1
|
Maharati A, Moghbeli M. Role of microRNA-505 during tumor progression and metastasis. Pathol Res Pract 2024; 258:155344. [PMID: 38744001 DOI: 10.1016/j.prp.2024.155344] [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: 12/23/2023] [Revised: 04/23/2024] [Accepted: 05/09/2024] [Indexed: 05/16/2024]
Abstract
Late diagnosis of cancer in advanced stages due to the lack of screening methods is considered as the main cause of poor prognosis and high mortality rate among these patients. Therefore, it is necessary to investigate the molecular tumor biology in order to introduce biomarkers that can be used in cancer screening programs and early diagnosis. MicroRNAs (miRNAs) have key roles in regulation of the cellular pathophysiological processes. Due to the high stability of miRNAs in body fluids, they are widely used as the non-invasive tumor markers. According to the numerous reports about miR-505 deregulation in a wide range of cancers, we investigated the role of miR-505 during tumor progression. It was shown that miR-505 mainly has the tumor suppressor functions through the regulation of signaling pathways, chromatin remodeling, and cellular metabolism. This review has an effective role in introducing miR-505 as a suitable marker for the early cancer diagnosis.
Collapse
Affiliation(s)
- Amirhosein Maharati
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Sun Y, Guo G, Zhang Y, Chen X, Lu Y, Hong R, Xiong J, Li J, Hu X, Wang S, Liu Y, Zhang Z, Yang X, Nan Y, Huang Q. IKBKE promotes the ZEB2-mediated EMT process by phosphorylating HMGA1a in glioblastoma. Cell Signal 2024; 116:111062. [PMID: 38242271 DOI: 10.1016/j.cellsig.2024.111062] [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: 08/08/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 01/21/2024]
Abstract
IKBKE (Inhibitor of Nuclear Factor Kappa-B Kinase Subunit Epsilon) is an important oncogenic protein in a variety of tumors, which can promote tumor growth, proliferation, invasion and drug resistance, and plays a critical regulatory role in the occurrence and progression of malignant tumors. HMGA1a (High Mobility Group AT-hook 1a) functions as a cofactor for proper transcriptional regulation and is highly expressed in multiple types of tumors. ZEB2 (Zinc finger E-box Binding homeobox 2) exerts active functions in epithelial mesenchymal transformation (EMT). In our current study, we confirmed that IKBKE can increase the proliferation, invasion and migration of glioblastoma cells. We then found that IKBKE can phosphorylate HMGA1a at Ser 36 and/or Ser 44 sites and inhibit the degradation process of HMGA1a, and regulate the nuclear translocation of HMGA1a. Crucially, we observed that HMGA1a can regulate ZEB2 gene expression by interacting with ZEB2 promoter region. Hence, HMGA1a was found to promote the ZEB2-related metastasis. Consequently, we demonstrated that IKBKE can exert its oncogenic functions via the IKBKE/HMGA1a/ZEB2 signalling axis, and IKBKE may be a prominent biomarker for the treatment of glioblastoma in the future.
Collapse
Affiliation(s)
- Yan Sun
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Department of Neurosurgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300052, China
| | - Gaochao Guo
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Department of Neurosurgery, Henan Provincial People's Hospital, Cerebrovascular Disease Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450003, China
| | - Yu Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300052, China
| | - Xingjie Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300052, China
| | - Yalin Lu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300052, China
| | - Rujun Hong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300052, China
| | - Jinbiao Xiong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300052, China
| | - Jiabo Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300052, China
| | - Xue Hu
- Department of Clinical Nutrition, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, China
| | - Shuaishuai Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300052, China
| | - Yang Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Department of Neurosurgery, Henan Provincial People's Hospital, Cerebrovascular Disease Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450003, China
| | - Zhimeng Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Department of Neurosurgery, Ningbo Hospital of Zhejiang University, Ningbo, Zhejiang 315000, China
| | - Xuejun Yang
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Yang Nan
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300052, China
| | - Qiang Huang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300052, China.
| |
Collapse
|
3
|
Samare-Najaf M, Samareh A, Savardashtaki A, Khajehyar N, Tajbakhsh A, Vakili S, Moghadam D, Rastegar S, Mohsenizadeh M, Jahromi BN, Vafadar A, Zarei R. Non-apoptotic cell death programs in cervical cancer with an emphasis on ferroptosis. Crit Rev Oncol Hematol 2024; 194:104249. [PMID: 38145831 DOI: 10.1016/j.critrevonc.2023.104249] [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: 08/30/2023] [Revised: 12/10/2023] [Accepted: 12/20/2023] [Indexed: 12/27/2023] Open
Abstract
BACKGROUND Cervical cancer, a pernicious gynecological malignancy, causes the mortality of hundreds of thousands of females worldwide. Despite a considerable decline in mortality, the surging incidence rate among younger women has raised serious concerns. Immortality is the most important characteristic of tumor cells, hence the carcinogenesis of cervical cancer cells pivotally requires compromising with cell death mechanisms. METHODS The current study comprehensively reviewed the mechanisms of non-apoptotic cell death programs to provide possible disease management strategies. RESULTS Comprehensive evidence has stated that focusing on necroptosis, pyroptosis, and autophagy for disease management is associated with significant limitations such as insufficient understanding, contradictory functions, dependence on disease stage, and complexity of intracellular pathways. However, ferroptosis represents a predictable role in cervix carcinogenesis, and ferroptosis-related genes demonstrate a remarkable correlation with patient survival and clinical outcomes. CONCLUSION Ferroptosis may be an appropriate option for disease management strategies from predicting prognosis to treatment.
Collapse
Affiliation(s)
- Mohammad Samare-Najaf
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Kerman Regional Blood Transfusion Center, Kerman, Iran.
| | - Ali Samareh
- Department of Biochemistry, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran; Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Nastaran Khajehyar
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Kerman Regional Blood Transfusion Center, Kerman, Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sina Vakili
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Delaram Moghadam
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medicinal Chemistry, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sanaz Rastegar
- Department of Microbiology and Virology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Majid Mohsenizadeh
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Kerman Regional Blood Transfusion Center, Kerman, Iran
| | | | - Asma Vafadar
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Zarei
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
4
|
Janin M, Davalos V, Esteller M. Cancer metastasis under the magnifying glass of epigenetics and epitranscriptomics. Cancer Metastasis Rev 2023; 42:1071-1112. [PMID: 37369946 PMCID: PMC10713773 DOI: 10.1007/s10555-023-10120-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023]
Abstract
Most of the cancer-associated mortality and morbidity can be attributed to metastasis. The role of epigenetic and epitranscriptomic alterations in cancer origin and progression has been extensively demonstrated during the last years. Both regulations share similar mechanisms driven by DNA or RNA modifiers, namely writers, readers, and erasers; enzymes responsible of respectively introducing, recognizing, or removing the epigenetic or epitranscriptomic modifications. Epigenetic regulation is achieved by DNA methylation, histone modifications, non-coding RNAs, chromatin accessibility, and enhancer reprogramming. In parallel, regulation at RNA level, named epitranscriptomic, is driven by a wide diversity of chemical modifications in mostly all RNA molecules. These two-layer regulatory mechanisms are finely controlled in normal tissue, and dysregulations are associated with every hallmark of human cancer. In this review, we provide an overview of the current state of knowledge regarding epigenetic and epitranscriptomic alterations governing tumor metastasis, and compare pathways regulated at DNA or RNA levels to shed light on a possible epi-crosstalk in cancer metastasis. A deeper understanding on these mechanisms could have important clinical implications for the prevention of advanced malignancies and the management of the disseminated diseases. Additionally, as these epi-alterations can potentially be reversed by small molecules or inhibitors against epi-modifiers, novel therapeutic alternatives could be envisioned.
Collapse
Affiliation(s)
- Maxime Janin
- Cancer Epigenetics Group, Josep Carreras Leukaemia Research Institute (IJC), IJC Building, Germans Trias I Pujol, Ctra de Can Ruti, Cami de Les Escoles S/N, 08916 Badalona, Barcelona, Spain
- Centro de Investigacion Biomedica en Red Cancer (CIBERONC), Madrid, Spain
| | - Veronica Davalos
- Cancer Epigenetics Group, Josep Carreras Leukaemia Research Institute (IJC), IJC Building, Germans Trias I Pujol, Ctra de Can Ruti, Cami de Les Escoles S/N, 08916 Badalona, Barcelona, Spain
| | - Manel Esteller
- Cancer Epigenetics Group, Josep Carreras Leukaemia Research Institute (IJC), IJC Building, Germans Trias I Pujol, Ctra de Can Ruti, Cami de Les Escoles S/N, 08916 Badalona, Barcelona, Spain.
- Centro de Investigacion Biomedica en Red Cancer (CIBERONC), Madrid, Spain.
- Institucio Catalana de Recerca I Estudis Avançats (ICREA), Barcelona, Catalonia, Spain.
- Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Catalonia, Spain.
| |
Collapse
|
5
|
Aswathy R, Sumathi S. Defining new biomarkers for overcoming therapeutical resistance in cervical cancer using lncRNA. Mol Biol Rep 2023; 50:10445-10460. [PMID: 37878205 DOI: 10.1007/s11033-023-08864-w] [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: 06/04/2023] [Accepted: 09/27/2023] [Indexed: 10/26/2023]
Abstract
Despite improvements in cervical cancer diagnosis and treatment, the prognosis for cervical cancer patients remains dismal due to the development of drug resistance, metastasis, and invasion resulting leading to treatment failure. Long non-coding RNAs (lncRNAs), a class of RNA transcripts have been reported in mediating carcinogenesis as well as drug, and radio-resistance in tumor cells. These lncRNAs regulate various cancer hallmarks and contribute to the development of therapeutic resistance. They regulates multiple signalling pathways, recruits polycomb group, function as miRNA sponge and scaffolds. Additionally, lncRNAs can act as oncogenes or tumor suppressors in cervical cancer. This comprehensive review outlines the biogenesis of lncRNA and its role in cancer development. It delves into the mechanisms through which various lncRNAs mediate chemoresistance and radioresistance in cervical cancer. By shedding into the light of mechanism, this review will also aids researchers in understanding lncRNAs as biomarkers and latest advancements in clinically targeting them with the help of Artificial Intelligence for overcoming chemoresistance and radioresistance, thereby improving cervical cancer treatment.
Collapse
Affiliation(s)
- Raghu Aswathy
- Department of Biochemistry, Biotechnology and Bioinformatics, Avinashilingam Institute for Home Science and Higher Education for Women, Bharathi Park Rd, near Forest College Campus, Saibaba Colony, Coimbatore, Tamil Nadu, 641043, India
| | - Sundaravadivelu Sumathi
- Department of Biochemistry, Biotechnology and Bioinformatics, Avinashilingam University for Home Science and Higher Education for Women, Bharathi Park Rd, near Forest College Campus, Saibaba Colony, Coimbatore, Tamil Nadu, 641043, India.
| |
Collapse
|
6
|
Rezaee A, Ahmadpour S, Jafari A, Aghili S, Zadeh SST, Rajabi A, Raisi A, Hamblin MR, Mahjoubin-Tehran M, Derakhshan M. MicroRNAs, long non-coding RNAs, and circular RNAs and gynecological cancers: focus on metastasis. Front Oncol 2023; 13:1215194. [PMID: 37854681 PMCID: PMC10580988 DOI: 10.3389/fonc.2023.1215194] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/28/2023] [Indexed: 10/20/2023] Open
Abstract
Gynecologic cancer is a significant cause of death in women worldwide, with cervical cancer, ovarian cancer, and endometrial cancer being among the most well-known types. The initiation and progression of gynecologic cancers involve a variety of biological functions, including angiogenesis and metastasis-given that death mostly occurs from metastatic tumors that have invaded the surrounding tissues. Therefore, understanding the molecular pathways underlying gynecologic cancer metastasis is critical for enhancing patient survival and outcomes. Recent research has revealed the contribution of numerous non-coding RNAs (ncRNAs) to metastasis and invasion of gynecologic cancer by affecting specific cellular pathways. This review focuses on three types of gynecologic cancer (ovarian, endometrial, and cervical) and three kinds of ncRNAs (long non-coding RNAs, microRNAs, and circular RNAs). We summarize the detailed role of non-coding RNAs in the different pathways and molecular interactions involved in the invasion and metastasis of these cancers.
Collapse
Affiliation(s)
- Aryan Rezaee
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sara Ahmadpour
- Biotechnology Department, Faculty of Chemistry, University of Kashan, Kashan, Iran
| | - Ameneh Jafari
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sarehnaz Aghili
- Department of Gynecology and Obstetrics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Ali Rajabi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Arash Raisi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, South Africa
| | - Maryam Mahjoubin-Tehran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Marzieh Derakhshan
- Shahid Beheshti Fertility Clinic, Department of Gynecology and Obsteterics, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
7
|
Mao Y, Jiang X, Guo P, Ouyang Y, Chen X, Xia M, Wu L, Tang Z, Liang T, Li Y, He M. ZXDC enhances cervical cancer metastasis through IGF2BP3-mediated activation of RhoA/ ROCK signaling. iScience 2023; 26:107447. [PMID: 37599824 PMCID: PMC10433122 DOI: 10.1016/j.isci.2023.107447] [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: 01/19/2023] [Revised: 04/17/2023] [Accepted: 07/18/2023] [Indexed: 08/22/2023] Open
Abstract
Metastasis in cervical cancer (CC) has a significant negative impact on patient survival, highlighting the urgent need for investigation in this area. In this study, we identified significant overexpression of zinc finger, X-linked, duplicated family member C (ZXDC) in CC tissue with metastasis, which correlates with poor outcomes for CC patients. We observed that overexpression of ZXDC promotes, while silencing of ZXDC inhibits the metastasis of CC cells both in vitro and in vivo. Additionally, our research demonstrated that ZXDC activated RhoA/ROCK signaling pathway, leading to enhanced cytoskeleton remodeling in CC cells. Besides, we found that IGF2BP3 plays an essential role in the activation of ZXDC on the RhoA/ROCK signaling pathway by stabilizing RhoA mRNA. These findings reveal a mechanism whereby ZXDC promotes the cervical cancer metastasis by targeting IGF2BP3/RhoA/ROCK pathway.
