discuss the molecular basis of the transition from hypo- to hyper-proliferation in ribosomopathies with elevated cancer risk, a paradox termed "Dameshek's riddle." Furthermore, we review the current treatments for ribosomopathies and prospective therapies targeting ribosomal defects. We also highlight recent advances in ribosome stress-based cancer therapeutics. Importantly, insights into the mechanisms of resistance to therapies targeting ribosome biogenesis bring new perspectives into the molecular basis of cancer susceptibility in ribosomopathies and new clinical implications for cancer therapy.
Collapse
Affiliation(s)
- Jian Kang
- grid.1055.10000000403978434Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC Australia ,grid.1008.90000 0001 2179 088XSir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC Australia
| | - Natalie Brajanovski
- grid.1055.10000000403978434Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC Australia
| | - Keefe T. Chan
- grid.1055.10000000403978434Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC Australia ,grid.1008.90000 0001 2179 088XSir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC Australia
| | - Jiachen Xuan
- grid.1055.10000000403978434Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC Australia ,grid.1008.90000 0001 2179 088XSir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC Australia
| | - Richard B. Pearson
- grid.1055.10000000403978434Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC Australia ,grid.1008.90000 0001 2179 088XSir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC Australia ,grid.1002.30000 0004 1936 7857Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, VIC Australia
| | - Elaine Sanij
- grid.1055.10000000403978434Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC Australia ,grid.1008.90000 0001 2179 088XSir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Clinical Pathology, University of Melbourne, Melbourne, VIC Australia ,grid.1073.50000 0004 0626 201XSt. Vincent’s Institute of Medical Research, Fitzroy, VIC Australia
| |
Collapse
|
34
|
Bian Z, Ji W, Xu B, Huo Z, Huang H, Huang J, Jiao J, Shao J, Zhang X. Noncoding RNAs involved in the STAT3 pathway in glioma. Cancer Cell Int 2021; 21:445. [PMID: 34425834 PMCID: PMC8381529 DOI: 10.1186/s12935-021-02144-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 08/11/2021] [Indexed: 01/03/2023] Open
Abstract
Glioma is the most common malignant primary brain tumour in adults. Despite improvements in neurosurgery and radiotherapy, the prognosis of glioma patients remains poor. One of the main limitations is that there are no proper clinical therapeutic targets for glioma. Therefore, it is crucial to find one or more effective targets. Signal transducer and activator of transcription 3 (STAT3) is a member of the STAT family of genes. Abnormal expression of STAT3 is involved in the process of cell proliferation, migration, invasion, immunosuppression, angiogenesis, dryness maintenance, and resistance to radiotherapy and chemotherapy in glioma. Therefore, STAT3 has been considered an ideal therapeutic target in glioma. Noncoding RNAs (ncRNAs) are a group of genes with limited or no protein-coding capacity that can regulate gene expression at the epigenetic, transcriptional and posttranscriptional level. In this review, we summarized the ncRNAs that are correlated with the ectopic expression of STAT3 in glioma.
Collapse
Affiliation(s)
- Zheng Bian
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, Wuxi, People's Republic of China
| | - Wei Ji
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, Wuxi, People's Republic of China
| | - Bin Xu
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, Wuxi, People's Republic of China
| | - Zhengyuan Huo
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, Wuxi, People's Republic of China
| | - Hui Huang
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, Wuxi, People's Republic of China
| | - Jin Huang
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, Wuxi, People's Republic of China
| | - Jiantong Jiao
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, Wuxi, People's Republic of China
| | - Junfei Shao
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, Wuxi, People's Republic of China.
| | - Xiaolu Zhang
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, Wuxi, People's Republic of China.
| |
Collapse
|
35
|
Zhao J, Jiang Y, Chen L, Ma Y, Zhang H, Zhou J, Li H, Jing Z. The EIF4A3/CASC2/RORA Feedback Loop Regulates the Aggressive Phenotype in Glioblastomas. Front Oncol 2021; 11:699933. [PMID: 34408982 PMCID: PMC8366401 DOI: 10.3389/fonc.2021.699933] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 07/16/2021] [Indexed: 12/25/2022] Open
Abstract
Glioblastoma (GBM) is a common and refractory subtype of high-grade glioma with a poor prognosis. The epithelial-mesenchymal transition (EMT) is an important cause of enhanced glioblastoma invasiveness and tumor recurrence. Our previous study found that retinoic acid receptor-related orphan receptor A (RORA) is a nuclear receptor and plays an important role in inhibiting proliferation and tumorigenesis of glioma. We further confirmed RORA was downregulated in GBM. Thus, we determined whether RORA was involved in the migration, invasion, and EMT of GBM. Human GBM cell lines, U87 and T98G, and patient-derived glioma stem cells (GSCs), GSC2C and GSC4D, were used for in vitro and in vivo experiments. The expressions of RORA, CASC2, and EIF4A3 in GBM cells and GSCs were detected by RT-qPCR and western blotting. The biological effects of RORA, CASC2, and EIF4A3 on GBM migration, invasion, and EMT were evaluated using the migration assay, transwell assay, immunofluorescence staining, and xenograft experiments. We found that RORA inhibited the migration, invasion, and EMT of GBM. CASC2 could bind to, maintain the stability, and promote the nuclear translocation of RORA protein. EIF4A3 could downregulate CASC2 expression via inducing its cleavage, while RORA transcriptionally inhibited EIF4A3 expression, which formed a feedback loop among EIF4A3/CASC2/RORA. Moreover, gene set enrichment analysis (GSEA) and in vitro and in vivo experiments showed RORA inhibited the aggressiveness of GBM by negatively regulating the TGF-β1/Smad signaling pathway. Therefore, The EIF4A3/CASC2/RORA feedback loop regulated TGF-β1/Smad signaling pathway might become a promising therapeutic strategy for GBM treatment.
