1
|
Lashkarboloki M, Jahanbakhshi A, Mowla SJ, Bjeije H, Soltani BM. Oncogenic roles of long non-coding RNAs in essential glioblastoma signaling pathways. J Neurogenet 2024; 38:62-78. [PMID: 39169886 DOI: 10.1080/01677063.2024.2390403] [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: 04/22/2024] [Accepted: 08/05/2024] [Indexed: 08/23/2024]
Abstract
Glioblastoma multiforme (GBM) is an aggressive and diffuse type of glioma with the lowest survival rate in patients. The recent failure of multiple treatments suggests that targeting several targets at once may be a different strategy to overcome GBM carcinogenesis. Normal function of oncogenes and tumor suppressor genes need for the preservation of regular cellular processes, so any defects in these genes' activity, operate the corresponding signaling pathways, which initiate carcinogenic processes. Long non-coding RNAs (lncRNAs) that can be found in the cytoplasm or nucleus of the cells, control the transcription and translation of genes. LncRNAs perform a variety of functions, including epigenetic alteration, protein modification and stability, transcriptional regulation, and competition for miRNA that regulate mRNA translation through sponging miRNAs. Identification of various oncogenic lncRNAs and their multiple roles in brain cancers making them potential candidates for use as glioma diagnostic, prognostic, and therapeutic targets in the future. This study highlighted multiple oncogenic lncRNAs and classified them into different signaling pathways based on the regulated target genes in glioblastoma.
Collapse
Affiliation(s)
- Mina Lashkarboloki
- Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amin Jahanbakhshi
- Skull Base Research Center, Rasool Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Javad Mowla
- Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hassan Bjeije
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Bahram M Soltani
- Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
2
|
Pokorná M, Černá M, Boussios S, Ovsepian SV, O’Leary VB. lncRNA Biomarkers of Glioblastoma Multiforme. Biomedicines 2024; 12:932. [PMID: 38790894 PMCID: PMC11117901 DOI: 10.3390/biomedicines12050932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/15/2024] [Accepted: 04/18/2024] [Indexed: 05/26/2024] Open
Abstract
Long noncoding RNAs (lncRNAs) are RNA molecules of 200 nucleotides or more in length that are not translated into proteins. Their expression is tissue-specific, with the vast majority involved in the regulation of cellular processes and functions. Many human diseases, including cancer, have been shown to be associated with deregulated lncRNAs, rendering them potential therapeutic targets and biomarkers for differential diagnosis. The expression of lncRNAs in the nervous system varies in different cell types, implicated in mechanisms of neurons and glia, with effects on the development and functioning of the brain. Reports have also shown a link between changes in lncRNA molecules and the etiopathogenesis of brain neoplasia, including glioblastoma multiforme (GBM). GBM is an aggressive variant of brain cancer with an unfavourable prognosis and a median survival of 14-16 months. It is considered a brain-specific disease with the highly invasive malignant cells spreading throughout the neural tissue, impeding the complete resection, and leading to post-surgery recurrences, which are the prime cause of mortality. The early diagnosis of GBM could improve the treatment and extend survival, with the lncRNA profiling of biological fluids promising the detection of neoplastic changes at their initial stages and more effective therapeutic interventions. This review presents a systematic overview of GBM-associated deregulation of lncRNAs with a focus on lncRNA fingerprints in patients' blood.
Collapse
Affiliation(s)
- Markéta Pokorná
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, Ruská 87, Vinohrady, 10000 Prague, Czech Republic; (M.Č.); (V.B.O.)
| | - Marie Černá
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, Ruská 87, Vinohrady, 10000 Prague, Czech Republic; (M.Č.); (V.B.O.)
| | - Stergios Boussios
- Department of Medical Oncology, Medway NHS Foundation Trust, Gillingham ME7 5NY, UK;
- Faculty of Medicine, Health, and Social Care, Canterbury Christ Church University, Canterbury CT2 7PB, UK
- Faculty of Life Sciences & Medicine, School of Cancer & Pharmaceutical Sciences, King’s College London, Strand, London WC2R 2LS, UK
- Kent Medway Medical School, University of Kent, Canterbury CT2 7LX, UK
- AELIA Organization, 9th Km Thessaloniki-Thermi, 57001 Thessaloniki, Greece
| | - Saak V. Ovsepian
- Faculty of Engineering and Science, University of Greenwich London, Chatham Maritime, Kent ME4 4TB, UK;
- Faculty of Medicine, Tbilisi State University, Tbilisi 0177, Georgia
| | - Valerie Bríd O’Leary
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, Ruská 87, Vinohrady, 10000 Prague, Czech Republic; (M.Č.); (V.B.O.)