Collapse
Affiliation(s)
- Yifang Mao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Xingyu Jiang
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Peng Guo
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Ying Ouyang
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xiangfu Chen
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Meng Xia
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Lixin Wu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Zihao Tang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Tianyi Liang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Yue Li
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Mian He
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
8
|
Miao R, Yao Z, Hu B, Jin T, Zhu D, Shi Y, Gong Y, Shao S, Shao C. A novel long non-coding RNA XLOC_004787, is associated with migration and promotes cancer cell proliferation by downregulating mir-203a-3p in gastric cancer. BMC Gastroenterol 2023; 23:280. [PMID: 37573302 PMCID: PMC10422700 DOI: 10.1186/s12876-023-02912-2] [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: 09/14/2022] [Accepted: 08/02/2023] [Indexed: 08/14/2023] Open
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) have been identified as important regulatory factors implicated in a wide array of diseases, including various forms of cancer. However, the roles of most lncRNAs in the progression of gastric cancer (GC) remain largely unexplored. This study investigates the biological function and underlying mechanism of a novel lncRNA, XLOC_004787 in GC. METHODS The location of XLOC_004787 in GES-1 cells and HGC-27 cells were detected by fluorescence in situ hybridization (FISH) assay. The expression levels of XLOC_004787 were assessed using quantitative real-time fluorescence PCR (qRT-PCR) in various cell lines, including GES-1, MGC-803, MKN-45, BGC-823, SGC-7901, and HGC-27 cells. Functional assays such as Transwell migration, cell counting kit-8 (CCK-8), and colony formation experiments were employed to analyze the effects of XLOC_004787 and miR-203a-3p on cell migration and proliferation. Protein levels associated with GC in these cell lines were examined by Western blotting. The intracellular localization of β-catenin and P-Smad2/3 was assessed using immunofluorescence (IF) assay. Additionally, the interaction between XLOC_004787 and miR-203a-3p was investigated using a dual luciferase assay. RESULTS XLOC_004787 was localized at both the cytoplasm and nucleus of GES-1 cells and HGC-27 cells. Compared to normal tissues and GES-1 cells, XLOC_004787 expression was significantly upregulated in GC tissues and cells, with the highest and lowest expression observed in SGC-7901 and HGC-27 cells, respectively. Furthermore, a reduced expression of XLOC_004787 was seen to inhibit migration and proliferation in SGC-7901 cells. Western blotting analysis revealed that a decrease in XLOC_004787 expression correspondingly decreased the expression of N-cadherin, mmp2, mmp9, Snail, Vimentin, β-catenin, C-myc, Cyclin D1, and TGF-β, while concurrently increasing E-cadherin expression. This was also associated with diminished expression of P-Smad2/3 in relation to Smad2/3, and reduced P-Gsk3β expression in comparison to Gsk3β. Additionally, the nuclear entry of P-Smad2/3 and β-catenin was reduced by lower XLOC_004787 expression. Amplifying XLOC_004787 expression via pcDNA_XLOC_004787 suggested a potential for cancer promotion. Notably, XLOC_004787 was found to negatively regulate mir-203a-3p expression, with potential binding sites identified between the two. Higher mir-203a-3p expression was observed to decrease migration and proliferation, and enhance E-cadherin expression. Conversely, suppression of mir-203a-3p expression suggested a potential promotion of proliferation and migration in GC cells. CONCLUSIONS These results suggest that XLOC_004787, found to be upregulated in GC tissues, potentially promotes proliferation and migration in GC cells. This occurs through the activation of TGF-β and Wnt/β-catenin signaling pathways and the expression of EMT-related proteins. Additionally, XLOC_004787 may influence cell migration and proliferation by modulating the signaling pathway via the adsorption and inhibition of mir-203a-3p.
Collapse
Affiliation(s)
- Renjie Miao
- Department of Clinical Laboratory, Affiliated Third Hospital of Zhenjiang to Jiangsu University, Zhenjiang, 212001, Jiangsu, China
| | - Zhendong Yao
- Department of Gastroenterology, The Affiliated Yixing Hospital of Jiangsu University, Yixing, 214200, Jiangsu, China
| | - Bingheng Hu
- The Affiliated Hospital of Jiangsu University, Yizheng Road, Zhenjiang, 212013, Jiangsu, China
| | - Tao Jin
- Department of Gastroenterology, The Affiliated Yixing Hospital of Jiangsu University, Yixing, 214200, Jiangsu, China
| | - Donglai Zhu
- Department of Clinical Laboratory, Affiliated Third Hospital of Zhenjiang to Jiangsu University, Zhenjiang, 212001, Jiangsu, China
| | - Yun Shi
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu, China
| | - Yuhua Gong
- Department of Clinical Laboratory, Affiliated Third Hospital of Zhenjiang to Jiangsu University, Zhenjiang, 212001, Jiangsu, China
| | - Shihe Shao
- Department of Clinical Laboratory, Affiliated Third Hospital of Zhenjiang to Jiangsu University, Zhenjiang, 212001, Jiangsu, China
| | - Chen Shao
- The Affiliated Hospital of Jiangsu University, Yizheng Road, Zhenjiang, 212013, Jiangsu, China.
| |
Collapse
|
9
|
Meng M, Yang L, Zhou H, Cheng Q, Peng R, Wang Z, Liang X, Wen J, Nie J, Hu Z, Zhang L, Liu Z. LINC00978 regulates metabolic rewiring to promote the malignancy of glioblastoma through AKR1B1. Cancer Lett 2023:216277. [PMID: 37336288 DOI: 10.1016/j.canlet.2023.216277] [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: 02/11/2023] [Revised: 06/08/2023] [Accepted: 06/09/2023] [Indexed: 06/21/2023]
Abstract
Glioma is a fatal primary brain tumor. Improved glioma treatment effectiveness depends on a better understanding of its underlying mechanisms. Herein, we reported LINC00978 overexpressed in gliomas. Downregulation of LINC00978 in glioblastoma cells inhibited cell proliferation, invasion, migration, and induced apoptosis. In vivo experiments confirmed that the CamK-A siRNA of LINC00978 could effectively inhibit the proliferation of glioma cells. The main pathway and genes regulated by LINC00978 were detected using RNA sequencing to elucidate the molecular mechanism. The results suggest that LINC00978 regulates the expression of genes related to metabolic pathways, including aldo-keto reductase family 1 member B (AKR1B1), which mediates the cytotoxicity of 2-deoxyglucose. LINC00978 positively regulated AKR1B1 expression, and 2-deoxyglucose induced AKR1B1 expression via a LINC00978-dependent mechanism. This research has revealed that LINC00978 promotes the sensitivity of glioma cells to 2DG. LINC00978 is highly expressed in most glioma patients. Thus, understanding the anticancer mechanism identified in this study may contribute to treating the majority of glioma patients. This study clarified the function and molecular mechanism of LINC00978 in glioblastoma and provided a study basis for LINC00978 to guide the clinical treatment of glioblastoma.
Collapse
Affiliation(s)
- Ming Meng
- Department of Neurosurgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China; Hypothalamic Pituitary Research Center, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China.
| | - Liting Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China; Hypothalamic Pituitary Research Center, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China.
| | - Hongshu Zhou
- Department of Neurosurgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China; Hypothalamic Pituitary Research Center, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China; Hypothalamic Pituitary Research Center, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China.
| | - Renjun Peng
- Department of Neurosurgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China; Hypothalamic Pituitary Research Center, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China.
| | - Zeyu Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China; Hypothalamic Pituitary Research Center, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China.
| | - Xisong Liang
- Department of Neurosurgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China; Hypothalamic Pituitary Research Center, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China.
| | - Jie Wen
- Department of Neurosurgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China; Hypothalamic Pituitary Research Center, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China.
| | - Jilin Nie
- Department of Radiology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China.
| | - Zhongliang Hu
- Clinical Diagnosis and Therapy Center for Glioma of Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China; Department of Radiology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China.
| | - Liyang Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China; Hypothalamic Pituitary Research Center, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China; Clinical Diagnosis and Therapy Center for Glioma of Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China.
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China; Hypothalamic Pituitary Research Center, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China.
| |
Collapse
|
10
|
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: 2] [Impact Index Per Article: 2.0] [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
|
11
|
Yin H, Chen L, Piao S, Wang Y, Li Z, Lin Y, Tang X, Zhang H, Zhang H, Wang X. M6A RNA methylation-mediated RMRP stability renders proliferation and progression of non-small cell lung cancer through regulating TGFBR1/SMAD2/SMAD3 pathway. Cell Death Differ 2023; 30:605-617. [PMID: 34628486 PMCID: PMC9984538 DOI: 10.1038/s41418-021-00888-8] [Citation(s) in RCA: 63] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 09/29/2021] [Accepted: 09/29/2021] [Indexed: 11/09/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) has the highest mortality rate among all malignancies worldwide. The role of long noncoding RNAs (lncRNAs) in the progression of cancers is a contemporary research hotspot. Based on an integrative analysis of The Cancer Genome Atlas database, we identified lncRNA-RNA Component of Mitochondrial RNA Processing Endoribonuclease (RMRP) as one of the most highly upregulated lncRNAs that are associated with poor survival in NSCLC. Furthermore, N(6)-methyladenosine (m6A) was highly enriched within RMRP and enhanced its RNA stability. In vitro and in vivo experiments showed that RMRP promoted NSCLC cell proliferation, invasion, and migration. In terms of mechanism, RMRP recruited YBX1 to the TGFBR1 promotor region, leading to upregulation of the transcription of TGFBR1. The TGFBR1/SMAD2/SMAD3 pathway was also regulated by RMRP. In addition, RMRP promoted the cancer stem cells properties and epithelial mesenchymal transition, which promote the resistance to radiation therapy and cisplatin. Clinical data further confirmed a positive correlation between RMRP and TGFBR1. In short, our work reveals that m6A RNA methylation-mediated RMRP stability renders proliferation and progression of NSCLC through regulating TGFBR1/SMAD2/SMAD3 pathway.
Collapse
Affiliation(s)
- Hang Yin
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, PR China
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang Province, PR China
| | - Lin Chen
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, PR China
| | - Shiqi Piao
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, PR China
| | - Yiru Wang
- Department of Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, PR China
| | - Zhange Li
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang Province, PR China
- Department of Pharmacology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, PR China
| | - Yuan Lin
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang Province, PR China
| | - Xueqing Tang
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang Province, PR China
| | - Huijuan Zhang
- Department of Oncology, Yuhuangding Hospital, Yantai, Shangdong Province, PR China
| | - Haiyang Zhang
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, PR China
| | - Xiaoyuan Wang
- Department of Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, PR China.
| |
Collapse
|
12
|
Chi C, Hou W, Zhang Y, Chen J, Shen Z, Chen Y, Li M. PDHB-AS suppresses cervical cancer progression and cisplatin resistance via inhibition on Wnt/β-catenin pathway. Cell Death Dis 2023; 14:90. [PMID: 36750722 PMCID: PMC9905568 DOI: 10.1038/s41419-022-05547-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 11/17/2022] [Accepted: 12/30/2022] [Indexed: 02/09/2023]
Abstract
Cervical cancer (CC) is the most prevalent gynecological malignancy occurring in the cervix. Long non-coding RNAs (lncRNAs) can act as oncogenes or anti-oncogenes in CC development. Here, we investigated the functional role and detailed mechanism of lncRNA pyruvate dehydrogenase E1 subunit beta antisense (PDHB-AS) in CC. At first, we found that PDHB-AS was significantly down-regulated in CC cells. Besides, overexpression of PDHB-AS repressed CC cell malignant behaviors. HKF-derived exosomes carried miR-4536-5p to CC cells and thereby inhibited PDHB-AS expression. Moreover, PDHB-AS inactivated the Wnt/β-catenin pathway via impeding the nuclear translocation of β-catenin in CC cells. In addition, miR-582-5p could bind with both PDHB-AS and Dickkopf-1 (DKK1). PDHB-AS recruited poly(A) binding protein cytoplasmic 1 (PABPC1) to inhibit Wnt7b expression. PDHB-AS interacted with RNA-binding motif protein X-linked (RBMX) to regulate cisplatin resistance in CC. Finally, we conducted in vivo experiments to confirm that HKF promoted CC tumor growth whereas PDHB-AS suppressed CC tumor growth. Collectively, PDHB-AS plays a tumor-suppressive role in the progression of CC, which suggests the therapeutic potential of PDHB-AS for CC.
