1
|
Li Z, Chen G, Tan G, Chen CYC. CoupleMDA: Metapath-Induced Structural-Semantic Coupling Network for miRNA-Disease Association Prediction. Int J Mol Sci 2025; 26:4948. [PMID: 40430088 PMCID: PMC12112494 DOI: 10.3390/ijms26104948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2025] [Revised: 05/18/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025] Open
Abstract
The prediction of microRNA-disease associations (MDAs) is crucial for understanding disease mechanisms and biomarker discovery. While graph neural networks have emerged as promising tools for MDA prediction, existing methods face critical limitations: (1) data leakage caused by improper use of Gaussian interaction profile (GIP) kernel similarity during feature construction, (2) self-validation loops in calculating miRNA functional similarity using known MDA data, and (3) information bottlenecks in conventional graph neural network (GNN) architectures that flatten heterogeneous relationships and employ over-simplified decoders. To address these challenges, we propose CoupleMDA, a metapath-guided heterogeneous graph learning framework coupling structural and semantic features. The model constructs a biological heterogeneous network using independent data sources to eliminate feature-target space coupling. Our framework implements a two-stage encoding strategy: (1) relational graph convolutional networks (RGCN) for pre-encoding and (2) metapath-guided semantic aggregation for secondary encoding. During decoding, common metapaths between node pairs structurally guide feature pooling, mitigating information bottlenecks. The comprehensive evaluation shows that CoupleMDA achieves a 2-5% performance improvement over the current state-of-the-art baseline methods in the heterogeneous graph link prediction task. Ablation studies confirm the necessity of each proposed component, while case analyses reveal the framework's capability to recover cancer-related miRNA-disease associations through biologically interpretable metapaths.
Collapse
Affiliation(s)
- Zhuojian Li
- School of Intelligent Systems Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (Z.L.); (G.C.)
| | - Guanxing Chen
- School of Intelligent Systems Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (Z.L.); (G.C.)
| | - Guang Tan
- School of Intelligent Systems Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (Z.L.); (G.C.)
| | - Calvin Yu-Chian Chen
- School of AI for Science, Peking University, Beijing 100871, China
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China
- Department of Medical Research, China Medical University Hospital, Taichung 40447, Taiwan
| |
Collapse
|
2
|
Piergentili R, Sechi S, De Paola L, Zaami S, Marinelli E. Building a Hand-Curated ceRNET for Endometrial Cancer, Striving for Clinical as Well as Medicolegal Soundness: A Systematic Review. Noncoding RNA 2025; 11:34. [PMID: 40407592 PMCID: PMC12101250 DOI: 10.3390/ncrna11030034] [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: 02/24/2025] [Revised: 03/31/2025] [Accepted: 04/27/2025] [Indexed: 05/26/2025] Open
Abstract
Background/Objectives: Competing endogenous RNAs (ceRNA) are molecules that compete for the binding to a microRNA (miR). Usually, there are two ceRNA, one of which is a protein-coding RNA (mRNA), with the other being a long non-coding RNA (lncRNA). The miR role is to inhibit mRNA expression, either promoting its degradation or impairing its translation. The lncRNA can "sponge" the miR, thus impeding its inhibitory action on the mRNA. In their easier configuration, these three molecules constitute a regulatory axis for protein expression. However, each RNA can interact with multiple targets, creating branched and intersected axes that, all together, constitute what is known as a competing endogenous RNA network (ceRNET). Methods: In this systematic review, we collected all available data from PubMed about experimentally verified (by luciferase assay) regulatory axes in endometrial cancer (EC), excluding works not using this test; Results: This search allowed the selection of 172 bibliographic sources, and manually building a series of ceRNETs of variable complexity showed the known axes and the deduced intersections. The main limitation of this search is the highly stringent selection criteria, possibly leading to an underestimation of the complexity of the networks identified. However, this work allows us not only to hypothesize possible gap fillings but also to set the basis to instruct artificial intelligence, using adequate prompts, to expand the EC ceRNET by comparing it with ceRNETs of other cancers. Moreover, these networks can be used to inform and guide research toward specific, though still unidentified, axes in EC, to complete parts of the network that are only partially described, or even to integrate low complexity subnetworks into larger more complex ones. Filling the gaps among the existing EC ceRNET will allow physicians to hypothesize new therapeutic strategies that may either potentiate or substitute existing ones. Conclusions: These ceRNETs allow us to easily visualize long-distance interactions, thus helping to select the best treatment, depending on the molecular profile of each patient, for personalized medicine. This would yield higher efficiency rates and lower toxicity levels, both of which are extremely relevant factors not only for patients' wellbeing, but also for the legal, regulatory, and ethical aspects of miR-based innovative treatments and personalized medicine as a whole. This systematic review has been registered in PROSPERO (ID: PROSPERO 2025 CRD420251035222).
Collapse
Affiliation(s)
- Roberto Piergentili
- Institute of Molecular Biology and Pathology, Italian National Research Council (CNR-IBPM), 00185 Rome, Italy;
| | - Stefano Sechi
- Institute of Molecular Biology and Pathology, Italian National Research Council (CNR-IBPM), 00185 Rome, Italy;
| | - Lina De Paola
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University of Rome, 00161 Rome, Italy; (L.D.P.); (S.Z.)
| | - Simona Zaami
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University of Rome, 00161 Rome, Italy; (L.D.P.); (S.Z.)
| | - Enrico Marinelli
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy;
| |
Collapse
|
3
|
Yang JL, Ma JJ, Qu TY, Dai Q, Leng J, Fang L, Wu J, Li YJ, Yu HF. Glycolysis-related lncRNA FTX upregulates YAP1 to facilitate colorectal cancer progression via sponging miR-215-3p. Sci Rep 2025; 15:9929. [PMID: 40121300 PMCID: PMC11929783 DOI: 10.1038/s41598-025-94638-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 03/17/2025] [Indexed: 03/25/2025] Open
Abstract
Increased evidence reveals that glycolysis is one of the key metabolic hallmarks of cancer cells. However, the roles of lncRNA FTX in energy metabolism and cancer progression remain unclear. In this study we aim to show that lncRNA FTX was significantly upregulated in cancer tissues and serum of CRC patients and CRC cell lines. Function study indicated that it could promote aerobic glycolysis, cell proliferation, migration and invasion in colorectal cancer cells. Further mechanistic studies showed, lncRNA FTX was found to function as a sponge for miR-215-3p, which reduced the ability of miR-215-3p to repress the YAP1 oncoprotein. Additionally, a negative correlation was observed between lncRNA FTX and miR-215-3p expression, and the knockdown of lncRNA FTX or miR-215-3p overexpression yielded opposite effects. In conclusion, this study demonstrates that FTX could directly combine with miR-215-3p as a competitive endogenous RNA, thus promoting the aerobic glycolysis and progression of CRC in vitro and in vivo.
