1
|
Jalali-Zefrei F, Mousavi SM, Delpasand K, Shourmij M, Farzipour S. Role of Non-coding RNAs on the Radiotherapy Sensitivity and Resistance in Cancer Cells. Curr Gene Ther 2025; 25:113-135. [PMID: 38676526 DOI: 10.2174/0115665232301727240422092311] [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: 12/31/2023] [Revised: 03/21/2024] [Accepted: 03/28/2024] [Indexed: 04/29/2024]
Abstract
Radiotherapy (RT) is an integral part of treatment management in cancer patients. However, one of the limitations of this treatment method is the resistance of cancer cells to radiotherapy. These restrictions necessitate the introduction of modalities for the radiosensitization of cancer cells. It has been shown that Noncoding RNAs (ncRNAs), along with modifiers, can act as radiosensitivity and radioresistant regulators in a variety of cancers by affecting double strand break (DSB), wnt signaling, glycolysis, irradiation induced apoptosis, ferroptosis and cell autophagy. This review will provide an overview of the latest research on the roles and regulatory mechanisms of ncRNA after RT in in vitro and preclinical researches.
Collapse
Affiliation(s)
- Fatemeh Jalali-Zefrei
- Department of Cardiology, Cardiovascular Diseases Research Center, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Seyed Mehdi Mousavi
- Department of Cardiology, Cardiovascular Diseases Research Center, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Kourosh Delpasand
- Razi Clinical Research Development Unit, Razi Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammad Shourmij
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Soghra Farzipour
- Department of Cardiology, Cardiovascular Diseases Research Center, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
2
|
Mosca N, Russo A, Potenza N. Making Sense of Antisense lncRNAs in Hepatocellular Carcinoma. Int J Mol Sci 2023; 24:8886. [PMID: 37240232 PMCID: PMC10219390 DOI: 10.3390/ijms24108886] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/14/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
Transcriptome complexity is emerging as an unprecedented and fascinating domain, especially by high-throughput sequencing technologies that have unveiled a plethora of new non-coding RNA biotypes. This review covers antisense long non-coding RNAs, i.e., lncRNAs transcribed from the opposite strand of other known genes, and their role in hepatocellular carcinoma (HCC). Several sense-antisense transcript pairs have been recently annotated, especially from mammalian genomes, and an understanding of their evolutionary sense and functional role for human health and diseases is only beginning. Antisense lncRNAs dysregulation is significantly involved in hepatocarcinogenesis, where they can act as oncogenes or oncosuppressors, thus playing a key role in tumor onset, progression, and chemoradiotherapy response, as deduced from many studies discussed here. Mechanistically, antisense lncRNAs regulate gene expression by exploiting various molecular mechanisms shared with other ncRNA molecules, and exploit special mechanisms on their corresponding sense gene due to sequence complementarity, thus exerting epigenetic, transcriptional, post-transcriptional, and translational controls. The next challenges will be piecing together the complex RNA regulatory networks driven by antisense lncRNAs and, ultimately, assigning them a function in physiological and pathological contexts, in addition to defining prospective novel therapeutic targets and innovative diagnostic tools.
Collapse
Affiliation(s)
| | | | - Nicoletta Potenza
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy; (N.M.); (A.R.)
| |
Collapse
|
3
|
Xu K, Guo H, Xia A, Wang Z, Wang S, Wang Q. Non-coding RNAs in radiotherapy resistance: Roles and therapeutic implications in gastrointestinal cancer. Biomed Pharmacother 2023; 161:114485. [PMID: 36917887 DOI: 10.1016/j.biopha.2023.114485] [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: 01/03/2023] [Revised: 02/19/2023] [Accepted: 03/07/2023] [Indexed: 03/14/2023] Open
Abstract
Radiotherapy has become an indispensable and conventional means for patients with advanced solid tumors including gastrointestinal cancer. However, innate or acquired radiotherapy resistance remains a significant challenge and greatly limits the therapeutic effect, which results in cancer relapse and poor prognosis. Therefore, it is an urgent need to identify novel biomarkers and therapeutic targets for clarify the biological characteristics and mechanism of radiotherapy resistance. Recently, lots of studies have revealed that non-coding RNAs (ncRNAs) are the potential indicators and regulators of radiotherapy resistance via the mediation of various targets/pathways in different cancers. These findings may serve as a potential therapeutic strategy to overcome radiotherapy resistance. In this review, we will shed light on the recent findings regarding the functions and regulatory mechanisms of ncRNAs following radiotherapy, and comprehensively discuss their potential as biomarkers and therapeutic targets in radiotherapy resistance of gastrointestinal cancer.
Collapse
Affiliation(s)
- Kaiyue Xu
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210000, China; Department of Radiation Oncology, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing University Medical School, Suzhou 215000, China
| | - Huimin Guo
- Department of Gastroenterology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210000, China
| | - Anliang Xia
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210000, China
| | - Zhangding Wang
- Department of Gastroenterology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210000, China.
| | - Shouyu Wang
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210000, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing 210093, China.
| | - Qiang Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230000, China; Medical Transformation Research Institute, The First Affiliated Hospital of Anhui Medical University, Hefei 230000, China.
