1
|
Kwas K, Szubert M, Wilczyński JR. Latest Update on lncRNA in Epithelial Ovarian Cancer-A Scoping Review. Cells 2025; 14:555. [PMID: 40214508 PMCID: PMC11988607 DOI: 10.3390/cells14070555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/22/2025] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
Long noncoding RNAs (lncRNAs) are RNA molecules exceeding 200 nucleotides that do not encode proteins yet play critical roles in regulating gene expression at multiple levels, such as chromatin modification and transcription. These molecules are significantly engaged in cancer progression, development, metastasis, and chemoresistance. However, the function of lncRNAs in epithelial ovarian cancer (EOC) has not yet been thoroughly studied. EOC remains challenging due to its complex molecular pathogenesis, characterized by genetic and epigenetic alterations. Emerging evidence suggests that lncRNAs, such as XIST, H19, NEAT1, and MALAT1, are involved in EOC by modulating gene expression and signaling pathways, influencing processes like cell proliferation, invasion, migration, and chemoresistance. Despite extensive research, the precise mechanism of acting of lncRNAs in EOC pathogenesis and treatment resistance still needs to be fully understood, highlighting the need for further studies. This review aims to provide an updated overview of the current understanding of lncRNAs in EOC, emphasizing their potential as biomarkers and therapeutic targets. We point out the gaps in the knowledge regarding lncRNAs' influence on epithelial ovarian cancer (EOC), deliberating on new possible research areas.
Collapse
Affiliation(s)
- Katarzyna Kwas
- Department of Surgical and Oncologic Gynaecology, 1st Department of Gynaecology and Obstetrics, Medical University of Lodz, 90-136 Łódź, Poland; (M.S.); (J.R.W.)
| | | | | |
Collapse
|
2
|
Yang J. Unveiling the multifaceted roles of long non-coding RNA CTBP1-DT in human diseases: Special attention to its microprotein-encoding potential. Pathol Res Pract 2025; 268:155870. [PMID: 40020329 DOI: 10.1016/j.prp.2025.155870] [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: 11/18/2024] [Revised: 01/30/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
C-terminal binding protein 1 divergent transcript (CTBP1-DT) is a novel long non-coding RNA (lncRNA) located on human chromosome 4p16.3. Numerous studies have shown that CTBP1-DT plays a critical regulatory role in various human malignancies and non-malignant diseases. In several cancers, the expression of CTBP1-DT is upregulated, closely associated with the risk of 12 types of cancer, and strongly correlated with the clinical pathological features and poor prognosis of 10 of these cancers. Mechanistically, CTBP1-DT is stimulated by the transcription factors ETV5 and Sp1, or methylated by YTHDC1. By competitively inhibiting 12 microRNAs, it activates 3 signaling pathways that influence malignant behaviors of tumor cells, including proliferation, apoptosis, cell cycle arrest, migration, invasion, immune evasion, and chemoresistance. Importantly, it also encodes the microprotein DNA damage up-regulated protein (DDUP), which mediates cisplatin resistance through sustained response to DNA damage signals. Furthermore, CTBP1-DT has been implicated in the progression of non-malignant diseases such as diabetes and related conditions, cardiovascular diseases, and osteoarthritis. This review summarizes the latest research on the RNA and protein functions of CTBP1-DT in human diseases, outlines various molecular regulatory networks centered around CTBP1-DT, and discusses the opportunities and challenges of its clinical applications.
Collapse
Affiliation(s)
- Jingjie Yang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Cancer Immunology and Biotherapy, Tianjin 300060, China.
