1
|
Gao X, Liu K, Luo S, Tang M, Liu N, Jiang C, Fang J, Li S, Hou Y, Guo C, Qu K. Comparative analysis of methodologies for detecting extrachromosomal circular DNA. Nat Commun 2024; 15:9208. [PMID: 39448595 PMCID: PMC11502736 DOI: 10.1038/s41467-024-53496-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
Extrachromosomal circular DNA (eccDNA) is crucial in oncogene amplification, gene transcription regulation, and intratumor heterogeneity. While various analysis pipelines and experimental methods have been developed for eccDNA identification, their detection efficiencies have not been systematically assessed. To address this, we evaluate the performance of 7 analysis pipelines using seven simulated datasets, in terms of accuracy, identity, duplication rate, and computational resource consumption. We also compare the eccDNA detection efficiency of 7 experimental methods through twenty-one real sequencing datasets. Here, we show that Circle-Map and Circle_finder (bwa-mem-samblaster) outperform the other short-read pipelines. However, Circle_finder (bwa-mem-samblaster) exhibits notable redundancy in its outcomes. CReSIL is the most effective pipeline for eccDNA detection in long-read sequencing data at depths higher than 10X. Moreover, long-read sequencing-based Circle-Seq shows superior efficiency in detecting copy number-amplified eccDNA over 10 kb in length. These results offer valuable insights for researchers in choosing the suitable methods for eccDNA research.
Collapse
Affiliation(s)
- Xuyuan Gao
- Department of Oncology, The First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ke Liu
- Department of Oncology, The First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Songwen Luo
- Department of Oncology, The First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Meifang Tang
- Department of Oncology, The First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
| | - Nianping Liu
- Department of Oncology, The First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Chen Jiang
- Department of Oncology, The First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
| | - Jingwen Fang
- Department of Oncology, The First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- HanGene Biotech, Xiaoshan Innovation Polis, Hangzhou, Zhejiang, China
| | - Shouzhen Li
- Department of Oncology, The First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yanbing Hou
- Department of Oncology, The First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Chuang Guo
- Department of Oncology, The First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- School of Pharmacy, Bengbu Medical University, Bengbu, China.
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| | - Kun Qu
- Department of Oncology, The First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China.
- School of Biomedical Engineering, Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, China.
| |
Collapse
|
2
|
Sheikhnia F, Fazilat A, Rashidi V, Azizzadeh B, Mohammadi M, Maghsoudi H, Majidinia M. Exploring the therapeutic potential of quercetin in cancer treatment: Targeting long non-coding RNAs. Pathol Res Pract 2024; 260:155374. [PMID: 38889494 DOI: 10.1016/j.prp.2024.155374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 05/11/2024] [Accepted: 05/28/2024] [Indexed: 06/20/2024]
Abstract
The escalating global incidence of cancer, which results in millions of fatalities annually, underscores the pressing need for effective pharmacological interventions across diverse cancer types. Long noncoding RNAs (lncRNAs), a class of RNA molecules that lack protein-coding capacity but profoundly impact gene expression regulation, have emerged as pivotal players in key cellular processes, including proliferation, apoptosis, metastasis, cellular metabolism, and drug resistance. Among natural compounds, quercetin, a phenolic compound abundantly present in fruits and vegetables has garnered attention due to its significant anticancer properties. Quercetin demonstrates the ability to inhibit cancer cell growth and induce apoptosis-a process often impaired in malignant cells. In this comprehensive review, we delve into the therapeutic potential of quercetin in cancer treatment, with a specific focus on its intricate interactions with lncRNAs. We explore how quercetin modulates lncRNA expression and function to exert its anticancer effects. Notably, quercetin suppresses oncogenic lncRNAs that drive cancer development and progression while enhancing tumor-suppressive lncRNAs that impede cancer growth and dissemination. Additionally, we discuss quercetin's role as a chemopreventive agent, which plays a crucial role in mitigating cancer risk. We address research challenges and future directions, emphasizing the necessity for in-depth mechanistic studies and strategies to enhance quercetin's bioavailability and target specificity. By synthesizing existing knowledge, this review underscores quercetin's promising potential as a novel therapeutic strategy in the ongoing battle against cancer, offering fresh insights and avenues for further investigation in this critical field.
Collapse
Affiliation(s)
- Farhad Sheikhnia
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran; Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Ahmad Fazilat
- Motamed Cancer Institute, Breast Cancer Research Center, ACECR, Tehran, Iran
| | - Vahid Rashidi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Bita Azizzadeh
- Department of Biochemistry, School of Medicine, Ilam University of Medical sciences, Ilam, Iran
| | - Mahya Mohammadi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Maghsoudi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran; Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Maryam Majidinia
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
3
|
Wu S, Dai X, Zhu Z, Fan D, Jiang S, Dong Y, Chen B, Xie Q, Yao Z, Li Q, Thorne RF, Lu Y, Gu H, Hu W. Reciprocal regulation of lncRNA MEF and c-Myc drives colorectal cancer tumorigenesis. Neoplasia 2024; 49:100971. [PMID: 38301392 PMCID: PMC10847691 DOI: 10.1016/j.neo.2024.100971] [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: 08/23/2023] [Revised: 01/09/2024] [Accepted: 01/09/2024] [Indexed: 02/03/2024]
Abstract
More than half of all cancers demonstrate aberrant c-Myc expression, making this arguably the most important human oncogene. Deregulated long non-coding RNAs (lncRNAs) are also commonly implicated in tumorigenesis, and some limited examples have been established where lncRNAs act as biological tuners of c-Myc expression and activity. Here, we demonstrate that the lncRNA denoted c-Myc Enhancing Factor (MEF) enjoys a cooperative relationship with c-Myc, both as a transcriptional target and driver of c-Myc expression. Mechanistically, MEF functions by binding to and stabilizing the expression of hnRNPK in colorectal cancer cells. The MEF-hnRNPK interaction serves to disrupt binding between hnRNPK and the E3 ubiquitin ligase TRIM25, which attenuates TRIM25-dependent hnRNPK ubiquitination and proteasomal destruction. In turn, the stabilization of hnRNPK through MEF enhances c-Myc expression by augmenting the translation c-Myc. Moreover, modulating the expression of MEF in shRNA-mediated knockdown and overexpression studies revealed that MEF expression is essential for colorectal cancer cell proliferation and survival, both in vitro and in vivo. From the clinical perspective, we show that MEF expression is differentially increased in colorectal cancer tissues compared to normal adjacent tissues. Further, correlations exist between MEF, c-Myc, and hnRNPK suggesting the MEF-c-Myc positive feedback loop is active in patients. Together these data demonstrate that MEF is a pivotal partner of the c-Myc network and propose MEF as a valuable therapeutic target for colorectal cancer.