Collapse
Affiliation(s)
- Jin-Lan Yang
- Department of Oncology, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, 563000, Guizhou Province, China
- Cancer Disease Research Institute, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), 98# Fenghuang Road, Zunyi, 563003, Guizhou Province, China
| | - Jing-Jing Ma
- Department of Clinical Laboratory, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, 563000, Guizhou Province, China
| | - Tian-Yin Qu
- Department of Oncology, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, 563000, Guizhou Province, China
- Cancer Disease Research Institute, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), 98# Fenghuang Road, Zunyi, 563003, Guizhou Province, China
| | - Qing Dai
- Department of Oncology, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, 563000, Guizhou Province, China
- Cancer Disease Research Institute, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), 98# Fenghuang Road, Zunyi, 563003, Guizhou Province, China
| | - Jing Leng
- Department of Oncology, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, 563000, Guizhou Province, China
- Cancer Disease Research Institute, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), 98# Fenghuang Road, Zunyi, 563003, Guizhou Province, China
| | - Lin Fang
- Department of Oncology, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, 563000, Guizhou Province, China
- Cancer Disease Research Institute, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), 98# Fenghuang Road, Zunyi, 563003, Guizhou Province, China
| | - Jie Wu
- Scientific Research Center, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou Province, China
| | - Ya-Jun Li
- Department of Oncology, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, 563000, Guizhou Province, China
| | - Huang-Fei Yu
- Department of Oncology, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, 563000, Guizhou Province, China.
- Cancer Disease Research Institute, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), 98# Fenghuang Road, Zunyi, 563003, Guizhou Province, China.
| |
Collapse
|
4
|
Márton É, Varga A, Domoszlai D, Buglyó G, Balázs A, Penyige A, Balogh I, Nagy B, Szilágyi M. Non-Coding RNAs in Cancer: Structure, Function, and Clinical Application. Cancers (Basel) 2025; 17:579. [PMID: 40002172 PMCID: PMC11853212 DOI: 10.3390/cancers17040579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 02/04/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
We are on the brink of a paradigm shift in both theoretical and clinical oncology. Genomic and transcriptomic profiling, alongside personalized approaches that account for individual patient variability, are increasingly shaping discourse. Discussions on the future of personalized cancer medicine are mainly dominated by the potential of non-coding RNAs (ncRNAs), which play a prominent role in cancer progression and metastasis formation by regulating the expression of oncogenic or tumor suppressor proteins at transcriptional and post-transcriptional levels; furthermore, their cell-free counterparts might be involved in intercellular communication. Non-coding RNAs are considered to be promising biomarker candidates for early diagnosis of cancer as well as potential therapeutic agents. This review aims to provide clarity amidst the vast body of literature by focusing on diverse species of ncRNAs, exploring the structure, origin, function, and potential clinical applications of miRNAs, siRNAs, lncRNAs, circRNAs, snRNAs, snoRNAs, eRNAs, paRNAs, YRNAs, vtRNAs, and piRNAs. We discuss molecular methods used for their detection or functional studies both in vitro and in vivo. We also address the challenges that must be overcome to enter a new era of cancer diagnosis and therapy that will reshape the future of oncology.
Collapse
Affiliation(s)
- Éva Márton
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (É.M.); (A.V.); (D.D.); (G.B.); (A.P.); (I.B.); (B.N.)
| | - Alexandra Varga
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (É.M.); (A.V.); (D.D.); (G.B.); (A.P.); (I.B.); (B.N.)
| | - Dóra Domoszlai
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (É.M.); (A.V.); (D.D.); (G.B.); (A.P.); (I.B.); (B.N.)
| | - Gergely Buglyó
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (É.M.); (A.V.); (D.D.); (G.B.); (A.P.); (I.B.); (B.N.)
| | - Anita Balázs
- Department of Integrative Health Sciences, Institute of Health Sciences, Faculty of Health Sciences, University of Debrecen, H-4032 Debrecen, Hungary;
| | - András Penyige
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (É.M.); (A.V.); (D.D.); (G.B.); (A.P.); (I.B.); (B.N.)
| | - István Balogh
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (É.M.); (A.V.); (D.D.); (G.B.); (A.P.); (I.B.); (B.N.)
- Division of Clinical Genetics, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Bálint Nagy
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (É.M.); (A.V.); (D.D.); (G.B.); (A.P.); (I.B.); (B.N.)
| | - Melinda Szilágyi
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (É.M.); (A.V.); (D.D.); (G.B.); (A.P.); (I.B.); (B.N.)
| |
Collapse
|
5
|
Afroze N, Sundaram MK, Haque S, Hussain A. Long non-coding RNA involved in the carcinogenesis of human female cancer - a comprehensive review. Discov Oncol 2025; 16:122. [PMID: 39912983 PMCID: PMC11803034 DOI: 10.1007/s12672-025-01848-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/22/2025] [Indexed: 02/07/2025] Open
Abstract
Recent years have seen an increase in our understanding of lncRNA and their role in various disease states. lncRNA molecules have been shown to contribute to carcinogenesis and influence the various cancer hallmarks and signalling pathways. It is pertinent to understand the specific contributions and mechanisms of action of these molecules in various cancers. This review provides an overview of the various lncRNA entities that influence and regulate the gynaecological cancers, namely, cervical, breast, ovarian and uterine cancers. The review curates a list of the key players and their effect on cellular processes. lncRNA molecules show immense potential to be used as diagnostic and prognostic indicators and in therapeutic strategies. Several phytochemicals, small molecules, RNA-based regulators, oligos and gene editing tools show promise as a therapeutic strategy. While this review highlights the promising developments in this field, it also underscores the necessity for further research to delineate the complex role of lncRNAs in cancer.
Collapse
Affiliation(s)
- Nazia Afroze
- School of Life Sciences, Manipal Academy of Higher Education, Dubai Campus, P.O. Box 345050, Dubai, United Arab Emirates
| | - Madhumitha K Sundaram
- School of Life Sciences, Manipal Academy of Higher Education, Dubai Campus, P.O. Box 345050, Dubai, United Arab Emirates
| | - Shafiul Haque
- Department of Nursing, College of Nursing and Health Sciences, Jazan University, Jazan, Saudi Arabia
- School of Medicine, Universidad Espiritu Santo, Samborondon, Ecuador
| | - Arif Hussain
- School of Life Sciences, Manipal Academy of Higher Education, Dubai Campus, P.O. Box 345050, Dubai, United Arab Emirates.
| |
Collapse
|
6
|
El-Daly SM, Abdelrahman SS, El-Bana MA, Abdel-Latif Y, Medhat D, Morsy SM, Wafay HA. Deciphering the Interplay of the PD-L1/MALT1/miR-200a Axis During Lung Cancer Development. Biotechnol Appl Biochem 2025. [PMID: 39910787 DOI: 10.1002/bab.2724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 01/13/2025] [Indexed: 02/07/2025]
Abstract
Lung cancer remains a leading cause of cancer-related mortality worldwide. Our study investigates the involvement of the PD-L1/MALAT1/miR-200a-3p axis in lung tumor progression using a murine model of lung carcinogenesis. Lung tumors were induced in rats, which were divided into groups and sacrificed at different stages of tumor development. A histopathological examination was performed to assess tumor progression. Immunohistochemistry was applied to evaluate the expression of Ki-67 and programmed death-ligand 1 (PD-L1). The level of carcinoembryonic antigen (CEA) and expression analysis of metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), miR-200a-3p, and zinc finger E-box binding homeobox 1 (ZEB1) were evaluated for each stage of induction. Immunohistochemical analysis demonstrated a progressive upregulation of the proliferative marker Ki-67 and the immune checkpoint protein PD-L1 during the induction process, indicative of enhanced tumor proliferation and immune evasion. Additionally, CEA levels revealed a progressive increase across induction stages, with a significant increase in advanced tumor stages, highlighting its clinical relevance as a biomarker for lung cancer progression. Expression analysis revealed dynamic upregulation of MALAT1 and downregulation of miR-200a during lung tumor induction, which correlated with advanced tumor stages and elevated PD-L1 expression, suggesting that the negative correlation between MALAT1 and miR-200a is involved in the development of lung tumors. ZEB1 expression exhibited a notable increase in the advanced stages of induction, consistent with its association with aggressive lung cancer. Our findings underscore the interplay between molecular pathways involved in lung tumor development and the potential diagnostic and therapeutic implications of the PD-L1/MALAT1/miR-200a-3p axis.