| |
Collapse
|
3
|
Soltani Khaboushan A, Salimian SN, Mehraban S, Bahramy A, Zafari N, Kajbafzadeh AM, Johnson J, Majidi Zolbin M. Prognostic significance of non-coding RNAs related to the tumorigenic epithelial-mesenchymal transition (EMT) process among ovarian cancer patients: A systematic review and meta-analysis. Heliyon 2024; 10:e35202. [PMID: 39253159 PMCID: PMC11382180 DOI: 10.1016/j.heliyon.2024.e35202] [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: 01/13/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 09/11/2024] Open
Abstract
Introduction Ovarian cancer is the seventh most prevalent cancer among women. It has high mortality and morbidity and imposes a great burden on healthcare systems worldwide. Unraveling the mechanisms behind the Epithelial-Mesenchymal Transition and finding a panel for predicting the prognosis of the disease may help find the appropriate treatment approaches for the management of the disease. The overarching aim of this systematic review was to define a panel of different types of EMT-associated non-coding RNAs (ncRNAs) with significant prognostic value in all types of ovarian cancers. Methods We searched PubMed, Web of Science, Scopus, and Embase till Jun 2024 to retrieve relevant papers. Two independent reviewers screened papers, and discrepancies were resolved by consensus. Publications related to the dysregulation of different types of ncRNAs, including microRNAs, lncRNAs, and circRNAs, only in patients with ovarian cancer were included. The participation of ncRNAs in epithelial-mesenchymal transformation should be assessed via methods evaluating different EMT-related proteins. To assess the quality and risk of bias for the included case-control and cohort studies, refined Newcastle-Ottawa Scale (NOS) and Quadas-2 were recruited. A bivariate meta-analysis was performed to analyze extracted data. Results A total of 37 studies with overall 42 non-coding RNAs (15 microRNA, 24 long non-coding RNAs, and 3 circular RNAs) were entered into the analysis. Overall diagnostic odds ratio for ncRNAs in lymph node metastasis, distant metastasis, TNM stage, and clinical stage were 4.19, 3.80, 6.52, and 3.97, respectively. Also, a hazard ratio of 1.39 (P = 0.32) for overall survival was observed. Bioinformatic analyses on the Pan-cancer database demonstrated a significant correlation between low expression of miRNA and high expression of lncRNAs with poor prognosis of ovarian cancer. Conclusion Based on the results, the defined panel of ncRNAs can properly predict prognostic factors related to EMT in ovarian cancer without involving potentially invasive methods.
Collapse
Affiliation(s)
- Alireza Soltani Khaboushan
- Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyedeh Nazanin Salimian
- Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Saghar Mehraban
- Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Afshin Bahramy
- Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Narges Zafari
- Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Abdol-Mohammad Kajbafzadeh
- Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Joshua Johnson
- Division of Reproductive Sciences, Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Colorado-Anschutz Medical Campus, Aurora, CO, USA
| | - Masoumeh Majidi Zolbin
- Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Li R, Liu Y, Liu J, Chen B, Ji Z, Xu A, Zhang T. CCL2 regulated by the CTBP1-AS2/miR-335-5p axis promotes hemangioma progression and angiogenesis. Immunopharmacol Immunotoxicol 2024; 46:385-394. [PMID: 38622049 DOI: 10.1080/08923973.2024.2330651] [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: 10/21/2022] [Accepted: 03/09/2024] [Indexed: 04/17/2024]
Abstract
CONTEXT Hemangioma (HA) is a benign vascular neoplasm that can lead to permanent scarring. C-C motif chemokine ligand 2 (CCL2) plays a crucial role in facilitating growth and angiogenesis during HA progression. However, the mechanism regulating CCL2 in HA remains poorly elucidated. OBJECTIVE To elucidate the mechanism regulating CCL2 in HA. METHODS Quantitative real-time polymerase chain reaction (RT-qPCR) was employed to determine the expression levels of CCL2, long noncoding RNA (lncRNA) CTBP1 divergent transcript (CTBP1-AS2), and microRNAs (miRNAs). Proliferation, migration, invasion, and angiogenic abilities of human HA endothelial cells (HemECs) were assessed using cell counting kit-8 (CCK-8), colony formation, flow cytometry, transwell, and tube formation assays. Bioinformatics analysis, RNA pull-down, and luciferase reporter assays were conducted to investigate whether CCL2 targets miR-335-5p. Additionally, rescue experiments were performed in this study. RESULTS CCL2 expression was markedly upregulated in HemECs. CCL2 promoted HA cell proliferation, migration, invasion, and angiogenesis while inhibiting apoptosis. CCL2 was directly targeted by miR-335-5p. Additionally, we found that CTBP1-AS2 could function as a competing endogenous RNA (ceRNA) to sponge miR-335-5p, thereby upregulating CCL2. CONCLUSION Our findings suggest that targeting the CTBP1-AS2/miR-335-5p/CCL2 axis may hold promise as a therapeutic strategy for HA.