Collapse
Affiliation(s)
- Shuang Wu
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230027, China
| | - Xiangyu Dai
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230027, China; Translational Research Institute, People's Hospital of Zhengzhou University, Academy of Medical Science, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Tianjian Laboratory of Advanced Biomedical Sciences, State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, 450003, China
| | - Zhipu Zhu
- Translational Research Institute, People's Hospital of Zhengzhou University, Academy of Medical Science, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Tianjian Laboratory of Advanced Biomedical Sciences, State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, 450003, China
| | - Dianhui Fan
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230027, China
| | - Su Jiang
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230027, China
| | - Yi Dong
- Translational Research Institute, People's Hospital of Zhengzhou University, Academy of Medical Science, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Tianjian Laboratory of Advanced Biomedical Sciences, State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, 450003, China
| | - Bing Chen
- Translational Research Institute, People's Hospital of Zhengzhou University, Academy of Medical Science, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Tianjian Laboratory of Advanced Biomedical Sciences, State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, 450003, China
| | - Qi Xie
- Translational Research Institute, People's Hospital of Zhengzhou University, Academy of Medical Science, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Tianjian Laboratory of Advanced Biomedical Sciences, State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, 450003, China
| | - Zhihui Yao
- Translational Research Institute, People's Hospital of Zhengzhou University, Academy of Medical Science, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Tianjian Laboratory of Advanced Biomedical Sciences, State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, 450003, China
| | - Qun Li
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230027, China
| | - Rick Francis Thorne
- Translational Research Institute, People's Hospital of Zhengzhou University, Academy of Medical Science, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Tianjian Laboratory of Advanced Biomedical Sciences, State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, 450003, China
| | - Yao Lu
- Department of Anesthesiology, the First Affiliated of Anhui Medical University, Anhui Medical University, Hefei 230022, China.
| | - Hao Gu
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230027, China.
| | - Wanglai Hu
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230027, China; Translational Research Institute, People's Hospital of Zhengzhou University, Academy of Medical Science, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Tianjian Laboratory of Advanced Biomedical Sciences, State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, 450003, China.
| |
Collapse
|
4
|
Xu X, Li H, Xie M, Zhou Z, Wang D, Mao W. LncRNAs and related molecular basis in malignant pleural mesothelioma: challenges and potential. Crit Rev Oncol Hematol 2023; 186:104012. [PMID: 37116816 DOI: 10.1016/j.critrevonc.2023.104012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 04/04/2023] [Accepted: 04/24/2023] [Indexed: 04/30/2023] Open
Abstract
Malignant pleural mesothelioma (MPM) is a rare but invasive cancer, which mainly arises from mesothelial tissues of pleura, peritoneum and pericardium. Despite significant advances in treatments, the prognosis of MPM patients remains poor, and the 5-year survival rate is less than 10%. Therefore, it is urgent to explore novel therapeutic targets for the treatment of MPM. Growing evidence has indicated that long non-coding RNAs (lncRNAs) potentially could be promising therapeutic targets for numerous cancers. In this regard, lncRNAs might also potentially therapeutic targets for MPM. Recent advances have been made to investigate the molecular basis of MPM. This review first provides a comprehensive overview of roles of lncRNAs in MPM and then discusses the relationship between molecular basis of MPM and MPM-related lncRNAs to implement them as promising therapeutic targets for MPM.
Collapse
Affiliation(s)
- Xiaoling Xu
- Key Laboratory on Diagnosis and Treatment Technology on Thoracic Cancer, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Huihui Li
- Key Laboratory on Diagnosis and Treatment Technology on Thoracic Cancer, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Mingying Xie
- Key Laboratory on Diagnosis and Treatment Technology on Thoracic Cancer, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Zichao Zhou
- Key Laboratory on Diagnosis and Treatment Technology on Thoracic Cancer, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Ding Wang
- Key Laboratory on Diagnosis and Treatment Technology on Thoracic Cancer, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Weimin Mao
- Key Laboratory on Diagnosis and Treatment Technology on Thoracic Cancer, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, China; Department of Thoracic Surgery, Zhejiang Cancer Hospital (Zhejiang Cancer Research Institute), Hangzhou, Zhejiang Province, China.
| |
Collapse
|
5
|
Ranga S, Yadav R, Chhabra R, Chauhan MB, Tanwar M, Yadav C, Kadian L, Ahuja P. Long non-coding RNAs as critical regulators and novel targets in cervical cancer: current status and future perspectives. Apoptosis 2023:10.1007/s10495-023-01840-6. [PMID: 37095313 PMCID: PMC10125867 DOI: 10.1007/s10495-023-01840-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/27/2023] [Indexed: 04/26/2023]
Abstract
Cervical cancer is among the leading causes of cancer-associated mortality in women. In spite of vaccine availability, improved screening procedures, and chemoradiation therapy, cervical cancer remains the most commonly diagnosed cancer in 23 countries and the leading cause of cancer deaths in 36 countries. There is, therefore, a need to come up with novel diagnostic and therapeutic targets. Long non-coding RNAs (lncRNAs) play a remarkable role in genome regulation and contribute significantly to several developmental and disease pathways. The deregulation of lncRNAs is often observed in cancer patients, where they are shown to affect multiple cellular processes, including cell cycle, apoptosis, angiogenesis, and invasion. Many lncRNAs are found to be involved in the pathogenesis as well as progression of cervical cancer and have shown potency to track metastatic events. This review provides an overview of lncRNA mediated regulation of cervical carcinogenesis and highlights their potential as diagnostic and prognostic biomarkers as well as therapeutic targets for cervical cancer. In addition, it also discusses the challenges associated with the clinical implication of lncRNAs in cervical cancer.
Collapse
Affiliation(s)
- Shalu Ranga
- Associate Professor, Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Ritu Yadav
- Associate Professor, Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana, 124001, India.
| | - Ravindresh Chhabra
- Assistant Professor, Department of Biochemistry, Central University of Punjab, Bathinda, Punjab, 151401, India.
| | - Meenakshi B Chauhan
- Department of Obstetrics and Gynaecology, Pandit Bhagwat Dayal Sharma University of Health Sciences, Rohtak, Haryana, 124001, India
| | - Mukesh Tanwar
- Associate Professor, Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Chetna Yadav
- Associate Professor, Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Lokesh Kadian
- School of Medicine, Indiana University, Indianapolis, IN, 46202, USA
| | - Parul Ahuja
- Associate Professor, Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| |
Collapse
|
6
|
Dong S, Wang W, Liao Z, Fan Y, Wang Q, Zhang L. MYC-activated LINC00607 promotes hepatocellular carcinoma progression by regulating the miR-584-3p/ROCK1 axis. J Gene Med 2023; 25:e3477. [PMID: 36740760 DOI: 10.1002/jgm.3477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 11/27/2022] [Accepted: 12/20/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND There have been many reports of long non-coding RNAs (lncRNAs) in tumors, and abnormally expressed lncRNA is closely related to hepatocellular carcinoma (HCC). The mechanism of LINC00607 in HCC has not been reported. METHODS We utilized qPCR to evaluate the RNA expression level. The mechanism of MYC binding to the LINC00607 promoter was revealed through chromatin immunoprecipitation assay and dual luciferase reporter assay. The proliferation and invasive ability were evaluated by CCK-8 and transwell assays. The relation between LINC00607 and miR-584-3p was assessed by RNA immunoprecipitation assay and dual luciferase reporter assay. The level of ROCK1 was evaluated by qPCR and western blot. RESULTS In this research, we found that the expression of LINC00607 was higher in HCC tissues when compared with that in the adjacent non-tumor tissues. Meanwhile, MYC was observed to interact with the LINC00607 promoter, leading to the upregulation of LINC00607 in HCC. We further revealed that LINC00607 functioned as a sponge for miR-584-3p. Cell proliferation and migration assays showed that miR-584-3p may inhibit the HCC progression. Moreover, we found that the miR-584-3p inhibitor could reverse the effects of LINC00607 downregulation in HCC through rescue experiments. Through verification, miR-584-3p bound to the 3' UTR of ROCK1 to downregulate its expression. CONCLUSION LINC00607 regulated by MYC can promote the proliferation, migration and invasion of HCC cells through the miR-584-3p/ROCK1 axis.