Collapse
Affiliation(s)
- Sherien M El-Daly
- Department of Medical Biochemistry, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
- Cancer Biology and Genetics Laboratory, Centre of Excellence for Advanced Sciences, National Research Centre, Giza, Egypt
| | - Sahar S Abdelrahman
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Mona A El-Bana
- Department of Medical Biochemistry, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Yasmin Abdel-Latif
- Department of Medical Biochemistry, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
- Faculty of Biotechnology, October University for Modern Sciences and Arts, 6th of October, Giza, Egypt
| | - Dalia Medhat
- Department of Medical Biochemistry, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Safaa M Morsy
- Department of Medical Biochemistry, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Hanaa A Wafay
- Department of Medical Biochemistry, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| |
Collapse
|
7
|
Sayed NH, Shaker OG, Abd-Elmawla MA, Gamal A, Fathy N. New insights into the interplay between MALAT1 and miRNA-155 to unravel potential diagnostic and prognostic biomarkers of Behçet's disease. Clin Rheumatol 2025; 44:775-787. [PMID: 39798064 PMCID: PMC11774967 DOI: 10.1007/s10067-024-07291-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/14/2024] [Accepted: 12/18/2024] [Indexed: 01/13/2025]
Abstract
The current study was deployed to evaluate the role of metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) and miR-155, along with the inflammatory markers, TNFα and IL-6, and the adhesion molecule, cluster of differentiation 106 (CD106), in Behçet's disease (BD) pathogenesis. The study also assessed MALAT1/miR-155 as promising diagnostic and prognostic biomarkers for BD. The current retrospective case-control study included 74 Egyptian BD patients and 50 age and sex-matched controls. Blood samples were collected, and then, serum samples were separated for further biochemical and molecular investigations. The gene expression of MALAT1 and miR-155 was measured using qRT-PCR, whereas the levels of TNFα, IL-6, and CD106 were estimated using ELISA technique. MALAT1 was significantly downregulated, whereas miR-155 was upregulated among BD patients, compared with control subjects. Levels of TNFα, IL-6, and CD106 were elevated in BD patients. Further downregulation in MALAT1 together with upregulation of miR-155 was observed in active BD patients, relative to the inactive group. Receiver-operating-characteristic analysis revealed that MALAT1 and miR-155 could discriminate BD patients from controls, on the one hand, and active from inactive BD patients, on the other hand. MALAT1 was negatively correlated with TNFα, IL-6, and CD106, while miR-155 was positively correlated with them. Logistic regression analyses demonstrated miR-155 as a significant independent predictor of BD susceptibility, and MALAT1 as an independent negative predictor of BD activity. For the first time, the current research enlightens the role of MALAT1 and miR-155 in BD pathogenesis via impacting IL-6/TNF-α/CD-106 signaling. As well, MALAT1 and miR-155 could be regarded as novel non-invasive biomarkers that may improve BD diagnosis and prognosis. Key Points •MALAT1/miR-155 exerts potential role in Behçet's disease. •MALAT1/miR-155 are promising biomarkers for Behçet's disease. •MALAT1/miR-155 targets IL-6/TNF-α/CD-106 signaling.
Collapse
Affiliation(s)
- Noha H Sayed
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Olfat G Shaker
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Cairo, 11562, Egypt
| | - Mai A Abd-Elmawla
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| | - Ahmed Gamal
- Andrology, Sexology and STI's Department, Faculty of Medicine, Cairo University, Cairo, 11562, Egypt
| | - Nevine Fathy
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| |
Collapse
|
8
|
Fan L, Lan M, Wei X, Wei L, Yang L, Nong L, Wei J, Li J, Huang W. Comprehensive analysis of ceRNA Networks in UCEC: Prognostic and therapeutic implications. PLoS One 2025; 20:e0314314. [PMID: 39883704 PMCID: PMC11781699 DOI: 10.1371/journal.pone.0314314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 11/06/2024] [Indexed: 02/01/2025] Open
Abstract
Endometrial cancer (UCEC) is the most prevalent gynecological malignancy in high-income countries, and its incidence is rising globally. Although early-stage UCEC can be treated with surgery, advanced cases have a poor prognosis, highlighting the need for effective molecular biomarkers to improve diagnosis and prognosis. In this study, we analyzed mRNA and miRNA sequencing data from UCEC tissues and adjacent non-cancerous tissues from the TCGA database. Differential expression analysis was conducted using the DESeq2 package, identifying differentially expressed lncRNAs, miRNAs, and mRNAs (DElncRNAs, DEmiRNAs, and DEmRNAs). Key molecules were screened using LASSO regression, and a ceRNA network was constructed by predicting lncRNA-miRNA and miRNA-mRNA interaction, which were visualized with Cytoscape. Functional enrichment analysis elucidated the roles and mechanisms of the network. The prognostic potential of the identified RNAs was assessed through survival and Cox regression analyses, while methylation and immune infiltration analyses explored regulatory mechanisms and immune interactions. We identified a prognostic lncRNA-miRNA-mRNA ceRNA network in UCEC, centered on the CDKN2B-AS1-hsa-miR-497-5p-IGF2BP3 axis. Survival analyses confirmed the prognostic significance of this network, with univariate Cox regression demonstrating a strong association between its aberrant expression and overall prognosis in UCEC. However, multivariate Cox regression suggested that other clinical factors may modulate this relationship. Methylation analysis revealed low methylation levels of IGF2BP3, possibly contributing to its overexpression. Furthermore, immune infiltration studies highlighted significant correlations between CDKN2B-AS1, IGF2BP3, and multiple immune cell types, suggesting that this axis regulates the tumor immune microenvironment. These findings suggest that the CDKN2B-AS1-hsa-miR-497-5p-IGF2BP3 axis is a key regulatory element in UCEC and a potential therapeutic target.