Collapse
Affiliation(s)
- Chi Chi
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, No. 188 Shizi Road, Suzhou, 215006, Jiangsu, China
| | - Wenjie Hou
- Department of Obstetrics and Gynecology, Dushu Lake Hospital Affiliated to Soochow University, 9#, Chongwen Rd. SIP, Suzhou, 215000, Jiangsu, China
| | - Yi Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, No. 188 Shizi Road, Suzhou, 215006, Jiangsu, China
| | - Jie Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, No. 188 Shizi Road, Suzhou, 215006, Jiangsu, China
| | - Zongji Shen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, No. 188 Shizi Road, Suzhou, 215006, Jiangsu, China
| | - Youguo Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, No. 188 Shizi Road, Suzhou, 215006, Jiangsu, China.
| | - Min Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, No. 188 Shizi Road, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
13
|
Hu Z, Liu Y, Liu M, Zhang Y, Wang C. Roles of TGF‑β signalling pathway‑related lncRNAs in cancer (Review). Oncol Lett 2023; 25:107. [PMID: 36817052 PMCID: PMC9932718 DOI: 10.3892/ol.2023.13693] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 01/13/2023] [Indexed: 02/04/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) are a class of RNAs that are >200 nucleotides in length that do not have the ability to be translated into protein but are associated with numerous diseases, including cancer. The involvement of lncRNAs in the signalling of certain signalling pathways can promote tumour progression; these pathways include the transforming growth factor (TGF)-β signalling pathway, which is related to tumour development. The expression of lncRNAs in various tumour tissues is specific, and their interaction with the TGF-β signalling pathway indicates that they may serve as new tumour markers and therapeutic targets. The present review summarized the role of TGF-β pathway-associated lncRNAs in regulating tumorigenesis in different types of cancer and their effects on the TGF-β signalling pathway.
Collapse
Affiliation(s)
- Zhizhong Hu
- Cancer Research Institute, Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yitong Liu
- Cancer Research Institute, Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Meiqi Liu
- Cancer Research Institute, Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yang Zhang
- Cancer Research Institute, Medical School, University of South China, Hengyang, Hunan 421001, P.R. China,Correspondence to: Dr Yang Zhang or Dr Chengkun Wang, Cancer Research Institute, Medical School, University of South China, 28 Chang Sheng Xi Avenue, Hengyang, Hunan 421001, P.R. China, E-mail:
| | - Chengkun Wang
- Cancer Research Institute, Medical School, University of South China, Hengyang, Hunan 421001, P.R. China,Correspondence to: Dr Yang Zhang or Dr Chengkun Wang, Cancer Research Institute, Medical School, University of South China, 28 Chang Sheng Xi Avenue, Hengyang, Hunan 421001, P.R. China, E-mail:
| |
Collapse
|
14
|
Long Non-coding RNA KTN1-AS1 Targets miR-505 to Promote Glioblastoma Progression. Behav Neurol 2023; 2023:4190849. [PMID: 36762036 PMCID: PMC9904930 DOI: 10.1155/2023/4190849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 12/19/2022] [Accepted: 01/13/2023] [Indexed: 02/04/2023] Open
Abstract
Glioblastoma (GBM) is a highly malignant cancer, the prognosis of which is pretty poor. Long non-coding RNAs (lncRNAs) are a class of non-coding RNAs, which play important roles in carcinogenesis process of many cancers including GBM. In this study, we want to clarify the expression, biological function, and molecular mechanism of lncRNA KTN1 antisense RNA 1 (KTN1-AS1) in GBM tumor progression. We found that KTN1-AS1 expression was upregulated in GBM tissues and cell lines. KTN1-AS1 played oncogenic roles to facilitate proliferation, migration, and invasion of GBM cells. Then, we revealed that miR-505 was a target of KTN1-AS1, and its expression was decreased in GBM. KTN1-AS1 contributed to GBM progression by mediating miR-505. Finally, we demonstrated that KTN1-AS1 upregulated some target oncogenes of miR-505 including ZEB2, HMGB1, and RUNX2 in GBM cells. All in all, we concluded that the highly expressed KTN1-AS1 in GBM played oncogenic roles to facilitate GBM progression by targeting miR-505.
Collapse
|
15
|
Wu S, Zhu H, Wu Y, Wang C, Duan X, Xu T. Molecular mechanisms of long noncoding RNAs associated with cervical cancer radiosensitivity. Front Genet 2023; 13:1093549. [PMID: 36685972 PMCID: PMC9846343 DOI: 10.3389/fgene.2022.1093549] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/16/2022] [Indexed: 01/06/2023] Open
Abstract
Despite advances in cervical cancer screening and human papilloma virus (HPV) vaccines, cervical cancer remains a global health burden. The standard treatment of cervical cancer includes surgery, radiation therapy, and chemotherapy. Radiotherapy (RT) is the primary treatment for advanced-stage disease. However, due to radioresistance, most patients in the advanced stage have an adverse outcome. Recent studies have shown that long noncoding RNAs (lncRNAs) participate in the regulation of cancer radiosensitivity by regulating DNA damage repair, apoptosis, cancer stem cells (CSCs), and epithelial-mesenchymal transition (EMT). In this review, we summarize the molecular mechanisms of long noncoding RNAs in cervical cancer and radiosensitivity, hoping to provide a theoretical basis and a new molecular target for the cervical cancer RT in the clinic.
Collapse
Affiliation(s)
| | | | | | | | | | - Tianmin Xu
- Department of Obstetrics and Gynecology, Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
16
|
The role and application of transcriptional repressors in cancer treatment. Arch Pharm Res 2023; 46:1-17. [PMID: 36645575 DOI: 10.1007/s12272-023-01427-4] [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: 09/30/2022] [Accepted: 01/03/2023] [Indexed: 01/17/2023]
Abstract
Gene expression is modulated through the integration of many regulatory elements and their associated transcription factors (TFs). TFs bind to specific DNA sequences and either activate or repress transcriptional activity. Through decades of research, it has been established that aberrant expression or functional abnormalities of TFs can lead to uncontrolled cell division and the development of cancer. Initial studies on transcriptional regulation in cancer have focused on TFs as transcriptional activators. However, recent studies have demonstrated several different mechanisms of transcriptional repression in cancer, which could be potential therapeutic targets for the development of specific anti-cancer agents. In the first section of this review, "Emerging roles of transcriptional repressors in cancer development," we summarize the current understanding of transcriptional repressors and their involvement in the molecular processes of cancer progression. In the subsequent section, "Therapeutic applications," we provide an updated overview of the available therapeutic targets for drug discovery and discuss the new frontier of such applications.
Collapse
|
17
|
Lin Y, Wang F, Zhong Y, Cheng NM, Xiong J, Zheng X, Zhou Y, Gao S, Li F, Wang Y, Yang X, Zhao B. Long non-coding RNA COX7C-5 promotes hepatocellular carcinoma progression via miR-581/ZEB2 axis. Cell Signal 2023; 101:110501. [PMID: 36273755 DOI: 10.1016/j.cellsig.2022.110501] [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: 09/06/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
Abstract
Long non-coding RNAs (lncRNA) play crucial roles in hepatocellular carcinoma (HCC) progression. However, the functional roles of lncRNAs in HCC still remain largely unknown. Our study aimed to investigate the biological function and potential molecular mechanism of lnc-COX7C-5 in HCC. Here, we show that Lnc-COX7C-5 was significantly upregulated in HCC tissues, which was correlated with poor prognosis in HCC patients. Lnc-COX7C-5 positively regulated proliferation, migration, and invasion of HCC cells. Mechanistically, lnc-COX7C-5 function as a competing endogenous RNA (ceRNA) for miR-581 in HCC cells. Over-expression or knockdown of miR-581 could alter cell phenotypes caused by Lnc-COX7C-5 in HCC. Further investigations indicated that ZEB2 was demonstrated as a downstream target of miR-581. In mouse model, over-expression of Lnc-COX7C-5 facilitate lung metastasis of HCC. Collectively, Lnc-COX7C-5 promote HCC tumorigenesis and progression by targeting the miR-581/ZEB2 axis. Lnc-COX7C-5 may be a potential therapeutic target for HCC.
Collapse
Affiliation(s)
- Yantin Lin
- Fuzhou Hospital of Traditional Chinese Medicine Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou 350001, China; School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, PR China
| | - Fei Wang
- The United Innovation of Meng chao Hepatobiliary Technology Key Laboratory of Fujian Province, Meng chao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China
| | - Yue Zhong
- The United Innovation of Meng chao Hepatobiliary Technology Key Laboratory of Fujian Province, Meng chao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China
| | - Niang Mei Cheng
- The United Innovation of Meng chao Hepatobiliary Technology Key Laboratory of Fujian Province, Meng chao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China
| | - Jiahui Xiong
- Fuzhou Hospital of Traditional Chinese Medicine Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou 350001, China
| | - Xiaoyuan Zheng
- The United Innovation of Meng chao Hepatobiliary Technology Key Laboratory of Fujian Province, Meng chao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China
| | - Yang Zhou
- The United Innovation of Meng chao Hepatobiliary Technology Key Laboratory of Fujian Province, Meng chao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China
| | - Shaodong Gao
- Fuzhou Hospital of Traditional Chinese Medicine Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou 350001, China; School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, PR China
| | - Fang Li
- Fuzhou Hospital of Traditional Chinese Medicine Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou 350001, China; School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, PR China
| | - Yingchao Wang
- The United Innovation of Meng chao Hepatobiliary Technology Key Laboratory of Fujian Province, Meng chao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China
| | - Xiaoyu Yang
- Fuzhou Hospital of Traditional Chinese Medicine Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou 350001, China; School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, PR China.
| | - Bixing Zhao
- The United Innovation of Meng chao Hepatobiliary Technology Key Laboratory of Fujian Province, Meng chao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China.
| |
Collapse
|
18
|
Hashemi M, Hajimazdarany S, Mohan CD, Mohammadi M, Rezaei S, Olyaee Y, Goldoost Y, Ghorbani A, Mirmazloomi SR, Gholinia N, Kakavand A, Salimimoghadam S, Ertas YN, Rangappa KS, Taheriazam A, Entezari M. Long non-coding RNA/epithelial-mesenchymal transition axis in human cancers: Tumorigenesis, chemoresistance, and radioresistance. Pharmacol Res 2022; 186:106535. [DOI: 10.1016/j.phrs.2022.106535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/22/2022] [Accepted: 10/30/2022] [Indexed: 11/07/2022]
|
19
|
Yuan C, Liu C, Zhao S, Zhang X, Jia H, Chen B, Zhang M, Zheng Y, Zhou J, Bo Y. The Role of N6-Methyladenosine-Associated lncRNAs in the Immune Microenvironment and Prognosis of Colorectal Cancer. JOURNAL OF ONCOLOGY 2022; 2022:4689396. [PMID: 36111241 PMCID: PMC9470360 DOI: 10.1155/2022/4689396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 06/17/2022] [Indexed: 11/30/2022]
Abstract
Background The role of N6-methyladenosine long noncoding RNAs (lncRNAs) in colorectal cancer (CRC) is elusive. Materials and Methods We identified m6A-associated lncRNAs by using the data gathered from The Cancer Genome Atlas (TCGA) and stratified CRC patients into different subgroups. Cox regression analysis was performed to construct an m6A-associated lncRNA signature. The role of this signature in the immune microenvironment and prognosis was dissected subsequently. Finally, a gene set enrichment analysis (GSEA) was conducted to predict the possible mechanisms based on the signature. Results Three m6A-associated clusters were constructed from 866 differentially expressed lncRNAs. Cluster 2 had poor prognosis and low immune cell infiltration. An m6A-associated lncRNA signature consisting of 14 lncRNAs was constructed and recognized as an independent prognostic indicator of CRC by using survival analysis and receiver operating characteristic (ROC) curves. The clinical features and immune cell infiltration status were significantly different in patients stratified by the risk score. Furthermore, GSEA showed that the P53 pathway and natural killer cell-mediated cytotoxicity were more enriched in the low-risk group. Conclusion Our data revealed that m6A-associated lncRNAs could be potential prognostic indicators of immunogenicity in CRC.
Collapse
Affiliation(s)
- Congfei Yuan
- Department of General Surgery, Lianshui County People's Hospital, Huai'an 223400, China
| | - Caidong Liu
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Shuli Zhao
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Xishan Zhang
- Department of General Surgery, Lianshui County People's Hospital, Huai'an 223400, China
| | - Haifeng Jia
- Department of General Surgery, Lianshui County People's Hospital, Huai'an 223400, China
| | - Baiyu Chen
- Department of General Surgery, Lianshui County People's Hospital, Huai'an 223400, China
| | - Maojin Zhang
- Department of General Surgery, Lianshui County People's Hospital, Huai'an 223400, China
| | - Yuan Zheng
- Department of General Surgery, Lianshui County People's Hospital, Huai'an 223400, China
| | - Jin Zhou
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Yanzhi Bo
- Department of General Surgery, Lianshui County People's Hospital, Huai'an 223400, China
| |
Collapse
|
20
|
Yi H, Han Y, Li S. Oncogenic circular RNA circ_0007534 contributes to paclitaxel resistance in endometrial cancer by sponging miR-625 and promoting ZEB2 expression. Front Oncol 2022; 12:985470. [PMID: 35992812 PMCID: PMC9386306 DOI: 10.3389/fonc.2022.985470] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 07/18/2022] [Indexed: 12/05/2022] Open
Abstract
Circular RNAs (circRNAs) and epithelial to mesenchymal transition (EMT) have been implicated in the development of human cancer and paclitaxel resistance. CircRNA circ_0007534 has been described as a key oncogenic circular RNA that is upregulated in a variety of cancer tissues. However, whether circ_0007534 causes EMT and paclitaxel resistance in endometrial cancer is still unknown. In this work, we revealed that circ_0007534 levels were significantly higher in endometrial cancer tissues, and that high circ_0007534 expression was associated with poor differentiation, advanced tumor stage, cancer invasion, cancer metastasis, and poor prognosis in endometrial cancer patients. Overexpression of circ_0007534 boosted endometrial cancer cell proliferation, invasion, EMT, and paclitaxel resistance. Knockdown of circ_0007534 restored paclitaxel sensitivity and reversed EMT in endometrial cancer cells. We also showed that circ_0007534 enhanced endometrial cancer aggressiveness, progression, and paclitaxel resistance by sponging microRNA-625 (miR-625) and subsequently increasing the expression of the miR-625 target gene ZEB2. Our cell functional studies demonstrated that inhibiting miR-625 or increasing ZEB2 mimicked the effects of circ_0007534 overexpression. Consequently, our data show that circ_0007534 plays a crucial role in EMT and paclitaxel resistance through miR-625/ZEB2 signaling. Targeting the circ_0007534/miR-625/ZEB2 pathway might be an effective strategy for overcoming paclitaxel resistance in endometrial cancer.