Collapse
Affiliation(s)
- Ruixuan Li
- Department of Cardiology, the Third Xiangya Hospital, Central South University, Changsha, PR China
| | - Ying Liu
- Department of Plastic Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Jianfeng Liu
- Department of Plastic Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Bo Chen
- Department of Plastic Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Zhongjie Ji
- Department of Plastic Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Aixia Xu
- Department of Endocrinology, Changsha Central Hospital, Changsha, PR China
| | - Tianhua Zhang
- Department of Vascular Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, PR China
| |
Collapse
|
5
|
Jin Y, Jiang D. GATA6-AS1 via Sponging miR-543 to Regulate PTEN/AKT Signaling Axis Suppresses Cell Proliferation and Migration in Gastric Cancer. Mediators Inflamm 2023; 2023:9340499. [PMID: 37273453 PMCID: PMC10238141 DOI: 10.1155/2023/9340499] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/16/2022] [Accepted: 04/05/2023] [Indexed: 06/06/2023] Open
Abstract
Gastric cancer (GC) is one of the most common and lethal cancers worldwide. In view of the prominent roles of long noncoding RNAs (lncRNAs) in cancers, we investigated the specific role and underlying mechanism of GATA binding protein 6 antisense RNA 1 (GATA6-AS1) in GC. Quantitative real-time polymerase chain reaction (qRT-PCR) detected GATA6-AS1 expression in GC cell lines. Functional assays were conducted to explore the role of GATA6-AS1 in GC. Furthermore, mechanism investigations were implemented to uncover the interaction among GATA6-AS1, microRNA-543 (miR-543), and phosphatase and tensin homolog (PTEN). In the present study, it was found that GATA6-AS1 expression is significantly downregulated in GC cell lines. Functionally, GATA6-AS1 markedly suppresses GC cell growth and migration in vitro and in vivo tumorigenesis. Besides tumor suppressor, GATA6-AS1 serves as a miR-543 sponge. Specifically speaking, GATA6-AS1 acts as a competing endogenous RNA (ceRNA) of miR-543 to upregulate the expression of PTEN, thus inactivating AKT signaling pathway to inhibit GC progression. In conclusion, this study has manifested that GATA6-AS1 inhibits GC cell proliferation and migration as a sponge of miR-543 by regulating PTEN/AKT signaling axis, offering new perspective into developing novel GC therapies.
Collapse
Affiliation(s)
- Yi Jin
- Department of Breast Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110001 Liaoning, China
| | - Daqing Jiang
- Department of Breast Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110001 Liaoning, China
| |
Collapse
|
6
|
Hamidi AA, Taghehchian N, Zangouei AS, Akhlaghipour I, Maharati A, Basirat Z, Moghbeli M. Molecular mechanisms of microRNA-216a during tumor progression. Cancer Cell Int 2023; 23:19. [PMID: 36740668 PMCID: PMC9899407 DOI: 10.1186/s12935-023-02865-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 02/02/2023] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs (miRNAs) as the members of non-coding RNAs family are involved in post-transcriptional regulation by translational inhibiting or mRNA degradation. They have a critical role in regulation of cell proliferation and migration. MiRNAs aberrations have been reported in various cancers. Considering the importance of these factors in regulation of cellular processes and their high stability in body fluids, these factors can be suggested as suitable non-invasive markers for the cancer diagnosis. MiR-216a deregulation has been frequently reported in different cancers. Therefore, in the present review we discussed the molecular mechanisms of the miR-216a during tumor progression. It has been reported that miR-216a mainly functioned as a tumor suppressor through the regulation of signaling pathways and transcription factors. This review paves the way to suggest the miR-216a as a probable therapeutic and diagnostic target in cancer patients.