Collapse
Affiliation(s)
- Junshuang Zhao
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Yang Jiang
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lian Chen
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Yue Ma
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Haiying Zhang
- International Education College, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Jinpeng Zhou
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Hao Li
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Zhitao Jing
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
36
|
Xia Y, Zhang X, Sun D, Gao Y, Zhang X, Wang L, Cai Q, Wang Q, Sun J. Effects of water-soluble components of atmospheric particulates from rare earth mining areas in China on lung cancer cell cycle. Part Fibre Toxicol 2021; 18:27. [PMID: 34340691 PMCID: PMC8330054 DOI: 10.1186/s12989-021-00416-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 06/02/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND This study aims to investigate the effects of water soluble particulate matter (WSPM) on the viability and protein expression profile of human lung adenocarcinoma cell A549 in the Bayou Obo rare earth mining area, and explore the influence of WSPM on the A549 cell cycle. RESULTS It was found that WSPM can inhibit the viability of A549 cells and induce cell arrest in the G2/M phase. Compared with controls, exposure to WSPM10 and WSPM2.5 induced 134 and 116 proteins to be differentially expressed in A549 cells, respectively. In addition, 33 and 31 differentially expressed proteins were further confirmed, and was consistent with the proteomic analysis. The most prominent enrichment in ribosome-associated proteins were presented. When RPL6, RPL13, or RPL18A gene expression was inhibited, A549 cells were arrested in the G1 phase, affecting the expression of Cyclin D1, p21, RB1, Cyclin A2, Cyclin B1, CDC25A, CDK2, CHEK2 and E2F1. Furthermore, the La3+, Ce3+, Nd3+ and F- in WSPM also inhibited the viability of A549 cells. After 24 h of exposure to 2 mM of NaF, A549 cells were also arrested in the G2/M phase, while the other three compounds did not have this effect. These four compounds affected the cell cycle regulatory factors in A549 cells, mainly focusing on effecting the expression of CDK2, CDK4, RB1, ATM, TP53 and MDM2 genes. These results are consistent with the those from WSPM exposure. CONCLUSIONS These results revealed that WSPM from rare earth mines decreased the viability of A549 cells, and induced cell cycle G2/M phase arrest, and even apoptosis, which may be independent of the NF-κB/MYD88 pathway, and be perceived by the TLR4 receptor. The dysfunction of the cell cycle is correlated to the down-expression of ribosomal proteins (RPs). However, it is not the direct reason for the A549 cell arrest in the G2/M phase. La3+, Ce3+, and F- are probably the main toxic substances in WSPM, and may be regulate the A549 cell cycle by affecting the expression of genes, such as MDM2, RB1, ATM, TP53, E2F1, CDK2 and CDK4. These results indicate the importance for further research into the relationship between APM and lung cancer.
Collapse
Affiliation(s)
- Yuan Xia
- School of Public Health, Inner Mongolia Autonomous Region, Jinshan Economic and Technological Development Zone, Inner Mongolia Medical University, Inner Mongolia Autonomous Region, 010010, Hohhot, China
| | - Xulong Zhang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Dejun Sun
- Inner Mongolia People's Hospital, Inner Mongolia Autonomous Region, Hohhot, China
| | - Yumin Gao
- School of Public Health, Inner Mongolia Autonomous Region, Jinshan Economic and Technological Development Zone, Inner Mongolia Medical University, Inner Mongolia Autonomous Region, 010010, Hohhot, China
| | - Xiaoe Zhang
- School of Public Health, Inner Mongolia Autonomous Region, Jinshan Economic and Technological Development Zone, Inner Mongolia Medical University, Inner Mongolia Autonomous Region, 010010, Hohhot, China
| | - Li Wang
- School of Public Health, Inner Mongolia Autonomous Region, Jinshan Economic and Technological Development Zone, Inner Mongolia Medical University, Inner Mongolia Autonomous Region, 010010, Hohhot, China
| | - Qingjun Cai
- School of Public Health, Inner Mongolia Autonomous Region, Jinshan Economic and Technological Development Zone, Inner Mongolia Medical University, Inner Mongolia Autonomous Region, 010010, Hohhot, China
| | - Qihao Wang
- School of Public Health, Inner Mongolia Autonomous Region, Jinshan Economic and Technological Development Zone, Inner Mongolia Medical University, Inner Mongolia Autonomous Region, 010010, Hohhot, China
| | - Juan Sun
- School of Public Health, Inner Mongolia Autonomous Region, Jinshan Economic and Technological Development Zone, Inner Mongolia Medical University, Inner Mongolia Autonomous Region, 010010, Hohhot, China.
| |
Collapse
|
37
|
Cai Y, Wang M, Cui Y, Tan Z, Jiang Y. Differential Expression Profile of lncRNA in Glioma Cells and the Effect of lncRNA NKX3-1 on Glioma Cells Through Fem1b/SPDEF Pathway. Front Oncol 2021; 11:706863. [PMID: 34350121 PMCID: PMC8328487 DOI: 10.3389/fonc.2021.706863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 06/28/2021] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE To investigate the differential expression of lncRNA in glioma cells, as well as the effect of lncRNA NKX3-1 on glioma cells. METHODS Glioma-related data were first downloaded from the TCGA database and analyzed using bioinformatics, after which the lncRNA NKX3-1 was chosen for further experiments. The expression of the lncRNA NKX3-1 in glioma tumor samples was detected using qRT-PCR. The subcellular localization of lncRNA NKX3-1 was determined using fluorescence in situ hybridization (FISH). CCK-8, flow cytometry, cell scratch, and transwell assays were used to detect cell proliferation, apoptosis, and invasion. The downstream pathway of lncRNA NKX3-1 was investigated using luciferase assays and detected using western blot, transwell, and cell scratch assays. RESULTS The differential expression profile of lncRNA in glioma was obtained. NKX3-1 lncRNA was found to be significantly increased in glioma tumor tissues. LncRNA NKX3-1 was found in the nucleus. Proliferation, invasion, and migration of glioma cells were significantly increased (P <0.05) in the lncRNA NKX3-1 overexpression group, while apoptosis ability was significantly decreased (P <0.05). Tumor volume and weight were significantly increased in the lncRNA NKX3-1 overexpression group in nude mice (P <0.05). LncRNA NKX3-1 significantly increased the luciferase activity of Fem1b 3'-UTR-WT reporter genes (P <0.05) as well as the levels of SPDEF protein (P <0.05). The protein level of FEM1B was significantly reduced. Cell invasion and migration were significantly increased (P <0.05) in the lncRNA NKX3-1 overexpression group plus SPDEF group. CONCLUSION We investigated the differential expression profile of lncRNAs in glioma and discovered that the lncRNA NKX3-1 plays an important role in cancer promotion via the Fem1b/SPDEF pathway.