| |
Collapse
|
3
|
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
|
4
|
Cheng M, Wang Q, Chen L, Zhao D, Tang J, Xu J, He Z. LncRNA UCA1/miR-182-5p/MGMT axis modulates glioma cell sensitivity to TMZ through MGMT-related DNA damage pathways. Hum Pathol 2022; 123:59-73. [DOI: 10.1016/j.humpath.2022.02.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/16/2022] [Accepted: 02/18/2022] [Indexed: 12/30/2022]
|
5
|
Song F, Chen Z, Lyu D, Gu Y, Lu B, Hao S, Xu Y, Jin X, Fu Q, Yao K. Expression profiles of long noncoding RNAs in human corneal epithelial cells exposed to fine particulate matter. CHEMOSPHERE 2022; 287:131955. [PMID: 34478962 DOI: 10.1016/j.chemosphere.2021.131955] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 08/06/2021] [Accepted: 08/18/2021] [Indexed: 06/13/2023]
Abstract
PURPOSE The aim of this study was to investigate the expression profiles of long noncoding RNAs (lncRNAs) in human corneal epithelial cells (HCECs) exposed to fine particulate matter (PM2.5) and to identify potential biological pathways involved in PM2.5-induced toxicity in HCECs. METHODS Using RNA sequencing (RNA-seq) and hierarchy clustering analysis, lncRNA expression profiles in PM2.5-treated and untreated HCECs were examined. Gene ontology (GO) enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were performed to predict the role of altered lncRNAs in biological processes and pathways. A quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR) assay was conducted to verify the RNA-seq results in HCECs and human corneal epithelial cell sheets. RESULTS In total, 65 lncRNAs were altered in the PM2.5-treated HCECs, including 41 upregulated and 24 downregulated lncRNAs. The results of the qRT-PCR assay were consistent with those of the RNA-seq analysis. The expression of two significantly upregulated lncRNAs was confirmed in human corneal epithelial cell sheets. The GO analysis demonstrated that altered lncRNAs in the PM2.5-treated HCECs were significantly enriched in three domains: cellular component, molecular function, and biological process. The KEGG pathway analysis revealed enriched pathways of lncRNA co-expressed mRNAs, including cancer, RNA transport, and Rap1 signaling. CONCLUSIONS Our results suggest that lncRNAs are involved in the pathogenesis of PM2.5-induced ocular diseases, exerting their effects through biological processes and pathogenic pathways. Among the altered lncRNAs, RP3-406P24.3 and RP11-285E9.5 may play significant roles in PM2.5-induced ocular surface injury.
Collapse
Affiliation(s)
- Fan Song
- Eye Center of the 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, Zhejiang Province, China
| | - Zhijian Chen
- Department of Environmental and Occupational Health, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, Zhejiang Province, China
| | - Danni Lyu
- Eye Center of the 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, Zhejiang Province, China
| | - Yuzhou Gu
- Eye Center of the 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, Zhejiang Province, China
| | - Bing Lu
- Eye Center of the 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, Zhejiang Province, China
| | - Shengjie Hao
- Eye Center of the 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, Zhejiang Province, China
| | - Yili Xu
- Eye Center of the 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, Zhejiang Province, China
| | - Xiuming Jin
- Eye Center of the 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, Zhejiang Province, China
| | - Qiuli Fu
- Eye Center of the 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, Zhejiang Province, China.
| | - Ke Yao
- Eye Center of the 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
6
|
Zhang L, Zhang L, Guo F. MiRNA-494-3p Regulates Bupivacaine-Induced Neurotoxicity by the CDK6-PI3K/AKT Signaling. Neurotox Res 2021; 39:2007-2017. [PMID: 34652691 DOI: 10.1007/s12640-021-00427-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 12/28/2022]
Abstract
Bupivacaine (BUP) is a long-acting amide local anesthetic that may induce strong neurotoxicity and neurological complications. In this study, we elucidate the influence of microRNA-494-3p (miR-494-3p) in BUP-induced neurotoxicity in primary mouse hippocampal neuronal cells. In this study, primary hippocampal neurons were isolated from neonatal C57BL/6 mice. The isolated neurons were treated with various doses of BUP. MTT assay was conducted to analyze neuronal viability. Gene expression measurement was done by RT-qPCR. The impact of miR-494-3p in BUP-mediated neural injury was examined using TUNEL, flow cytometry, western blotting, and ROS activity detection. The regulatory relationship between miR-494-3p and cyclin-dependent kinases 4 and 6 (CDK6) was identified using a luciferase reporter assay. BUP treatment led to neurotoxicity and miR-494-3p upregulation in primary cultured hippocampal neurons. Functionally, miR-494-3p depletion alleviated neuronal apoptosis and oxidative damage induced by BUP. We verified that miR-494-3p targeted and negatively modulated CDK6. MiR-494-3p depletion also activated PI3K/AKT signaling by elevating CDK6 expression in BUP-treated neurons. Furthermore, CDK6 knockdown or PI3K/AKT inactivation attenuated the neuroprotective role of miR-494-3p depletion. Silencing miR-494-3p exerts neuroprotective function in hippocampal neuronal cells against BUP-induced injury by the CDK6-PI3K/AKT pathway.