Collapse
Affiliation(s)
- Shuilin Dong
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China
| | - Wei Wang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China
| | - Zhibin Liao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China
| | - Yawei Fan
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China
| | - Qi Wang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China
| | - Lei Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China.,Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Shanxi Medical University; Shanxi Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Taiyuan, China
| |
Collapse
|
7
|
Li R, Wang X, Zhu C, Wang K. lncRNA PVT1: a novel oncogene in multiple cancers. Cell Mol Biol Lett 2022; 27:84. [PMID: 36195846 PMCID: PMC9533616 DOI: 10.1186/s11658-022-00385-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 09/07/2022] [Indexed: 12/01/2022] Open
Abstract
Long noncoding RNAs are involved in epigenetic gene modification, including binding to the chromatin rearrangement complex in pre-transcriptional regulation and to gene promoters in gene expression regulation, as well as acting as microRNA sponges to control messenger RNA levels in post-transcriptional regulation. An increasing number of studies have found that long noncoding RNA plasmacytoma variant translocation 1 (PVT1) plays an important role in cancer development. In this review of a large number of studies on PVT1, we found that PVT1 is closely related to tumor onset, proliferation, invasion, epithelial–mesenchymal transformation, and apoptosis, as well as poor prognosis and radiotherapy and chemotherapy resistance in some cancers. This review comprehensively describes PVT1 expression in various cancers and presents novel approaches to the diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Ruiming Li
- Department of Urology, Shengjing Hospital of China Medical University, #36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning, China
| | - Xia Wang
- Department of Urology, Shengjing Hospital of China Medical University, #36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning, China
| | - Chunming Zhu
- Department of Family Medicine, Shengjing Hospital of China Medical University, #36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning, China.
| | - Kefeng Wang
- Department of Urology, Shengjing Hospital of China Medical University, #36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning, China.
| |
Collapse
|
8
|
Cheng Z, Guo Y, Sun J, Zheng L. Four-copy number alteration (CNA)-related lncRNA prognostic signature for liver cancer. Sci Rep 2022; 12:14261. [PMID: 35995822 PMCID: PMC9395537 DOI: 10.1038/s41598-022-17927-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 08/02/2022] [Indexed: 11/26/2022] Open
Abstract
The objective of this study was to identify CNA-related lncRNAs that can better evaluate the prognosis of patients with liver cancer. Prognostic molecular subtypes were identified, followed by tumor mutation and differential expression analyses. Genomic copy number anomalies and their association with lncRNAs were also evaluated. A risk model was built based on lncRNAs, as well as a nomogram, and the differences in the tumor immune microenvironment and drug sensitivity between the High_ and Low_risk groups were compared. Weighted gene co-expression network analysis was used to identify modules with significant enrichment in prognostic-related lncRNAs. In total, two subtypes were identified, TP53 and CTNNB1 were common high-frequency mutated genes in the two subtypes. A total of 8,372 differentially expressed (DE) mRNAs and 798 DElncRNAs were identified between cluster1 and cluster2. In addition, a four-lncRNA signature was constructed, and statistically significant differences between the Low_ and High_risk groups were found in terms of CD8 T cells, resting memory CD4 T cells, etc. Enrichment analysis showed that prognostic-related lncRNAs were involved in the cell cycle, p53 signaling pathway, non-alcoholic fatty liver disease, etc. A prognostic prediction signature, based on four-CNA-related lncRNAs, could contribute to a more accurate prognosis of patients with liver cancer.
Collapse
Affiliation(s)
- Zhenyun Cheng
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, No. 1 Jian She East Road, Zhengzhou, Henan, People's Republic of China, 450052.,Key Clinical Laboratory of Henan Province, NO.1 Jian She East Road, Zhengzhou, Henan, People's Republic of China, 450052
| | - Yan Guo
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, No. 1 Jian She East Road, Zhengzhou, Henan, People's Republic of China, 450052.,Key Clinical Laboratory of Henan Province, NO.1 Jian She East Road, Zhengzhou, Henan, People's Republic of China, 450052
| | - Jingjing Sun
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, No. 1 Jian She East Road, Zhengzhou, Henan, People's Republic of China, 450052.,Key Clinical Laboratory of Henan Province, NO.1 Jian She East Road, Zhengzhou, Henan, People's Republic of China, 450052
| | - Lei Zheng
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, No. 1 Jian She East Road, Zhengzhou, Henan, People's Republic of China, 450052. .,Key Clinical Laboratory of Henan Province, NO.1 Jian She East Road, Zhengzhou, Henan, People's Republic of China, 450052.
| |
Collapse
|
9
|
Zhang J, Shen Q, Xia L, Zhu X, Zhu X. DYNLT3 overexpression induces apoptosis and inhibits cell growth and migration via inhibition of the Wnt pathway and EMT in cervical cancer. Front Oncol 2022; 12:889238. [PMID: 35965516 PMCID: PMC9372440 DOI: 10.3389/fonc.2022.889238] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 07/01/2022] [Indexed: 12/24/2022] Open
Abstract
The role of the dynein light chain Tctex-type 3 (DYNLT3) protein in the biological behavior of cervical cancer and its relative molecular mechanisms were investigated. Immunohistochemical staining was used to detect DYNLT3 protein expression in cervical cancer tissues. Cell proliferation and apoptosis rates and invasiveness and migratory capacities were determined by CCK-8 assays, BrdU staining assays and colony formation assays, fluorescence activated cell sorting (FACS), wound healing assays, and Transwell invasion assays of cervical cancer cells after DYNLT3 modulation. The expression levels of Wnt signaling pathway- and EMT-related proteins were examined by Western blotting. Furthermore, the effects of DYNLT3 on the tumorigenicity and metastasis of cervical cancer in nude mice were analyzed by performing immunohistochemistry, and we found that the expression level of the DYNLT3 protein was higher in human normal cervical tissues than in cervical cancer tissues. Overexpression of DYNLT3 obviously attenuated the proliferation, migration and invasion of CaSki and SiHa cells, and promoted cell apoptosis. Upregulation of DYNLT3 expression markedly decreased the expression of Wnt signaling pathway-related proteins (Dvl2, Dvl3, p-LRP6, Wnt3a, Wnt5a/b, Naked1, Naked2, β-catenin and C-Myc) and EMT-related proteins (N-cadherin, SOX2, OCT4, vimentin and Snail), and increased the expression of E-cadherin and Axin1. However, the opposite results were observed after down-regulation of DYNLT3 expression. Up-regulation of DYNLT3 expression significantly inhibited tumor growth in a nude mouse model, while downregulation of DYNLT3 showed the opposite results. In addition, the major metastatic site of cervical cancer cells in mice was the lung, and downregulation of DYNLT3 expression increased cancer metastasis in vivo. DYNLT3 exerted inhibitory effects on cervical cancer by inhibiting cell proliferation, migration and invasion, promoting cell apoptosis in vitro, and inhibiting tumor growth and metastasis in vivo, possibly by suppressing the Wnt signaling pathway and the EMT.