| |
Collapse
|
4
|
Hashemi M, Mirzaei S, Zandieh MA, Rezaei S, Amirabbas Kakavand, Dehghanpour A, Esmaeili N, Ghahremanzade A, Saebfar H, Heidari H, Salimimoghadam S, Taheriazam A, Entezari M, Ahn KS. Long non-coding RNAs (lncRNAs) in hepatocellular carcinoma progression: Biological functions and new therapeutic targets. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 177:207-228. [PMID: 36584761 DOI: 10.1016/j.pbiomolbio.2022.12.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 11/29/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022]
Abstract
Liver is an important organ in body that performs vital functions such as detoxification. Liver is susceptible to development of cancers, and hepatocellular carcinoma (HCC) is among them. 75-85% of liver cancer cases are related to HCC. Therefore, much attention has been directed towards understanding factors mediating HCC progression. LncRNAs are epigenetic factors with more than 200 nucleotides in length located in both nucleus and cytoplasm and they are promising candidates in cancer therapy. Directing studies towards understanding function of lncRNAs in HCC is of importance. LncRNAs regulate cell cycle progression and growth of HCC cells, and they can also induce/inhibit apoptosis in tumor cells. LncRNAs affect invasion and metastasis in HCC mainly by epithelial-mesenchymal transition (EMT) mechanism. Revealing the association between lncRNAs and downstream signaling pathways in HCC is discussed in the current manuscript. Infectious diseases can affect lncRNA expression in mediating HCC development and then, altered expression level of lncRNA is associated with drug resistance and radio-resistance. Biomarker application of lncRNAs and their role in prognosis and diagnosis of HCC are also discussed to pave the way for treatment of HCC patients.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Mohammad Arad Zandieh
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Sahar Rezaei
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Amirabbas Kakavand
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Amir Dehghanpour
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Negin Esmaeili
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Azin Ghahremanzade
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hamidreza Saebfar
- European University Association, League of European Research Universities, University of Milan, Italy
| | - Hajar Heidari
- Department of Biomedical Sciences, School of Public Health University at Albany State University of New York, Albany, NY, 12208, USA
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Kwang Seok Ahn
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
5
|
Shishido K, Reinders A, Asuthkar S. Epigenetic regulation of radioresistance: insights from preclinical and clinical studies. Expert Opin Investig Drugs 2022; 31:1359-1375. [PMID: 36524403 DOI: 10.1080/13543784.2022.2158810] [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: 12/23/2022]
Abstract
INTRODUCTION Oftentimes, radiation therapy (RT) is ineffective due to the development of radioresistance (RR). However, studies have shown that targeting epigenetic modifiers to enhance radiosensitivity represents a promising direction of clinical investigation. AREAS COVERED This review discusses the mechanisms by which epigenetic modifiers alter radiosensitivity through dysregulation of MAPK-ERK and AKT-mTOR signaling. Finally, we discuss the clinical directions for targeting epigenetic modifiers and current radiology techniques used in the clinic. METHODOLOGY We searched PubMed and ScienceDirect databases from April 4th, 2022 to October 18th, 2022. We examined 226 papers related to radioresistance, epigenetics, MAPK, and PI3K/AKT/mTOR signaling. 194 papers were selected for this review. Keywords used for this search include, 'radioresistance,' 'radiosensitivity,' 'radiation,' 'radiotherapy,' 'particle radiation,' 'photon radiation,' 'epigenetic modifiers,' 'MAPK,' 'AKT,' 'mTOR,' 'cancer,' and 'PI3K.' We examined 41 papers related to clinical trials on the aforementioned topics. Outcomes of interest were safety, overall survival (OS), dose-limiting toxicities (DLT), progression-free survival (PFS), and maximum tolerated dose (MTD). EXPERT OPINION Current studies focusing on epigenetic mechanisms of RR strongly support the use of targeting epigenetic modifiers as adjuvants to standard cancer therapies. To further the success of such treatments and their clinical benefit , both preclinical and clinical studies are needed to broaden the scope of known radioresistant mechanisms.
Collapse
Affiliation(s)
- Katherine Shishido
- Department of Cancer Biology and Pharmacology and Department of Pediatrics, University of Illinois College of Medicine Peoria, Peoria, IL, United States of America
| | - Alexis Reinders
- Department of Cancer Biology and Pharmacology and Department of Pediatrics, University of Illinois College of Medicine Peoria, Peoria, IL, United States of America
| | - Swapna Asuthkar
- Department of Cancer Biology and Pharmacology and Department of Pediatrics, University of Illinois College of Medicine Peoria, Peoria, IL, United States of America
| |
Collapse
|
6
|
[Overexpression of miR-607 inhibits hepatocellular carcinoma cell growth and metastasis by down-regulating TRPC5]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2022; 42:1587-1593. [PMID: 36504050 PMCID: PMC9742782 DOI: 10.12122/j.issn.1673-4254.2022.11.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To investigate the clinical implications of abnormal expression of miR-607 in hepatocellular carcinoma (HCC) and its influence on HCC cell proliferation and migration. METHODS The expression of miR-607 in 45 pairs of HCC and adjacent tissues were detected with real-time PCR, and the correlation between miR-607 expression and clinicopathological features of the patients was analyzed. The effects of transfection with miR-607 mimics on proliferation, apoptosis, migration and invasion of two HCC cell lines (Huh-7 and HCCLM3) were evaluated using CCK-8 assay, flow cytometry, wound-healing assay and Transwell assay. A dual-luciferase reporter system was used to detect the direct binding between miR-607 and 3'-UTR of TRPC5 mRNA. Western blotting was used to measure the expressions of TRPC5, CCND1, MMP2 and phosphorylated Akt in the HCC cells. RESULTS The expression of miR-607 was significantly decreased in HCC tissues (P=0.029) and HCC cell lines (P < 0.05). In HCC patients, a low expression of miR-607 was associated with a larger tumor size (>5 cm, P=0.031), vascular invasion (P=0.027) and advanced TNM stages (Ⅲ + Ⅳ, P=0.015). In the two HCC cell line, overexpression of miR-607 significantly inhibited cell proliferation, migration, and invasion and enhanced cell apoptosis (P < 0.05). The results of dualluciferase reporter assay confirmed that TRPC5 was a direct target of miR- 607 in HCC cells. Overexpression of miR-607 significantly inhibited the expressions of TRPC5, CCND1, and MMP2 and suppressed Akt phosphorylation in HCC cells (P < 0.05). CONCLUSION A low expression of miR-607 in HCC is associated with poor clinicopathological phenotypes of HCC. Overexpression of miR-607 inhibits HCC growth and metastasis possibly by down- regulating TRPC5, which causes Akt signaling inactivation.