Collapse
Affiliation(s)
- Licheng Zhang
- Deparment of Anesthesia Resuscitation Room, Zhongshan Hospital Xiamen University, Xiamen, Fujian, 361004, China
| | - Lifeng Zhang
- Deparment of Surgical Anesthesiology, Zhongshan Hospital Xiamen University, Xiamen, Fujian, 361004, China
| | - Fengying Guo
- Department of Tumor Radiotherapy, Zhongshan Hospital Xiamen University, Siming District, 1854 Xinjing Garden, Luling Road, Xiamen, 361004, Fujian, China.
| |
Collapse
|
7
|
Fu J, Pan J, Yang X, Zhang Y, Shao F, Chen J, Huang K, Wang Y. Mechanistic study of lncRNA UCA1 promoting growth and cisplatin resistance in lung adenocarcinoma. Cancer Cell Int 2021; 21:505. [PMID: 34544452 PMCID: PMC8454127 DOI: 10.1186/s12935-021-02207-0] [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: 03/11/2021] [Accepted: 09/08/2021] [Indexed: 02/07/2023] Open
Abstract
Aim This study aimed to explore the mechanism of LncRNA urothelial carcinoma-associated 1 (UCA1) promoting cisplatin resistance in lung adenocarcinoma (LUAD). Method The UCA1 expression level in LUAD cell lines was detected by reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR). We overexpressed UCA1 in A549 cells and downregulated UCA1 in A549/DDP cells by the lentivirus‑mediated technique. Subsequently, in vitro, and in vivo functional experiments were performed to investigate the functional roles of UCA1 in the growth and metastasis of LUAD cell lines. Furthermore, RNA pulldown, mass spectrometry, and RNA immunoprecipitation technique were performed to analyze various downstream target factors regulated by UCA1. Results The results revealed a higher UCA1 expression level in A549/DDP cells and LUAD tissues than in A549 cells and adjacent cancer tissues. UCA1 expression was significantly associated with distant metastasis, clinical stage, and survival time of patients with LUAD. UCA1 overexpression significantly increased the proliferation, invasion, clone formation, and cisplatin resistance ability and enhanced the expression levels of proliferating cell nuclear antigen and excision repair cross-complementing gene 1 in A549 cells. However, these trends were mostly reversed after the knockdown of UCA1 in A549/DDP cells. Tumorigenic assays in nude mice showed that UCA1 knockdown significantly inhibited tumor growth and reduced cisplatin resistance. Enolase 1 was the RNA-binding protein (RBP) of UCA1. Conclusion Based on the results, we concluded that UCA1 promoted LUAD progression and cisplatin resistance and hence could be a potential diagnostic marker and therapeutic target in patients with LUAD. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02207-0.
Collapse
Affiliation(s)
- Jiali Fu
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Jingjing Pan
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xiang Yang
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yan Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Fanggui Shao
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Jie Chen
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Kate Huang
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| | - Yumin Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
8
|
Long Non-coding RNAs: Potential Players in Cardiotoxicity Induced by Chemotherapy Drugs. Cardiovasc Toxicol 2021; 22:191-206. [PMID: 34417760 DOI: 10.1007/s12012-021-09681-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 07/24/2021] [Indexed: 10/20/2022]
Abstract
One of the most important side effects of chemotherapy is cardiovascular complications, such as cardiotoxicity. Many factors are involved in the pathogenesis of cardiotoxicity; one of the most important of which is long non-coding RNAs (lncRNAs). lncRNA has 200-1000 nucleotides. It is involved in important processes such as cell proliferation, regeneration and apoptosis; today it is used as a prognostic and diagnostic factor. A, various drugs by acting on lncRNAs can affect cells. Therefore, by accurately identifying IncRNAs function, we can play an effective role in preventing the development of cardiotoxicity-induced chemotherapy drugs, and use them as a therapeutic strategy to improve clinical symptoms and increase patient survival.