Collapse
Affiliation(s)
- Jianan Zhang
- Center of Uterine Cancer Diagnosis and Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qi Shen
- Center of Uterine Cancer Diagnosis and Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lu Xia
- Center of Uterine Cancer Diagnosis and Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xueqiong Zhu
- Center of Uterine Cancer Diagnosis and Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xuejie Zhu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
10
|
Wang D, Liu X, Cao L, Gong S, He Y, Jiang X, Wang Z. miR-486-3p Controls the Apoptosis of Endometrial Carcinoma Cells. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.2985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Our study aimed to discuss the mechanism of miR-486-3p in controlling the apoptosis of endometrial carcinoma (EC) cells. EC cells were divided into NC group, miR-486-3p mimic and miR-486-3p inhibitor group followed by analysis of miR-486-3p level by Real-time PCR, cell proliferation
by spectrophotometric method, apoptosis by FCM, cell migration and invasion by Transwell analysis. EC cells showed reduced miR-486-3p level. The EC malignant biological behaviors could be prompted through retraining miR-486-3p level with increased EC cell invasive capacity. DDR1 was a target
of miR-486-3p. The variation of tumor activity could be regulated through controlling DDR1 expression. In conclusion, the apoptotic and invasive characteristic of EC cells are restrained after overexpression of miR-486-3p in EC cells through targeting DDR1, indicating that miR-486-3p could
be considered to be one kind of brand-new target for the treatment of EC.
Collapse
Affiliation(s)
- Donghua Wang
- Department of Obstetrics and Gynecology, Wuhan No.1 Hospital, Wuhan, Hubei, 430022, China
| | - Xiaoli Liu
- Department of Obstetrics and Gynecology, Wuhan No.1 Hospital, Wuhan, Hubei, 430022, China
| | - Lirong Cao
- Department of Obstetrics and Gynecology, Wuhan No.1 Hospital, Wuhan, Hubei, 430022, China
| | - Shixiong Gong
- Department of Obstetrics and Gynecology, Wuhan No.1 Hospital, Wuhan, Hubei, 430022, China
| | - Yi He
- Department of Obstetrics and Gynecology, Wuhan No.1 Hospital, Wuhan, Hubei, 430022, China
| | - Xiangbin Jiang
- Department of Obstetrics and Gynecology, Wuhan No.1 Hospital, Wuhan, Hubei, 430022, China
| | - Zhongxian Wang
- Department of Obstetrics and Gynecology, Wuhan No.1 Hospital, Wuhan, Hubei, 430022, China
| |
Collapse
|
11
|
Li X, Yuan Y, Wang Y, Xie K, Lu S, Chen F, Zhou M, Zhen P. MicroRNA-486-3p promotes the proliferation and metastasis of cutaneous squamous cell carcinoma by suppressing flotillin-2. J Dermatol Sci 2022; 105:18-26. [PMID: 34930675 DOI: 10.1016/j.jdermsci.2021.11.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/26/2021] [Accepted: 11/11/2021] [Indexed: 01/22/2023]
Abstract
BACKGROUND Dysregulation of miR-486-3p was related to the growth and development of a variety of cancers, but the specific function of miR-486-3p in cutaneous squamous cell carcinoma (cSCC) is not to be confirmed yet. OBJECTIVE Our present study aimed to validate the potential molecular mechanisms of miR-486-3p in cSCC and the potential of miR-486-3p as a novel target for future treatment. METHODS Human cSCC samples and normal skin tissues were applied to determine the expression level of miR-486-3p and FLOT2 by fluorescence in situ hybridization (FISH) and quantitative reverse transcription PCR (qRT-PCR), respectively. As well as BALB/C nude mouse tumor model, three cSCC cells lines including HSC-1, HSC-5 and A431 were utilized to demonstrate the potential function of miR-486-3p and FLOT2 in tumorigenesis. RESULTS Our experimental results showed that miR-486-3p was highly expressed both in tumor samples and cell lines of cSCC. Upregulation of miR-486-3p enhanced the proliferation and migration ability of cSCC cell lines and promoted tumorigenicity in vivo. Furthermore, we confirmed that FLOT2 was a direct targeted gene of miR-486-3p. In contrary to the expression level of miR-486-3p, FLOT2 was low expressed in cSCC patient specimens and cell lines. Knockdown of FLOT2 promoted tumorigenesis of cSCC; whereas FLOT2 reversed the tumor-promoting effect of miR-486-3p. CONCLUSION Our data exhibited that miR-486-3p exerted its effects on carcinogenesis as an oncogene in cSCC via suppression of FLOT2. This discovery will develop new therapeutic targets of cSCC.
Collapse
Affiliation(s)
- Xiangzhi Li
- Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, China; Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yawen Yuan
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yimeng Wang
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Kaisheng Xie
- Department of Pathology, The Second Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, Liuzhou, China
| | - Sheng Lu
- The First School of Clinical Medicine, Southern Medical University, Guangdong, China
| | - Fuqiang Chen
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Meijuan Zhou
- Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, China; Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China.
| | - Peilin Zhen
- Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, China.
| |
Collapse
|
12
|
Pan J, Huang G, Yin Z, Cai X, Gong E, Li Y, Xu C, Ye Z, Cao Z, Cheng W. Circular RNA FLNA acts as a sponge of miR-486-3p in promoting lung cancer progression via regulating XRCC1 and CYP1A1. Cancer Gene Ther 2022; 29:101-121. [PMID: 33500536 PMCID: PMC8761575 DOI: 10.1038/s41417-021-00293-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 11/18/2020] [Accepted: 01/05/2021] [Indexed: 01/29/2023]
Abstract
Significantly high-expressed circFLNA has been found in various cancer cell lines, but not in lung cancer. Therefore, this study aimed to explore the role of circFLNA in the progression of lung cancer. The target gene of circFLNA was determined by bioinformatics and luciferase reporter assay. Viability, proliferation, migration, and invasion of the transfected cells were detected by CCK-8, colony formation, wound-healing, and transwell assays, respectively. A mouse subcutaneous xenotransplanted tumor model was established, and the expressions of circFLNA, miR-486-3p, XRCC1, CYP1A1, and related genes in the cancer cells and tissues were detected by RT-qPCR, Western blot, or immunohistochemistry. The current study found that miR-486-3p was low-expressed in lung cancer. MiR-486-3p, which has been found to target XRCC1 and CYP1A1, was regulated by circFLNA. CircFLNA was located in the cytoplasm and had a high expression in lung cancer cells. Cancer cell viability, proliferation, migration, and invasion were promoted by overexpressed circFLNA, XRCC1, and CYP1A1 but inhibited by miR-486-3p mimic and circFLNA knockdown. The weight of the xenotransplanted tumor was increased by circFLNA overexpression yet reduced by miR-486-3p mimic. Furthermore, miR-486-3p mimic reversed the effect of circFLNA overexpression on promoting lung cancer cells and tumors and regulating the expressions of miR-486-3p, XRCC1, CYP1A1, and metastasis/apoptosis/proliferation-related factors. However, overexpressed XRCC1 and CYP1A1 reversed the inhibitory effect of miR-486-3p mimic on cancer cells and tumors. In conclusion, circFLNA acted as a sponge of miR-486-3p to promote the proliferation, migration, and invasion of lung cancer cells in vitro and in vivo by regulating XRCC1 and CYP1A1.