Collapse
|
7
|
Abstract
The tumor microenvironment (TME) is a well-recognized system that plays an essential role in tumor initiation, development, and progression. Intense intercellular communication between tumor cells and other cells (especially macrophages) occurs in the TME and is mediated by cell-to-cell contact and/or soluble messengers. Emerging evidence indicates that noncoding RNAs (ncRNAs) are critical regulators of the relationship between cells within the TME. In this review, we provide an update on the regulation of ncRNAs (primarily micro RNAs [miRNAs], long ncRNAs [lncRNAs], and circular RNAs [circRNAs]) in the crosstalk between macrophages and tumor cells in hepatocellular carcinoma (HCC). These ncRNAs are derived from macrophages or tumor cells and act as oncogenes or tumor suppressors, contributing to tumor progression not only by regulating the physiological and pathological processes of tumor cells but also by controlling macrophage infiltration, activation, polarization, and function. Herein, we also explore the options available for clinical therapeutic strategies targeting crosstalk-related ncRNAs to treat HCC. A better understanding of the relationship between macrophages and tumor cells mediated by ncRNAs will uncover new diagnostic biomarkers and pharmacological targets in cancer.
Collapse
|
8
|
Lin P, Xing W, Ren Q, Wang Q, Yan J, Mao G. LncRNAs as Theragnostic Biomarkers for Predicting Radioresistance in Cancer: A Systematic Review and Meta-Analysis. Front Oncol 2022; 12:767750. [PMID: 35692742 PMCID: PMC9176206 DOI: 10.3389/fonc.2022.767750] [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: 09/15/2021] [Accepted: 04/21/2022] [Indexed: 11/23/2022] Open
Abstract
Background Radioresistance is the major obstacle after cancer radiotherapy. The dysregulation of long non-coding RNAs (lncRNAs) was closely related the radioresistance response. This meta-analysis was aimed to interpret the relationship between lncRNAs and radiotherapy responses in different cancers. Method The studies were selected from databases including PubMed, ISI Web of Science, Embase, Google Scholar, PMC, and CNKI (China National Knowledge Infrastructure). The publication time was limited to before March 20, 2021. The hazard ratios (HRs) and 95% confidence interval were calculated with random-effects models. Subgroup analyses, sensitivity analyses, and publication bias were also conducted. Result Twenty-seven lncRNAs in 14 cancer types were investigated, in which 23 lncRNAs were upregulated and four lncRNAs were downregulated. Dysregulation of these lncRNAs were found to be related to radioresistance response. The pooled HR and 95% confidence interval for the combined up-regulated lncRNAs was 1.73 (95% CI=1.50-2.00; P< 0.01) and down-regulated lncRNAs was 2.09 (95% CI= 1.60-2.72; P< 0.01). The HR values of the subgroup analysis for glioma (HR= 2.22, 95% CI= 1.79-2.74; p< 0.01), non-small cell lung cancer (HR=1.48, 95% CI=1.18-1.85; P<0.01), nasopharyngeal carcinoma (HR=4.26; 95% CI= 1.58-11.46; P< 0.01), and breast cancer (HR=1.29; 95% CI= 1.08-1.54; P< 0.01) were obtained. Moreover, the expression of lncRNAs was significantly related to overall survival of patients no matter if the sample size was >50 or not. In addition, the HR values of the subgroup analysis for lncRNA H19 (HR=2.68; 95% CI= 1.92-3.74; P <0.01), lncRNA FAM201A (HR=2.15; 95% CI= 1.15-3.99; P <0.01), and lncRNA HOTAIR (HR=1.22; 95% CI= 0.98-1.54; P =0.08) were also obtained. Conclusion LncRNAs can induce cancer radioresistance by regulating cell death-related signaling pathways. Results indicated that lncRNAs, especially lncRNA H19, FAM201A, and HOTAIR, could be considered as a predictive theragnostic biomarker to evaluate radiotherapy response.
Collapse
Affiliation(s)
- Ping Lin
- Department of Geriatrics, The Third People’s Hospital of Hangzhou, Hangzhou, China
| | - Wenmin Xing
- Zhejiang Provincial Key Laboratory of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou, China
| | - Qian Ren
- Department of Geriatrics, The Third People’s Hospital of Hangzhou, Hangzhou, China
| | - Qin Wang
- Department of Geriatrics, The Third People’s Hospital of Hangzhou, Hangzhou, China
| | - Jing Yan
- Zhejiang Provincial Key Laboratory of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou, China
- *Correspondence: Genxiang Mao, ; Jing Yan,
| | - Genxiang Mao
- Zhejiang Provincial Key Laboratory of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou, China
- *Correspondence: Genxiang Mao, ; Jing Yan,
| |
Collapse
|
9
|
Wu W, Zhang S, He J. The Mechanism of Long Non-coding RNA in Cancer Radioresistance/Radiosensitivity: A Systematic Review. Front Pharmacol 2022; 13:879704. [PMID: 35600868 PMCID: PMC9117703 DOI: 10.3389/fphar.2022.879704] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 04/04/2022] [Indexed: 12/15/2022] Open
Abstract
Background and purpose: Radioresistance remains a significant challenge in tumor therapy. This systematic review aims to demonstrate the role of long non-coding RNA (lncRNA) in cancer radioresistance/radiosensitivity. Material and methods: The electronic databases Pubmed, Embase, and Google Scholar were searched from January 2000 to December 2021 to identify studies addressing the mechanisms of lncRNAs in tumor radioresistance/sensitivity, each of which required both in vivo and in vitro experiments. Results: Among the 87 studies identified, lncRNAs were implicated in tumor radioresistance/sensitivity mainly in three paradigms. 1) lncRNAs act on microRNA (miRNA) by means of a sponge, and their downstream signals include some specific molecular biological processes (DNA repair and chromosome stabilization, mRNA or protein stabilization, cell cycle and proliferation, apoptosis-related pathways, autophagy-related pathways, epithelial-mesenchymal transition (EMT), cellular energy metabolism) and some signaling mediators (transcription factors, kinases, some important signal transduction pathways) that regulate various biological processes. 2) lncRNAs directly interact with proteins, affecting the cell cycle and autophagy to contribute to tumor radioresistance. 3) lncRNAs act like transcription factors to initiate downstream signaling pathways and participate in tumor radioresistance. Conclusion: lncRNAs are important regulators involved in tumor radioresistance\sensitivity. Different lncRNAs may participate in the radioresistance with the same regulatory paradigm, and the same lncRNAs may also participate in the radioresistance in different ways. Future research should focus more on comprehensively characterizing the mechanisms of lncRNAs in tumor radioresistance to help us identify corresponding novel biomarkers and develop new lncRNA-based methods to improve radioresistance.