Collapse
Affiliation(s)
- Li Fan
- Department of Reproductive Medicine, Guangzhou Women and Children’s Medical center Liuzhou Hospital, Liuzhou, Guangxi, China
- Reproductive Medicine Center, Liuzhou Maternity and Child Health Care Hospital, Liuzhou, China
- Guangxi Maternal and Obstetric Disease Research Center, Liuzhou, China
- Liuzhou Institute of Reproduction and Genetics, Liuzhou Maternity and Child Health Care Hospital, Liuzhou, China
- Liuzhou Key Laboratory of Gynecologic Tumor, Zhengzhou, China
| | - Mengqiu Lan
- Liuzhou Municipal Liutie Central Hospital, Liuzhou, Guangxi, China
| | - Xiaohua Wei
- Department of Reproductive Medicine, Guangzhou Women and Children’s Medical center Liuzhou Hospital, Liuzhou, Guangxi, China
- Reproductive Medicine Center, Liuzhou Maternity and Child Health Care Hospital, Liuzhou, China
- Guangxi Maternal and Obstetric Disease Research Center, Liuzhou, China
- Liuzhou Institute of Reproduction and Genetics, Liuzhou Maternity and Child Health Care Hospital, Liuzhou, China
- Liuzhou Key Laboratory of Gynecologic Tumor, Zhengzhou, China
| | - Lili Wei
- Department of Reproductive Medicine, Guangzhou Women and Children’s Medical center Liuzhou Hospital, Liuzhou, Guangxi, China
- Reproductive Medicine Center, Liuzhou Maternity and Child Health Care Hospital, Liuzhou, China
- Guangxi Maternal and Obstetric Disease Research Center, Liuzhou, China
- Liuzhou Institute of Reproduction and Genetics, Liuzhou Maternity and Child Health Care Hospital, Liuzhou, China
- Liuzhou Key Laboratory of Gynecologic Tumor, Zhengzhou, China
| | - Liuhong Yang
- Department of Reproductive Medicine, Guangzhou Women and Children’s Medical center Liuzhou Hospital, Liuzhou, Guangxi, China
- Reproductive Medicine Center, Liuzhou Maternity and Child Health Care Hospital, Liuzhou, China
- Guangxi Maternal and Obstetric Disease Research Center, Liuzhou, China
- Liuzhou Institute of Reproduction and Genetics, Liuzhou Maternity and Child Health Care Hospital, Liuzhou, China
- Liuzhou Key Laboratory of Gynecologic Tumor, Zhengzhou, China
| | - Liuying Nong
- Department of Reproductive Medicine, Guangzhou Women and Children’s Medical center Liuzhou Hospital, Liuzhou, Guangxi, China
- Reproductive Medicine Center, Liuzhou Maternity and Child Health Care Hospital, Liuzhou, China
| | - Jiajia Wei
- Department of Reproductive Medicine, Guangzhou Women and Children’s Medical center Liuzhou Hospital, Liuzhou, Guangxi, China
- Reproductive Medicine Center, Liuzhou Maternity and Child Health Care Hospital, Liuzhou, China
- Guangxi Maternal and Obstetric Disease Research Center, Liuzhou, China
- Liuzhou Institute of Reproduction and Genetics, Liuzhou Maternity and Child Health Care Hospital, Liuzhou, China
- Liuzhou Key Laboratory of Gynecologic Tumor, Zhengzhou, China
| | - Jingjing Li
- Department of Reproductive Medicine, Guangzhou Women and Children’s Medical center Liuzhou Hospital, Liuzhou, Guangxi, China
- Reproductive Medicine Center, Liuzhou Maternity and Child Health Care Hospital, Liuzhou, China
- Guangxi Maternal and Obstetric Disease Research Center, Liuzhou, China
- Liuzhou Institute of Reproduction and Genetics, Liuzhou Maternity and Child Health Care Hospital, Liuzhou, China
- Liuzhou Key Laboratory of Gynecologic Tumor, Zhengzhou, China
| | - Wenjie Huang
- Department of Reproductive Medicine, Guangzhou Women and Children’s Medical center Liuzhou Hospital, Liuzhou, Guangxi, China
- Reproductive Medicine Center, Liuzhou Maternity and Child Health Care Hospital, Liuzhou, China
- Guangxi Maternal and Obstetric Disease Research Center, Liuzhou, China
- Liuzhou Institute of Reproduction and Genetics, Liuzhou Maternity and Child Health Care Hospital, Liuzhou, China
- Liuzhou Key Laboratory of Gynecologic Tumor, Zhengzhou, China
| |
Collapse
|
9
|
Ye L, Dimitriadis E. Endometrial Receptivity-Lessons from "Omics". Biomolecules 2025; 15:106. [PMID: 39858500 PMCID: PMC11764156 DOI: 10.3390/biom15010106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/03/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
The window of implantation (WOI) is a critical phase of the menstrual cycle during which the endometrial lining becomes receptive and facilitates embryo implantation. Drawing on findings from various branches of "omics", including genomics, epigenomics, transcriptomics, proteomics, lipidomics, metabolomics, and microbiomics, this narrative review aims to (1) discuss mechanistic insights on endometrial receptivity and its implication in infertility; (2) highlight advances in investigations for endometrial receptivity; and (3) discuss novel diagnostic and therapeutic strategies that may improve reproductive outcomes.
Collapse
Affiliation(s)
- Louie Ye
- Reproductive Service Unit, The Royal Women’s Hospital, Melbourne, VIC 3052, Australia
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne, Melbourne, VIC 3052, Australia;
| | - Evdokia Dimitriadis
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne, Melbourne, VIC 3052, Australia;
| |
Collapse
|
10
|
Hussein MA, Valinezhad K, Adel E, Munirathinam G. MALAT-1 Is a Key Regulator of Epithelial-Mesenchymal Transition in Cancer: A Potential Therapeutic Target for Metastasis. Cancers (Basel) 2024; 16:234. [PMID: 38201661 PMCID: PMC10778055 DOI: 10.3390/cancers16010234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 12/29/2023] [Accepted: 01/01/2024] [Indexed: 01/12/2024] Open
Abstract
Metastasis-associated lung adenocarcinoma transcript-1 (MALAT-1) is a long intergenic non-coding RNA (lncRNA) located on chr11q13. It is overexpressed in several cancers and controls gene expression through chromatin modification, transcriptional regulation, and post-transcriptional regulation. Importantly, MALAT-1 stimulates cell proliferation, migration, and metastasis and serves a vital role in driving the epithelial-to-mesenchymal transition (EMT), subsequently acquiring cancer stem cell-like properties and developing drug resistance. MALAT-1 modulates EMT by interacting with various intracellular signaling pathways, notably the phosphoinositide 3-kinase (PI3K)/Akt and Wnt/β-catenin pathways. It also behaves like a sponge for microRNAs, preventing their interaction with target genes and promoting EMT. In addition, we have used bioinformatics online tools to highlight the disparities in the expression of MALAT-1 between normal and cancer samples using data from The Cancer Genome Atlas (TCGA). Furthermore, the intricate interplay of MALAT-1 with several essential targets of cancer progression and metastasis renders it a good candidate for therapeutic interventions. Several innovative approaches have been exploited to target MALAT-1, such as short hairpin RNAs (shRNAs), antisense oligonucleotides (ASOs), and natural products. This review emphasizes the interplay between MALAT-1 and EMT in modulating cancer metastasis, stemness, and chemoresistance in different cancers.