Collapse
Affiliation(s)
- Amir Abbas Hamidi
- grid.411583.a0000 0001 2198 6209Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negin Taghehchian
- grid.411583.a0000 0001 2198 6209Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Sadra Zangouei
- grid.411583.a0000 0001 2198 6209Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Iman Akhlaghipour
- grid.411583.a0000 0001 2198 6209Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhosein Maharati
- grid.411583.a0000 0001 2198 6209Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Basirat
- grid.411583.a0000 0001 2198 6209Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- grid.411583.a0000 0001 2198 6209Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran ,grid.411583.a0000 0001 2198 6209Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
7
|
Wang G, Li C, Zhang X, Tang L, Li Y. Long non-coding PRNCR1 regulates the proliferation and apoptosis of synoviocytes in osteoarthritis by sponging miR-377-3p. J Orthop Surg Res 2022; 17:238. [PMID: 35422021 PMCID: PMC9008967 DOI: 10.1186/s13018-022-03035-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 02/23/2022] [Indexed: 11/21/2022] Open
Abstract
Background LncRNA PRNCR1 has been reported to be involved in LPS-induced inflammation, which contributes to osteoarthritis (OA). We predicted that miR-377-3p could bind to PRNCR1.MiR-377-3p can suppress OA development. We therefore analyzed the potential interaction between them in OA. Methods Expression of miR-377-3p and PRNCR1 in both OA (n = 40) and control (n = 40) samples were analyzed by RT-qPCR. MiR-377-3p or PRNCR1 were overexpressed in synoviocytes to explore their potential interaction. The subcellular location of PRNCR1 was analyzed by nuclear fractionation assay. The direct interaction between miR-377-3p and PRNCR1 was analyzed by RNA-pull down assay. The proliferation and apoptosis of synoviocytes were analyzed by BrdU and apoptosis assay, respectively. Results PRNCR1 was overexpressed in OA, while miR-377-3p was downexpressed in OA. PRNCR1 was detected in the cytoplasm and directly interacted with miR-377-3p. Interestingly, overexpression of PRNCR1 and miR-377-3p showed no regulatory role in each other’s expression. LPS treatment increased PRNCR1 expression and decreased miR-377-3p expression. PRNCR1 overexpression decreased LPS-induced synoviocyte proliferation and increased LPS-induced synoviocyte apoptosis. MiR-377-3p played opposite roles in cell proliferation and apoptosis. Moreover, PRNCR1 suppressed the role of miR-377-3p. Conclusions Therefore, PRNCR1 is was detected in cytoplasm and regulates synoviocyte proliferation and apoptosis in OA by sponging miR-377-3p. Supplementary Information The online version contains supplementary material available at 10.1186/s13018-022-03035-2.
Collapse
|
8
|
Feng S, Yin H, Zhang K, Shan M, Ji X, Luo S, Shen Y. Integrated clinical characteristics and omics analysis identifies a ferroptosis and iron-metabolism-related lncRNA signature for predicting prognosis and therapeutic responses in ovarian cancer. J Ovarian Res 2022; 15:10. [PMID: 35057848 PMCID: PMC8772079 DOI: 10.1186/s13048-022-00944-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 01/03/2022] [Indexed: 12/17/2022] Open
Abstract
Background Ferroptosis and iron-metabolism are regulated by Long non-coding RNAs (lncRNAs) in ovarian cancer (OC). Therefore, a comprehensive analysis of ferroptosis and iron-metabolism related lncRNAs (FIRLs) in OC is crucial for proposing therapeutic strategies and survival prediction. Methods In multi-omics data from OC patients, FIRLs were identified by calculating Pearson correlation coefficients with ferroptosis and iron-metabolism related genes (FIRGs). Cox-Lasso regression analysis was performed on the FIRLs to screen further the lncRNAs participating in FIRLs signature. In addition, all patients were divided into two robust risk subtypes using the FIRLs signature. Receiver operator characteristic (ROC) curve, Kaplan–Meier analysis, decision curve analysis (DCA), Cox regression analysis and calibration curve were used to confirm the clinical benefits of FIRLs signature. Meanwhile, two nomograms were constructed to facilitate clinical application. Moreover, the potential biological functions of the signature were investigated by genes function annotation. Finally, immune microenvironment, chemotherapeutic sensitivity, and the response of PARP inhibitors were compared in different risk groups using diversiform bioinformatics algorithms. Results The raw data were randomized into a training set (n = 264) and a testing set (n = 110). According to Pearson coefficients between FIRGs and lncRNAs, 1075 FIRLs were screened for univariate Cox regression analysis, and then LASSO regression analysis was used to construct 8-FIRLs signature. It is worth mentioning that a variety of analytical methods indicated excellent predictive performance for overall survival (OS) of FIRLs signature (p < 0.05). The multivariate Cox regression analysis showed that FIRLs signature was an independent prognostic factor for OS (p < 0.05). Moreover, significant differences in the abundance of immune cells, immune-related pathways, and drug response were excavated in different risk subtypes (p < 0.05). Conclusion The FIRLs signature can independently predict overall survival and therapeutic effect in OC patients. Supplementary Information The online version contains supplementary material available at 10.1186/s13048-022-00944-y.