Collapse
Affiliation(s)
- Jiongwei Pan
- Department of Respiratory, the Sixth Affiliated Hospital of Wenzhou Medical University/Lishui People's Hospitlal, Lishui, Zhejiang, 323000, China
| | - Gang Huang
- Department of Chinese Medicine, the Sixth Affiliated Hospital of Wenzhou Medical University/Lishui People's Hospitlal, Lishui, Zhejiang, 323000, China
| | - Zhangyong Yin
- Department of Respiratory, the Sixth Affiliated Hospital of Wenzhou Medical University/Lishui People's Hospitlal, Lishui, Zhejiang, 323000, China
| | - Xiaoping Cai
- Department of Respiratory, the Sixth Affiliated Hospital of Wenzhou Medical University/Lishui People's Hospitlal, Lishui, Zhejiang, 323000, China
| | - Enhui Gong
- Department of Respiratory, the Sixth Affiliated Hospital of Wenzhou Medical University/Lishui People's Hospitlal, Lishui, Zhejiang, 323000, China
| | - Yuling Li
- Department of Respiratory, the Sixth Affiliated Hospital of Wenzhou Medical University/Lishui People's Hospitlal, Lishui, Zhejiang, 323000, China
| | - Cunlai Xu
- Department of Respiratory, the Sixth Affiliated Hospital of Wenzhou Medical University/Lishui People's Hospitlal, Lishui, Zhejiang, 323000, China
| | - Zaiting Ye
- Department of Radiology, the Sixth Affiliated Hospital of Wenzhou Medical University/Lishui People's Hospitlal, Lishui, Zhejiang, 323000, China
| | - Zhuo Cao
- The Sixth Affiliated Hospital of Wenzhou Medical University; Longquan Branch, Lishui People's Hospitlal, Lishui, China.
| | - Wei Cheng
- Department of Anesthesiology, the Affiliated Hospital of Xuzhou Medical University, Jiangsu Province Key Laboratory of Anesthesiology and Center for Pain Research and Treatment, Xuzhou, Jiangsu, 221002, China.
| |
Collapse
|
13
|
Ji Z, Tang T, Chen M, Dong B, Sun W, Wu N, Chen H, Feng Q, Yang X, Jin R, Jiang L. C-Myc-activated long non-coding RNA LINC01050 promotes gastric cancer growth and metastasis by sponging miR-7161-3p to regulate SPZ1 expression. J Exp Clin Cancer Res 2021; 40:351. [PMID: 34749766 PMCID: PMC8573944 DOI: 10.1186/s13046-021-02155-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 10/25/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Growing evidence shows that long non-coding RNAs (lncRNAs) play significant roles in cancer development. However, the functions of most lncRNAs in human gastric cancer are still not fully understood. Here, we explored the role of a novel c-Myc-activated lncRNA, LINC01050, in gastric cancer progression. METHODS The expression of LINC01050 in the context of gastric cancer was assessed using The Cancer Genome Atlas datasets. Its functions in gastric cancer were investigated through gain- and loss-of-function experiments combined with the Cell Counting Kit-8 assays, colony-forming assays, Transwell assays, flow cytometry, Western blot analyses, and xenograft tumor and mouse metastasis models. Potential LINC01050 transcription activators were screened via bioinformatics and validated by chromatin immunoprecipitation and luciferase assays. The interaction between LINC01050 and miR-7161-3p and the targets of miR-7161-3p were predicted by bioinformatics analysis and confirmed by a luciferase assay, RNA immunoprecipitation, RNA pull-down, and rescue experiments. RESULTS LINC01050 was significantly up-regulated in gastric cancer, and its high expression was positively correlated with a poor prognosis. The transcription factor c-Myc was found to directly bind to the LINC01050 promoter region and activate its transcription. Furthermore, overexpression of LINC01050 was confirmed to promote gastric cancer cell proliferation, migration, invasion, and epithelial-mesenchymal transition in vitro and tumor growth in vivo. At the same time, its knockdown inhibited gastric cancer cell proliferation, migration, invasion, and epithelial-mesenchymal transition in vitro along with tumor growth and metastasis in vivo. Moreover, mechanistic investigations revealed that LINC01050 functions as a molecular sponge to absorb cytosolic miR-7161-3p, which reduces the miR-7161-3p-mediated translational repression of SPZ1, thus contributing to gastric cancer progression. CONCLUSIONS Taken together, our results identified a novel gastric cancer-associated lncRNA, LINC01050, which is activated by c-Myc. LINC01050 may be considered a potential therapeutic target for gastric cancer.
Collapse
Affiliation(s)
- Ziwei Ji
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Tianbin Tang
- Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Mengxia Chen
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Buyuan Dong
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Wenjing Sun
- Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Nan Wu
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Hao Chen
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Qian Feng
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xingyi Yang
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Rong Jin
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| | - Lei Jiang
- Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
14
|
Deng Y, Li J, Zhou M, Liang Z, Zhao L. c-Myc affects hedgehog pathway via KCNQ1OT1/RAC1: A new mechanism for regulating HSC proliferation and epithelial-mesenchymal transition. Dig Liver Dis 2021; 53:1458-1467. [PMID: 33451909 DOI: 10.1016/j.dld.2020.11.035] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/24/2020] [Accepted: 11/27/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND This study aimed to probe into the potential mechanism of KCNQ1OT1 in liver fibrosis. METHODS The pathological changes in liver tissues were observed by Masson and hematoxylin-eosin (HE) staining. The proliferation or cell cycle of hepatic stellate cells (HSCs) was analyzed by MTT or flow cytometry. The expressions of epithelial markers E-cadherin, interstitial markers Snail and Vimentin, and hedgehog signaling pathway-related molecules Hhip, Shh, and Gli2 were detected by Western blot. The interaction or binding of c-Myc with the KCNQ1OT1 promoter was analyzed by dual-luciferase reporter gene or Chromatin immunoprecipitation (ChIP)-qPCR, and the interaction between KCNQ1OT1 and RAC1 was assessed by RNA immunoprecipitation and RNA pull-down. Moreover, the stability of RAC1 protein was detected by cycloheximide-chase and ubiquitination. RESULTS c-Myc and KCNQ1OT1 were up-regulated in liver fibrosis tissues and cells. After the interference with c-Myc in primary-1-Day HSCs, the down-regulated KCNQ1OT1 restrained HSC proliferation and EMT by down-regulating RAC1 expression and restraining the hedgehog pathway. CONCLUSION Our results indicated that the interference with c-Myc down-regulated RAC1 expression and restrained the hedgehog pathway by down-regulating KCNQ1OT1, thus restraining HSC proliferation and EMT in liver fibrosis.