Collapse
Affiliation(s)
- Wenhan Wu
- Department of General Surgery (Gastrointestinal Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Shijian Zhang
- School of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Jia He
- Faculty Affairs and Human Resources Management Department, Southwest Medical University, Luzhou, China
| |
Collapse
|
10
|
Shafabakhsh R, Arianfar F, Vosough M, Mirzaei HR, Mahjoubin-Tehran M, Khanbabaei H, Kowsari H, Shojaie L, Azar MEF, Hamblin MR, Mirzaei H. Autophagy and gastrointestinal cancers: the behind the scenes role of long non-coding RNAs in initiation, progression, and treatment resistance. Cancer Gene Ther 2021; 28:1229-1255. [PMID: 33432087 DOI: 10.1038/s41417-020-00272-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 10/06/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023]
Abstract
Gastrointestinal (GI) cancers comprise a heterogeneous group of complex disorders that affect different organs, including esophagus, stomach, gallbladder, liver, biliary tract, pancreas, small intestine, colon, rectum, and anus. Recently, an explosion in nucleic acid-based technologies has led to the discovery of long non-coding RNAs (lncRNAs) that have been found to possess unique regulatory functions. This class of RNAs is >200 nucleotides in length, and is characterized by their lack of protein coding. LncRNAs exert regulatory effects in GI cancer development by affecting different functions such as the proliferation and metastasis of cancer cells, apoptosis, glycolysis and angiogenesis. Over the past few decades, considerable evidence has revealed the important role of autophagy in both GI cancer progression and suppression. In addition, recent studies have confirmed a significant correlation between lncRNAs and the regulation of autophagy. In this review, we summarize how lncRNAs play a behind the scenes role in the pathogenesis of GI cancers through regulation of autophagy.
Collapse
Affiliation(s)
- Rana Shafabakhsh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Farzaneh Arianfar
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Massoud Vosough
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 1665659911, Iran
| | - Hamid Reza Mirzaei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Mahjoubin-Tehran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hashem Khanbabaei
- Medical Physics Department, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hamed Kowsari
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Layla Shojaie
- Research Center for Liver Diseases, Keck School of Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa.
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
11
|
Ni J, Huang Z, Wang D. LncRNA TP73-AS1 promotes oxidized low-density lipoprotein-induced apoptosis of endothelial cells in atherosclerosis by targeting the miR-654-3p/AKT3 axis. Cell Mol Biol Lett 2021; 26:27. [PMID: 34103010 PMCID: PMC8188714 DOI: 10.1186/s11658-021-00264-x] [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: 01/18/2021] [Accepted: 05/12/2021] [Indexed: 11/10/2022] Open
Abstract
Background Although lncRNA TP73-AS1 has been shown to play important roles in various human diseases, its function in atherosclerosis (AS) remains unclear. Methods Human aortic endothelial cells (HAECs) were treated with 50 μg/ml oxidized low-density lipoprotein (ox-LDL) to establish an atherosclerotic cell model. The expression of TP73-AS1, miR-654-3p and AKT3 was detected by qRT-PCR. Cell functions were evaluated CCK-8 assay and flow cytometry. The protein levels of apoptosis-related proteins were evaluated by western blot. The binding relationship among TP73-AS1, miR-654-3p and AKT3 was determined by bioinformatics analysis and luciferase reporter assay. Results TP73-AS1 was upregulated and miR-654-3p was downregulated in ox-LDL treated HAECs. TP73-AS1 silencing and miR-654-3p mimics decreased the viability and inhibited apoptosis of ox-LDL treated HAECs, decreased the expression levels of c-caspase-9, c-caspase-3 and Bax, and increased Bcl-2 expression. In addition, miR-654-3p inhibitor significantly reversed the inhibitory effects of si-TP73-AS1 on cell viability and apoptosis. TP73-AS1 could positively regulate AKT3 through directly sponging miR-654-3p. Conclusion TP73-AS1 promoted apoptosis of ox-LDL stimulated endothelial cells by targeting the miR-654-3p/AKT3 axis, suggesting that TP73-AS1 might be a potential target for AS treatment.
Collapse
Affiliation(s)
- Jia Ni
- Stomatological Hospital, Southern Medical University, No. 366 Jiangnan Avenue South, Haizhu District, Guangzhou City, Guangdong Province, People's Republic of China.
| | - Zhen Huang
- Peking University School of Stomatology, Beijing, 100081, People's Republic of China
| | - Dan Wang
- Stomatological Hospital, Southern Medical University, No. 366 Jiangnan Avenue South, Haizhu District, Guangzhou City, Guangdong Province, People's Republic of China.
| |
Collapse
|
12
|
Liu F, Deng W, Wan Z, Xu D, Chen J, Yang X, Xu J. lncRNA MAGI2-AS3 overexpression had antitumor effect on Hepatic cancer via miRNA-23a-3p/PTEN axis. Food Sci Nutr 2021; 9:2517-2530. [PMID: 34026068 PMCID: PMC8116851 DOI: 10.1002/fsn3.2199] [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: 12/15/2020] [Revised: 02/02/2021] [Accepted: 02/06/2021] [Indexed: 11/16/2022] Open
Abstract
The present study aimed to evaluate the antitumor effects of MAGI2-AS3 and its mechanism in liver cancer. Cancer tissues and adjacent nontumor tissues were collected, and lncRNAs were analyzed via chip assay. The correlation between MAGEI2-AS3 and patient pathology and prognosis was then analyzed. Bel-7402 and Huh-7 cell lines were also used in our study. For the in vitro study, MTT assay, flow cytometry, transwell assay, and wound healing assay were conducted to evaluate hepatic cancer cell (Bel-7402 and Huh-7) proliferation, apoptosis, invasion, and migration. The relative mechanisms were evaluated by Western blot (WB) and cellular immunofluorescence. The correlation among MAGI2-AS3, miRNA-23a-3p, and PTEN was determined by a dual-luciferase reporter assay. The expression of lncRNA MAGI2-AS3 was significantly downregulated in tumor tissues. MAGI2-AS3 expression was closely correlation with HCC patient's clinicopathology and prognosis and prognosis. In the cell experiment, compared with the negative control (NC) group, MAGI2-AS3 overexpression reduced cell proliferation, invasion, and migration and increased cell apoptosis in Bel-7402 and Huh-7 cell lines. However, when Bel-7402 and Huh-7 cells were transfected with miRNA-23a-3p, their biological activities (proliferation, invasion, and migration) were significantly increased. Through WB assay, MAGI2-AS3 could increase PTEN and depress p-AKT and MMP-9 protein expressions via miRNA-23a-3p suppression. The dual-luciferase reporter assay revealed that MAGI2-AS3 directly targeted miRNA-23a-3p and that miRNA-23a-3p could target PTEN. MAGI2-AS3 might be a potential therapeutic target for liver cancer owing to its regulation by the miRNA-23a-3p/PTEN axis.