Collapse
|
9
|
Momtazmanesh S, Rezaei N. Long Non-Coding RNAs in Diagnosis, Treatment, Prognosis, and Progression of Glioma: A State-of-the-Art Review. Front Oncol 2021; 11:712786. [PMID: 34322395 PMCID: PMC8311560 DOI: 10.3389/fonc.2021.712786] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 06/25/2021] [Indexed: 12/12/2022] Open
Abstract
Glioma is the most common malignant central nervous system tumor with significant mortality and morbidity. Despite considerable advances, the exact molecular pathways involved in tumor progression are not fully elucidated, and patients commonly face a poor prognosis. Long non-coding RNAs (lncRNAs) have recently drawn extra attention for their potential roles in different types of cancer as well as non-malignant diseases. More than 200 lncRNAs have been reported to be associated with glioma. We aimed to assess the roles of the most investigated lncRNAs in different stages of tumor progression and the mediating molecular pathways in addition to their clinical applications. lncRNAs are involved in different stages of tumor formation, invasion, and progression, including regulating the cell cycle, apoptosis, autophagy, epithelial-to-mesenchymal transition, tumor stemness, angiogenesis, the integrity of the blood-tumor-brain barrier, tumor metabolism, and immunological responses. The well-known oncogenic lncRNAs, which are upregulated in glioma, are H19, HOTAIR, PVT1, UCA1, XIST, CRNDE, FOXD2-AS1, ANRIL, HOXA11-AS, TP73-AS1, and DANCR. On the other hand, MEG3, GAS5, CCASC2, and TUSC7 are tumor suppressor lncRNAs, which are downregulated. While most studies reported oncogenic effects for MALAT1, TUG1, and NEAT1, there are some controversies regarding these lncRNAs. Expression levels of lncRNAs can be associated with tumor grade, survival, treatment response (chemotherapy drugs or radiotherapy), and overall prognosis. Moreover, circulatory levels of lncRNAs, such as MALAT1, H19, HOTAIR, NEAT1, TUG1, GAS5, LINK-A, and TUSC7, can provide non-invasive diagnostic and prognostic tools. Modulation of expression of lncRNAs using antisense oligonucleotides can lead to novel therapeutics. Notably, a profound understanding of the underlying molecular pathways involved in the function of lncRNAs is required to develop novel therapeutic targets. More investigations with large sample sizes and increased focus on in-vivo models are required to expand our understanding of the potential roles and application of lncRNAs in glioma.
Collapse
Affiliation(s)
- Sara Momtazmanesh
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Ni H, Zhi R, Zuo J, Liu W, Xie P, Zhi Z. Pseudogene ANXA2P2 knockdown shows tumor-suppressive function by inhibition of the PI3K/PKB pathway in glioblastoma cells. J Biochem Mol Toxicol 2021; 35:e22824. [PMID: 34047431 DOI: 10.1002/jbt.22824] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 02/04/2021] [Accepted: 05/18/2021] [Indexed: 01/15/2023]
Abstract
The pseudogene annexin A2 pseudogene 2 (ANXA2P2) is highly expressed in glioblastoma (GBM). However, its role and mechanism involved in the progression of GBM remain poorly understood. ANXA2P2 messenger RNA expression was measured by quantitative reverse transcription-polymerase chain reaction. The protein levels were detected by Western blot. Cell viability was evaluated by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and lactate dehydrogenase (LDH) release assays. Cell invasive ability was investigated by the transwell assay and by epithelial-mesenchymal transition (EMT). Cell apoptosis was examined by flow cytometry. The results showed that ANXA2P2 expression was increased in GBM tissues and cells. Silencing of ANXA2P2 inhibited the activation of the phosphoinositide 3-kinase (PI3K)/protein kinase B (PKB) pathway in GBM cells. Knockdown of ANXA2P2 decreased cell viability, promoted LDH release, suppressed cell invasive ability, and EMT, and induced cell apoptosis in GBM cells. The addition of the PI3K/PKB activator 740Y-P abrogated the effects of ANXA2P2 knockdown on cell viability, LDH release, invasive ability, and apoptosis. In conclusion, knockdown of ANXA2P2 inhibited cell viability and invasion but promoted the apoptotic rate by suppressing the PI3K/PKB pathway in GBM cells. ANXA2P2 may represent a new target for the treatment of GBM.