Collapse
Affiliation(s)
- Yilei Deng
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China.
| | - Jian Li
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Menghao Zhou
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Zhiwei Liang
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Longshuan Zhao
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| |
Collapse
|
15
|
Liu H, Zhang Q, Song Y, Hao Y, Cui Y, Zhang X, Zhang X, Qin Y, Zhu G, Wang F, Dang J, Ma S, Zhang Y, Guo W, Li S, Guan F, Fan T. Long non-coding RNA SLC2A1-AS1 induced by GLI3 promotes aerobic glycolysis and progression in esophageal squamous cell carcinoma by sponging miR-378a-3p to enhance Glut1 expression. J Exp Clin Cancer Res 2021; 40:287. [PMID: 34517880 PMCID: PMC8436487 DOI: 10.1186/s13046-021-02081-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 08/20/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Emerging evidence demonstrates that lncRNAs play pivotal roles in tumor energy metabolism; however, the detailed mechanisms of lncRNAs in the regulation of tumor glycolysis remain largely unknown. METHODS The expression of SLC2A1-AS1 was investigated by TCGA, GEO dataset and qRT-PCR. The binding of GLI3 to SLC2A1-AS1 promoter was detected by Luciferase Reporter Assay System and Ago2-RIP assay. FISH was performed to determine the localization of SLC2A1-AS1 in ESCC cells. Double Luciferase Report assay was used to investigate the interaction of miR-378a-3p with SLC2A1-AS1 and Glut1. Gain-of-function and Loss-of-function assay were performed to dissect the function of SLC2A1-AS1/miR-378a-3p/Glut1 axis in ESCC progression in vitro and in vivo. RESULTS We identified a novel lncRNA SLC2A1-AS1 in ESCC. SLC2A1-AS1 was frequently overexpressed in ESCC tissues and cells, and its overexpression was associated with TNM stage, lymph node metastasis and poor prognosis of ESCC patients. Importantly, GLI3 and SLC2A1-AS1 formed a regulatory feedback loop in ESCC cells. SLC2A1-AS1 promoted cell growth in vitro and in vivo, migration and invasion, and suppressed apoptosis, leading to EMT progression and increased glycolysis in ESCC cells. SLC2A1-AS1 functioned as ceRNA for sponging miR-378a-3p, resulting in Glut1 overexpression in ESCC cells. MiR-378a-3p inhibited cell proliferation and invasion as well as induced apoptosis, resulting in reduced glycolysis, which was partly reversed by SLC2A1-AS1 or Glut1 overexpression in ESCC cells. CONCLUSION SLC2A1-AS1 plays important roles in ESCC development and progression by regulating glycolysis, and SLC2A1-AS1/miR-378a-3p/Glut1 regulatory axis may be a novel therapeutic target in terms of metabolic remodeling of ESCC patients.
Collapse
Affiliation(s)
- Hongtao Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China.
| | - Qing Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China.,Translational Medicine Research Center, Zhengzhou People's Hospital, Zhengzhou, 450003, Henan, China
| | - Yinsen Song
- Translational Medicine Research Center, Zhengzhou People's Hospital, Zhengzhou, 450003, Henan, China
| | - Yibin Hao
- Translational Medicine Research Center, Zhengzhou People's Hospital, Zhengzhou, 450003, Henan, China
| | - Yunxia Cui
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Xin Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Xueying Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Yue Qin
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Guangzhao Zhu
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Feng Wang
- Institute of Genomic Medicine, College of Pharmacy, Jinan University, Guangzhou, 510632, Guangdong, China.,International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of pharmacy, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Jinghan Dang
- Department of Clinical Medicine, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Shanshan Ma
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Yanting Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Wenna Guo
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Shenglei Li
- Department of Pathology, the First Affiliated Hospital of Zhengzhou University, 40 Daxue Road, Zhengzhou, 450052, Henan, China.
| | - Fangxia Guan
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China.
| | - Tianli Fan
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, 100 Kexue Road, Zhengzhou, 450001, Henan, China.
| |
Collapse
|
16
|
Lan H, Wang H, Gao M, Luo G, Zhang J, Yi E, Liang C, Xiong X, Chen X, Wu Q, Chen R, Lin B, Qian D, Hong W. Analysis and Construction of a Competitive Endogenous RNA Regulatory Network of Baicalin-Induced Apoptosis in Human Osteosarcoma Cells. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9984112. [PMID: 34337069 PMCID: PMC8315844 DOI: 10.1155/2021/9984112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/06/2021] [Accepted: 06/14/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND Baicalin is an extract from the traditional Chinese herb Scutellaria baicalensis and has the potential to treat osteosarcoma (OS). However, the transcriptome-level mechanism of baicalin-mediated antitumor effects in OS has not yet been investigated. The aim of this study was to analyze the competitive endogenous RNA (ceRNA) regulatory network involved in baicalin-induced apoptosis of OS cells. METHODS In this study, CCK-8 and flow cytometry assays were used to detect the antitumor effects of baicalin on human OS MG63 cells. Furthermore, transcriptome sequencing was employed to establish the long noncoding RNA (lncRNA), microRNA (miRNA), and mRNA profiles. RESULTS Baicalin inhibited MG63 cell proliferation and induced apoptosis. Totals of 58 lncRNAs, 31 miRNAs, and 2136 mRNAs in the baicalin-treated MG63 cells were identified as differentially expressed RNAs compared to those in control cells. Of these, 2 lncRNAs, 3 miRNAs, and 18 mRNAs were included in the ceRNA regulatory network. The differentially expressed RNAs were confirmed by quantitative real-time PCR (qRT-PCR). CONCLUSIONS By identifying the ceRNA network, our results provide new information about the possible molecular basis of baicalin, which has potential applications in OS treatment.
Collapse
Affiliation(s)
- Haifeng Lan
- Department of Orthopaedic Surgery, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Haiyan Wang
- Guangzhou Key Laboratory of Basic and Applied Research in Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Mi Gao
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Guan Luo
- Guangzhou Key Laboratory of Basic and Applied Research in Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jiahuan Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Erkang Yi
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Chunxiao Liang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaoxiao Xiong
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xing Chen
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Qinghua Wu
- The Third Clinical School of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ruikun Chen
- The Third Clinical School of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Biting Lin
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Dongyang Qian
- Department of Orthopaedics, The First Affiliated Hospital, Guangzhou Medical University/Guangdong Key Laboratory of Orthopaedic Technology and Implant Materials, Guangzhou, Guangdong, China
| | - Wei Hong
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
17
|
Huang W, Song W, Jiang Y, Chen L, Lu H. c-Myc-induced circ-NOTCH1 promotes aggressive phenotypes of nasopharyngeal carcinoma cells by regulating the miR-34c-5p/c-Myc axis. Cell Biol Int 2021; 45:1436-1447. [PMID: 33675278 DOI: 10.1002/cbin.11582] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 02/06/2021] [Accepted: 02/27/2021] [Indexed: 01/17/2023]
Abstract
Nasopharyngeal carcinoma (NPC) is the subclass of head and neck cancer with the highest incidence among otolaryngology malignancies. A growing amount of evidence has proven that circular RNAs (circRNAs) play key roles in the progression of multiple cancers. It has been reported that circ-NOTCH1 is a novel circRNA and functions as an oncogene in gastric cancer, while the regulatory mechanism of circ-NOTCH1 in NPC remains unknown. In the present research, our findings revealed that circ-NOTCH1 was overexpressed in NPC tissues and cells. Circ-NOTCH1 knockdown suppressed NPC cell proliferation, invasion, and migration. Subsequently, we discovered that c-Myc can activate circ-NOTCH1 by binding to the NOTCH1 promoter. c-Myc functioned as a tumor promoter in NPC cells. Mechanistically, circ-NOTCH1 served as a competitive endogenous RNA to modulate c-Myc expression by sponging miR-34c-5p. Additionally, overexpression of c-Myc reversed the circ-NOTCH1 knockdown-mediated inhibition of NPC cellular progression. Overall, this study suggested that c-Myc-induced circ-NOTCH1 promoted malignant phenotypes of NPC cells by regulating the miR-34c-5p/c-Myc axis.