Collapse
Affiliation(s)
| | | | | | | | - Yugang Jiang
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
38
|
Sun Y, Wang X, Bu X. LINC02381 contributes to cell proliferation and hinders cell apoptosis in glioma by transcriptionally enhancing CBX5. Brain Res Bull 2021; 176:121-129. [PMID: 34274429 DOI: 10.1016/j.brainresbull.2021.07.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/25/2021] [Accepted: 07/12/2021] [Indexed: 01/11/2023]
Abstract
Glioma, featured with high incidence and low survival rate, is the most common type of primary brain tumor, severely affecting human life worldwide. LINC02381 is an interesting lncRNA functioning as oncogenic lncRNA in some cancers but as tumor-suppressor in others, but no report demonstrates its association with and function in glioma. Intriguingly, we found in a bioinformatics website LncRNADisease that LINC02381 was closely related to malignant glioma, so this study aimed to figure out the expression and function of LINC02381 in glioma. By RT-qPCR, we confirmed LINC02381 upregulation in glioma cells. Functional experiments demonstrated that LINC02381 knockdown repressed glioma cell proliferation and induced apoptosis. Boinformatics tools and RT-qPCR revealed the positive correlation between LINC02381 and CBX5 in glioma cells. More importantly, we confirmed that LINC02381 could interact and work synergistically with CEBPβ to bind to CBX5 promoter and activate CBX5 transcriptionally. Additionally, rescue experiments indicated that CBX5 up-regulation reversed the decline in cell proliferation and the augment in cell apoptosis caused by LINC02381 knockdown. To conclude, LINC02381 could facilitate CBX5 transcription via interaction with CEBPβ, thus exerting its oncogenic role in glioma cells, which could contribute to better understanding of glioma.
Collapse
Affiliation(s)
- Yong Sun
- Department of Neurosurgery, Henan Provincial People's Hospital, No.7 Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan, China
| | - Xinjun Wang
- Department of Neurosurgery, Fifth Affiliated Hospital of Zhengzhou University, No.3 Kangfuqian Street, Erqi District, Zhengzhou, Henan, 450052, China
| | - Xingyao Bu
- Department of Neurosurgery, Henan Provincial People's Hospital, No.7 Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan, China.
| |
Collapse
|
39
|
Kai H, Wu Q, Yin R, Tang X, Shi H, Wang T, Zhang M, Pan C. LncRNA NORAD Promotes Vascular Endothelial Cell Injury and Atherosclerosis Through Suppressing VEGF Gene Transcription via Enhancing H3K9 Deacetylation by Recruiting HDAC6. Front Cell Dev Biol 2021; 9:701628. [PMID: 34307380 PMCID: PMC8301222 DOI: 10.3389/fcell.2021.701628] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 06/08/2021] [Indexed: 11/15/2022] Open
Abstract
Coronary artery disease (CAD) is a major atherosclerotic cardiovascular disease and the leading cause of mortality globally. Long non-coding RNAs (lncRNAs) play crucial roles in CAD development. To date, the effect of lncRNA non-coding RNA activated by DNA damage (NORAD) on atherosclerosis in CAD remains unclear. The primary aim of this study was to investigate the effect of lncRNA NORAD on vascular endothelial cell injury and atherosclerosis. Here, ox-LDL-treated human umbilical vein endothelial cells (HUVECs) and high-fat-diet (HFD)-fed ApoE–/– mice were utilized as in vitro and in vivo models. The present study found that lncRNA NORAD expression was increased in ox-LDL-treated HUVECs and thoracic aorta of atherosclerotic mice, and knockdown of lncRNA NORAD alleviated vascular endothelial cell injury and atherosclerosis development in vitro and in vivo. Knockdown of lncRNA NORAD aggravated ox-LDL-reduced or atherosclerosis-decreased vascular endothelial growth factor (VEGF) expression in HUVECs and thoracic aorta of mice to ameliorate vascular endothelial cell injury and atherosclerosis development. Moreover, nucleus lncRNA NORAD suppressed VEGF gene transcription through enhancing H3K9 deacetylation via recruiting HDAC6 to the VEGF gene promoter in ox-LDL-treated HUVECs. In addition, VEGF reduced FUS (FUS RNA binding protein) expression by a negative feedback regulation in HUVECs. In summary, lncRNA NORAD enhanced vascular endothelial cell injury and atherosclerosis through suppressing VEGF gene transcription via enhancing H3K9 deacetylation by recruiting HDAC6. The findings could facilitate discovering novel diagnostic markers and therapeutic targets for CAD.