Collapse
Affiliation(s)
- Hongzao Ni
- Department of Neurosurgery, The Second People's Hospital of Huai'an, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| | - Rongrong Zhi
- Department of Gastroenterology, Lianshui County People's Hospital Affiliated to Kangda College of Nanjing Medical University, Huai'an, China
| | - Jiandong Zuo
- Department of Neurosurgery, The Second People's Hospital of Huai'an, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| | - Wenguang Liu
- Department of Neurosurgery, The Second People's Hospital of Huai'an, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| | - Peng Xie
- Department of Neurosurgery, The Second People's Hospital of Huai'an, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| | - Zhongwen Zhi
- Department of Neurosurgery, The Second People's Hospital of Huai'an, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| |
Collapse
|
11
|
Guo X, Wang T, Huang G, Li R, Da Costa C, Li H, Lv S, Li N. Rediscovering potential molecular targets for glioma therapy through the analysis of the cell of origin, microenvironment, and metabolism. Curr Cancer Drug Targets 2021; 21:558-574. [PMID: 33949933 DOI: 10.2174/1568009621666210504091722] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 01/12/2021] [Accepted: 01/15/2021] [Indexed: 11/22/2022]
Abstract
Gliomas are the most common type of malignant brain tumors. Despite significant medical advances, gliomas remain incurable and are associated with high mortality. Although numerous biomarkers of diagnostic value have been identified and significant progress in the prognosis of the outcome has been made, the treatment has not been parallelly improved during the last three decades. This review summarizes and discusses three aspects of recent discoveries related to glioma, with the objective to highlight the advantages of glioma-specific drugs targeting the cell of origin, microenvironment, and metabolism. Given the heterogeneous nature of gliomas, various cell populations have been implicated as likely sources of the tumor. Depending on the mutation(s) acquired by the cells, it is believed that neuronal stem/progenitor cells, oligodendrocyte progenitor cells, mature neurons, and glial cells can initiate cell transformation into a malignant phenotype. The level of tumorigenicity appears to be inversely correlated with the maturation of a given cell population. The microenvironment of gliomas includes non-cancer cells such as immune cells, fibroblasts, and cells of blood vessels, as well as secreted molecules and the extracellular matrix, and all these components play a vital role during tumor initiation and progression. We will discuss in detail how the tumor microenvironment can stimulate and drive the transformation of non-tumor cell populations into tumor-supporting cells or glioma cells. Metabolic reprogramming is a key feature of gliomas and is thought to reflect the adaptation to the increased nutritional requirements of tumor cell proliferation, growth, and survival. Mutations in the IDH gene can shape metabolic reprogramming and may generate some vulnerabilities in glioma cells, such as abnormal lipid metabolism and sensitivity to endoplasmic reticulum stress (ERS). We will analyze the prominent metabolic features of malignant gliomas and the key pathways regulating glioma metabolism. This review is intended to provide a conceptual background for the development of glioma therapies based on the properties of tumor cell populations, microenvironment, and metabolism.
Collapse
Affiliation(s)
- Xiaoran Guo
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University (SYSU), No.628, Zhenyuan Rd, Guangming Dist., Shenzhen 518107. China
| | - Tao Wang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University (SYSU), No.628, Zhenyuan Rd, Guangming Dist., Shenzhen 518107. China
| | - Guohao Huang
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, No. 183 Xinqiao Street, Shapingba District, Chongqing City 400037. China
| | - Ruohan Li
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University (SYSU), No.628, Zhenyuan Rd, Guangming Dist., Shenzhen 518107. China
| | - Clive Da Costa
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT. United Kingdom
| | - Huafu Li
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University (SYSU), No.628, Zhenyuan Rd, Guangming Dist., Shenzhen 518107. China
| | - Shengqing Lv
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, No. 183 Xinqiao Street, Shapingba District, Chongqing City 400037. China
| | - Ningning Li
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University (SYSU), No.628, Zhenyuan Rd, Guangming Dist., Shenzhen 518107. China
| |
Collapse
|
12
|
Hosseini NF, Manoochehri H, Khoei SG, Sheykhhasan M. The Functional Role of Long Non-coding RNA UCA1 in Human Multiple Cancers: a Review Study. Curr Mol Med 2021; 21:96-110. [PMID: 32560605 DOI: 10.2174/1566524020666200619124543] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 02/08/2023]
Abstract
In various cancers, high-grade tumor and poor survival rate in patients with upregulated lncRNAs UCA1 have been confirmed. Urothelial carcinoma associated 1 (UCA1) is an oncogenic non-coding RNA with a length of more than 200 nucleotides. The UCA1 regulate critical biological processes that are involved in cancer progression, including cancer cell growth, invasion, migration, metastasis, and angiogenesis. So It should not surprise that UCA1 overexpresses in variety of cancers type, including pancreatic cancer, ovarian cancer, gastric cancer, colorectal cancer, breast cancer, prostate cancer, endometrial cancer, cervical cancer, bladder cancer, adrenal cancer, hypopharyngeal cancer, oral cancer, gallbladder cancer, nasopharyngeal cancer, laryngeal cancer, osteosarcoma, esophageal squamous cell carcinoma, renal cell carcinoma, cholangiocarcinoma, leukemia, glioma, thyroid cancer, medulloblastoma, hepatocellular carcinoma and multiple myeloma. In this article, we review the biological function and regulatory mechanism of UCA1 in several cancers and also, we will discuss the potential of its as cancer biomarker and cancer treatment.