Collapse
Affiliation(s)
- Wei Huang
- Department of Radiation Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.,School of Clinical, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Wei Song
- Department of Radiation Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yunfei Jiang
- Department of Emergency, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Lejun Chen
- School of Clinical, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Hong Lu
- Department of Radiation Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
18
|
Zheng Y, Duan L, Yang Y, Luo D, Yan B. Circ_0023028 contributes to the progression of laryngeal squamous cell carcinoma by upregulating LASP1 through miR-486-3p. Mol Cell Biochem 2021; 476:2951-2961. [PMID: 33755879 DOI: 10.1007/s11010-021-04129-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 03/04/2021] [Indexed: 10/21/2022]
Abstract
Circular RNAs (circRNAs) are implicated in the tumorigenesis of human cancers. However, the effects of circRNAs on laryngeal squamous cell carcinoma (LSCC) are largely unknown. Here, we aimed to explore the roles of circ_0023028 in LSCC development. Quantitative real-time polymerase chain reaction (qRT-PCR) was performed to measure circ_0023028, miR-486-3p, and Lin-Isl-Mec (LIM) and SH3 domain protein 1 (LASP1) mRNA. The characteristics of circ_0023028 were determined by RNase R digestion assay and Actinomycin D assay. Cell Counting Kit-8 (CCK-8) assay and colony formation assay were utilized for cell proliferation. Transwell assay was adopted for cell invasion and migration. Flow cytometry analysis was carried out to analyze cell cycle and apoptosis. RNA pull-down assay and dual-luciferase reporter assay were used to explore the association between miR-486-3p and circ_0023028 or LASP1. Western blot assay was adopted to measure LASP1 protein level. Murine xenograft model was executed to investigate the function of circ_0023028 in vivo. High expression of circ_0023028 was observed in LSCC tissues and cells. Circ_0023028 was stable and possessed a loop structure. Circ_0023028 silencing suppressed LSCC cell proliferation, metastasis and cell cycle process and induced apoptosis in vitro and hampered tumor growth in vivo. Further mechanism analysis demonstrated that circ_0023028 could sponge miR-486-3p to regulate LASP1 expression in LSCC cells. The malignant behaviors of LSCC cells mediated by circ_0023028 knockdown were rescued by the inhibition of miR-486-3p. Moreover, miR-486-3p suppressed LSCC cell progression via binding to LASP1. Circ_0023028 knockdown might impede the progression of LSCC by regulating miR-486-3p/LASP1 axis, which could provide a novel insight on the mechanism of LSCC progression.
Collapse
Affiliation(s)
- Yuebin Zheng
- Department of Otorhinolaryngology, Head and Neck Surgery, Zigong First People's Hospital, No.42, Shangyihao Yi Zhi Road, Ziliujing, Zigong, 643000, Sichuan, China
| | - Lifu Duan
- Department of Otorhinolaryngology, Head and Neck Surgery, Zigong First People's Hospital, No.42, Shangyihao Yi Zhi Road, Ziliujing, Zigong, 643000, Sichuan, China
| | - Yirong Yang
- Department of Otorhinolaryngology, Head and Neck Surgery, Zigong First People's Hospital, No.42, Shangyihao Yi Zhi Road, Ziliujing, Zigong, 643000, Sichuan, China
| | - Dengyao Luo
- Department of Otorhinolaryngology, Head and Neck Surgery, Zigong First People's Hospital, No.42, Shangyihao Yi Zhi Road, Ziliujing, Zigong, 643000, Sichuan, China
| | - Bincheng Yan
- Department of Otorhinolaryngology, Head and Neck Surgery, Zigong First People's Hospital, No.42, Shangyihao Yi Zhi Road, Ziliujing, Zigong, 643000, Sichuan, China.
| |
Collapse
|
19
|
Zhong Q, Lu M, Yuan W, Cui Y, Ouyang H, Fan Y, Wang Z, Wu C, Qiao J, Hang J. Eight-lncRNA signature of cervical cancer were identified by integrating DNA methylation, copy number variation and transcriptome data. J Transl Med 2021; 19:58. [PMID: 33557879 PMCID: PMC8045209 DOI: 10.1186/s12967-021-02705-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 01/12/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Copy number variation (CNV) suggests genetic changes in malignant tumors. Abnormal expressions of long non-coding RNAs (lncRNAs) resulted from genomic and epigenetic abnormalities play a driving role in tumorigenesis of cervical cancer. However, the role of lncRNAs-related CNV in cervical cancer remained largely unclear. METHODS The data of messenger RNAs (mRNAs), DNA methylation, and DNA copy number were collected from 292 cervical cancer specimens. The prognosis-related subtypes of cervical cancer were determined by multi-omics integration analysis, and protein-coding genes (PCGs) and lncRNAs with subtype-specific expressions were identified. The CNV pattern of the subtype-specific lncRNAs was analyzed to identify the subtype-specific lncRNAs. A prognostic risk model based on lncRNAs was established by least absolute shrinkage and selection operator (LASSO). RESULTS Multi-omics integration analysis identified three molecular subtypes incorporating 617 differentially expressed lncRNAs and 1395 differentially expressed PCGs. The 617 lncRNAs were found to intersect with disease-related lncRNAs. Functional enrichment showed that 617 lncRNAs were mainly involved in tumor metabolism, immunity and other pathways, such as p53 and cAMP signaling pathways, which are closely related to the development of cervical cancer. Finally, according to CNV pattern consistent with differential expression analysis, we established a lncRNAs-based signature consisted of 8 lncRNAs, namely, RUSC1-AS1, LINC01990, LINC01411, LINC02099, H19, LINC00452, ADPGK-AS1, C1QTNF1-AS1. The interaction of the 8 lncRNAs showed a significantly poor prognosis of cervical cancer patients, which has also been verified in an independent dataset. CONCLUSION Our study expanded the network of CNVs and improved the understanding on the regulatory network of lncRNAs in cervical cancer, providing novel biomarkers for the prognosis management of cervical cancer patients.