Collapse
Affiliation(s)
- Fei Liu
- Jiangxi Province Hospital of Integrated Chinese and Western MedicineNanchangChina
| | - Wenwen Deng
- Jiangxi Province Hospital of Integrated Chinese and Western MedicineNanchangChina
| | - Zhenda Wan
- Jiangxi Province Hospital of Integrated Chinese and Western MedicineNanchangChina
| | - Dajin Xu
- Jiangxi Province Hospital of Integrated Chinese and Western MedicineNanchangChina
| | - Jun Chen
- Jiangxi Province Hospital of Integrated Chinese and Western MedicineNanchangChina
| | - Xin Yang
- Jiangxi Province Hospital of Integrated Chinese and Western MedicineNanchangChina
| | - Jianhua Xu
- Jiangxi Province Hospital of Integrated Chinese and Western MedicineNanchangChina
| |
Collapse
|
13
|
Ghafouri-Fard S, Gholipour M, Hussen BM, Taheri M. The Impact of Long Non-Coding RNAs in the Pathogenesis of Hepatocellular Carcinoma. Front Oncol 2021; 11:649107. [PMID: 33968749 PMCID: PMC8097102 DOI: 10.3389/fonc.2021.649107] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 03/22/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is among the utmost deadly human malignancies. This type of cancer has been associated with several environmental, viral, and lifestyle risk factors. Among the epigenetic factors which contribute in the pathogenesis of HCC is dysregulation of long non-coding RNAs (lncRNAs). These transcripts modulate expression of several tumor suppressor genes and oncogenes and alter the activity of cancer-related signaling axes. Several lncRNAs such as NEAT1, MALAT1, ANRIL, and SNHG1 have been up-regulated in HCC samples. On the other hand, a number of so-called tumor suppressor lncRNAs namely CASS2 and MEG3 are down-regulated in HCC. The interaction between lncRNAs and miRNAs regulate expression of a number of mRNA coding genes which are involved in the pathogenesis of HCC. H19/miR-15b/CDC42, H19/miR-326/TWIST1, NEAT1/miR-485/STAT3, MALAT1/miR-124-3p/Slug, MALAT1/miR-195/EGFR, MALAT1/miR-22/SNAI1, and ANRIL/miR-144/PBX3 axes are among functional axes in the pathobiology of HCC. Some genetic polymorphisms within non-coding regions of the genome have been associated with risk of HCC in certain populations. In the current paper, we describe the recent finding about the impact of lncRNAs in HCC.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Gholipour
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bashdar Mahmud Hussen
- Pharmacognosy Department, College of Pharmacy, Hawler Medical University, Erbil, Iraq
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Qiu H, Zhang L, Yi T, Yang K, Gong Y, Xie C. Retracted: Long non-coding RNA TP73-AS1 facilitates progression and radioresistance in lung cancer cells by the miR-216a-5p/CUL4B axis with exosome involvement. Thorac Cancer 2021; 12:409. [PMID: 32841514 PMCID: PMC7862789 DOI: 10.1111/1759-7714.13602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/13/2020] [Accepted: 07/15/2020] [Indexed: 12/24/2022] Open
Abstract
Retraction: Qiu, H., Zhang, L., Yi, T., Yang, K., Gong, Y. and Xie, C. (2020), Long non-coding RNA TP73-AS1 facilitates progression and radioresistance in lung cancer cells by the miR-216a-5p/CUL4B axis with exosome involvement. Thorac Cancer. https://doi.org/10.1111/1759-7714.13602 The above article, published online on 25 August 2020 in Wiley Online Library (wileyonlinelibrary.com), has been retracted by agreement among the authors, the journal Editor in Chief Qinghua Zhou, and John Wiley & Sons Ltd. The retraction has been agreed after the results reported in Figures 3B, 3C, 3I, and 3J were found to be not repeatable in authors' further study.
Collapse
Affiliation(s)
- Huibing Qiu
- Department of Radiation and Medical OncologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Xiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
| | - Lingyun Zhang
- Xiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
| | - Tienan Yi
- Xiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
| | - Kai Yang
- Xiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
| | - Yan Gong
- Department of Biological RepositoriesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Conghua Xie
- Department of Radiation and Medical OncologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Key Laboratory of Tumor Biological BehaviorsZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Cancer Clinical Study CenterZhongnan Hospital of Wuhan UniversityWuhanChina
| |
Collapse
|
15
|
Yuan D, Chen Y, Li X, Li J, Zhao Y, Shen J, Du F, Kaboli PJ, Li M, Wu X, Ji H, Cho CH, Wen Q, Li W, Xiao Z, Chen B. Long Non-Coding RNAs: Potential Biomarkers and Targets for Hepatocellular Carcinoma Therapy and Diagnosis. Int J Biol Sci 2021; 17:220-235. [PMID: 33390845 PMCID: PMC7757045 DOI: 10.7150/ijbs.50730] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 11/01/2020] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related death worldwide. Increasing studies showed that long non-coding RNAs (lncRNAs), a novel class of RNAs that are greater than 200 nucleotides in length but lack the ability to encode proteins, exert crucial roles in the occurrence and progression of HCC. LncRNAs promote the proliferation, migration, invasion, autophagy, and apoptosis of tumor cells by regulating downstream target gene expression and cancer-related signaling pathways. Meanwhile, lncRNA can be used as biomarkers to predict the efficacy of HCC treatment strategies, such as surgery, radiotherapy, chemotherapy, and immunotherapy, and as a potential individualized tool for HCC diagnosis and treatment. In this review, we overview up-to-date findings on lncRNAs as potential biomarkers for HCC surgery, radiotherapy, chemotherapy resistance, target therapy, and immunotherapy, and discuss the potential clinical application of lncRNA as tools for HCC diagnosis and treatment.