Collapse
Affiliation(s)
- Huihua Kai
- Department of Radiology, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Qiyong Wu
- Department of Thoracic and Cardiac Surgery, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Ruohan Yin
- Department of Radiology, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Xiaoqiang Tang
- Department of Radiology, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Haifeng Shi
- Department of Radiology, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Tao Wang
- Department of Radiology, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Ming Zhang
- Department of Radiology, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Changjie Pan
- Department of Radiology, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| |
Collapse
|
40
|
Ataei A, Arab SS, Zahiri J, Rajabpour A, Kletenkov K, Rizvanov A. Filtering of the Gene Signature as the Predictors of Cisplatin-Resistance in Ovarian Cancer. IRANIAN JOURNAL OF BIOTECHNOLOGY 2021; 19:e2643. [PMID: 34825010 PMCID: PMC8590720 DOI: 10.30498/ijb.2021.209370.2643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
BACKGROUND Gene expression profiling and prediction of drug responses based on the molecular signature indicate new molecular biomarkers which help to find the most effective drugs according to the tumor characteristics. OBJECTIVES In this study two independent datasets, GSE28646 and GSE15372 were subjected to meta-analysis based on Affymetrix microarrays. MATERIAL AND METHODS In-silico methods were used to determine differentially expressed genes (DEGs) in the previously reported sensitive and resistant A2780 cell lines to Cisplatin. Gene Fuzzy Scoring (GFS) and Principle Component Analysis (PCA) were then used to eliminate batch effects and reduce data dimension, respectively. Moreover, SVM method was performed to classify sensitive and resistant data samples. Furthermore, Wilcoxon Rank sum test was performed to determine DEGs. Following the selection of drug resistance markers, several networks including transcription factor-target regulatory network and miRNA-target network were constructed and Differential correlation analysis was performed on these networks. RESULTS The trained SVM successfully classified sensitive and resistant data samples. Moreover, Performing DiffCorr analysis on the sensitive and resistant samples resulted in detection of 27 and 25 significant (with correlation ≥|0.9|) pairs of genes that respectively correspond to newly constructed correlations and loss of correlations in the resistant samples. CONCLUSIONS Our results indicated the functional genes and networks in Cisplatin resistance of ovarian cancer cells and support the importance of differential expression studies in ovarian cancer chemotherapeutic agent responsiveness.
Collapse
Affiliation(s)
- Atousa Ataei
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia
| | - Seyed Shahriar Arab
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Javad Zahiri
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Azam Rajabpour
- Department of Molecular medicine, Pasteur Institute of Iran, Tehran, Iran
| | - Konstantin Kletenkov
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia
| | - Albert Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia
| |
Collapse
|
41
|
Li C, Yao Y, Long D, Lin X. KDELC1 and TRMT1 Serve as Prognosis-Related SARS-CoV-2 Proteins Binding Human mRNAs and Promising Biomarkers in Clear Cell Renal Cell Carcinoma. Int J Gen Med 2021; 14:2475-2490. [PMID: 34163216 PMCID: PMC8214210 DOI: 10.2147/ijgm.s312416] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 05/17/2021] [Indexed: 12/13/2022] Open
Abstract
Background SARS-CoV-2 proteins binding human mRNAs (SPBRs) have been proven to regulate a variety of tumor-related functions in different types of cancer. However, their biological roles and potential mechanisms in clear cell renal cell carcinoma (ccRCC) are still elusive. Herein, we investigate the expression and prognostic value of SPBRs in ccRCC through bioinformatics methods. Methods Data downloaded from the Cancer Genome Atlas (TCGA) database was used to screen differentially expressed SPBRs (DE-SPBRs) between ccRCC samples and noncancerous samples. Metascape was utilized to perform function and pathway enrichment analyses of these DE-SPBRs. Kaplan–Meier method of overall survival (OS) was used to assess the prognostic value of DE-SPBRs in ccRCC patients. Univariate and multivariate Cox regression analyses were applied to identify candidate SPBRs, which were independently associated with overall survival of ccRCC patients. Subsequently, several internationally renowned databases were employed to conduct a comprehensive analysis of candidate SPBRs to further investigate their roles and mechanisms in ccRCC. Results A total of 33 DE-SPBRs, including 18 upregulated SPBRs and 17 downregulated SPBRs, were screened between ccRCC samples and noncancerous samples. Among them, two candidate SPBRs, KDELC1 and TRMT1, were identified. Additionally, we observed that upregulated KDELC1/TRMT1 expression in ccRCC at both gene and protein levels was significantly associated with clinicopathological features. Furthermore, we found that KDELC1/TRMT1 genetic mutation has an unfavorable influence on prognosis of patients with ccRCC. Functional enrichment analysis revealed that KDELC1/TRMT1 was closely enriched in several vital biological processes and pathways. Finally, we noticed that KDELC1/TRMT1 was remarkably associated with immune infiltrates. Conclusion In summary, we screened DE-SPBRs of ccRCC, which were enriched mainly in various biological and signaling pathways with tumor progression. Furthermore, we identified two candidate DE-SPBRs (KDELC1 and TRMT1), which could serve as promising biomarkers and therapeutic targets of patients with ccRCC.