Collapse
Affiliation(s)
- Qihang Zhong
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Peking University, HaiDian District, No. 38 XueYuan Road, Beijing, 100191, China.,Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, HaiDian District, No. 49 North HuaYuan Road, Beijing, 100191, China
| | - Minzhen Lu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, HaiDian District, No. 49 North HuaYuan Road, Beijing, 100191, China.,National Clinical Research Center for Obstetrics and Gynecology, Beijing, 100191, China.,Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Wanqiong Yuan
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100091, China.,Beijing Key Laboratory of Spinal Disease Research, Beijing, 100191, China
| | - Yueyi Cui
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, HaiDian District, No. 49 North HuaYuan Road, Beijing, 100191, China
| | - Hanqiang Ouyang
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100091, China
| | - Yong Fan
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Zhaohui Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Congying Wu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Peking University, HaiDian District, No. 38 XueYuan Road, Beijing, 100191, China.
| | - Jie Qiao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, HaiDian District, No. 49 North HuaYuan Road, Beijing, 100191, China. .,National Clinical Research Center for Obstetrics and Gynecology, Beijing, 100191, China. .,Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China. .,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China.
| | - Jing Hang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, HaiDian District, No. 49 North HuaYuan Road, Beijing, 100191, China. .,National Clinical Research Center for Obstetrics and Gynecology, Beijing, 100191, China. .,Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China. .,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China.
| |
Collapse
|
20
|
Amjadi-Moheb F, Paniri A, Akhavan-Niaki H. Insights into the Links between MYC and 3D Chromatin Structure and Epigenetics Regulation: Implications for Cancer Therapy. Cancer Res 2021; 81:1925-1936. [PMID: 33472888 DOI: 10.1158/0008-5472.can-20-3613] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/21/2020] [Accepted: 01/06/2021] [Indexed: 11/16/2022]
Abstract
MYC is embedded in the transcriptional oasis of the 8q24 gene desert. A plethora of genomic elements has roles in MYC aberrant expression in cancer development by interacting with transcription factors and epigenetics regulators as well as altering the structure of chromatin at the MYC locus and tissue-specific long-range enhancer-promoter contacts. Furthermore, MYC is a master regulator of several human cancers by modulating the transcription of numerous cancer-related genes through epigenetic mechanisms. This review provides a comprehensive overview of the three-dimensional genomic organization around MYC and the role of epigenetic machinery in transcription and function of MYC as well as discusses various epigenetic-targeted therapeutic strategies in MYC-driven cancers.
Collapse
Affiliation(s)
- Fatemeh Amjadi-Moheb
- Department of Genetics, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Alireza Paniri
- Department of Genetics, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Haleh Akhavan-Niaki
- Department of Genetics, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
21
|
Ashrafizadeh M, Zarabi A, Hushmandi K, Moghadam ER, Hashemi F, Daneshi S, Hashemi F, Tavakol S, Mohammadinejad R, Najafi M, Dudha N, Garg M. C-Myc Signaling Pathway in Treatment and Prevention of Brain Tumors. Curr Cancer Drug Targets 2021; 21:2-20. [PMID: 33069197 DOI: 10.2174/1568009620666201016121005] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/26/2020] [Accepted: 09/07/2020] [Indexed: 12/16/2022]
Abstract
Brain tumors are responsible for high morbidity and mortality worldwide. Several factors such as the presence of blood-brain barrier (BBB), sensitive location in the brain, and unique biological features challenge the treatment of brain tumors. The conventional drugs are no longer effective in the treatment of brain tumors, and scientists are trying to find novel therapeutics for brain tumors. In this way, identification of molecular pathways can facilitate finding an effective treatment. c-Myc is an oncogene signaling pathway capable of regulation of biological processes such as apoptotic cell death, proliferation, survival, differentiation, and so on. These pleiotropic effects of c-Myc have resulted in much fascination with its role in different cancers, particularly brain tumors. In the present review, we aim to demonstrate the upstream and down-stream mediators of c-Myc in brain tumors such as glioma, glioblastoma, astrocytoma, and medulloblastoma. The capacity of c-Myc as a prognostic factor in brain tumors will be investigated. Our goal is to define an axis in which the c-Myc signaling pathway plays a crucial role and to provide direction for therapeutic targeting in these signaling networks in brain tumors.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Universite Caddesi No. 27, Orhanli, Tuzla, 34956 Istanbul, Turkey
| | - Ali Zarabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956, Istanbul, Turkey
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Ebrahim Rahmani Moghadam
- Department of Anatomical sciences, School of Medicine, Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farid Hashemi
- DVM. Graduated, Young Researcher and Elite Club, Kazerun Branch, Islamic Azad University, Kazeroon, Iran
| | - Salman Daneshi
- Department of Public Health, School of Health, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Fardin Hashemi
- Student Research Committee, Department of physiotherapy, Faculty of rehabilitation, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Reza Mohammadinejad
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 7619813159, Iran
| | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Namrata Dudha
- Department of Biotechnology and Microbiology, School of Sciences, Noida International University, Gautam Budh Nagar, Uttar Pradesh, India
| | - Manoj Garg
- Amity of Molecular Medicine and Stem cell Research (AIMMSCR), Amity University Uttar Pradesh, Noida-201313, India
| |
Collapse
|
22
|
Li M, Chi C, Zhou L, Chen Y, Tang X. Circular PVT1 regulates cell proliferation and invasion via miR-149-5p/FOXM1 axis in ovarian cancer. J Cancer 2021; 12:611-621. [PMID: 33391456 PMCID: PMC7738991 DOI: 10.7150/jca.52234] [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: 08/21/2020] [Accepted: 09/28/2020] [Indexed: 12/14/2022] Open
Abstract
Long non-coding RNA plasmacytoma variant translocation 1 (PVT1) is a dysregulated gene in malignancy and is associated with oncogenesis. In this study, we found PVT1 RNA was an ovarian specific expressing gene, and overexpressed in multiple cancer types, including ovarian cancer (OV). Higher expression levels of PVT1 are related to shorter survival time in OV patients, especially in patients with advanced stage and grade. Recent studies indicated circular PVT1 also had an important role in cancer progression, whose roles in OV remain unclear. Knockdown of circular PVT1 significantly suppressed OV cell proliferation, migration and invasion. Bioinformatics analysis demonstrated that circular PVT1 was involved in regulating angiogenesis, osteoblast differentiation, regulation of cell growth, type B pancreatic cell proliferation, negative regulation of apoptotic process, phospholipid homeostasis, regulation of neurogenesis, definitive hemopoiesis, cell migration, regulation of glucose metabolic process, central nervous system development and type 2 immune response. Our data showed miR-149-5p targeted FOXM1, which was regulated by circular PVT1. Forkhead Box M1 (FOXM1) expression in ovarian cancer exhibited high level when compared with normal tissues, and had relation with relatively poor survival. FOXM1 promoted cell viability and reduced FOXM1 could rescue circular influence of circular PVT1-caused carcinoma induction. In conclusion, circular PVT1 increased FOXM1 level via binding to miR-149-5p and thus affected ovarian cancer cell viability and migration.
Collapse
Affiliation(s)
- Min Li
- Department of Gynecology & Obstetrics, the First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Chi Chi
- Department of Gynecology & Obstetrics, the First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Liqin Zhou
- Department of Gynecology & Obstetrics, Suzhou Xiangcheng People's Hospital, Suzhou 215006, Jiangsu Province, China
| | - Youguo Chen
- Department of Gynecology & Obstetrics, the First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Xiuwu Tang
- Department of Gynecology & Obstetrics, the First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| |
Collapse
|