Collapse
Affiliation(s)
- Hanjie Yi
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yongqing Han
- Department of Oncology, ShangRao People’s Hospital, Shangrao, China
| | - Shanfeng Li
- Department of Nosocomial Infection Management, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- *Correspondence: Shanfeng Li,
| |
Collapse
|
21
|
Li C, Li X, Zhang Y, Wu L, He J, Jiang N, Zhao H, Liu W. DSCAM-AS1 promotes cervical carcinoma cell proliferation and invasion via sponging miR-338-3p. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:58906-58914. [PMID: 35378649 DOI: 10.1007/s11356-022-19962-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 04/01/2020] [Indexed: 06/14/2023]
Abstract
Deregulated lncRNA DSCAM-AS1 expression was found in several tumors. However, mechanism and functional role of DSCAM-AS1 in cervical carcinoma remain unknown. DSCAM-AS1 was detected in cervical carcinoma specimens and cells by RT-qPCR. CCK-8, Matrigel transwell, and flow cytometry were conducted to determine cell functions. In this research, we firstly we explored DSCAM-AS1 expression in cervical carcinoma cells and specimens. We revealed that DSCAM-AS1 was upregulated in cervical carcinoma lines (C4-1, Caski, Hela, and Siha) compared to GH329 cells. DSCAM-AS1 was upregulated in cervical carcinoma specimens compared to control no-tumor specimens. Overexpression of DSCAM-AS1 induced cervical carcinoma cell growth and cycle. Moreover, our data revealed that miR-338-3p expression was downregulated in cervical carcinoma cells and specimens. There was a negative correlation between miR-338-3p expression and DSCAM-AS1 expression in cervical carcinoma specimens. Elevated expression of miR-338-3p decreased cervical carcinoma cell growth and cycle and invasion. Furthermore, luciferase reporter analysis revealed that miR-338-3p overexpression suppressed luciferase activity of WT-DSCAM-AS1 vector but not the mut-DSCAM-AS1. Ectopic expression of DSCAM-AS1 decreased miR-338-3p expression in the Siha cell. Overexpression of DSCAM-AS1 promoted cervical carcinoma cell growth and cycle via regulating miR-338-3p. These results suggested that DSCAM-AS1 functions as one oncogene through sponging miR-338-3p in cervical carcinoma.
Collapse
Affiliation(s)
- Chunmei Li
- International Medical Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China.
- Department of Traditional Chinese Medicine, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China.
| | - Xiaoqiong Li
- International Medical Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
- Department of Traditional Chinese Medicine, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Yi Zhang
- Department of Traditional Chinese Medicine, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
- Department of Gynecology, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Liangzhi Wu
- International Medical Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
- Department of Traditional Chinese Medicine, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Jingjun He
- International Medical Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
- Department of Traditional Chinese Medicine, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Nan Jiang
- Department of Traditional Chinese Medicine, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
- Department of Gynecology, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Hui Zhao
- Department of Traditional Chinese Medicine, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
- Department of Gynecology, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Wenwen Liu
- Department of Traditional Chinese Medicine, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
- Department of Gynecology, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| |
Collapse
|
22
|
LncRNA FAM13A-AS1 Regulates Proliferation and Apoptosis of Cervical Cancer Cells by Targeting miRNA-205-3p/DDI2 Axis. JOURNAL OF ONCOLOGY 2022; 2022:8411919. [PMID: 35783157 PMCID: PMC9246599 DOI: 10.1155/2022/8411919] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 06/02/2022] [Indexed: 11/17/2022]
Abstract
The aim of this study was to explore the function of long noncoding RNA (lncRNA) FAM13A-AS1 and its associated mechanism in cervical cancer. A total of 30 cervical cancer tissues and adjacent tissues were collected. Cervical cancer cell lines, including SiHa and HeLa, were transfected with constructs expressing LV-FAM13A-AS1, silencing RNA LV-siFAM13A-AS1, miRNA mimics, and miRNA inhibitors. RT-qPCR was used to detect the expression of FAM13A-AS1 in cervical cancer tissues, including SiHa, HeLa, and HUCEC cells. MTT, flow cytometry, and transwell assays were performed to explore the influence of FAM13A-AS1 on cervical cancer cell proliferation, apoptosis, invasion, and migration. A bioinformatics analysis and a dual-luciferase assay were carried to confirm the target relationship between FAM13A-AS1 or DDI2 and miRNA-205-3p. Finally, in vivo tumorigenesis experiments were performed in nude mice to explore the effect of FAM13A-AS1 expression on cervical cancer. Low FAM13A-AS1 expression and high miRNA-205-3p expression were observed in cervical cancer tissues and cell lines (SiHa and HeLa). Upregulating the expression of FAM13A-AS1 inhibited proliferation, migration, and invasion of SiHa and HeLa cells, while the apoptosis of SiHa and HeLa cells was increased. More importantly, LV-FAM13A-AS1 could improve tumor development in vivo. In addition, FAM13A-AS1 negatively regulated the expression of miRNA-205-3p, while miRNA-205-3p reduced DDI2 expression, and miRNA-205-3p mimic reversed the effects of FAM13A-AS1 overexpression in vitro. In conclusion, FAM13A-AS1 inhibits the progression of cervical cancer by targeting the miRNA-205-3p/DDI2 axis, suggesting that FAM13A-AS1 might be a potential target for cancer cell treatment.
Collapse
|
23
|
LncRNA LINC01116 Regulates the Proliferation, Migration, and Invasion of Cervical Cancer Cells by Targeting miR-744-5p. Cell Microbiol 2022. [DOI: 10.1155/2022/2615523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Objective. To investigate the effects and potential molecular mechanisms of LncRNA LINC01116 on proliferation, migration, and invasion of cervical cancer cells. Method(s). The content of miR-744-5p and LINC01116 in cervical cancer cells HeLa, SiHa, and C33a was detected by RT-PCR, the proliferative activity and clone number of SiHa cells were determined by MTT and clone formation assay, the number of invaded and migrated cells was determined by Transwell assay, the expressions of Cyclin D1 and MMP-2 in cells were detected by Western blot, and the activity of luciferase detected by dual-luciferase reporting system verified the regulatory relationship between LINC01116 and miR-744-5p. Result(s). Compared with human normal cervical epithelial cells Ect1/E6E7, the content of LINC01116 in cervical cancer cells HeLa, SiHa, and C33a was increased significantly [(
) vs. (
)/(
)/(
)] (
), the content of miR-744-5p was decreased significantly [(
) vs. (
)/(
)/(
)] (
). Silencing LINC01116 could inhibit the protein expression of Cyclin D1 [(
) vs. (
)] and MMP-2 [(
) vs. (
)] in SiHa cells, and inhibition of cell proliferation [(
)% vs. (
)%], clone formation ability [(
) vs. (
)], migration [(
) vs. (
)], and invasion [(
) vs. (
)]. LINC01116 targeted and negatively regulates the expression of miR-744-5p. Overexpression of miR-744-5p could inhibit the proliferation of cervical cancer SiHa cells [(
)% vs. (
)%], clone formation ability [(
) vs. (
)], migration [(
) vs. (
)], and invasion [(
) vs. (
)]. Inhibition of miR-744-5p reversed the effects of silencing LINC01116 on SiHa cell proliferation [(
)% vs. (
)%], clone formation ability [(
) vs. (
)], migration [(
) vs. (
)], and invasion [(69.12 ± 5.56) vs (94.31 ± 7.44)]. Conclusion(s). LncRNA LINC01116 inhibits proliferation, migration, and invasion of SiHa cells by targeting miR-744-5p, and LINC01116 is a potential molecular target for cervical cancer.
Collapse
|
24
|
Li W, Hu X, Huang X. Long intergenic non-protein coding RNA 847 promotes laryngeal squamous cell carcinoma progression through the microRNA-181a-5p/zinc finger E-box binding homeobox 2 axis. Bioengineered 2022; 13:9987-10000. [PMID: 35435130 PMCID: PMC9161931 DOI: 10.1080/21655979.2022.2062531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The present study is targeted at investigating the effects of long intergenic non-protein coding RNA 847 (LINC00847) on the malignant biological behaviors of laryngeal squamous cell carcinoma (LSCC) cells, and the mechanisms. Quantitative real-time PCR and Western blotting were conducted for detecting the expressions of LINC00847, microRNA-181a-5p (miR-181a-5p) and zinc finger E-box binding homeobox 2 (ZEB2) in LSCC cell lines and tissue samples. BrdU, cell counting kit-8, scratch wound healing, Transwell and flow cytometry assays were utilized for detecting cell proliferation, migration, invasion, and cell cycle progression. Dual-luciferase reporter gene, RNA binding protein immunoprecipitation (RIP), and RNA pull-down assays were utilized to investigate the interaction among LINC00847, miR-181a-5p, and ZEB2. The subcellular location of LINC00847 was determined by RNA fluorescence in situ hybridization (RNA-FISH) assay. Tumor growth was evaluated using a xenograft model of nude mice. It was revealed that LINC00847 expression was increased in LSCC tissues, and its high expression was associated with lymph node metastasis and poor differentiation. LINC00847 was mainly located in the cytoplasm of LSCC cells, and LINC00847 overexpression promoted LSCC cell proliferation, migration, invasion, and accelerated the cell cycle progression while knocking down LINC00847 had the opposite effects in vitro and inhibited the tumor growth in vivo. LINC00847 directly targeted miR-181a-5p and negatively modulated miR-181a-5p expression. ZEB2 was a target gene of miR-181a-5p, and was positively and indirectly modulated by LINC00847. Our data suggest that LINC00847 promotes LSCC progression by regulating the miR-181a-5p/ZEB2 axis.
Collapse
Affiliation(s)
- Wei Li
- Department of Otolaryngology, Ezhou Central Hospital, Hubei, Ezhou Province, P.R. China
| | - Xionghui Hu
- Department of Otolaryngology, Tongxiang First People’s Hospital, Jiaxing, Zhejiang Province, P.R. China
| | - Xiaolin Huang
- Department of Otolaryngology, Ezhou Central Hospital, Hubei, Ezhou Province, P.R. China
| |
Collapse
|
25
|
Wang H, Wang D, Wei Q, Li C, Li C, Yang J. Long non-coding RNAs PGM5-AS1 upregulates Decorin (DCN) to inhibit cervical cancer progression by sponging miR-4284. Bioengineered 2022; 13:9872-9884. [PMID: 35420507 PMCID: PMC9161867 DOI: 10.1080/21655979.2022.2062088] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) have been widely studied and play crucial roles in cervical cancer (CC) progression. Here, we investigated the function and mechanism of lncRNA PGM5-AS1 action in CC cells. Using real-time quantitative polymerase chain reaction or western blotting, PGM5-AS1 and decorin (DCN) were downregulated in CC tissues and cells, whereas miR-4284 was upregulated. Luciferase assay, RNA pull-down assay, and western blotting showed that PGM5-AS1 could sponge miR-4284 to upregulate DCN expression in CC cells. Additionally, cell functional experiments showed that PGM5-AS1 overexpression led to decreased proliferation, migration, and invasion of CC cells. However, the inhibitory effect of PGM5-AS1 overexpression on CC cells was partly relieved by DCN knockdown because of the targeting interaction between PGM5-AS1, miR-4284, and DCN. In summary, this study identified that PGM5-AS1 negatively regulates CC cell malignancy by targeting miR-4284/DCN.
Collapse
Affiliation(s)
- Huimin Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Department of Obstetrics and Gynecology, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, Hubei, China
| | - Dan Wang
- Department of Obstetrics and Gynecology, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qiong Wei
- Department of Obstetrics and Gynecology, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, Hubei, China
| | - Chun Li
- Department of Obstetrics and Gynecology, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, Hubei, China
| | - Chunyan Li
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jing Yang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
26
|
Chen X, Liu Y, Liu H, Wang ZW, Zhu X. Unraveling diverse roles of noncoding RNAs in various human papillomavirus negative cancers. Pharmacol Ther 2022; 238:108188. [PMID: 35421419 DOI: 10.1016/j.pharmthera.2022.108188] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/02/2022] [Accepted: 04/07/2022] [Indexed: 12/17/2022]
Abstract
Human papillomavirus (HPV)-negative tumors distinguish from cancers associated with HPV infection. Due to its high rate of lymph node metastasis and difficulty in inchoate discover and diagnosis, the treatment efficacy of HPV-negative cancers is unsatisfactory. Epidemiological evidence suggests that HPV-negative tumor patients have a poor prognosis, and the mortality is higher than that of cancer patients caused by HPV infection. Evidence has demonstrated that noncoding RNAs (ncRNAs) play a crucial role in regulation of physiological and developmental processes. Therefore, dysregulated ncRNAs are involved in the occurrence of diversified diseases, including cancer. In cumulative studies, ncRNAs are concerned with pathogenetic mechanisms of HPV-negative tumors via regulating gene expression and signal transduction. It is important to decipher the functions of ncRNAs in HPV-negative cancers and identify the potential biomarkers, which will bring new treatment strategies for improving outcome of cancer therapy. In this review, we demonstrated the effects of ncRNAs via regulating the development and progression of HPV- negative tumors by directly or indirectly acting on target molecules, which provide a basis for future tumor targeted therapy by targeting ncRNAs for HPV-negative cancers.