Collapse
Affiliation(s)
- Nashmin Fayazi Hosseini
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hamed Manoochehri
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | - Mohsen Sheykhhasan
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
13
|
Yadav B, Pal S, Rubstov Y, Goel A, Garg M, Pavlyukov M, Pandey AK. LncRNAs associated with glioblastoma: From transcriptional noise to novel regulators with a promising role in therapeutics. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 24:728-742. [PMID: 33996255 PMCID: PMC8099481 DOI: 10.1016/j.omtn.2021.03.018] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Glioblastoma multiforme (GBM) is the most widespread and aggressive subtype of glioma in adult patients. Numerous long non-coding RNAs (lncRNAs) are deregulated or differentially expressed in GBM. These lncRNAs possess unique regulatory functions in GBM cells, ranging from high invasion/migration to recurrence. This review outlines the present status of specific involvement of lncRNAs in GBM pathogenesis, with a focus on their association with key molecular and cellular regulatory mechanisms. Also, we highlighted the potential of different novel RNA-based strategies that may be beneficial for therapeutic purposes.
Collapse
Affiliation(s)
- Bhupender Yadav
- Amity Institute of Biotechnology, Amity University Haryana, Panchgaon, Manesar, Haryana 122413, India
| | - Sonali Pal
- Amity Institute of Biotechnology, Amity University Haryana, Panchgaon, Manesar, Haryana 122413, India
| | - Yury Rubstov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, GSP-7, Ulitsa Miklukho-Maklaya, 16/10, 117997 Moscow, Russian Federation.,Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Vavilova Street 7, 117312 Moscow, Russian Federation
| | - Akul Goel
- La Canada High School, La Canada Flintridge, CA 91011, USA
| | - Manoj Garg
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Uttar Pradesh, Sector 125, Noida 201313, India
| | - Marat Pavlyukov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, GSP-7, Ulitsa Miklukho-Maklaya, 16/10, 117997 Moscow, Russian Federation
| | - Amit Kumar Pandey
- Amity Institute of Biotechnology, Amity University Haryana, Panchgaon, Manesar, Haryana 122413, India
| |
Collapse
|
14
|
Liu Z, Wang Y, Yuan S, Wen F, Liu J, Zou L, Zhang J. Regulatory role of long non-coding RNA UCA1 in signaling pathways and its clinical applications. Oncol Lett 2021; 21:404. [PMID: 33777227 PMCID: PMC7988699 DOI: 10.3892/ol.2021.12665] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 02/11/2021] [Indexed: 12/15/2022] Open
Abstract
Long non-coding RNA metastasis-associated urothelial carcinoma associated 1 (UCA1) plays a pivotal role in various human diseases. Its gene expression is regulated by several factors, including transcription factors, chromatin remodeling and epigenetic modification. UCA1 is involved in the regulation of the PI3K/AKT, Wnt/β-catenin, MAPK, NF-κB and JAK/STAT signaling pathways, affecting a series of cellular biological functions, such as cell proliferation, apoptosis, migration, invasion and tumor drug resistance. Furthermore, UCA1 is used as a novel potential biomarker for disease diagnosis and prognosis, as well as a target for clinical gene therapy. The present review systematically summarizes and elucidates the mechanisms of upstream transcriptional regulation of UCA1, the regulatory role of UCA1 in multiple signaling pathways in the occurrence and development of several diseases, and its potential applications in clinical treatment.
Collapse
Affiliation(s)
- Zhaoping Liu
- Department of Rheumatology, The First Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yanyan Wang
- Department of Rheumatology, The First Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Shunling Yuan
- Department of Rheumatology, The First Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Feng Wen
- Department of Hematology, The First Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Jing Liu
- Molecular Biology Research Center and Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410078, P.R. China
| | - Liheng Zou
- Department of Rheumatology, The First Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Ji Zhang
- Department of Rheumatology, The First Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China.,Department of Clinical Laboratory, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518033, P.R. China
| |
Collapse
|
15
|
Li J, Zhou Y, Yan Y, Zheng Z, Hu Y, Wu W. Sulforaphane-cysteine downregulates CDK4 /CDK6 and inhibits tubulin polymerization contributing to cell cycle arrest and apoptosis in human glioblastoma cells. Aging (Albany NY) 2020; 12:16837-16851. [PMID: 32860670 PMCID: PMC7521484 DOI: 10.18632/aging.103537] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 04/27/2020] [Indexed: 01/24/2023]
Abstract
Here we demonstrated that sulforaphane-cysteine (SFN-Cys) regulated cell cycle-related protein expressions in G0/G1 and G2/M phases of U87MG cells via High Performance Liquid Chromatography-Mass Spectrometry/Mass Spectrometry (HPLC-MS/MS) and proteomics analysis. Further, mRNA products of CDK4, CDK6 and α-tubulin were significantly higher in glioblastoma than those in normal tissues, and these results were significantly correlated to pathological grades and clinical prognosis via analyzing TCGA and CGGA databases. Furthermore, Western blot showed that SFN-Cys downregulated CDK4, CDK6 and p-Rb in a dose-dependent manner and these results were reversed by p-ERK1/2 blocker PD98059 in U87MG and U373MG cells. The reductions of CDK4, CDK6 and p-Rb were reversed by proteasome inhibitor MG132; similarly, the upregulation of 26S proteasome by SFN-Cys was reversed by PD98059. Interestingly, SFN-Cys decreased CDK4 and CDK6 by phosphorylated ERK1/2-caused proteasomal degradation resulting in decreased Rb phosphorylation contributing to cell cycle arrest in G0/G1 phase. Besides, Western blot showed that SFN-Cys downregulated α-tubulin resulting in microtubule disruption and aggregation, and cell cycle arrest in G2/M phase and apoptosis. These results might help us understand the molecular etiology of glioblastoma progression to establish brand-new anti-cancer therapies.