Collapse
Affiliation(s)
- Donghong Yuan
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.,South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Yu Chen
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xiaobing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Jing Li
- Department of Oncology and Hematology, Hospital (T.C.M) Affiliated to Southwest Medical University, Luzhou, Sichuan, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.,South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.,South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Fukuan Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.,South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Parham Jabbarzadeh Kaboli
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.,South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.,South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.,South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Huijiao Ji
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.,South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Chi Hin Cho
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.,South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Qinglian Wen
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China.,Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Wanping Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.,South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Bo Chen
- Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
16
|
Ebahimzadeh K, Shoorei H, Mousavinejad SA, Anamag FT, Dinger ME, Taheri M, Ghafouri-Fard S. Emerging role of non-coding RNAs in response of cancer cells to radiotherapy. Pathol Res Pract 2020; 218:153327. [PMID: 33422780 DOI: 10.1016/j.prp.2020.153327] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 12/23/2020] [Accepted: 12/23/2020] [Indexed: 01/03/2023]
Abstract
Radiotherapy is an effective method for treatment of a large proportion of human cancers. Yet, the efficacy of this method is precluded by the induction of radioresistance in tumor cells and the radiation-associated injury of normal cells surrounding the field of radiation. These restrictions necessitate the introduction of modalities for either radiosensitization of cancer cells or protection of normal cells against adverse effects of radiation. Non-coding RNAs (ncRNAs) have essential roles in the determination of radiosensitivity. Moreover, ncRNAs can modulate radiation-induced side effects in normal cells. Several microRNAs (miRNAs) such as miR-620, miR-21 and miR-96-5p confer radioresistance, while other miRNAs including miR-340/ 429 confer radiosensitivity. The expression levels of a number of miRNAs are associated with radiation-induced complications such as lung fibrosis or oral mucositis. The expression patterns of several long non-coding RNAs (lncRNAs) such as MALAT1, LINC00630, HOTAIR, UCA1 and TINCR are associated with response to radiotherapy. Taken together, lncRNAs and miRNAs contribute both in modulation of response of cancer cells to radiotherapy and in protection of normal cells from the associated side effects. The current review provides an overview of the roles of these transcripts in these aspects.
Collapse
Affiliation(s)
- Kaveh Ebahimzadeh
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Shoorei
- Department of Anatomical Sciences, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Seyed Ali Mousavinejad
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Marcel E Dinger
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Soudeh Ghafouri-Fard
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
17
|
Pei S, Chen Z, Tan H, Fan L, Zhang B, Zhao C. SLC16A1-AS1 enhances radiosensitivity and represses cell proliferation and invasion by regulating the miR-301b-3p/CHD5 axis in hepatocellular carcinoma. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:42778-42790. [PMID: 32748357 DOI: 10.1007/s11356-020-09998-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 07/02/2020] [Indexed: 06/11/2023]
Abstract
Hepatocellular carcinoma (HCC), a common type of human malignancies, leads to increasing incidence and fairly high mortality. An increasing number of studies have verified that long noncoding RNAs (lncRNAs) played key roles in the development of multiple human cancers. As a biomarker, SLC16A1-AS1 has been reported in non-small cell lung cancer (NSCLC) and oral squamous cell carcinoma (OSCC). Thus, we decided to investigate whether SLC16A1-AS1 exerts its biological function in HCC. In this study, we discovered that SLC16A1-AS1 was obviously downregulated in HCC tissues and cells. Overexpression of SLC16A1-AS1 inhibited HCC cell proliferation, invasion, and epithelial-mesenchymal transition (EMT) process as well as promoted cell apoptosis. Moreover, SLC16A1-AS1 was confirmed to enhance the radiosensitivity of HCC cells. Molecular mechanism exploration suggested that SLC16A1-AS1 served as a sponge for miR-301b-3p and CHD5 was the downstream target gene of miR-301b-3p in HCC cells. Rescue assays implied that CHD5 knockdown could recover the effects of SLC16A1-AS1 overexpression on HCC cellular processes. In brief, our study clarified that SLC16A1-AS1 acted as a tumor suppressor in HCC by targeting the miR-301b-3p/CHD5 axis, which may be a promising diagnostic biomarker and provide promising treatment for HCC patients.
Collapse
Affiliation(s)
- Shenglin Pei
- Department of Anesthesiology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Zuyi Chen
- Department of Intervention, Affiliated Tumor Hospital of Guangxi Medical University, No. 71 Hedi Road, Qingxiu District, Nanning, 530021, Guangxi, China
| | - Huajun Tan
- Department of Intervention, Affiliated Tumor Hospital of Guangxi Medical University, No. 71 Hedi Road, Qingxiu District, Nanning, 530021, Guangxi, China
| | - Liwei Fan
- Department of Intervention, Affiliated Tumor Hospital of Guangxi Medical University, No. 71 Hedi Road, Qingxiu District, Nanning, 530021, Guangxi, China
| | - Baina Zhang
- Department of Intervention, Affiliated Tumor Hospital of Guangxi Medical University, No. 71 Hedi Road, Qingxiu District, Nanning, 530021, Guangxi, China
| | - Chang Zhao
- Department of Intervention, Affiliated Tumor Hospital of Guangxi Medical University, No. 71 Hedi Road, Qingxiu District, Nanning, 530021, Guangxi, China.