Collapse
Affiliation(s)
- Xin Chen
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Yi Liu
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Hejing Liu
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Zhi-Wei Wang
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China; Department of Research and Development, Beijing Zhongwei Research Center of Biological and Translational Medicine, Beijing 100161, China.
| | - Xueqiong Zhu
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China.
| |
Collapse
|
27
|
Wu C, Wei W, Li J, Peng S. The Impacts of Bone Marrow Mesenchymal Stem Cells (BMSCs)-Derived Periostin on Epithelial-Mesenchymal Transition (EMT) of Cervical Cancer Cells. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.2958] [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
Epithelial-mesenchymal transition (EMT) is closely related to the migrating and invading behaviors of cells. Periostin is one of the essential components in the extracellular matrix and can induce EMT of cells and their sequential metastasis. But its underlying mechanism is unclear.
The Hela and BMSC cell lines were assigned into Periostin-mimic group, Periostin-Inhibitor group and Periostin-NC group followed by analysis of cell migration and invasion, expression of E-Cadherin, Vimentin, β-Catenin, Snail, MMP-2, MMP-9, PTEN, and p-PTEN. Cells in Periostin-mimic
group exhibited lowest migration, least number of invaded cells, as well as lowest levels of Vimentin, β-Catenin, Snail, MMP-2, MMP-9, p-PTEN, Akt, p-Akt, p-GSK-3β, p-PDK1 and p-cRcf, along with highest levels of E-cadherin and PTEN. Moreover, cells in Periostin-NC
group had intermediate levels of these above indicators, while, the Periostin-Inhibitor group exhibited the highest migration rate, the most number of invaded cells, and the highest levels of these proteins (P < 0.05). In conclusion, BMSCs-derived Periostin can influence the EMT
of cervical cancer cells possibly through restraining the activity of the PI3K/AKT signal transduction pathway, indicating that Periostin might be a target of chemotherapy in clinics for the treatment of cervical cancer.
Collapse
Affiliation(s)
- Chengyong Wu
- Reproductive Medicine Center, Yichun People’s Hospital, Yichun, Jiangxi, 336000, China
| | - Weifeng Wei
- Department of Obstetrics and Gynecology, Meizhou People’s Hospital, Meizhou, Guangdong, 514000, China
| | - Jing Li
- Department of Obstetrics and Gynecology, Meizhou People’s Hospital, Meizhou, Guangdong, 514000, China
| | - Shenglin Peng
- Reproductive Medicine Center, Yichun People’s Hospital, Yichun, Jiangxi, 336000, China
| |
Collapse
|
28
|
Zinc Finger E-Box Binding Homeobox 2 as a Prognostic Biomarker in Various Cancers and Its Correlation with Infiltrating Immune Cells in Ovarian Cancer. Curr Issues Mol Biol 2022; 44:1203-1214. [PMID: 35723302 PMCID: PMC8947672 DOI: 10.3390/cimb44030079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/25/2022] [Accepted: 02/27/2022] [Indexed: 11/17/2022] Open
Abstract
This study investigated the expression of zinc finger E-box binding homeobox 2 (ZEB2), its prognostic significance in various cancers, and the correlation between ZEB2 and infiltrating immune cells and ZEB2-related proteins in ovarian cancer (OV). The Gene Expression Profiling Interactive Analysis tool was used to analyze RNA sequencing data and cancer survival rates, based on normal and tumor tissue data available in The Cancer Genome Atlas (TCGA) database. The Kaplan–Meier plotter and PrognoScan databases were used to analyze the prognostic value of ZEB2 in OV (n = 1144). The Tumor Immune Estimation Resource was used to investigate the correlation between ZEB2 and infiltrating immune cells in various cancers, including OV. High ZEB2 expression was associated with a poorer prognosis in OV. In OV, ZEB2 is positively correlated with CD8+T cells, neutrophils, macrophages, and dendritic cell invasion; and ZEB2 is negatively correlated with tumor-infiltrating B cells. The STRING database was used to investigate the correlations with ZEB2-related proteins. The results reveal that ZEB2 was positively correlated with SMAD1 and SMAD2 in OV. Our findings may serve as a potential prognostic biomarker, and provide novel insights into the tumor immunology in OV. Thus, ZEB2 may be a potential diagnostic and therapeutic target in OV.
Collapse
|
29
|
LncRNA MAGI2-As3 Suppresses the Proliferation and Invasion of Cervical Cancer by Sponging MiR-15b. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:9707206. [PMID: 35126958 PMCID: PMC8808199 DOI: 10.1155/2022/9707206] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/06/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Cervical cancer is the leading cause of cancer deaths among women, and more than 85% of cervical cancer deaths occur in low and middle-income countries. The purpose of this study is to investigate the functions of MAGI2-AS3 and miR-15b in cervical cancer. MATERIALS AND METHODS The mRNA levels of MAGI2-AS3, miR-15b, and CCNE1 were evaluated using RT-qPCR assay. Dual-luciferase reporter gene assay was used to confirm whether miR-15b binds to CCNE1. RESULTS LncRNA MAGI2-AS3 was downregulated, while miR-15b was upregulated in cervical cancer. Cervical cancer patients with low expression of MAGI2-AS3 have a poor prognosis. Upregulation of MAGI2-AS3 inhibited proliferative and invasive abilities of HeLa cells via regulating the expression of miRNA-15b. MiR-15b inhibitor suppressed cell proliferation and invasion. CCNE1 was a direct target gene of miR-15b, which binds to the 3'-UTR of its mRNA. MiR-15b partially reversed the inhibitory effect of overexpression of MAGI2-AS3 on the proliferation and invasion of HeLa cells. MAGI2-AS3 mediated the expression of CCNE1 in HeLa cells. CONCLUSION LncRNA MAGI2-AS3 inhibits the proliferation and invasion of cervical cancer cells via the miRNA-15/CCNE1 axis. Our results illustrates that MAGI2-AS3 can be used as a useful clinical predictor for early diagnosis and prognosis assessment of cervical cancer.
Collapse
|
30
|
lncRNA SNHG15 Induced by SOX12 Promotes the Tumorigenic Properties and Chemoresistance in Cervical Cancer via the miR-4735-3p/HIF1a Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8548461. [PMID: 35069980 PMCID: PMC8769851 DOI: 10.1155/2022/8548461] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 11/02/2021] [Accepted: 11/08/2021] [Indexed: 12/12/2022]
Abstract
Cervical cancer (CC) is one of the most common malignancies in females, with high prevalence and mortality globally. Despite advances in diagnosis and therapeutic strategies developed in recent years, CC is still a major health burden worldwide. The molecular mechanisms underlying the development of CC need to be understood. In this study, we aimed to demonstrate the role of lncRNA SNHG15 in CC progression. Using qRT-PCR, we determined that lncRNA SNHG15 is highly expressed in CC tumor tissues and cells. lncRNA SNHG15 knockdown also reduces the tumorigenic properties of CC in vitro, as determined using the MTT, EdU, flow cytometry, and transwell assays. Using bioinformatics analysis, RNA pull-down, ChIP, and luciferase reporter assays, we verified the molecular mechanisms of lncRNA SNHG15 in CC progression and found that lncRNA SNHG15 expression in CC cells is transcriptionally regulated by SOX12; moreover, lncRNA SNHG15 promotes CC progression via the miR-4735-3p/HIF1a axis. This study can provide a potential target for CC diagnosis or therapeutic strategies in the future.
Collapse
|
31
|
CTI-2 Inhibits Metastasis and Epithelial-Mesenchymal Transition of Breast Cancer Cells by Modulating MAPK Signaling Pathway. Int J Mol Sci 2021; 22:ijms222212229. [PMID: 34830111 PMCID: PMC8622910 DOI: 10.3390/ijms222212229] [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: 09/23/2021] [Revised: 10/26/2021] [Accepted: 10/29/2021] [Indexed: 02/07/2023] Open
Abstract
Although some breast cancer patients die due to tumor metastasis rather than from the primary tumor, the molecular mechanism of metastasis remains unclear. Therefore, it is necessary to inhibit breast cancer metastasis during cancer treatment. In this case, after designing and synthesizing CTI-2, we found that CTI-2 treatment significantly reduced breast cancer cell metastasis in vivo and in vitro. Notably, with the treatment of CTI-2 in breast cancer cells, the expression level of E-cadherin increased, while the expression level of N-cadherin and vimentin decreased. In addition, after CTI-2 treatment, those outflow levels for p-ERK, p-p38, and p-JNK diminished, while no significant changes in the expression levels of ERK, JNK, or p38 were observed. Our conclusion suggested that CTI-2 inhibits the epithelial-mesenchymal transition (EMT) of breast carcinoma cells by inhibiting the activation of the mitogen-activated protein kinase (MAPK) signaling pathway, thereby inhibiting the metastasis of breast tumor cells. Therefore, we believe that CTI-2 is another candidate for breast tumor medication.
Collapse
|
32
|
Lei GL, Li Z, Li YY, Hong ZX, Wang S, Bai ZF, Sun F, Yan J, Yu LX, Yang PH, Yang ZY. Long noncoding RNA FAM66C promotes tumor progression and glycolysis in intrahepatic cholangiocarcinoma by regulating hsa-miR-23b-3p/KCND2 axis. ENVIRONMENTAL TOXICOLOGY 2021; 36:2322-2332. [PMID: 34418280 DOI: 10.1002/tox.23346] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/04/2021] [Accepted: 08/04/2021] [Indexed: 06/13/2023]
Abstract
Long noncoding RNAs (lncRNAs) are known to be the important regulators in cancer progression. However, the role of lncRNA FAM66C (FAM66C) is yet to be investigated in intrahepatic cholangiocarcinoma (ICC). This study aimed to investigate the effects and related mechanisms of FAM66C in ICC. Human ICC tissues and cell lines were collected. The expression levels of FAM66C, hsa-miR-23b-3p (miR-23b-3p), and KCND2 were detected by qRT-RCR. The transfection experiments were employed to measure the effect of FAM66C on cell viabilities, migration, and invasion in ICC cells by CCK-8, transwell assays. Glycolysis was investigated by glucose consumption, lactate production and ATP levels. The dual-luciferase reporter and RNA pull down assays were conducted as a means of confirming the interactions between FAM66C, miR-23b-3p, and KCND2. Furthermore, the levels of the EMT-associated proteins (KCND2, GLUT1, PKM2, and LDHA) in ICC cells were detected by western blot. FAM66C was increased in ICC tissues and cells, increased cell viability, glycolysis, migration and invasion, and decreased apoptosis were shown in FAM66C overexpressing cells. Mechanistic analyses revealed that FAM66C regulated the downstream target gene KCND2 by sponging miR-23b-3p. FAM66C effect on ICC was further validated in murine xenograft assays. FAM66C knockdown cells gave rise to tumors that were smaller in size, consistent with the role of FAM66C as a promoter of in vivo tumor growth. These data revealed that FAM66C was able to drive ICC tumor progression and glycolytic activity via the miR-23b-3p/KCND2 axis, indicating FAM66C may be a viable target for treating ICC.
Collapse
Affiliation(s)
- Guang-Lin Lei
- Medical School of Chinese PLA, Beijing, China
- Senior Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zhi Li
- Medical School of Chinese PLA, Beijing, China
- Senior Department of Hepato-Pancreato-Biliary Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yuan-Yuan Li
- Senior Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zhi-Xian Hong
- Senior Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Sen Wang
- Senior Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zhi-Fang Bai
- Senior Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Fang Sun
- Senior Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jin Yan
- Senior Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Ling-Xiang Yu
- Senior Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Peng-Hui Yang
- Medical School of Chinese PLA, Beijing, China
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zhan-Yu Yang
- Medical School of Chinese PLA, Beijing, China
- Senior Department of Hepato-Pancreato-Biliary Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
33
|
Tie W, Ge F. MALAT1 Inhibits Proliferation of HPV16-Positive Cervical Cancer by Sponging miR-485-5p to Promote Expression of MAT2A. DNA Cell Biol 2021; 40:1407-1417. [PMID: 34610246 DOI: 10.1089/dna.2020.6205] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cervical cancer is the leading cause of morbidity and mortality in women throughout the world, human papillomavirus 16 (HPV16) is the main type of HPV causing invasive cervical cancer. However, the underlying mechanism of the high carcinogenicity of HPV16 remains unclear. In the current study, we documented that metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), a long noncoding RNA, is upregulated in HPV16-positive cervical cancer tissue and cell lines. The results of immunohistochemistry and immunofluorescence showed that MALAT1 was mainly localized in the cytoplasm. To clarify the biological functions of MALAT1 in cervical cancer cells, we performed gain- and loss-of-function experiments to explore the underlying molecular mechanism. Functionally, the proliferation of cervical cancer was detected by Cell Counting Kit-8 (CCK-8) and colony formation assay in MALAT1 overexpression or knockdown cells, our data showed that MALAT1 promotes the proliferation of cervical cancer cells. Mechanistically, our results suggested that MALAT1 upregulates Methionine adenosyltransferase 2A (MAT2A) by sponging miR-485-5p. Moreover, the gain-of-function assay validated the function of MAT2A in HPV16-positive cervical cancer proliferation. Taken together, our results demonstrated that MALAT1 acts as a competitive endogenous RNA (ceRNA) to regulate MAT2A by sponging miR-485-5p in HPV16-positive cervical cancer, suggesting that MALAT1 may act as a potential therapeutic target for HPV16-positive cervical cancer.