Collapse
Affiliation(s)
- Juntao Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China,Beijing Key Laboratory for Tumor Invasion and Metastasis, Capital Medical University, Beijing, China
| | - Yan Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China,Beijing Key Laboratory for Tumor Invasion and Metastasis, Capital Medical University, Beijing, China
| | - Yuting Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China,Beijing Key Laboratory for Tumor Invasion and Metastasis, Capital Medical University, Beijing, China
| | - Zhongnan Zheng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China,Beijing Key Laboratory for Tumor Invasion and Metastasis, Capital Medical University, Beijing, China
| | - Yabin Hu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China,Beijing Key Laboratory for Tumor Invasion and Metastasis, Capital Medical University, Beijing, China
| | - Wei Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China,Beijing Key Laboratory for Tumor Invasion and Metastasis, Capital Medical University, Beijing, China
| |
Collapse
|
16
|
Long non-coding RNA CRNDE promotes malignant progression of hepatocellular carcinoma through the miR-33a-5p/CDK6 axis. J Physiol Biochem 2020; 76:469-481. [DOI: 10.1007/s13105-020-00754-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 06/15/2020] [Indexed: 02/06/2023]
|
17
|
Han W, Shi J, Cao J, Dong B, Guan W. Current advances of long non-coding RNAs mediated by wnt signaling in glioma. Pathol Res Pract 2020; 216:153008. [PMID: 32703485 DOI: 10.1016/j.prp.2020.153008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/14/2020] [Accepted: 05/10/2020] [Indexed: 12/21/2022]
Abstract
Glioma is the most common and aggressive brain tumor in the central nervous system (CNS), in which Wnt signaling pathway has been verified to play a pivotal role in regulating the initiation and progression. Currently, numerous studies have indicated that long non-coding RNAs (lncRNAs) have critical functions across biological processes including cell proliferation, colony formation, migration, invasion and apoptosis via Wnt signaling pathway in glioma. This review depicts canonical and non-canonical Wnt/β-catenin signaling pathway properties and relative processing mechanisms in gliomas, and summarizes the function and regulation of lncRNAs mediated by Wnt signaling pathway in the development and progression of glioma. Ultimately, we hope to seek out promising biomarkers and reliable therapeutic targets for glioma.
Collapse
Affiliation(s)
- Wei Han
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jia Shi
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jiachao Cao
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Bo Dong
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Wei Guan
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China.
| |
Collapse
|
18
|
Duan Q, Xu M, Wu M, Zhang X, Gan M, Jiang H. Long noncoding RNA UCA1 promotes cell growth, migration, and invasion by targeting miR-143-3p in oral squamous cell carcinoma. Cancer Med 2020; 9:3115-3129. [PMID: 32130788 PMCID: PMC7196043 DOI: 10.1002/cam4.2808] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 12/10/2019] [Accepted: 12/14/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The long noncoding RNA (lncRNA) urothelial carcinoma-associated 1 (UCA1) is dysregulated in many types of tumors; however, its role in oral squamous cell carcinoma (OSCC) remains unclear. This study aims to determine the effect of lncRNA UCA1 on OSCC. METHODS Fifty-six paired OSCC and adjacent nontumorous tissues were collected and the levels of UCA1, miR-143-3p, and MYO6 in the tissues were evaluated by qRT-PCR. In in vitro experiments, cell viability, migration, and invasion were measured by, respectively, performing CCK-8, wound healing, and transwell assays. The target relationships among UCA1, miR-143-3p, and MYO6 were verified by dual-luciferase assay. Western blot and immunohistochemistry were carried out to determine the protein levels. Xenograft mouse model was established to explore the effects of UCA1 in vivo. RESULTS Levels of UCA1 and MYO6 were increased significantly in OSCC, while the level of miR-143-3p was decreased compared with the adjacent nontumorous tissues. UCA1 promoted OSCC cell growth, migration, and invasion both in vitro and in vivo, while miR-143-3p reversed the progression. MYO6 was validated as a target for miR-143-3p, and MYO6 overexpression reversed the effects of miR-143-3p mimic on OSCC cells. CONCLUSION LncRNA UCA1 contributes to the proliferation and metastasis of OSCC cells by targeting miR-143-3p and upregulating its downstream gene MYO6. UCA1 could serve as a promising novel target therapy for treatment of OSCC.