Collapse
Affiliation(s)
- Canxuan Li
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, People's Republic of China
| | - Yuzhi Yao
- Department of Breast Surgery and General Surgery, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong, People's Republic of China.,Guangzhou Women and Children's Medical Center, Department of Paediatric Surgery Clinic, Guangzhou, Guangdong, People's Republic of China
| | - Dan Long
- Respiratory medicine, Shenshan Central Hospital, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Shanwei, Guangdong, People's Republic of China
| | - Xiaobin Lin
- Department of Breast Surgery and General Surgery, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong, People's Republic of China.,Department of Breast Surgery, The First Affiliated Hospital of Jinan University, Guangdong, People's Republic of China
| |
Collapse
|
42
|
Zhang Y, Guo H, Zhang H. SNHG10/DDX54/PBX3 Feedback Loop Contributes to Gastric Cancer Cell Growth. Dig Dis Sci 2021; 66:1875-1884. [PMID: 32712782 DOI: 10.1007/s10620-020-06488-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 07/11/2020] [Indexed: 12/09/2022]
Abstract
BACKGROUND The importance of long noncoding RNAs (lncRNAs) has been identified in human cancers, such as emerged as tumor facilitator or tumor suppressor. Small nucleolar RNA host gene 10 (SNHG10) has been reported as an oncogenic lncRNA in hepatocellular carcinoma. However, its functional role and underlying mechanism in gastric cancer (GC) need to be further explored. AIMS Our study was conducted to investigate the function and molecular mechanism of SNHG10 in GC. METHODS SNHG10 expression was detected by qRT-PCR. The effect of SNHG10 on GC cell growth was assessed by colony formation, EdU, JC-1, flow cytometry, and wound-healing assays. The interaction between SNHG10 and PBX3 was confirmed through ChIP and luciferase reporter assay. RIP and RNA pull down assays was used to define the binding of DEAD-box helicase 54 (DDX54) to SNHG10 or PBX homeobox 3 (PBX3). RESULTS SNHG10 was expressed at a high level in GC cells. SNHG10 knockdown resulted in the inhibition on GC cell proliferation, migration but induced cell apoptosis. PBX3 could interact with SNHG10 promoter and thereby activate the expression of SNHG10. Subsequently, it was confirmed that SNHG10 positively modulated the expression of PBX3. Based on this, we found that DDX54 could bind to SNHG10 and PBX3, suggesting that SNHG10 maintained PBX3 mRNA stability through recruiting DDX54. Restoration assays indicated that PBX3 overexpression recovered SNHG10 silencing-induced inhibition on GC cell growth. CONCLUSIONS SNHG10 facilitates cell growth by affecting DDX54-mediated PBX3 mRNA stability in GC.
Collapse
Affiliation(s)
- Yunfei Zhang
- Deparetment of Pathology, the Affiliated Hospital of Northwest University for Nationalities, Second Province People's Hospital of Gansu, Lanzhou, 730000, Gansu, China.
| | - Hongyan Guo
- Deparetment of Pathology, the Affiliated Hospital of Northwest University for Nationalities, Second Province People's Hospital of Gansu, Lanzhou, 730000, Gansu, China
| | - Hong Zhang
- Deparetment of Pathology, the Affiliated Hospital of Northwest University for Nationalities, Second Province People's Hospital of Gansu, Lanzhou, 730000, Gansu, China
| |
Collapse
|
43
|
Wang C, Yu G, Xu Y, Liu C, Sun Q, Li W, Sun J, Jiang Y, Ye L. Knockdown of Long Non-Coding RNA HCP5 Increases Radiosensitivity Through Cellular Senescence by Regulating microRNA-128 in Gliomas. Cancer Manag Res 2021; 13:3723-3737. [PMID: 33994812 PMCID: PMC8113609 DOI: 10.2147/cmar.s301333] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/16/2021] [Indexed: 12/17/2022] Open
Abstract
Introduction Glioma is the most common malignant brain tumor in adults. Radiation is a key therapy in glioma. However, the radioresistance of glioma was a big challenge. HLA complex P5 (HCP5) has been reported dysregulated in several types of malignant tumor, including glioma. The role of HCP5 in the radiosensitivity of glioma is so far unknown. The present study aimed to investigate the effect of HCP5 on radiosensitivity in gliomas. Methods The levels of HCP5 and microRNA (miR)-128 were detected using qRT-PCR. The cell growth curve was used to show the cell proliferation and evaluate the radiosensitivity of glioma cells following exposure to X-ray. Senescence-associated β-galactosidase (SA-β-Gal) staining was used to test the cellular senescence. Luciferase reporter and RNA immunoprecipitation (RIP) assays were performed to determine the correlation between HCP5 and miR-128. Results HCP5 level of glioma cells was significantly higher than human astrocytes, whereas miR-128 level was lower in glioma cells. Besides, the HCP5 expression was increased in glioma tissues compared to normal brain tissues (NBTs). Knockdown of HCP5 inhibited cell proliferation and increased radiosensitivity in glioma cells. MiR-128 was predicted to be a target of HCP5. It was demonstrated that HCP5 directly bound to miR-128 and regulated its expression in glioma cells. Furthermore, the effects of HCP5 knockdown on radiosensitivity of glioma cells were attenuated by the inhibitor of miR-128. Conclusion These findings suggested that interaction between lncRNA HCP5 and microRNA-128 could regulate the radiosensitivity of glioma cells by intervening in cellular senescence. This might be used as the potential radio-sensitization targets for glioma therapy.