Collapse
Affiliation(s)
- Weiwei Tie
- Department of Gynaecology, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Fenfen Ge
- Department of Gynaecology, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| |
Collapse
|
34
|
Wang J, Liu Y, Cai H, Jiang H, Li W, Shi Y. Long coding RNA CCAT2 enhances the proliferation and epithelial-mesenchymal transition of cervical carcinoma cells via the microRNA-493-5p/CREB1 axis. Bioengineered 2021; 12:6264-6274. [PMID: 34499007 PMCID: PMC8806934 DOI: 10.1080/21655979.2021.1969834] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Cervical cancer (CC) is one of the most common malignancies among women. It has been demonstrated that long coding RNAs (lncRNAs) play a crucial role in CC. The purpose of this study was to investigate the role of the colon cancer associated transcript 2 (CCAT2) lncRNA in CC and elucidate its possible mechanisms of action. The expression of CCAT2, the miR-493-5p microRNA (miRNA), and mRNA was detected using qRT-PCR. Cell viability, proliferation, and migration and invasion were determined using the MTT, colony formation, and transwell assays, respectively. The interactions between miR-493-5p and CCAT2 or cAMP response element-binding protein 1 (CREB1) were verified using the luciferase and RNA pull-down assays. The effects of CCAT2 knockdown on in vivo tumor growth were determined using tumor xenografts and immunohistochemistry assays. The expression of CCAT2 was upregulated in CC cells and tissues. However, the knockdown of CCAT2 inhibited the proliferation and epithelial-mesenchymal transition (EMT) of CC cells in vitro and suppressed tumor growth in vivo. Mechanistically, CCAT2 functions as a competing endogenous RNA (ceRNA) to upregulate the expression of CREB1 by binding to miR-493-5p. The overexpression of CREB1 or downregulation of miR-493-5p antagonized the effect of CCAT2 knockdown on the proliferation and EMT of CC cells. The knockdown of CCAT2 suppressed the aggressiveness of CC via the miR-493-5p/CREB1 axis. Therefore, CCAT2 is likely to be a promising therapeutic target for CC.
Collapse
Affiliation(s)
- Jing Wang
- Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, P.R. China.,Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, P.R. China.,Hubei Key Laberatory of Tumor Biological Behavirs, P.R. China.,Hubei Cancer Clinical Study Center, P.R. China
| | - Yan Liu
- Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, P.R. China
| | - Hongbing Cai
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, P.R. China.,Hubei Key Laberatory of Tumor Biological Behavirs, P.R. China.,Hubei Cancer Clinical Study Center, P.R. China
| | - Hong Jiang
- Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, P.R. China
| | - Wei Li
- Department of Obstetrics and Gynecology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, P.R. China
| | - Yuying Shi
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, P.R. China.,Hubei Key Laberatory of Tumor Biological Behavirs, P.R. China.,Hubei Cancer Clinical Study Center, P.R. China
| |
Collapse
|
35
|
Liang M, Li Y, Dai T, Chen C. lncRNA FEZF1-AS1 regulates biological behaviors of cervical cancer by targeting miRNA-1254. Food Sci Nutr 2021; 9:4722-4737. [PMID: 34531986 PMCID: PMC8441442 DOI: 10.1002/fsn3.2315] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 11/21/2022] Open
Abstract
AIM The purpose of this research was to evaluate lncRNA FEZF1-AS1 in cervical cancer development and clinical significance. MATERIALS AND METHODS Collecting cervical cancer tissues, measuring FEZF1-AS1 expression, and analysis correlation between FEZF1-AS1 and prognosis. In cell vitro study, using MTT assay to measure cell proliferation, evaluating cell apoptosis by flow cytometry, measuring cell invasion and migration by Transwell and wound healing assay; lncRNA FEZF1-AS1 and miR-1254 gene expressions were evaluated by RT-qPCR assay; relative protein (Smurf1, E-cadherin, Vimentin, N-cadherin, AKT, p-AKT, c-Myc, and ZEB1) expressions were measured by Western blot assay. The correlation among FEZF1-AS1, miR-1254, and Smurf1 were analysis by dual luciferase reporter gene assay. RESULTS By clinical analysis, lncRNA FEZF1-AS1 was high expression in cervical cancer tissues and high expression was closely correlated with poor prognosis in cervical cancer patients. In vitro study, the SiHa and HeLa cell biologically including cell proliferation, migration, and invasion of si-FEZF1-AS1 group which knockdown lncRNA FEZF1-AS1 were significantly depressed (p < .001, respectively). However, with miR-1254 expression inhibiting, the cell biological activities were significantly increased in si-FEZF1-AS1+miRNA inhibitor groups (p < .001, respectively). CONCLUSION lncRNA FEZF1-AS1 might be an oncological role in cervical cancer; lncRNA FEZF1-AS1 knockdown had antitumor effects with miR-1254 activating in cervical cancer by in vitro study.
Collapse
Affiliation(s)
- Miao Liang
- Department of gynaecology and obstetricsChongqing General HospitalUniversity of Chinese Academy of SciencesChongqingChina
| | - Yongkang Li
- Department of gynaecology and obstetricsChongqing General HospitalUniversity of Chinese Academy of SciencesChongqingChina
| | - Tingting Dai
- Department of gynaecology and obstetricsChongqing General HospitalUniversity of Chinese Academy of SciencesChongqingChina
| | - Cheng Chen
- Department of gynaecology and obstetricsChongqing General HospitalUniversity of Chinese Academy of SciencesChongqingChina
| |
Collapse
|
36
|
Lamsisi M, Wakrim L, Bouziyane A, Benhessou M, Oudghiri M, Laraqui A, Elkarroumi M, Ennachit M, El Mzibri M, Ennaji MM. The Biological Significance of Long noncoding RNAs Dysregulation and their Mechanism of Regulating Signaling Pathways in Cervical Cancer. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2021; 10:75-101. [PMID: 34703793 PMCID: PMC8496250 DOI: 10.22088/ijmcm.bums.10.2.75] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 08/01/2021] [Indexed: 12/19/2022]
Abstract
Despite the remarkable decrease in cervical cancer incidence due to the availability of the HPV vaccine and implementation of screening programs for early detection in developed countries, this cancer remains a major health problem globally, especially in developing countries where most of the cases and mortality occur. Therefore, more understanding of molecular mechanisms of cervical cancer development might lead to the discovery of more effective diagnosis and treatment options. Research on long noncoding RNAs (lncRNAs) demonstrates the important roles of these molecules in many physiological processes and diseases, especially cancer. In the present review, we discussed the significance of lncRNAs altered expression in cervical cancer, highlighting their roles in regulating highly conserved signaling pathways, such as mitogen-activated protein kinase (MAPK), Wnt/β-catenin, Notch, and phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) pathways and their association with the progression of cervical cancer in order to bring more insight and understanding of this disease and their potential implications in cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Maryame Lamsisi
- Team of Virology, Oncology and Medical Biotechnologies, Laboratory of Virology, Microbiology, Quality, and Biotechnologies/ ETB. Faculty of Science and Techniques Mohammedia, Hassan II University of Casablanca, Morocco.
| | - Lahcen Wakrim
- Laboratory of Virology, Pasteur Institute of Morocco. Casablanca, Morocco.
| | - Amal Bouziyane
- Team of Virology, Oncology and Medical Biotechnologies, Laboratory of Virology, Microbiology, Quality, and Biotechnologies/ ETB. Faculty of Science and Techniques Mohammedia, Hassan II University of Casablanca, Morocco.
- University Mohammed VI of Health Science, Casablanca, Morocco.
| | - Mustapha Benhessou
- Team of Virology, Oncology and Medical Biotechnologies, Laboratory of Virology, Microbiology, Quality, and Biotechnologies/ ETB. Faculty of Science and Techniques Mohammedia, Hassan II University of Casablanca, Morocco.
- School of Medicine and Pharmacy, University Hassan II of Casablanca, Morocco.
| | - Mounia Oudghiri
- Immunology and Biodiversity laboratory, Faculty of Sciences Ain chock, Hassan II University of Casablanca, Morocco.
| | - Abdelilah Laraqui
- Research and Biosafety Laboratory, Mohammed V Military Hospital, University Mohammed V of Rabat, Morocco.
| | - Mohamed Elkarroumi
- School of Medicine and Pharmacy, University Hassan II of Casablanca, Morocco.
| | - Mohammed Ennachit
- School of Medicine and Pharmacy, University Hassan II of Casablanca, Morocco.
| | | | - Moulay Mustapha Ennaji
- Corresponding author: Faculty of Science and Techniques Mohammedia, University Hassan II of Casablanca, Morocco. E-mail:
| |
Collapse
|
37
|
Wei M, Chen Y, Du W. LncRNA LINC00858 enhances cervical cancer cell growth through miR-3064-5p/ VMA21 axis. Cancer Biomark 2021; 32:479-489. [PMID: 34275889 DOI: 10.3233/cbm-200033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Cervical cancer (CC) is the most common form of gynecological malignancy. Long intergenic non-protein coding RNA 858 (LINC00858) has been identified to participate in multiple cancers. However, the role and mechanism of LINC00858 in CC cells are still elusive. AIM The aim of this study is to explore the biological functions and mechanisms of LINC00858 in CC cells. METHODS RT-qPCR analysis was used to examine the expression of LINC00858 in CC cells. EdU and colony formation assay were utilized to assess cell proliferation. TUNEL assay and flow cytometry assay were conducted to assess cell apoptosis. The mechanism regarding LINC00858 was certified through RNA pull down, RIP and luciferase reporter assays. RESULTS The up-regulated LINC00858 was detected in CC cells. Reduction of LINC00858 effectively subdued CC cells proliferation and stimulated cell apoptosis. LINC00858 was determined to bind with miR-3064-5p and up-regulate VMA21 in CC cells. In rescue assays, miR-3064-5p down-regulation and VMA21 up-regulation were able to counteract the effect caused by LINC00858 decrease on CC cell proliferation and apoptosis. CONCLUSION LINC00858 enhances cell proliferation, while restraining cell apoptosis in CC through targeting miR-3064-5p/VMA21 axis, implying that LINC00858 may serve as a promising therapeutic target for CC.
Collapse
Affiliation(s)
- Min Wei
- Department of Gynecology, 1st Affiliated Hospital, Soochow University, Gusu District, Suzhou, Jiangsu, China.,Department of Gynecology, The Affiliated Hospital of Xuzhou Medical University, Quanshan District, Xuzhou, Jiangsu, China
| | - Youguo Chen
- Department of Gynecology, 1st Affiliated Hospital, Soochow University, Gusu District, Suzhou, Jiangsu, China
| | - Wensheng Du
- Department of Gynecology, The Affiliated Hospital of Xuzhou Medical University, Quanshan District, Xuzhou, Jiangsu, China
| |
Collapse
|
38
|
Shayimu P, Yusufu A, Rehemutula A, Redati D, Jiapaer R, Tuerdi R. MTBP promoted the proliferation, migration and invasion of colon cancer cells by activating the expression of ZEB2. Anim Cells Syst (Seoul) 2021; 25:152-160. [PMID: 34262658 PMCID: PMC8253212 DOI: 10.1080/19768354.2021.1938218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Colon cancer is a malignant tumor that seriously affects human health. Recently, studies revealed that the expression of MTBP enhanced the proliferation and metastasis of many types of cancer cells. And the data also showed that MTBP has the potential to regulate the expression of ZEB2. However, it is unclear whether MTBP can affect the proliferation, migration and invasion of colon cancer cells by modulating the expression of ZEB2. In this study, we established the MTBP overexpression and knockdown colon cancer cells with the transfection. Next, CCK-8 and transwell assays were carried out to determine the changes of the proliferation and invasion of colon cancer cells, respectively. After that, we overexpressed the ZEB2 in these MTBP knockdown colon cancer cells. Finally, the invasion and migration of these cells were detected with the same methods. We revealed that overexpression of MTBP enhanced the proliferation and invasion of colon cancer cells. Moreover, suppression of MTBP repressed the proliferation, migration and invasion of colon cancer cells. Furthermore, MTBP promoted the expression of ZEB2. The overexpression of ZEB2 abolished the MTBP knockdown induced inhibition of the migration and invasion of colon cancer cells. These results implied that MTBP enhanced the proliferation, migration and invasion of colon cancer cells by activating the expression of ZEB2.
Collapse
Affiliation(s)
- Paerhati Shayimu
- Department of Gastrointestinal Surgery, Cancer Hospital of Xinjiang Medical University, Urumqi, People's Republic of China
| | - Aikeremu Yusufu
- Department of Gastrointestinal Surgery, Cancer Hospital of Xinjiang Medical University, Urumqi, People's Republic of China
| | - Aizimaiti Rehemutula
- Department of Gastrointestinal Surgery, Cancer Hospital of Xinjiang Medical University, Urumqi, People's Republic of China
| | - Darebai Redati
- B-Ultrasound Room, Cancer Hospital of Xinjiang Medical University, Urumqi, People's Republic of China
| | - Rexida Jiapaer
- Department of Gastrointestinal Surgery, Cancer Hospital of Xinjiang Medical University, Urumqi, People's Republic of China
| | - Rousidan Tuerdi
- Central Laboratory, Xinjiang Medical University, Urumqi, People's Republic of China
| |
Collapse
|
39
|
Competing Endogenous RNAs in Cervical Carcinogenesis: A New Layer of Complexity. Processes (Basel) 2021. [DOI: 10.3390/pr9060991] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
MicroRNAs (miRNAs) regulate gene expression by binding to complementary sequences within target mRNAs. Apart from working ‘solo’, miRNAs may interact in important molecular networks such as competing endogenous RNA (ceRNA) axes. By competing for a limited pool of miRNAs, transcripts such as long noncoding RNAs (lncRNAs) and mRNAs can regulate each other, fine-tuning gene expression. Several ceRNA networks led by different lncRNAs—described here as lncRNA-mediated ceRNAs—seem to play essential roles in cervical cancer (CC). By conducting an extensive search, we summarized networks involved in CC, highlighting the major impacts of such dynamic molecular changes over multiple cellular processes. Through the sponging of distinct miRNAs, some lncRNAs as HOTAIR, MALAT1, NEAT1, OIP5-AS1, and XIST trigger crucial molecular changes, ultimately increasing cell proliferation, migration, invasion, and inhibiting apoptosis. Likewise, several lncRNAs seem to be a sponge for important tumor-suppressive miRNAs (as miR-140-5p, miR-143-3p, miR-148a-3p, and miR-206), impairing such molecules from exerting a negative post-transcriptional regulation over target mRNAs. Curiously, some of the involved mRNAs code for important proteins such as PTEN, ROCK1, and MAPK1, known to modulate cell growth, proliferation, apoptosis, and adhesion in CC. Overall, we highlight important lncRNA-mediated functional interactions occurring in cervical cells and their closely related impact on cervical carcinogenesis.