Collapse
|
9
|
Zhang M, Ma S, Li X, Yu H, Tan Y, He J, Wei X, Ma J. Long non‑coding RNA CTBP1‑AS2 upregulates USP22 to promote pancreatic carcinoma progression by sponging miR‑141‑3p. Mol Med Rep 2022; 25:86. [PMID: 35039872 PMCID: PMC8809116 DOI: 10.3892/mmr.2022.12602] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 12/09/2021] [Indexed: 11/23/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) feature prominently in pancreatic carcinoma progression. The present study aimed to clarify the biological functions, clinical significance and underlying mechanism of lncRNA CTBP1 antisense RNA 2 (CTBP1-AS2) in pancreatic carcinoma. Reverse transcription-quantitative PCR was performed to assess the expression levels of CTBP1-AS2, microRNA (miR)-141-3p and ubiquitin-specific protease 22 (USP22) mRNA in pancreatic carcinoma tissues and cell lines. Western blotting was used to examine USP22 protein expression in pancreatic carcinoma cell lines. Loss-of-function experiments were used to analyze the regulatory effects of CTBP1-AS2 on proliferation, apoptosis, migration and invasion of pancreatic carcinoma cells. Dual-luciferase reporter assay was used to examine the binding relationship between CTBP1-AS2 and miR-141-3p, as well as between miR-141-3p and USP22. It was demonstrated that CTBP1-AS2 expression was markedly increased in pancreatic carcinoma tissues and cell lines. High CTBP1-AS2 expression was associated with advanced clinical stage and lymph node metastasis of patients. Functional experiments confirmed that knocking down CTBP1-AS2 significantly inhibited pancreatic carcinoma cell proliferation, migration and invasion, and promoted cell apoptosis. In terms of mechanism, it was found that CTBP1-AS2 adsorbed miR-141-3p as a molecular sponge to upregulate the expression level of USP22. In conclusion, lncRNA CTBP1-AS2 may be involved in pancreatic carcinoma progression by regulating miR-141-3p and USP22 expressions; in addition, CTBP1-AS2 may be a diagnostic biomarker and treatment target for pancreatic carcinoma.
Collapse
Affiliation(s)
- Mingliang Zhang
- Department of General Surgery, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Hui Autonomous Region, Yinchuan 750021, P.R. China
| | - Songbo Ma
- Department of General Surgery, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Hui Autonomous Region, Yinchuan 750021, P.R. China
| | - Xuzhao Li
- Department of General Surgery, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Hui Autonomous Region, Yinchuan 750021, P.R. China
| | - Henghai Yu
- Department of General Surgery, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Hui Autonomous Region, Yinchuan 750021, P.R. China
| | - Yizheng Tan
- Department of Hepatopancreatobiliary Surgery, The 2nd Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Jun He
- Department of Hepatopancreatobiliary Surgery, The 2nd Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Xiaoping Wei
- Department of General Surgery, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Hui Autonomous Region, Yinchuan 750021, P.R. China
| | - Junming Ma
- Department of General Surgery, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Hui Autonomous Region, Yinchuan 750021, P.R. China
| |
Collapse
|
10
|
ETV5-mediated upregulation of lncRNA CTBP1-DT as a ceRNA facilitates HGSOC progression by regulating miR-188-5p/MAP3K3 axis. Cell Death Dis 2021; 12:1146. [PMID: 34887384 PMCID: PMC8660778 DOI: 10.1038/s41419-021-04256-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 09/22/2021] [Accepted: 10/04/2021] [Indexed: 12/26/2022]
Abstract