Collapse
Affiliation(s)
- Mohamed Ali Hussein
- Department of Pharmaceutical Services, Children’s Cancer Hospital Egypt, Cairo 57357, Egypt;
- Department of Biology, School of Sciences and Engineering, American University in Cairo, New Cairo 11835, Egypt;
| | - Kamyab Valinezhad
- Department of Biomedical Sciences, College of Medicine, University of Illinois, Rockford, IL 61107, USA;
| | - Eman Adel
- Department of Biology, School of Sciences and Engineering, American University in Cairo, New Cairo 11835, Egypt;
| | - Gnanasekar Munirathinam
- Department of Biomedical Sciences, College of Medicine, University of Illinois, Rockford, IL 61107, USA;
| |
Collapse
|
11
|
Shaker O, El Amir M, Elfatah YA, Elwi HM. Expression patterns of lncRNA MALAT-1 in SARS-COV-2 infection and its potential effect on disease severity via miR-200c-3p and SIRT1. Biochem Biophys Rep 2023; 36:101562. [PMID: 37965063 PMCID: PMC10641570 DOI: 10.1016/j.bbrep.2023.101562] [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: 07/19/2023] [Revised: 10/14/2023] [Accepted: 10/17/2023] [Indexed: 11/16/2023] Open
Abstract
Downregulating Angiotensin Converting Enzyme2 (ACE2) expression may be a shared mechanism for RNA viruses. Aim Evaluate the expressions of ACE2 effectors: the long non-coding RNA 'MALAT-1', the micro-RNA 'miR-200c-3p' and the histone deacetylase 'SIRT1' in SARS-COV-2 patients and correlate to disease severity. Sera samples from 98 SARS-COV-2 patients and 30 healthy control participants were collected. qRT-PCR was used for MALAT-1 and miR-200c-3p expression. SIRT1 was measured using ELISA. Results In sera of COVID-19 patients, gene expression of miR-200c-3p is increased while MALAT-1 is decreased. SIRT1 protein level is decreased (P value < 0.001). Findings are accentuated with increased disease severity. Serum MALAT-1, miR-200c-3p and SIRT1 could be used as diagnostic markers at cut off values of 0.04 (95.9 % sensitivity), 5.59 (94.9 % sensitivity, 99 % specificity), and 7.4 (98 % sensitivity) respectively. A novel MALAT-1-miR-200c-3p-SIRT1 pathway may be involved in the regulation of SARS-COV-2 severity.
Collapse
Affiliation(s)
- Olfat Shaker
- Medical Biochemistry and Molecular Biology, Kasr Alainy Faculty of Medicine, Cairo University, Kasralainy st, Cairo, 11562, Egypt
| | - Monica El Amir
- Medical Biochemistry and Molecular Biology, Kasr Alainy Faculty of Medicine, Cairo University, Kasralainy st, Cairo, 11562, Egypt
| | - Yasmine Abd Elfatah
- Internal Medicine, Kasr Alainy Faculty of Medicine, Cairo University, Kasralainy st, Cairo, 11562, Egypt
| | - Heba M. Elwi
- Medical Biochemistry and Molecular Biology, Kasr Alainy Faculty of Medicine, Cairo University, Kasralainy st, Cairo, 11562, Egypt
| |
Collapse
|
12
|
Grafanaki K, Grammatikakis I, Ghosh A, Gopalan V, Olgun G, Liu H, Kyriakopoulos GC, Skeparnias I, Georgiou S, Stathopoulos C, Hannenhalli S, Merlino G, Marie KL, Day CP. Noncoding RNA circuitry in melanoma onset, plasticity, and therapeutic response. Pharmacol Ther 2023; 248:108466. [PMID: 37301330 PMCID: PMC10527631 DOI: 10.1016/j.pharmthera.2023.108466] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/24/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023]
Abstract
Melanoma, the cancer of the melanocyte, is the deadliest form of skin cancer with an aggressive nature, propensity to metastasize and tendency to resist therapeutic intervention. Studies have identified that the re-emergence of developmental pathways in melanoma contributes to melanoma onset, plasticity, and therapeutic response. Notably, it is well known that noncoding RNAs play a critical role in the development and stress response of tissues. In this review, we focus on the noncoding RNAs, including microRNAs, long non-coding RNAs, circular RNAs, and other small RNAs, for their functions in developmental mechanisms and plasticity, which drive onset, progression, therapeutic response and resistance in melanoma. Going forward, elucidation of noncoding RNA-mediated mechanisms may provide insights that accelerate development of novel melanoma therapies.
Collapse
Affiliation(s)
- Katerina Grafanaki
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Department of Dermatology, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Ioannis Grammatikakis
- Cancer Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Arin Ghosh
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Vishaka Gopalan
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Gulden Olgun
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Huaitian Liu
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - George C Kyriakopoulos
- Department of Biochemistry, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Ilias Skeparnias
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Sophia Georgiou
- Department of Dermatology, School of Medicine, University of Patras, 26504 Patras, Greece
| | | | - Sridhar Hannenhalli
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Glenn Merlino
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kerrie L Marie
- Division of Molecular and Cellular Function, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| | - Chi-Ping Day
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
13
|
Sun M, Chen D, Chen Y, Wu Y. LncRNA SOX21-AS1 accelerates endometrial carcinoma progression through the miR-7-5p/RAF1 pathway. World J Surg Oncol 2023; 21:217. [PMID: 37481582 PMCID: PMC10362562 DOI: 10.1186/s12957-023-03114-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/13/2023] [Indexed: 07/24/2023] Open
Abstract
BACKGROUND Endometrial carcinoma (EC) is one of the world's typical female reproductive tract malignancies, mostly occurring in postmenopausal women. Many reports have confirmed that long non-coding RNA SOX21 antisense RNA1 (lncRNA SOX21-AS1) is associated with the progressions of various cancer. However, the mechanism of SOX21-AS1 in EC remains unclear. Our study is intended to probe the mechanisms of SOX21-AS1 on EC progression. METHODS The CCK-8 assay and colony formation detected cell proliferation. Cell migration and invasion were assessed by transwell analysis. Apoptosis was measured by flow cytometry assay. Bioinformatics software predicted target binding and confirmed using a luciferase reporter analysis. RESULTS SOX21-AS1 expression was upregulated in EC tumor tissues and cells. High expression of SOX21-AS1 was associated with poor overall survival. Silencing of SOX21-AS1 restrained cell proliferation, migration, invasion, and increased apoptosis in HEC-1A and Ishikawa cells. Additionally, bioinformatics analysis demonstrated that SOX21-AS1 modulated RAF1 expression by competitively binding to miR-7-5p. Functionally, silencing of RAF1 reversed the functions of miR-7-5p inhibitor in the proliferation, invasion, and apoptosis of HEC-1A/sh-SOX21-AS1 and Ishikawa/sh-SOX21-AS1 cells. CONCLUSIONS SOX21-AS1 promoted the pathological development of EC by regulating the miR-7-5p/RAF1 pathway. This research may provide a novel target for EC therapy.