| |
Collapse
|
18
|
Han Y, Li F, Xie J, Wang Y, Zhang H. PVT1 Mediates Cell Proliferation, Apoptosis and Radioresistance in Nasopharyngeal Carcinoma Through Regulating miR-515-5p/PIK3CA Axis. Cancer Manag Res 2020; 12:10077-10090. [PMID: 33116864 PMCID: PMC7568593 DOI: 10.2147/cmar.s257583] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 08/12/2020] [Indexed: 01/06/2023] Open
Abstract
Background Radioresistance greatly hinders the treatment of nasopharyngeal carcinoma (NPC). Long noncoding RNA (lncRNA) plasmacytoma variant translocation 1 (PVT1) has been corroborated to participate in diverse cancers, including NPC. Our aim was to investigate the underlying molecular mechanism of PVT1 in NPC radioresistance. Methods Quantitative real-time polymerase chain reaction (qRT-PCR) was utilized to measure the expression levels of PVT1, microRNA (miR)-515-5p and phosphatidylinositol-4, 5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) in NPC tissues and cells. Cell counting kit-8 (CCK8) assay, colony formation assay and flow cytometry assay were employed to detect cell proliferation, radiosensitivity and apoptosis, respectively. The protein levels of Cyclin D1, B-cell lymphoma 2 associated X (Bax), Cleaved-caspase-3, PIK3CA, protein kinase B (AKT) and phosphorylated AKT (p-AKT) in samples were measured by Western blot. The starBase was used to predict the binding sites between miR-515-5p and PVT1 or PIK3CA. Dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay were performed to verify the interaction. Xenograft tumor model was established to investigate the biological role of PVT1 in vivo. Results The levels of PVT1 and PIK3CA were upregulated in NPC tissues and cells, opposite to the expression of miR-515-5p. Knockdown of PVT1 inhibited cell proliferation, radioresistance and promoted cell apoptosis in NPC cells. Meanwhile, PVT1 silencing downregulated Cyclin D1, and upregulated Bax and Cleaved-casp-3 in NPC cells after radiotherapy. Besides, miR-515-5p interacted with PVT1 and targeted PIK3CA in NPC cells. Further studies indicated that PVT1 regulated radioresistance via miR-515-5p/PIK3CA axis and modulated the AKT pathway by interacting with miR-515-5p. Moreover, knockdown of PVT1 suppressed tumor growth in vivo. Conclusion Downregulation of PVT1 inhibited proliferation, radioresistance and promoted apoptosis by downregulating PIK3CA via sponging miR-515-5p in NPC cells.
Collapse
Affiliation(s)
- Yanyan Han
- Department of Otolaryngology, The First Affiliate Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People's Republic of China
| | - Fang Li
- Department of Otolaryngology, Urumqi Eye and ENT Specialist Hospital, Urumqi, Xinjiang, People's Republic of China
| | - Jun Xie
- Department of Otolaryngology, Urumqi Eye and ENT Specialist Hospital, Urumqi, Xinjiang, People's Republic of China
| | - Yi Wang
- Department of Otolaryngology, Urumqi Eye and ENT Specialist Hospital, Urumqi, Xinjiang, People's Republic of China
| | - Hua Zhang
- Department of Otolaryngology, The First Affiliate Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People's Republic of China
| |
Collapse
|
19
|
Wu Y, Zhang Y, Qin X, Geng H, Zuo D, Zhao Q. PI3K/AKT/mTOR pathway-related long non-coding RNAs: roles and mechanisms in hepatocellular carcinoma. Pharmacol Res 2020; 160:105195. [PMID: 32916254 DOI: 10.1016/j.phrs.2020.105195] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 08/30/2020] [Accepted: 09/03/2020] [Indexed: 01/27/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common tumors worldwide with high prevalence and lethality. The oncogenic phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) pathway is a classic dysregulated pathway involved in the pathogenesis of HCC. However, the underlying mechanism for how PI3K/AKT/mTOR pathway aberrantly activates HCC has not been entirely elucidated. The recognition of the functional roles of long non-coding RNAs (lncRNAs) in PI3K/AKT/mTOR signaling axis sheds light on a new dimension to our understanding of hepatocarcinogenesis. In this review, we comprehensively summarize 67 dysregulated PI3K/AKT/mTOR pathway-related lncRNAs in HCC. Many studies have indicated that the 67 dysregulated lncRNAs show oncogenic or anti-oncogenic effects in HCC by regulation on epigenetic, transcriptional and post-transcriptional levels and they play pivotal roles in the initiation of HCC in diverse biological processes like proliferation, metastasis, drug resistance, radio-resistance, energy metabolism, autophagy and so on. Besides, many of these lncRNAs are associated with clinicopathological features and clinical prognosis in HCC, which may provide a potential future application in the diagnosis and therapy of HCC.
Collapse
Affiliation(s)
- Yuting Wu
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China.
| | - Yingshi Zhang
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China.
| | - Xiaochun Qin
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China.
| | - Haobin Geng
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China.
| | - Daiying Zuo
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China.
| | - Qingchun Zhao
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China; Department of Pharmacy, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenhe District, Shenyang 110840, China.
| |
Collapse
|
20
|
Podralska M, Ciesielska S, Kluiver J, van den Berg A, Dzikiewicz-Krawczyk A, Slezak-Prochazka I. Non-Coding RNAs in Cancer Radiosensitivity: MicroRNAs and lncRNAs as Regulators of Radiation-Induced Signaling Pathways. Cancers (Basel) 2020; 12:E1662. [PMID: 32585857 PMCID: PMC7352793 DOI: 10.3390/cancers12061662] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/19/2020] [Accepted: 06/21/2020] [Indexed: 02/07/2023] Open
Abstract
Radiotherapy is a cancer treatment that applies high doses of ionizing radiation to induce cell death, mainly by triggering DNA double-strand breaks. The outcome of radiotherapy greatly depends on radiosensitivity of cancer cells, which is determined by multiple proteins and cellular processes. In this review, we summarize current knowledge on the role of microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), in determining the response to radiation. Non-coding RNAs modulate ionizing radiation response by targeting key signaling pathways, including DNA damage repair, apoptosis, glycolysis, cell cycle arrest, and autophagy. Additionally, we indicate miRNAs and lncRNAs that upon overexpression or inhibition alter cellular radiosensitivity. Current data indicate the potential of using specific non-coding RNAs as modulators of cellular radiosensitivity to improve outcome of radiotherapy.