High-grade serous ovarian cancer (HGSOC) is a common and lethal cancer of the female reproductive system. Long non-coding RNAs (lncRNAs) are aberrantly expressed in various cancers and play crucial roles in tumour progression. However, their function and molecular mechanism in HGSOC remain largely unknown. Based on public databases and bioinformatics analyses, the overexpression of lncRNA CTBP1-DT in HGSOC tissues was detected and validated in a cohort of HGSOC tissues. High expression of lncRNA CTBP1-DT was associated with poor prognosis and was an independent risk factor for survival. Overexpression of lncRNA CTBP1-DT promoted malignant biological behaviour of HGSOC cells, whereas its depletion induced growth arrest of HGSOC cells by vitro and in vivo assays. Mechanistically, lncRNA CTBP1-DT could competitively bind to miR-188-5p to protect MAP3K3 from degradation. Moreover, our results revealed that ETV5 could specifically interact with the promoter of lncRNA CTBP1-DT and activate its transcription. Collectively, these results reveal a novel ETV5/lncRNA CTBP1-DT/miR-188-5p/MAP3K3 pathway for HGSOC progression and suggest that lncRNA CTBP1-DT might be a potential biomarker and therapeutic target for HGSOC. ![]()
Collapse
|
11
|
Dou L, Zhang Y. miR-4461 Regulates the Proliferation and Metastasis of Ovarian Cancer Cells and Cisplatin Resistance. Front Oncol 2021; 11:614035. [PMID: 33767986 PMCID: PMC7985457 DOI: 10.3389/fonc.2021.614035] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 02/16/2021] [Indexed: 12/13/2022] Open
Abstract
microRNAs (miRNAs) are of great significance in cancer treatment, which may have a desirable result on the regulation of tumorigenesis, progression, recurrence, and chemo-resistance of ovarian cancer. However, the research on the further potential application of miR-4461 in ovarian cancer is little and limited. Therefore, the study in this paper focus on the investigation of the of miR-4461 in ovarian cancer progression and chemo-resistance. The phenomenon that the proliferation and metastasis of ovarian cancer cells can be promoted by miR4461 is revealed in functional assays. Through the bioinformatics and luciferase reporter analysis, the PTEN is validated to be the direct target of miR-4461 in ovarian. The association between the expression of miR-4461 and PTEN is negative in in human ovarian cancer tissues. The distinction of growth and metastasis capacity between miR-4461 knockdown ovarian cancer cells and control cells is partially abolished by si-PTEN. Moreover, it was found that cisplatin treatment has obvious effect on the miR-4461 knockdown ovarian cancer cells. In summary, the data given in this paper indicate that the miR-4461 can be regarded as a potential onco-miRNA in ovarian cancer by targeting PTEN.
Collapse
Affiliation(s)
- Lei Dou
- Department of Gynecology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yi Zhang
- Department of Gynecology, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
12
|
Ghafouri-Fard S, Abak A, Shoorei H, Mohaqiq M, Majidpoor J, Sayad A, Taheri M. Regulatory role of microRNAs on PTEN signaling. Biomed Pharmacother 2020; 133:110986. [PMID: 33166764 DOI: 10.1016/j.biopha.2020.110986] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 10/28/2020] [Accepted: 11/03/2020] [Indexed: 12/13/2022] Open
Abstract
Phosphatase and tensin homolog (PTEN) gene encodes a tumor suppressor protein which is altered in several malignancies. This protein is a negative regulator of the PI3K/AKT signaling. Several transcription factors regulate the expression of PTEN in positive or negative directions. Moreover, numerous microRNAs (miRNAs) have functional interactions with PTEN and inhibit its expression. Suppression of PTEN can attenuate the response of cancer cells to chemotherapeutic agents. Based on the critical role of this tumor suppressor gene, the identification of negative regulators of its expression has practical significance particularly in the prevention and management of cancer. Meanwhile, the interaction between miRNAs and PTEN has functional consequences in non-malignant disorders including myocardial infarction, osteoporosis, cerebral ischemic stroke, and recurrent abortion. In the present review, we describe the role of miRNAs in the regulation of expression and activity of PTEN.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atefe Abak
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Shoorei
- Department of Anatomical Sciences, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Mahdi Mohaqiq
- Wake Forest Institute for Regenerative Medicine, School of Medicine, Wake Forest University, Winston-Salem, NC, USA
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Arezou Sayad
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|