Collapse
Affiliation(s)
- Meng Sun
- Department of Gynecology, 1St Affiliated Hospital, Soochow University, 188#, Shizi Street, Gusu District, Suzhou, 215000, Jiangsu, China
- Department of Gynecology, Affiliated Hospital of Jiangnan University, Wuxi, 214000, Jiangsu, China
| | - Dongxu Chen
- Department of Nuclear Medicine, Wuxi Branch of Ruijin Hospital, Wuxi, 214000, Jiangsu, China
| | - Youguo Chen
- Department of Gynecology, 1St Affiliated Hospital, Soochow University, 188#, Shizi Street, Gusu District, Suzhou, 215000, Jiangsu, China.
| | - Yibo Wu
- Human Reproductive and Genetic Center, Affiliated Hospital of Jiangnan University, 200 Huihe Road, Wuxi, 214000, Jiangsu, China
| |
Collapse
|
14
|
Jin Y, Qiu Y, Li Y, Jiang Z, Hu S, Dai H. A novel epithelial-mesenchymal transition-related lncRNA signature predicts prognosis and immune status in endometrioid endometrial cancer. Medicine (Baltimore) 2023; 102:e34126. [PMID: 37390286 PMCID: PMC10313257 DOI: 10.1097/md.0000000000034126] [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: 04/09/2023] [Accepted: 06/07/2023] [Indexed: 07/02/2023] Open
Abstract
The pathogenesis and progression of endometrial cancer (EC) are associated with epithelial-mesenchymal transition (EMT) and long noncoding RNAs (lncRNAs). In the present study, we aimed to identify an EMT-related lncRNA signature and evaluate its prognostic value in EC. We obtained the expression profile of lncRNAs and clinical information of patients with endometrioid EC from The Cancer Genome Atlas database (N = 401). We identified a signature of 5 EMT-related lncRNAs and calculated the risk score of each patient. Next, we validated the independence of the prognostic value of the EMT-related lncRNA signature. Furthermore, we performed Gene Set Enrichment Analysis to identify potential molecular function and Kyoto Encyclopedia of Genes and Genomes pathways related to the EMT-related lncRNA signature. Tumor microenvironment analysis and immune checkpoint blockade (ICB) response prediction were also assessed. Survival analysis revealed that the high-risk group, based on the EMT-related lncRNA signature, had a poorer prognosis than the low-risk group in the training, testing, and entire sets. The predictive value of the EMT-related lncRNA signature was independent of age, The International Federation of Gynecology and Obstetrics stage, tumor grade, and body mass index. Time-dependent receiver operating characteristic curves also demonstrate the prognostic accuracy of this risk model. Gene Set Enrichment Analysis showed that cytokine-cytokine receptor interaction, glycolysis/gluconeogenesis, and IL-17 signaling pathway were significantly enriched. Furthermore, tumor microenvironment analysis indicated a significant negative correlation between the immune score and EMT-related lncRNA signature risks core, while the low-risk group was more likely to respond to ICB therapy than the high-risk group. A reliable EMT-related lncRNA signature of endometrioid EC was identified that could be utilized as an independent prognostic biomarker to predict patient survival outcomes and provide references for the option of ICB therapy.
Collapse
Affiliation(s)
- Yichao Jin
- Department of Gynecology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Yixuan Qiu
- Department of Gynecology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Yujing Li
- Department of Gynecology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Ziwei Jiang
- Department of Gynecology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Siwen Hu
- Department of Gynecology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Huihua Dai
- Department of Gynecology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| |
Collapse
|
15
|
Duan Y, Yue K, Ye B, Chen P, Zhang J, He Q, Wu Y, Lai Q, Li H, Wu Y, Jing C, Wang X. LncRNA MALAT1 promotes growth and metastasis of head and neck squamous cell carcinoma by repressing VHL through a non-canonical function of EZH2. Cell Death Dis 2023; 14:149. [PMID: 36813772 PMCID: PMC9946937 DOI: 10.1038/s41419-023-05667-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 02/03/2023] [Accepted: 02/08/2023] [Indexed: 02/24/2023]
Abstract
Long non-coding RNAs (LncRNAs) are implicated in malignant progression of human cancers. Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), a well-known lncRNA, has been reported to play crucial roles in multiple malignancies including head and neck squamous cell carcinoma (HNSCC). However, the underlying mechanisms of MALAT1 in HNSCC progression remain to be further investigated. Here, we elucidated that compared with normal squamous epithelium, MALAT1 was notably upregulated in HNSCC tissues, especially in which was poorly differentiated or with lymph nodes metastasis. Moreover, elevated MALAT1 predicted unfavorable prognosis of HNSCC patients. The results of in vitro and in vivo assays showed that targeting MALAT1 could significantly weaken the capacities of proliferation and metastasis in HNSCC. Mechanistically, MALAT1 inhibited von Hippel-Lindau tumor suppressor (VHL) by activating EZH2/STAT3/Akt axis, then promoted the stabilization and activation of β-catenin and NF-κB which could play crucial roles in HNSCC growth and metastasis. In conclusion, our findings reveal a novel mechanism for malignant progression of HNSCC and suggest that MALAT1 might be a promising therapeutic target for HNSCC treatment.
Collapse
Affiliation(s)
- Yuansheng Duan
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Kai Yue
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Beibei Ye
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Peng Chen
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Jin Zhang
- Department of Bone and Soft Tissue Tumors, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Qinghua He
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Yue Wu
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Qingchuan Lai
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Hong Li
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Yansheng Wu
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
| | - Chao Jing
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
| | - Xudong Wang
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
| |
Collapse
|
16
|
Hao C, Lin S, Liu P, Liang W, Li Z, Li Y. Potential serum metabolites and long-chain noncoding RNA biomarkers for endometrial cancer tissue. J Obstet Gynaecol Res 2023; 49:725-743. [PMID: 36510632 DOI: 10.1111/jog.15494] [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: 06/26/2022] [Revised: 10/05/2022] [Accepted: 10/28/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Endometrial carcinoma (EC) is one of the most common tumors in the female reproductive system. There are nearly 200 000 new cases every year. It is the third most common gynecological malignant tumor leading to female death. The incidence rate is closely related to lifestyle, and the incidence rate varies in different regions. The incidence rate of EC is ranking the first in the female reproductive system cancer just second only to breast, lung, and colorectal cancer in North America and Europe and the incidence rate of EC is only second, followed by breast cancer and cervical cancer in China. PURPOSE The potential metabolic markers of endometrial cancer were screened by liquid chromatograph mass spectrometer (LC-MS), and the tissues of patients with hysteromyoma and endometrial cancer were sequenced to explore the relationship between the disease and change in the content of long-chain noncoding RNA (lncRNA). METHODS Serum and tissue samples were collected from patients with endometrial dysplasia, endometrial cancer stage I, and endometrial cancer stage III. The metabolites in all serum samples were extracted, and the metabolites in all samples were detected by LC-MS/MS technology. The Pareto-scaling method was used for normalization, and the MetaboAnalyst 4.0 software was used for different analyses. The T test between groups showed that p ≤ 0.05 was regarded as the metabolite with a difference. Further, the function of differential metabolites was determined by metabolite function enrichment and co-expression analysis. Meanwhile, the differentially expressed lncRNA was detected by Illumina second-generation high-throughput sequencing technology, and the expression was analyzed by DEGseq software. Different lncRNA were screened according to p < 0.05. LncRNA with significant differences were screened by p < 0.01, q < 0.001, fold change ≥2, and false discovery rate (FDR) ≤0.001. RESULTS Through synthesis of T test, cluster heatmap, and ROC curve analysis, five biomarkers with potential diagnostic ability were obtained, including 2,3-Pyridinedicarboxylic acid (area under the curve (AUC) = 0.69), Hematommic acid, ethyl ester (AUC = 0.69), Maltitol (AUC = 0.69), 13(S)-HODE (AUC = 0.88), and D-Mannitol (AUC = 0.69) had potential diagnostic ability between EC phase I versus EC phase III. At the same time, lncRNA sequencing results showed that when endometrial atypical hyperplasia continued to change, including LINC00511, PVT1, and IQCH-AS1 (downregulated), and only changed significantly in the endometrial dysplasia group, including MALAT1, CARMN (downregulated) and LINC00648, BISPR, LINC01534, and LINC00930 (upregulated). Moreover, both differential metabolites and differential lncRNA were annotated to the lipid metabolism pathway, suggesting that this pathway played an important role in the occurrence and development of endometrial carcinoma. CONCLUSIONS It can combine the results of metabolomics and lncRNA sequencing to assist in the early diagnosis of endometrial precancerous lesions and endometrial cancer patients, to enhance the sensitivity and specificity of diagnosis, which has a certain clinical application prospect.