Collapse
Affiliation(s)
- Qingyun Duan
- Jiangsu Key Laboratory of Oral DiseasesNanjing Medical UniversityNanjingJiangsuChina
- Department of Oral and Maxillofacial SurgeryAffiliated Hangzhou First People's HospitalMedical College of Zhejiang UniversityHangzhouZhejiangChina
| | - Mei Xu
- Department of OphtalmologyHangzhou Jianggan District People's HospitalHangzhouZhejiangChina
| | - Meng Wu
- Department of Oral and Maxillofacial SurgeryThe Affiliated Huaian No.1 People's Hospital of Nanjing Medical UniversityHuaianJiangsuChina
| | - Xiong Zhang
- Department of Oral and Maxillofacial SurgeryAffiliated Hangzhou First People's HospitalMedical College of Zhejiang UniversityHangzhouZhejiangChina
| | - Min Gan
- Department of Oral and Maxillofacial SurgeryAffiliated Hangzhou First People's HospitalMedical College of Zhejiang UniversityHangzhouZhejiangChina
| | - Hongbing Jiang
- Jiangsu Key Laboratory of Oral DiseasesNanjing Medical UniversityNanjingJiangsuChina
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Nanjing Medical UniversityNanjingJiangsuChina
| |
Collapse
|
19
|
Wang P, Liu X, Han G, Dai S, Ni Q, Xiao S, Huang J. Downregulated lncRNA UCA1 acts as ceRNA to adsorb microRNA-498 to repress proliferation, invasion and epithelial mesenchymal transition of esophageal cancer cells by decreasing ZEB2 expression. Cell Cycle 2019; 18:2359-2376. [PMID: 31387451 PMCID: PMC6738578 DOI: 10.1080/15384101.2019.1648959] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/04/2019] [Accepted: 07/07/2019] [Indexed: 12/24/2022] Open
Abstract
Objective: Esophageal cancer (EC) is one of the most general malignant tumors in humans. There were few studies researching the connections between lncRNA UCA1 and EC. This study is to research the effect of lncRNA UCA1 adsorbing microRNA-498 (miR-498) as a ceRNA to regulate ZEB2 expression on epithelial mesenchymal transition (EMT), invasion and migration of EC cells. Methods: UCA1, miR-498 and ZEB2 expression in EC tissues and cells was detected by RT-qPCR or western blot analysis. EC cells were transfected with siRNA-UCA1, miR-498 mimics or their controls to determine cell colony, proliferation, cycle distribution, apoptosis, migration and invasion by colony formation assay, CCK-8 assay, flow cytometry, and Transwell assay, respectively. The protein expression of PCNA, c-Myc, E-cadherin, N-cadherin, MMP-2 and MMP-9 was detected by Western blot analysis. The growth rate and weight of transplanted tumor in nude mice were observed. Results: There were overly expressed UCA1 and ZEB2 and lowly expressed miR-498 in EC tissues and cells. LncRNA UCA1 acted as ceRNA to inhibit miR-498 expression and thereby increasing ZEB2 expression. With down-regulated UCA1 and up-regulated miR-498, ZEB2 expression, cell proliferation, colony formation, invasion, migration ability, EMT, tumor growth rate and weight in nude mice were apparently reduced. Conclusion: This study demonstrates that inhibited UCA1 up-regulated miR-498 and down-regulated ZEB2, thereby repressing proliferation activity, invasion, migration, and EMT of EC cells.
Collapse
Affiliation(s)
- Peng Wang
- Department of Oncology, Taizhou People’s Hospital, Taizhou, PR. China
| | - Xinfa Liu
- Anesthesiology Department, Taizhou People’s Hospital, Taizhou, PR. China
| | - Gaohua Han
- Department of Oncology, Taizhou People’s Hospital, Taizhou, PR. China
| | - Shengbin Dai
- Department of Oncology, Taizhou People’s Hospital, Taizhou, PR. China
| | - Qingtao Ni
- Department of Oncology, Taizhou People’s Hospital, Taizhou, PR. China
| | - Shujun Xiao
- Department of Gerontology, Taizhou People’s Hospital, Taizhou, PR. China
| | - Junxing Huang
- Department of Oncology, Taizhou People’s Hospital, Taizhou, PR. China
| |
Collapse
|