Collapse
Affiliation(s)
- Cuihong Wang
- Cancer Center, The Second Hospital of Shandong University, Jinan, Shandong, 250033, People's Republic of China
| | - Guanying Yu
- Department of Gastrointestinal Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, 250013, People's Republic of China
| | - Ying Xu
- Cancer Center, The Second Hospital of Shandong University, Jinan, Shandong, 250033, People's Republic of China
| | - Chengfei Liu
- Cancer Center, The Second Hospital of Shandong University, Jinan, Shandong, 250033, People's Republic of China
| | - Qian Sun
- Cancer Center, The Second Hospital of Shandong University, Jinan, Shandong, 250033, People's Republic of China
| | - Wenqing Li
- Cancer Center, The Second Hospital of Shandong University, Jinan, Shandong, 250033, People's Republic of China
| | - Junhua Sun
- Cancer Center, The Second Hospital of Shandong University, Jinan, Shandong, 250033, People's Republic of China
| | - Yuhua Jiang
- Cancer Center, The Second Hospital of Shandong University, Jinan, Shandong, 250033, People's Republic of China
| | - Lan Ye
- Cancer Center, The Second Hospital of Shandong University, Jinan, Shandong, 250033, People's Republic of China
| |
Collapse
|
44
|
Zhou H, Cao J, Yang F, Fan D, Li H, Fan T, Sun P. Member Domain 3 (LRIG3) Activates Hypoxia-Inducible Factor-1 α/Vascular Endothelial Growth Factor (HIF-1α/VEGF) Pathway to Inhibit the Growth of Bone Marrow Mesenchymal Stem Cells in Glioma. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2629] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Member domain 3 (LRIG3) of the LRIG gene family is down-regulated in several cancers. However, its role in bone marrow mesenchymal stem cells (BMSCs) in gliomas and the related mechanisms is unknown. The qRT-PCR assessed LRIG3 mRNA level. Rat BMSCs were randomly assigned into glioma
group (BMSCs cultured in glioma microenvironment); LRIG3 overexpression group; and si-LRIG3 inhibitor group followed by analysis of LRIG3 expression, cell proliferation, PCNA and Ki-67 apoptosis, TNF-α; and HIF-1α/VEGF mRNA level. LRIG3 mRNA expression was decreased
in gliomas patients (P < 0.05). BMSCs cultured in glioma microenvironment showed decreased LRIG3, increased cell proliferation, decreased PCNA, Ki-67 and TNF-α secretion as well as elevated HIF-1α and VEGF level (P < 0.05). Transfection of LRIG3
siRNA further promoted the above changes. Conversely, LRIG3 plasmid transfection significantly promoted its expression in glioma BMSCs (P < 0.05), inhibited cell proliferation, promoted PCNA, Ki-67, and TNF-α secretion, and increased HIF-1α and VEGF level
(P < 0.05). LRIG3 in rat BMSCs cultured in the glioma microenvironment is decreased. Down-regulation of LRIG3 inhibits TNF-α secretion by activating HIF-1α/VEGF pathway regulating BMSCs proliferation and apoptosis.
Collapse
Affiliation(s)
- Haiyan Zhou
- Department of Emergency Medicine, Baoding Second Hospital, Baoding, Hebei, 071052, China
| | - Jing Cao
- Department of Emergency Medicine, Baoding Second Hospital, Baoding, Hebei, 071052, China
| | - Fan Yang
- Department of Emergency Medicine, Baoding Second Hospital, Baoding, Hebei, 071052, China
| | - Duojiao Fan
- Department of Science and Education, Baoding Second Hospital, Baoding, Hebei, 071052, China
| | - Hengzhou Li
- Department of Emergency Medicine, Baoding Second Hospital, Baoding, Hebei, 071052, China
| | - Tao Fan
- Department of Neurosurgery, Beijing Sanbo Brain Hospital of Capital Medical University, Beijing, 100093, China
| | - Peng Sun
- Department of Emergency Medicine, Baoding Second Hospital, Baoding, Hebei, 071052, China
| |
Collapse
|
45
|
Noncoding RNAs in Glioblastoma: Emerging Biological Concepts and Potential Therapeutic Implications. Cancers (Basel) 2021; 13:cancers13071555. [PMID: 33800703 PMCID: PMC8037102 DOI: 10.3390/cancers13071555] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 02/28/2021] [Accepted: 03/19/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Since the completion of the Human Genome Project, noncoding RNAs (ncRNAs) have emerged as an important class of genetic regulators. Several classes of ncRNAs, which include microRNAs (miRNAs), long noncoding RNAs (lncRNAs), circular RNAs (circRNAs), and piwi-interacting RNAs (piRNAs), have been shown to play important roles in controlling developmental and disease processes. In this article, we discuss the potential roles of ncRNAs in regulating glioblastoma (GBM) formation and progression as well as potential strategies to exploit the diagnostic and therapeutic potential of ncRNAs in GBM. Abstract Noncoding RNAs (ncRNAs) have emerged as a novel class of genomic regulators, ushering in a new era in molecular biology. With the advent of advanced genetic sequencing technology, several different classes of ncRNAs have been uncovered, including microRNAs (miRNAs), long noncoding RNAs (lncRNAs), circular RNAs (circRNAs), and piwi-interacting RNAs (piRNAs), which have been linked to many important developmental and disease processes and are being pursued as clinical and therapeutic targets. Molecular phenotyping studies of glioblastoma (GBM), the most common and lethal cancer of the adult brain, revealed that several ncRNAs are frequently dysregulated in its pathogenesis. Additionally, ncRNAs regulate many important aspects of glioma biology including tumour cell proliferation, migration, invasion, apoptosis, angiogenesis, and self-renewal. Here, we present an overview of the biogenesis of the different classes of ncRNAs, discuss their biological roles, as well as their relevance to gliomagenesis. We conclude by discussing potential approaches to therapeutically target the ncRNAs in clinic.
Collapse
|
46
|
Fang Y, Zong Q, He Z, Liu C, Wang YF. Knockdown of RpL36 in testes impairs spermatogenesis in Drosophila melanogaster. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2021; 336:417-430. [PMID: 33734578 DOI: 10.1002/jez.b.23040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 02/06/2021] [Accepted: 02/27/2021] [Indexed: 01/03/2023]
Abstract
Many ribosomal proteins (RPs) not only play essential roles in ribosome biogenesis, but also have "extraribosomal" functions in various cellular processes. RpL36 encodes ribosomal protein L36, a component of the 60S subunit of ribosomes in Drosophila melanogaster. We report here that RpL36 is required for spermatogenesis in D. melanogaster. After showing the evolutionary conservation of RpL36 sequences in animals, we revealed that the RpL36 expression level in fly testes was significantly higher than in ovaries. Knockdown RpL36 in fly testes resulted in a significantly decreased egg hatch rate when these males mated with wild-type females. Furthermore, 76.67% of the RpL36 knockdown fly testes were much smaller in comparison to controls. Immunofluorescence staining exhibited that in the RpL36 knockdown testis hub cell cluster was enlarged, while the number of germ cells, including germ stem cells, was reduced. Knockdown of RpL36 in fly testis caused much fewer or no mature sperms in seminal vesicles. The terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) signal was stronger in RpL36 knockdown fly testes than in the control testes, but the TUNEL-positive cells could not be stained by Vasa antibody, indicating that apoptotic cells are not germ cells. The percentage of pH3-positive cells among the Vasa-positive cells was significantly reduced. The expression of genes involved in cell death, cell cycle progression, and JAK/STAT signaling pathway was significantly changed by RpL36 knockdown in fly testes. These results suggest that RpL36 plays an important role in spermatogenesis, likely through JAK/STAT pathway, thus resulting in defects in cell-cycle progression and cell death in D. melanogaster testes.