Collapse
|
40
|
Liu Y, Qiu S, Zheng X, Qiu Y, Yao S, Ge Y, Zhou C. LINC00662 modulates cervical cancer cell proliferation, invasion, and apoptosis via sponging miR-103a-3p and upregulating PDK4. Mol Carcinog 2021; 60:365-376. [PMID: 33819358 DOI: 10.1002/mc.23294] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 02/07/2021] [Accepted: 02/22/2021] [Indexed: 11/11/2022]
Abstract
Cervical cancer (CC) is one of the most common cancers among women with high recurrence rates all over the world. Recently, the molecular mechanism of CC has been gradually uncovered in accumulating reports. This study aimed to investigate the function and upstream regulation mechanism of pyruvate dehydrogenase kinase 4 (PDK4) in CC cells, which was verified as an oncogene in several cancers. Through RT-qPCR assay, we discovered that PDK4 was highly expressed in CC cells. Then, it was demonstrated in function assays that PDK4 facilitated CC cell proliferation and invasion, but inhibited CC cell apoptosis. Next, we sought to determine the upstream genes of PDK4, and miR-103a-3p was identified to target PDK4. Then, through bioinformatics tools and a range of mechanism assays, long intergenic non-protein coding RNA 662 (LINC00662) was verified as the sponge of miR-103a-3p. Moreover, LINC00662 positively modulated PDK4 expression via competitively binding to miR-103a-3p in CC cells. Subsequently, rescue assays demonstrated that LINC00662 accelerated CC cell proliferation and inhibited cell apoptosis through upregulating PDK4. Furthermore, forkhead box A1 (FOXA1) was verified to activate transcription of both LINC00662 and PDK4. Taken together, our study revealed a novel ceRNA pattern of LINC00662/miR-103a-3p/PDK4 with FOXA1 as a transcription factor of LINC00662 and PDK4 in CC cells.
Collapse
Affiliation(s)
- Yongli Liu
- Gynecology Department, Xuzhou First People's Hospital, Xuzhou, Jiangsu, China
- Gynecology Department, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Shuang Qiu
- Gynecology Department, Xuzhou First People's Hospital, Xuzhou, Jiangsu, China
- Gynecology Department, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiaoli Zheng
- Gynecology Department, Xuzhou First People's Hospital, Xuzhou, Jiangsu, China
- Gynecology Department, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yingying Qiu
- Gynecology Department, Xuzhou First People's Hospital, Xuzhou, Jiangsu, China
- Gynecology Department, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Shenghui Yao
- Gynecology Department, Xuzhou First People's Hospital, Xuzhou, Jiangsu, China
- Gynecology Department, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yan Ge
- Gynecology Department, Xuzhou First People's Hospital, Xuzhou, Jiangsu, China
- Gynecology Department, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Caixia Zhou
- Gynecology Department, Xuzhou First People's Hospital, Xuzhou, Jiangsu, China
- Gynecology Department, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
41
|
Huang SK, Ni RX, Wang WJ, Wang D, Zhao M, Lei CZ, Sun XJ, Huang CZ, Bai P, Che YQ, Xu JP. Overexpression of LINC00673 Promotes the Proliferation of Cervical Cancer Cells. Front Oncol 2021; 11:669739. [PMID: 34094965 PMCID: PMC8176101 DOI: 10.3389/fonc.2021.669739] [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: 02/19/2021] [Accepted: 04/30/2021] [Indexed: 12/29/2022] Open
Abstract
Objective To study the expression of LINC00673 in cervical cancer and cervical intraepithelial neoplasia (CIN) and to explore the role of LINC00673 in the development of cervical cancer. Methods The expression of LINC00673 in serum from cervical cancer patients, CIN patients, and healthy participants was detected by RT-qPCR. The function of LINC00673 in cervical cancer cells was analyzed using in vitro and in vivo experiments. Results Our results revealed that serum LINC00673 levels were highest in cervical cancer patients, followed by patients with CIN and healthy controls. In vitro experiments demonstrated that overexpression of LINC00673 enhanced the proliferation and cell cycle progression of HeLa and SiHa cells. In vivo experiments showed that the tumor weight and volume of nude mice subcutaneously injected with LINC00673-overexpressing HeLa cells were larger than those of nude mice injected with control cells (P < 0.05). Western blotting showed that cell cycle-related proteins cyclin A2 and cyclin E and interstitial-associated proteins Snail and N-cadherin were upregulated and p53 signaling pathway-related proteins were downregulated in LINC00673-overexpressing HeLa and SiHa cells. Conclusion LINC00673 plays an important role in the development of cervical cancer and may serve as a new therapeutic target for cervical cancer.
Collapse
Affiliation(s)
- Sheng-Kai Huang
- Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ruo-Xuan Ni
- Department of Etiology and Carcinogenesis, State Key Laboratory of Molecular Oncology, Beijing Key Laboratory for Carcinogenesis and Cancer Prevention, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wen-Jie Wang
- Department of Etiology and Carcinogenesis, State Key Laboratory of Molecular Oncology, Beijing Key Laboratory for Carcinogenesis and Cancer Prevention, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,The Center for Disease Control and Prevention of Huai Rou, Beijing, China
| | - Di Wang
- Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mei Zhao
- Department of Etiology and Carcinogenesis, State Key Laboratory of Molecular Oncology, Beijing Key Laboratory for Carcinogenesis and Cancer Prevention, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Cheng-Zhi Lei
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao-Jie Sun
- Department of Biochemistry, Qiqihar Medical University, Qiqihar, China
| | - Chang-Zhi Huang
- Department of Etiology and Carcinogenesis, State Key Laboratory of Molecular Oncology, Beijing Key Laboratory for Carcinogenesis and Cancer Prevention, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ping Bai
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi-Qun Che
- Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jian-Ping Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
42
|
Interplay between SOX9 transcription factor and microRNAs in cancer. Int J Biol Macromol 2021; 183:681-694. [PMID: 33957202 DOI: 10.1016/j.ijbiomac.2021.04.185] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/29/2021] [Accepted: 04/29/2021] [Indexed: 02/07/2023]
Abstract
SOX transcription factors are critical regulators of development, homeostasis and disease progression and their dysregulation is a common finding in various cancers. SOX9 belongs to SOXE family located on chromosome 17. MicroRNAs (miRNAs) possess the capacity of regulating different transcription factors in cancer cells by binding to 3'-UTR. Since miRNAs can affect differentiation, migration, proliferation and other physiological mechanisms, disturbances in their expression have been associated with cancer development. In this review, we evaluate the relationship between miRNAs and SOX9 in different cancers to reveal how this interaction can affect proliferation, metastasis and therapy response of cancer cells. The tumor-suppressor miRNAs can decrease the expression of SOX9 by binding to the 3'-UTR of mRNAs. Furthermore, the expression of downstream targets of SOX9, such as c-Myc, Wnt, PI3K/Akt can be affected by miRNAs. It is noteworthy that other non-coding RNAs including lncRNAs and circRNAs regulate miRNA/SOX9 expression to promote/inhibit cancer progression and malignancy. The pre-clinical findings can be applied as biomarkers for diagnosis and prognosis of cancer patients.
Collapse
|
43
|
Yang X, Du H, Bian W, Li Q, Sun H. FOXD3‑AS1/miR‑128‑3p/LIMK1 axis regulates cervical cancer progression. Oncol Rep 2021; 45:62. [PMID: 33760158 PMCID: PMC8020211 DOI: 10.3892/or.2021.8013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 02/05/2021] [Indexed: 02/07/2023] Open
Abstract
Long non‑coding RNA forkhead box D3 antisense RNA 1 (FOXD3‑AS1) functions as an oncogenic regulator in several types of cancer, including breast cancer, glioma and cervical cancer. However, the effects and mechanisms underlying FOXD3‑AS1 in cervical cancer (CC) are not completely understood. The present study aimed to investigate the biological functions and potential molecular mechanisms underlying FOXD3‑AS1 in CC progression. Reverse transcription‑quantitative PCR was performed to detect FOXD3‑AS1, microRNA (miR)‑128‑3p and LIM domain kinase 1 (LIMK1) expression levels in CC tissues and cells. Immunohistochemical staining and western blotting were conducted to assess LIMK1 protein expression levels in CC tissues and cells, respectively. Cell Counting Kit‑8 and BrdU assays were used to determine the role of FOXD3‑AS1 in regulating cell proliferation. CC cell migration and invasion were assessed by performing Transwell assays. Dual‑luciferase reporter assays were conducted to verify the binding between miR‑128‑3p and FOXD3‑AS1. FOXD3‑AS1 expression was significantly increased in CC tissues and cell lines compared with adjacent healthy tissues and normal cervical epithelial cells, respectively. High FOXD3‑AS1 expression was significantly associated with poor differentiation of tumor tissues, increased tumor size and positive lymph node metastasis. FOXD3‑AS1 overexpression significantly increased CC cell proliferation, migration and invasion compared with the negative control (NC) group, whereas FOXD3‑AS1 knockdown resulted in the opposite effects compared with the small interfering RNA‑NC group. Moreover, the results demonstrated that FOXD3‑AS1 targeted and negatively regulated miR‑128‑3p, which indirectly upregulated LIMK1 expression. Therefore, the present study demonstrated that FOXD3‑AS1 upregulated LIMK1 expression via competitively sponging miR‑128‑3p in CC cells, promoting CC progression.
Collapse
Affiliation(s)
- Xiufang Yang
- Department of Gynecology, Hengshui People's Hospital, Hengshui, Hebei 053000, P.R. China
| | - Huilan Du
- Department of Gynecology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050000, P.R. China
| | - Wenhui Bian
- Department of Gynecology, Chinese Medicine Hospital of Hebei, Shijiazhuang, Hebei 050000, P.R. China
| | - Qingxue Li
- Department of Gynecology, The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei 050000, P.R. China
| | - Hairu Sun
- Department of Gynecology, Hengshui People's Hospital, Hengshui, Hebei 053000, P.R. China
| |
Collapse
|
44
|
Cheng T, Huang S. Roles of Non-Coding RNAs in Cervical Cancer Metastasis. Front Oncol 2021; 11:646192. [PMID: 33777808 PMCID: PMC7990873 DOI: 10.3389/fonc.2021.646192] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 02/03/2021] [Indexed: 12/24/2022] Open
Abstract
Metastasis remains to be a huge challenge in cancer therapy. The mechanism underlying cervical cancer metastasis is not well understood and needs to be elucidated. Recent studies have highlighted the diverse roles of non-coding RNAs in cancer progression and metastasis. Increasing numbers of miRNAs, lncRNAs and circRNAs are found to be dysregulated in cervical cancer, associated with metastasis. They have been shown to regulate metastasis through regulating metastasis-related genes, epithelial-mesenchymal transition, signaling pathways and interactions with tumor microenvironment. Moreover, miRNAs can interact with lncRNAs and circRNAs respectively during this complex process. Herein, we review literatures up to date involving non-coding RNAs in cervical cancer metastasis, mainly focus on the underlying mechanisms and highlight the interaction network between miRNAs and lncRNAs, as well as circRNAs. Finally, we discuss the therapeutic prospects.
Collapse
Affiliation(s)
- Tanchun Cheng
- Department of Obstetrics and Gynecology, Affiliated Haikou Hospital, Xiangya Medical College of Central South University, Haikou, China
| | - Shouguo Huang
- Department of Obstetrics and Gynecology, Affiliated Haikou Hospital, Xiangya Medical College of Central South University, Haikou, China
| |
Collapse
|
45
|
Chen Y, Chen D, Wang J, Zhang Y, Zhang J, Chen B, Chen Y, Zhang Y, Ma C. Dysregulated LncRNAs Act as Competitive Endogenous RNAs and Are Associated With Cervical Cancer Development in UYGHUR Women. Technol Cancer Res Treat 2021; 20:1533033821989711. [PMID: 33596784 PMCID: PMC7897819 DOI: 10.1177/1533033821989711] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cervical cancer is one of the most malignant tumors in women, particularly those in rural and remote areas. Its underlying molecular mechanisms, including the functions of non-coding RNA (ncRNAs), require more extensive investigation. In this study, high throughput transcriptome sequencing (RNA-seq) was used to identify differentially expressed lncRNAs and mRNAs in normal, cervical intraepithelial neoplasia and cervical cancer tissues from Uyghur women in western China. Dysregulated lncRNAs were found to extensively participate in cervical cancer development, including viral carcinogenesis, cell cycle and cytokine-cytokine receptor signaling. Two miRNA-host lncRNAs, LINC00925 and MIR155HG, showed elevated expression in cervical cancer samples, but prolonged the survival time of cervical cancer patients. The 2 mature miRNAs of the above 2 lncRNAs, miR-9 and miR-155, also showed similar features in cervical cancer. In addition, we identified 545 lncRNAs with potential functions in regulating these 2 miRNAs as competing endogenous RNAs (ceRNAs). In summary, our study demonstrated the dysregulated lncRNAs/miRNAs, particularly LINC00925/miR-9 and MIR155HG/miR-155, regulate the development of cervical cancer by forming a interaction network with mRNAs, highlighting the importance of elucidating the underlying mechanisms of ncRNAs in cervical cancer development.