Collapse
Affiliation(s)
- Chenjun Hao
- Gynaecology and Obstetrics Department, Maternal and Child Health Hospital of PanYu District, Guangzhou, China
| | - Shaodan Lin
- Gynaecology and Obstetrics Department, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Ping Liu
- Gynaecology and Obstetrics Department, Maternal and Child Health Hospital of PanYu District, Guangzhou, China
| | - Weiguo Liang
- Gynaecology and Obstetrics Department, Maternal and Child Health Hospital of PanYu District, Guangzhou, China
| | - Zhi Li
- Gynaecology and Obstetrics Department, Maternal and Child Health Hospital of PanYu District, Guangzhou, China
| | - Yanqiu Li
- Gynaecology and Obstetrics Department, Maternal and Child Health Hospital of PanYu District, Guangzhou, China
| |
Collapse
|
17
|
Functional roles of long noncoding RNA MALAT1 in gynecologic cancers. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023; 25:48-65. [PMID: 36042115 DOI: 10.1007/s12094-022-02914-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 07/25/2022] [Indexed: 02/06/2023]
Abstract
Gynecologic cancers are reproductive disorders characterized by pelvic pain and infertility. The identification of new predictive markers and therapeutic targets for the treatment of gynecologic cancers is urgently necessary. One of the recent successes in gynecologic cancers research is identifying the role of signaling pathways in the pathogenesis of the disease. Recent experiments showed long noncoding RNAs (lncRNA) can be novel therapeutic approaches for the diagnosis and treatment of gynecologic cancers. LncRNA are transcribed RNA molecules that play pivotal roles in multiple biological processes by regulating the different steps of gene expression. Metastasis-associated lung adenocarcinoma transcript-1 (MALAT1) is a well-known lncRNA that plays functional roles in gene expression, RNA processing, and epigenetic regulation. High expression of MALAT1 is closely related to numerous human diseases. It is generally believed that MALAT1 expression is associated with cancer cell growth, autophagy, invasion, and metastasis. MALAT1 by targeting multiple signaling pathways and microRNAs (miRNAs) could contribute to the pathogenesis of gynecologic cancers. In this review, we will summarize functional roles of MALAT1 in the most common gynecologic cancers, including endometrium, breast, ovary, and cervix.
Collapse
|
18
|
Lih Yuan T, Sulaiman N, Nur Azurah AG, Maarof M, Rabiatul Adawiyah Razali, Yazid MD. Oestrogen-induced epithelial-mesenchymal transition (EMT) in endometriosis: Aetiology of vaginal agenesis in Mayer-Rokitansky-Küster-Hauser (MRKH) syndrome. Front Physiol 2022; 13:937988. [PMID: 36582359 PMCID: PMC9793092 DOI: 10.3389/fphys.2022.937988] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022] Open
Abstract
Endometriosis occurs when endometrial-like tissue forms and grows outside the uterus due to oestrogen-induced epithelial-mesenchymal transition in the female reproductive tract. Factors that suppress this event could become potential therapeutic agents against disease occurrence and progression. However, an overview of these studies is still lacking. This review assessed the impact of a number factors on oestrogen-mediated epithelial-mesenchymal transition in the emergence of several diseases in the female reproductive tract, primarily endometriosis. The association between epithelial-mesenchymal transition and Mayer-Rokitansky-Küster-Hauser (MRKH) syndrome was also investigated. Oestrogen, Wnt4 and epithelial-mesenchymal transition were chosen as keywords in Scopus, PubMed, and Web of Science searches performed on 28th June 2021. Study selection was refined to cancer-irrelevant, English, original articles published between years 2011-2021. The full-text assessment was carried out for topic-related articles after title and abstract screening. Included studies were summarised and assessed for their risk of bias using the Office of Health Assessment and Translation tool. In this review, 10 articles investigating oestrogen and epithelial-mesenchymal transition in the female reproductive tract were summarised and classified into two groups: seven studies under 'factor'-modulated epithelial-mesenchymal transition and three studies under 'factor'-manipulated oestrogen-induced epithelial-mesenchymal transition. The current evidence proposes that epithelial-mesenchymal transition is one of the prime causes of reproductive-related disease. This event could be mediated by distinct stimuli, specifically oestrogen and Wnt4 aberration. The results of this review suggest that oestrogen and Wnt4 participate in epithelial-mesenchymal transition in vaginal epithelial cells in MRKH syndrome, adopting from the theories of endometriosis development, which could therefore serve as a foundation for novel target treatment, specifically related to vaginal epithelialisation, to ensure better surgical outcomes.
Collapse
Affiliation(s)
- Too Lih Yuan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Cheras, Malaysia
| | - Nadiah Sulaiman
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Cheras, Malaysia
| | - Abdul Ghani Nur Azurah
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Cheras, Malaysia
| | - Manira Maarof
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Cheras, Malaysia
| | - Rabiatul Adawiyah Razali
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Cheras, Malaysia
| | - Muhammad Dain Yazid
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Cheras, Malaysia,*Correspondence: Muhammad Dain Yazid,
| |
Collapse
|
19
|
Shetty A, Venkatesh T, Kabbekodu SP, Tsutsumi R, Suresh PS. LncRNA-miRNA-mRNA regulatory axes in endometrial cancer: a comprehensive overview. Arch Gynecol Obstet 2022; 306:1431-1447. [PMID: 35182183 DOI: 10.1007/s00404-022-06423-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 01/24/2022] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Recent research on tumorigenesis and progression has opened up an array of novel molecular mechanisms in the form of interactions between cellular non-coding RNAs (long non-coding RNA[lncRNA]/microRNA [miRNA]) and coding transcripts that regulate health and disease. Endometrial cancer (EC) is a prominent gynecological malignancy with a high incidence rate and poorly known etiology and prognostic factors that hinder the success of disease management. The emerging role of lncRNA-miRNA-mRNA interactions and their dysregulation in the pathophysiology of EC has been elucidated in many recent studies. METHODS A thorough literature review was conducted to explore information about lncRNA-miRNA-mRNA axes in EC. RESULTS Several lncRNAs act as molecular sponges that sequester various tumor suppressor miRNAs to inhibit their function, leading to the dysregulation of their target mRNA transcripts that contribute to the EC regulation. CONCLUSIONS This review summarizes these networks of molecular mechanisms and their contribution to different aspects of endometrial carcinogenesis, leading to a better conceptualization of the molecular pathways that underlie the disease and helping establish novel diagnostic biomarkers and therapeutic intervention points to aid the curative intent of EC.