Collapse
Affiliation(s)
- Yang Fang
- School of Life Sciences, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, Central China Normal University, Wuhan, China
| | - Qiong Zong
- School of Life Sciences, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, Central China Normal University, Wuhan, China
| | - Zhen He
- School of Life Sciences, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, Central China Normal University, Wuhan, China
| | - Chen Liu
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Yu-Feng Wang
- School of Life Sciences, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, Central China Normal University, Wuhan, China
| |
Collapse
|
47
|
Cao J, Zhou H, Yang F, Fan D, Li H, Fan T, Sun P. Zinc Finger E-Box Binding Homeobox 1 Regulates the Biological Behavior of Glioma Cells via iNOS/NF- κB Signaling. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The gliomas pathogenesis is complex and effective molecular targets are still unclear. ZEB1 regulates epithelial mesenchymal transition (EMT) and participates in tumors. Our study intends to analyze ZEB1’s role in glioma cells. qRT-PCR detected ZEB1 mRNA expression in normal group
and tumor group. ZEB1 siRNA was transfected into glioma cells followed by measuring ZEB1, E-cadherin and Vimentin expression, cell proliferation, Capase-3 activity as well as NF-κB and iNOS changes by immunoblotting. Upregulation of ZEB1 was found in glioma tumor tissue and correlated
with glioma clinicopathological characteristics. Interfering with ZEB1 by siRNA significantly down-regulated ZEB1, inhibited cell proliferation, increased Capase-3 activity, down regulated NF-κB and iNOS proteins in glioma cells, elevated E-cadherin and decreased Vimentin level
(P <0.05). ZEB1 down regulation in glioma cells can change the expression of NF-κB/iNOS, regulate cell apoptosis and inhibit cell proliferation, thereby delaying EMT process.
Collapse
Affiliation(s)
- Jing Cao
- Department of Emergency Medicine, Baoding Second Hospital, Baoding City, Hebei Province, 071000, China
| | - Haiyan Zhou
- Department of Emergency Medicine, Baoding Second Hospital, Baoding City, Hebei Province, 071000, China
| | - Fan Yang
- Department of Emergency Medicine, Baoding Second Hospital, Baoding City, Hebei Province, 071000, China
| | - Duojiao Fan
- Department of Science and Education, Baoding Second Hospital, Baoding City, Hebei Province, 071000, China
| | - Hengzhou Li
- Department of Emergency Medicine, Baoding Second Hospital, Baoding City, Hebei Province, 071000, China
| | - Tao Fan
- Department of Neurosurgery, Beijing Sanbo Brain Hospital of Capital Medical University, Beijing, 100093, China
| | - Peng Sun
- Department of Emergency Medicine, Baoding Second Hospital, Baoding City, Hebei Province, 071000, China
| |
Collapse
|
48
|
Jin L, Huang S, Guan C, Chang S. ETS1-activated SNHG10 exerts oncogenic functions in glioma via targeting miR-532-3p/FBXL19 axis. Cancer Cell Int 2020; 20:589. [PMID: 33298070 PMCID: PMC7725120 DOI: 10.1186/s12935-020-01649-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 11/09/2020] [Indexed: 12/18/2022] Open
Abstract
Background In past few years, long non-coding RNAs (lncRNAs) have been reported to play regulatory roles during cancer progression. LncRNA SNHG10 has been explored in several sorts of cancers. However, its detailed role and mechanism are still not well understood in glioma. Methods Expression levels of genes were evaluated by RT-qPCR. EdU, TUNEL, sphere formation, wound healing and transwell assays appraised the effect of SNHG10 on glioma cellular processes. The interaction between molecules was examined by ChIP, RIP, RNA pull down and luciferase reporter assays. Results High level of SNHG10 was detected in glioma cells. Functional assay confirmed that SNHG10 promoted the proliferation, migration, invasion and stemness of glioma cells. Moreover, miR-532-3p was validated to bind with SNHG10 and expressed at a low level in glioma cells. Importantly, miR-532-3p exerted inhibitory functions in glioma. Furthermore, it was found that FBXL19 targeted by miR-532-3p facilitated cell growth and stemness in glioma, and that SNHG10 worked in glioma by increasing FBXL19 expression through sequestering miR-532-3p. More importantly, ETS1 promoted the transcription of SNHG10 and it mediated contribution to the malignant behaviors of glioma cells by SNHG10/miR-532-3p/FBXL19 signaling. Conclusion SNHG10 was transcriptionally activated by ETS1 and played an oncogenic role in glioma by sponging miR-532-3p and up-regulating FBXL19. ![]()
Collapse
Affiliation(s)
- Lide Jin
- The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, No.157 Jinbi Road, Kunming, 650032, Yunnan, China
| | - Shengquan Huang
- The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, No.157 Jinbi Road, Kunming, 650032, Yunnan, China
| | - Congjin Guan
- The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, No.157 Jinbi Road, Kunming, 650032, Yunnan, China.
| | - Shun Chang
- The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, No.157 Jinbi Road, Kunming, 650032, Yunnan, China.