Collapse
Affiliation(s)
- Marta Podralska
- Institute of Human Genetics, Polish Academy of Sciences, 60-479 Poznań, Poland;
| | - Sylwia Ciesielska
- Department of Systems Biology and Engineering, Faculty of Automatic Control, Electronics and Computer Science, Silesian University of Technology, 44-100 Gliwice, Poland;
| | - Joost Kluiver
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center, Groningen, 9700RB Groningen, The Netherlands; (J.K.); (A.v.d.B.)
| | - Anke van den Berg
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center, Groningen, 9700RB Groningen, The Netherlands; (J.K.); (A.v.d.B.)
| | | | | |
Collapse
|
21
|
Liu J, Zhao SY, Jiang Q, Qu Y, Huang X, Du J, Sun W, Ye Q. Long noncoding RNA MYLK-AS1 promotes growth and invasion of hepatocellular carcinoma through the EGFR/HER2-ERK1/2 signaling pathway. Int J Biol Sci 2020; 16:1989-2000. [PMID: 32398965 PMCID: PMC7211179 DOI: 10.7150/ijbs.43062] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 03/24/2020] [Indexed: 12/16/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) family members EGFR and HER2 play pivotal roles in oncogenesis and tumor progression. Anticancer drugs targeting EGFR and HER2 have been developed. Long noncoding RNAs (lncRNAs) have been reported to regulate cancer development and progression through signaling pathways. However, lncRNAs that regulate EGFR and HER2 expression remain unknown. Here, we show that lncRNA myosin light chain kinase-antisense RNA 1 (MYLK-AS1) promotes EGFR and HER2 expression and activates their downstream signaling pathway. MYLK-AS1 increases hepatocellular carcinoma (HCC) cell proliferation, migration, and invasion in vitro. Consistently, MYLK-AS1 knockdown hinders tumor growth in vivo. Mechanistically, MYLK-AS1 enhances HCC cell proliferation, migration, and invasion through stimulating the EGFR/HER2-extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathway. In addition, MYLK-AS1 is overexpressed in HCC patients and negatively correlated with HCC prognosis. Thus, MYLK-AS1 is an upstream regulator of EGFR/HER2, and acts as an oncogene, suggesting an additional target for cancer therapeutics.
Collapse
Affiliation(s)
- Juan Liu
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing 100850, China.,Department of Hematology, PLA Rocket Force Characteristic Medical Center, Beijing 100088, China
| | - Si-Yuan Zhao
- Medical unit, 91638 Troops, PLA, Beijing 102202, China
| | - Qiwei Jiang
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Yuanyuan Qu
- Department of Hematology, PLA Rocket Force Characteristic Medical Center, Beijing 100088, China
| | - Xiaomei Huang
- Department of Hematology, PLA Rocket Force Characteristic Medical Center, Beijing 100088, China
| | - Jundong Du
- Department of Surgery, Hebei Yanda Hospital, Hebei 065201, China
| | - Wanjun Sun
- Department of Hematology, PLA Rocket Force Characteristic Medical Center, Beijing 100088, China
| | - Qinong Ye
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing 100850, China
| |
Collapse
|
22
|
Long non-coding RNA TP73-AS1 in cancers. Clin Chim Acta 2020; 503:151-156. [DOI: 10.1016/j.cca.2019.12.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 12/26/2019] [Accepted: 12/30/2019] [Indexed: 02/07/2023]
|
23
|
Zhang X, Xie K, Zhou H, Wu Y, Li C, Liu Y, Liu Z, Xu Q, Liu S, Xiao D, Tao Y. Role of non-coding RNAs and RNA modifiers in cancer therapy resistance. Mol Cancer 2020; 19:47. [PMID: 32122355 PMCID: PMC7050132 DOI: 10.1186/s12943-020-01171-z] [Citation(s) in RCA: 177] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 02/24/2020] [Indexed: 02/08/2023] Open
Abstract
As the standard treatments for cancer, chemotherapy and radiotherapy have been widely applied to clinical practice worldwide. However, the resistance to cancer therapies is a major challenge in clinics and scientific research, resulting in tumor recurrence and metastasis. The mechanisms of therapy resistance are complicated and result from multiple factors. Among them, non-coding RNAs (ncRNAs), along with their modifiers, have been investigated to play key roles in regulating tumor development and mediating therapy resistance within various cancers, such as hepatocellular carcinoma, breast cancer, lung cancer, gastric cancer, etc. In this review, we attempt to elucidate the mechanisms underlying ncRNA/modifier-modulated resistance to chemotherapy and radiotherapy, providing some therapeutic potential points for future cancer treatment.
Collapse
Affiliation(s)
- Xinyi Zhang
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, School of Basic Medicine, Central South University, Changsha, 410078, Hunan, China
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Department of Cardiovascular Medicine, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Kai Xie
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Honghua Zhou
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Department of Cardiovascular Medicine, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Yuwei Wu
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Department of Cardiovascular Medicine, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Chan Li
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yating Liu
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, School of Basic Medicine, Central South University, Changsha, 410078, Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute, Central South University, Changsha, 410078, Hunan, China
- Hunan Key Laboratory of Early Diagnosis and Precision Therapy, Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Zhaoya Liu
- Department of Geriatrics, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Qian Xu
- Department of Cardiovascular Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Shuang Liu
- Department of Oncology, Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Desheng Xiao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, School of Basic Medicine, Central South University, Changsha, 410078, Hunan, China.
| | - Yongguang Tao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, School of Basic Medicine, Central South University, Changsha, 410078, Hunan, China.
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute, Central South University, Changsha, 410078, Hunan, China.
- Hunan Key Laboratory of Early Diagnosis and Precision Therapy, Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| |
Collapse
|