Collapse
Affiliation(s)
- Yanxia Chen
- Department of Gynaecology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Dong Chen
- Center for Genome Analysis, ABLife Inc., Wuhan, Hubei, China.,ABLife BioBigData Institute, Wuhan, Hubei, China
| | - Jing Wang
- Department of Gynaecology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Yu Zhang
- Center for Genome Analysis, ABLife Inc., Wuhan, Hubei, China
| | - Ji Zhang
- Shanghai Ruijin Hospital, Shanghai, China
| | - Bing Chen
- Department of Gynaecology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Yaru Chen
- Department of Gynaecology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Yi Zhang
- Center for Genome Analysis, ABLife Inc., Wuhan, Hubei, China.,ABLife BioBigData Institute, Wuhan, Hubei, China
| | - Cailing Ma
- Department of Gynaecology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China.,State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| |
Collapse
|
46
|
Ming H, Li B, Zhou L, Goel A, Huang C. Long non-coding RNAs and cancer metastasis: Molecular basis and therapeutic implications. Biochim Biophys Acta Rev Cancer 2021; 1875:188519. [PMID: 33548345 DOI: 10.1016/j.bbcan.2021.188519] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 01/29/2021] [Accepted: 01/29/2021] [Indexed: 02/08/2023]
Abstract
Cancer metastasis, defined by the epithelial to mesenchymal transition (EMT) of tumor cells, disseminates from the primary site to progressively colonize in distant tissues, and accounts for most cancer-associated deaths. However, studies on the molecular basis of cancer metastasis are still in their infancy. Besides genetic mutations, accumulating evidence indicates that epigenetic alterations also contribute in a major way to the refractory nature of cancer metastasis. Considered as one of the essential epigenetic regulators, long non-coding RNAs (lncRNAs) can act as signaling regulators, decoys, guides and scaffolds, modulating key molecules in every step of cancer metastasis including dissemination of carcinoma cells, intravascular transit, and metastatic colonization. Although still having limited clinical application, it is encouraging to witness that several lncRNAs, including CCAT1 and HOTAIR, are under clinical evaluation as potential biomarkers for cancer staging and assessment of metastatic potential. In this review, we focus on the molecular mechanisms underlying lncRNAs in the regulation of cancer metastasis and discuss their clinical potential as novel therapeutic targets as well as their diagnostic and prognostic significance for cancer treatment. Gaining clear insights into the detailed molecular basis underlying lncRNA-modulated cancer metastasis may provide previously unrecognized diagnostic and therapeutic strategies for metastatic patients.
Collapse
Affiliation(s)
- Hui Ming
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Bowen Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Li Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, 1218 S. Fifth Avenue, Suite 2226, Biomedical Research Center, Monrovia, CA 91016, USA.
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China; School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
47
|
Shi H, Xie J, Wang K, Li W, Yin L, Wang G, Wu Z, Ni J, Mao W, Guo C, Peng B. LINC01451 drives epithelial-mesenchymal transition and progression in bladder cancer cells via LIN28/TGF-β/Smad pathway. Cell Signal 2021; 81:109932. [PMID: 33516780 DOI: 10.1016/j.cellsig.2021.109932] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 01/04/2021] [Accepted: 01/20/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND The pathogenesis of bladder cancer (BLCa) is still unclear. Long non-coding RNAs (lncRNAs) participate in diverse biological processes across every branch of life, especially in cancer. Dysregulated lncRNAs in BLCa and their biological significance require further investigations. METHODS Herein, a differential expression profile of lncRNAs in BLCa was conducted by microarray data. The expression level of lncRNA LINC01451 in 70 pairs of BLCa tissue samples and different BLCa cell lines were analyzed via real-time quantitative PCR. The CRISPR-CAS9 technique was employed to establish the LINC01451 stably transfected cell lines. Loss-of-function, as well as gain-of-function assays were carried out to evaluate the effects of LINC01451 on cell proliferation, migration, and invasion. Patient-derived xenograft (PDX) mouse models were adopted in the in vivo experiments. Western blot, biotinylated RNA probe pull-down assay, fluorescence in situ hybridization, and immunohistochemistry were utilized to assess the underlying molecular mechanisms of LINC01451 in BLCa. RESULTS LINC01451 was identified a novel functional lncRNA, whose expression level in BLCa tissues was significantly higher compared with the normal tissues. Furthermore, it was found that LINC01451 directly docked LIN28A and LIN28B, and promoted the proliferation, invasion, and metastasis of BLCa. Mechanistically, LINC0145 was shown to depend on LIN28A and LIN28B, facilitated epithelial-mesenchymal transition (EMT) through activating the TGF-β/Smad signaling pathway, which subsequently aggravated BLCa progression. CONCLUSIONS We demonstrates that LINC01451 drives EMT-induced BLCa progression by activating the LIN28/TGF-β/Smad signaling pathway. Promisingly, LINC01451 acts as a prognostic biomarker and a novel therapeutic target for BLCa.
Collapse
Affiliation(s)
- Heng Shi
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China; Department of Urology, Nantong Tongzhou People's Hospital, Nantong 226000, China
| | - Jinbo Xie
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Keyi Wang
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Weiyi Li
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Lei Yin
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Guangchun Wang
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Zonglin Wu
- Department of Urology, Shidong Hospital of Shanghai, Shanghai 200438, China
| | - Jinliang Ni
- Shanghai Clinical College, Anhui Medical University, Shanghai 200072, China
| | - Weipu Mao
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing 210009, China
| | - Changcheng Guo
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China.
| | - Bo Peng
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China.
| |
Collapse
|
48
|
Xu Z, Zhang D, Zhang Z, Luo W, Shi R, Yao J, Li D, Wang L, Liao B. MicroRNA-505, Suppressed by Oncogenic Long Non-coding RNA LINC01448, Acts as a Novel Suppressor of Glycolysis and Tumor Progression Through Inhibiting HK2 Expression in Pancreatic Cancer. Front Cell Dev Biol 2021; 8:625056. [PMID: 33520999 PMCID: PMC7843961 DOI: 10.3389/fcell.2020.625056] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 12/21/2020] [Indexed: 01/10/2023] Open
Abstract
Background: MicroRNAs (miRNAs) and long non-coding RNAs (lncRNAs) play vital regulatory roles in pancreatic cancer (PC) initiation and progression. We aimed to explore the biological functions and underlying mechanisms of miR-505-3p (miR-505) in PC. Methods: We first screened miRNA expression profiles using microarray in PC tissues and normal tissues, and then studied the function and underlying mechanism of miR-505. Moreover, we evaluated the regulatory effect of lncRNA LINC01448 on miR-505. Results: We demonstrated miR-505 that was significantly downregulated in PC tissues. We further revealed that miR-505 significantly inhibited cell proliferation, invasion, sphere formation, glucose consumption, and lactate production by targeting HK2. In addition, overexpression of miR-505 led to tumor growth inhibition in vivo, demonstrating that it acts as a tumor suppressor in PC. LINC01448 was identified as an oncogenic lncRNA that could reduce miR-505 expression. Subsequent studies confirmed that LINC01448 enhanced cell proliferation, invasion, sphere formation, glucose consumption, and lactate production by regulating the miR-505/HK2 pathway. Conclusions: These findings demonstrated that miR-505, suppressed by LINC01448, could function as a key tumor suppressor by targeting HK2 in PC, elucidating an important role of the LINC01448/miR-505/HK2 pathway in regulating PC glycolysis and progression.
Collapse
Affiliation(s)
- Zhenglei Xu
- The Second Clinical Medical College, Jinan University, Department of Gastroenterology, Shenzhen People's Hospital, Shenzhen, China
| | - Dingguo Zhang
- The Second Clinical Medical College, Jinan University, Department of Gastroenterology, Shenzhen People's Hospital, Shenzhen, China
| | - Zhuliang Zhang
- The Second Clinical Medical College, Jinan University, Department of Gastroenterology, Shenzhen People's Hospital, Shenzhen, China
| | - Weixiang Luo
- Nursing Department, Shenzhen People's Hospital, The Second Affiliated Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Ruiyue Shi
- The Second Clinical Medical College, Jinan University, Department of Gastroenterology, Shenzhen People's Hospital, Shenzhen, China
| | - Jun Yao
- The Second Clinical Medical College, Jinan University, Department of Gastroenterology, Shenzhen People's Hospital, Shenzhen, China
| | - Defeng Li
- The Second Clinical Medical College, Jinan University, Department of Gastroenterology, Shenzhen People's Hospital, Shenzhen, China
| | - Lisheng Wang
- The Second Clinical Medical College, Jinan University, Department of Gastroenterology, Shenzhen People's Hospital, Shenzhen, China
| | - Bihong Liao
- Department of Cardiology, Shenzhen People's Hospital, The Second Affiliated Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
49
|
Ding Y, Yuan X, Gu W. Circular RNA RBM33 contributes to cervical cancer progression via modulation of the miR-758-3p/PUM2 axis. J Mol Histol 2021; 52:173-185. [PMID: 33398465 DOI: 10.1007/s10735-020-09933-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 11/24/2020] [Indexed: 02/01/2023]
Abstract
Cervical cancer (CC) is a gynecological malignant tumor. Circular RNA (hsa_circ_0001772) (circRBM33) is implicated in the tumorigenesis of cancers. Nevertheless, the role of circRBM33 in CC is indistinct. Quantitative real-time polymerase chain reaction (qRT-PCR) was employed to evaluate the levels of circRBM33, miR-758-3p, and pumilio RNA binding family member 2 (PUM2) mRNA in tissue samples and cells. Cell proliferation, apoptosis, migration, invasion, and glycolysis were assessed using 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay, flow cytometry assay, transwell assay, or special commercial kits. Relative protein levels were examined via western blotting. The targeting relationship between circRBM33 or PUM2 and miR-758-3p was verified via dual-luciferase reporter or RNA pull-down assays. The role of circRBM33 was confirmed via tumor formation experiments. CircRPPH1 and PUM2 were upregulated while miR-758-3p was downregulated in CC tissues and cells. Functionally, circRBM33 knockdown constrained tumor growth in vivo and cured CC cell proliferation, migration, invasion, glycolysis, and fostered CC cell apoptosis in in vitro. Mechanistically, circRBM33 sponged miR-758-3p to modulate PUM2 expression. MiR-758-3p inhibitor neutralized circRBM33 silencing-mediated effects on the malignant behaviors of CC cells. PUM2 elevation overturned the suppressive influence of miR-758-3p upregulation on the malignant behaviors of CC cells. CircRBM33 fostered CC advancement via absorbing miR-758-3p and upregulating PUM2, indicating that circRBM33 was a possible target for CC treatment.
Collapse
Affiliation(s)
- Yi Ding
- Department of Obstetrics & Gynaecology, the Affiliated Jintan Hospital of Jiangsu University, Changzhou, 213200, People's Republic of China.
- Department of Obstetrics & Gynaecology, The Affiliated Jintan Hospital of Jiangsu University, No. 16, Nanmen Street, Jintan District, Changzhou City, Jiangsu Province, People's Republic of China.
| | - Xia Yuan
- Department of Obstetrics & Gynaecology, the Affiliated Jintan Hospital of Jiangsu University, Changzhou, 213200, People's Republic of China
| | - Wenwen Gu
- Department of Obstetrics & Gynaecology, the Affiliated Jintan Hospital of Jiangsu University, Changzhou, 213200, People's Republic of China
| |
Collapse
|
50
|
Circ_0001247 functions as a miR-1270 sponge to accelerate cervical cancer progression by up-regulating ZEB2 expression level. Biotechnol Lett 2021; 43:745-755. [PMID: 33386495 DOI: 10.1007/s10529-020-03059-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 12/10/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND There is increasing evidence that circular RNA (circRNA) disorders have an impact on the progression of various malignancies. The expression characteristics, function and underlying mechanism of circ_0001247 in cervical cancer (CC) have not been confirmed. METHODS GSE147483 datasets of circRNAs expression in CC cell line and normal cervical cell line were retrieved from GEO database, and the circRNA with significant difference was selected; circ_0001247, miR-1270, and Zinc finger E-box binding homeobox 2 (ZEB2) expressions in CC tissues and cell lines were analyzed by quantitative real-time polymerase chain reaction (qRT-PCR) assay; cell counting kit-8 (CCK-8) assay and BrdU assay were applied to monitor the proliferative ability of CC cells; Transwell assay was conducted to examine the migration and invasion of CC cells, and flow cytometry was used to evaluate the apoptosis; Western blot assay was adopted to detect ZEB2 protein expressions; dual-luciferase report gene assay was used to verify the targeting relationship between circ_0001247 and miR-1270, and miR-1270 and the 3'UTR of ZEB2. RESULTS Analysis of GSE147483 suggested that circ_0001247 could probably be an oncogenic circRNA in CC. Compared with that in adjacent tissues and normal cervical epithelial cells, circ_0001247 expression in CC tissues and cell lines was significantly increased; knocking down circ_0001247 expression could inhibit the proliferation and metastasis of CC cells, and promote apoptosis, while circ_0001247 overexpression worked oppositely; circ_0001247 sponged miR-1270 in CC cells; miR-1270 diminished the promoting effect of circ_0001247 by inactivating the ZEB2. CONCLUSION Circ_0001247 promotes progression of CC by sponging miR-1270 to upregulate ZEB2 expression level.
Collapse
|