| |
Collapse
|
49
|
Liu H, Zhang L, Ding X, Sui X. LINC00861 inhibits the progression of cervical cancer cells by functioning as a ceRNA for miR‑513b‑5p and regulating the PTEN/AKT/mTOR signaling pathway. Mol Med Rep 2020; 23:24. [PMID: 33179755 PMCID: PMC7673320 DOI: 10.3892/mmr.2020.11662] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 10/01/2020] [Indexed: 12/24/2022] Open
Abstract
Long non‑coding RNAs (lncRNAs) have been discovered to serve important roles in a variety of types of cancer, including cervical cancer. The low expression of lncRNA long intergenic non‑protein coding RNA 861 (LINC00861) is related to poor prognosis in ovarian cancer. However, the effects and underlying mechanisms of LINC00861 in cervical cancer remain largely unknown. The present study aimed to examine the role of LINC00861 in the development and progression of ovarian cancer and its underlying mechanisms. The expression levels of LINC00861 and microRNA (miR)‑513b‑5p were analyzed using reverse transcription‑quantitative PCR analysis. Cell proliferation, migration and invasion were measured by using Cell Counting Kit‑8, colony formation, wound healing and Transwell assays, respectively. A luciferase assay was used to determine whether miR‑513b‑5p targeted LINC00861 and PTEN. The expression of protein was measured by using western blot assay. The results of the present study discovered that LINC00861 expression levels were significantly downregulated in cervical cancer tissues and CaSki and ME‑180 cell lines. Downregulated LINC00861 expression levels were identified to be associated with an advanced‑stage, lymph node metastasis and the poor survival of patients with cervical cancer. Gene Set Enrichment Analysis revealed that the PI3K/AKT/mTOR signaling pathway was significantly enriched in cervical tumors expressing low expression levels of LINC00861 compared with tumors expressing high levels of LINC00861. The overexpression of LINC00861 reduced cervical cancer cell proliferation, migration, invasion and epithelial‑mesenchymal transition (EMT) processes, upregulated PTEN protein expression levels and downregulated phosphorylated (p)‑AKT and p‑mTOR protein expression levels. The regulatory relationship between LINC00861, microRNA (miR)‑513b‑5p and PTEN was validated using a dual luciferase reporter gene assay. PTEN expression levels were significantly downregulated in the miR‑513b‑5p mimic group and significantly upregulated in the miR‑513b‑5p inhibitor group compared with the mimic NC and inhibitor NC in both cell lines. Furthermore, LINC00861 was suggested to serve as a competing endogenous RNA by sponging miR‑513b‑5p and consequently upregulating the expression levels of PTEN in cervical cancer cells. The expression of PTEN, the phosphorylation of Akt and mTOR and and the EMT phenotype were rescued following co‑transfection with LINC00861 and miR‑513b‑5p mimics. In conclusion, the findings of the present study indicated that the LINC00861/miR‑513b‑5p axis may inhibit the progression of cervical cancer cells through the PTEN/AKT/mTOR signaling pathway to suppress the EMT process.
Collapse
Affiliation(s)
- Hui Liu
- Department of Gynaecology and Obstetrics, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong 264013, P.R. China
| | - Ling Zhang
- Department of Gynaecology and Obstetrics, Yidu Central Hospital of Weifang, Qingzhou, Shandong 262500, P.R. China
| | - Xiaoling Ding
- Department of Gynaecology and Obstetrics, Maternal and Child Health Hospital of Qingzhou, Qingzhou, Shandong 264000, P.R. China
| | - Xuezuo Sui
- Department of Gynaecology and Obstetrics, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong 264013, P.R. China
| |
Collapse
|
50
|
Liu H, Hu K. The Long Intergenic Noncoding RNA 00707 Sponges MicroRNA-613 (miR-613) to Promote Proliferation and Invasion of Gliomas. Technol Cancer Res Treat 2020; 19:1533033820962092. [PMID: 33107401 PMCID: PMC7607719 DOI: 10.1177/1533033820962092] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background: Glioma is one of the most deadly malignant tumors in humans. Long non-coding RNA (lncRNA) plays a key role in the occurrence, development and invasion of tumors by regulating oncogenic and tumor suppressor pathways. However, the role and action mechanism of long intergenic non-coding RNA 00707 (LINC00707) in gliomas have not been elucidated. This study aimed to investigate the interaction between LINC00707 and miR-613 as well as its role in gliomas. Materials and Methods: The expression levels of LINC00707 and miR-613 were detected by qRT-PCR. The chi-square test was used to analyze the correlation between LINC00707 expression and clinicopathological parameters. CCK-8 and colony formation assays were used to detect glioma cell proliferation; and wound healing and transwell assays were used to detect glioma cell migration and invasion. The relationship between LINC00707 and miR-613 was predicted by Starbase, and verified by qRT-PCR and dual luciferase reporter gene assay. Results: LINC00707 was up-regulated in gliomas. Up-regulated LINC00707 increased the proliferation, migration and invasion of glioma cells, and silenced LINC00707 reduced these abilities. The increase of the expression level of LINC00707 down-regulated miR-613 in glioma cells, while the inhibition of the expression level of LINC00707 up-regulated miR-613 in glioma cells. The high expression of LINC00707 was related to the Karnofsky performance status (KPS) score and WHO staging. LINC00707 could offset the ability of miR-613 to inhibit glioma proliferation and invasion. Conclusion: LINC00707 promotes proliferation and invasion of glioma cells by sponging miR-613. The regulatory axis of LINC00707/miR-613 provides new insights into the mechanism and treatment of gliomas.
Collapse
Affiliation(s)
- Handong Liu
- Department of Neurosurgery, Xiangyang Center Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Keqi Hu
- Department of Neurosurgery, Xiangyang Center Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| |
Collapse
|