Collapse
Affiliation(s)
- Abhishek Shetty
- Department of Biosciences, Mangalore University, Mangalagangothri, Mangalore, 574 199, Karnataka, India
| | - Thejaswini Venkatesh
- Department of Biochemistry and Molecular Biology, Central University of Kerala, Kasargod, 671316, Kerala, India
| | - Shama Prasada Kabbekodu
- Department of Cell and Molecular Biology, School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Rie Tsutsumi
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15, Kuramoto-cho, Tokushima City, 770-8503, Japan
| | - Padmanaban S Suresh
- School of Biotechnology, National Institute of Technology, Calicut, 673601, Kerala, India.
| |
Collapse
|
20
|
Klicka K, Grzywa TM, Mielniczuk A, Klinke A, Włodarski PK. The role of miR-200 family in the regulation of hallmarks of cancer. Front Oncol 2022; 12:965231. [PMID: 36158660 PMCID: PMC9492973 DOI: 10.3389/fonc.2022.965231] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
MiRNAs are short non-coding RNAs that regulate gene expression post-transcriptionally contributing to the development of different diseases including cancer. The miR-200 family consists of five members, miR-200a, miR-200b, miR-200c, miR-141, and miR-429. Their expression is dysregulated in cancer tissue and their level is altered in the body fluids of cancer patients. Moreover, the levels of miR-200 family members correlate with clinical parameters such as cancer patients' survival which makes them potentially useful as diagnostic and prognostic biomarkers. MiRNAs can act as either oncomiRs or tumor suppressor miRNAs depending on the target genes and their role in the regulation of key oncogenic signaling pathways. In most types of cancer, the miR-200 family acts as tumor suppressor miRNA and regulates all features of cancer. In this review, we summarized the expression pattern of the miR-200 family in different types of cancer and their potential utility as biomarkers. Moreover, we comprehensively described the role of miR-200 family members in the regulation of all hallmarks of cancer proposed by Hanahan and Weinberg with the focus on the epithelial-mesenchymal transition, invasiveness, and metastasis of tumor cells.
Collapse
Affiliation(s)
- Klaudia Klicka
- Department of Methodology, Medical University of Warsaw, Warsaw, Poland
- Doctoral School, Medical University of Warsaw, Warsaw, Poland
| | - Tomasz M. Grzywa
- Department of Methodology, Medical University of Warsaw, Warsaw, Poland
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
- Laboratory of Experimental Medicine, Medical University of Warsaw, Warsaw, Poland
| | | | - Alicja Klinke
- Department of Methodology, Medical University of Warsaw, Warsaw, Poland
| | | |
Collapse
|
21
|
Cao Y, Ye D, Shen Z, Li Z, Li Q, Rong H. The Expression Profile, Clinical Application and Potential Tumor Suppressing Mechanism of hsa_circ_0001675 in Head and Neck Carcinoma. Front Oncol 2022; 12:769666. [PMID: 35600372 PMCID: PMC9121769 DOI: 10.3389/fonc.2022.769666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 04/07/2022] [Indexed: 11/13/2022] Open
Abstract
Purpose This study sought to identify circular RNAs (circRNA) that participate in the regulation of head and neck cancer (HNC), analyze their clinical application, and predict their molecular mechanism during HNC. Materials and Methods High-throughput sequencing was used to analyze circRNA expression in 18 matched HNC and adjacent normal tissues. Target circRNAs with significantly differential expression were obtained. In 103 HNC and adjacent normal tissues, real-time fluorescent quantitative PCR (qRT-PCR) was used to verify the differential expression of target circRNAs. This data was combined with clinicopathological information to analyze the diagnostic value of target circRNA. Bioinformatics was used to find target circRNAs that acted as competitive endogenous RNA (ceRNA) and construct a circRNA-miRNA-mRNA regulatory network. mRNA expression was verified by immunohistochemistry (IHC). Results A total of 714 differentially expressed circRNAs were detected in HNC, and the low expression of hsa_circ_0001675 was particularly significant (fold change [FC] = -4.85, P = 6.305E-05). hsa_circ_0001675 had significantly lower expression in HNC than in normal tissue (P < 0.01). Low hsa_circ_0001675 expression was positively associated with tumor invasion and clinical staging (P < 0.05), and its area under the ROC curve (AUC) was 0.7776. Low hsa_circ_0001675 expression also correlated with the overall survival (OS) rate and the progression-free survival (PFS) rate of HNC patients (P < 0.001). Bioinformatics was used to construct a ceRNA network of hsa_circ_0001675 with six differentially expressed miRNAs (hsa-miR-330-5p, hsa-miR-498, hsa-miR-532-3p, hsa-miR-577, hsa-miR-1248, and hsa-miR-1305) and 411 differentially expressed mRNAs and found that the neuroactive ligand-receptor interaction, and the cAMP and calcium signaling pathways were particularly enriched. Further bioinformatics and IHC analysis showed that miR577/TESC is the likely downstream signaling pathway for hsa_circ_0001675. Conclusion This study showed that hsa_circ_0001675 is downregulated in HNC and could be an effective biomarker for HNC diagnosis. In addition, hsa_circ_0001675 may have a potential ceRNA mechanism and suppress HNC disease progression through the hsa_circ_0001675-miRNA-mRNA axis.
Collapse
Affiliation(s)
- Yujie Cao
- Department of Otorhinolaryngology Head and Neck Surgery, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
- Department of Otorhinolaryngology Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital , Ningbo, China
- Medical School of Ningbo University, Ningbo, China
| | - Dong Ye
- Department of Otorhinolaryngology Head and Neck Surgery, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
- Department of Otorhinolaryngology Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital , Ningbo, China
| | - Zhisen Shen
- Department of Otorhinolaryngology Head and Neck Surgery, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
- Department of Otorhinolaryngology Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital , Ningbo, China
- *Correspondence: Zhisen Shen, ; Zan Li,
| | - Zan Li
- The Affiliated Cancer Hospital of Xiangya School of Medical, Central South University, Changsha, China
- *Correspondence: Zhisen Shen, ; Zan Li,
| | - Qun Li
- Department of Otorhinolaryngology Head and Neck Surgery, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
- Department of Otorhinolaryngology Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital , Ningbo, China
| | - Hao Rong
- Medical School of Ningbo University, Ningbo, China
| |
Collapse
|
22
|
Ye L, Pan K, Fang S, Wu SN, Chen S, Tang S, Wang N, Zhang H, Tong X, Shi X, Feng S, Xiang D, Zou R, Hu Y, Xue X, Guo G. Four Types of RNA Modification Writer-Related lncRNAs Are Effective Predictors of Prognosis and Immunotherapy Response in Serous Ovarian Carcinoma. Front Immunol 2022; 13:863484. [PMID: 35585970 PMCID: PMC9108167 DOI: 10.3389/fimmu.2022.863484] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/31/2022] [Indexed: 12/26/2022] Open
Abstract
Serous ovarian carcinoma (SOC) is a gynecological malignancy with high mortality rates. Currently, there is a lack of reliable biomarkers for accurate SOC patient prognosis. Here, we analyzed SOC RNA-Seq data from The Cancer Genome Atlas (TCGA) to identify prognostic biomarkers. Through the pearson correlation analysis, univariate Cox regression analysis, and LASSO-penalized Cox regression analysis, we identified nine lncRNAs significantly associated with four types of RNA modification writers (m6A, m1A, APA, and A-I) and with the prognosis of SOC patients (P <0.05). Six writer-related lncRNAs were ultimately selected following multivariate Cox analysis. We established a risk prediction model based on these six lncRNAs and evaluated its prognostic value in multiple groups (training set, testing set, and entire set). Our risk prediction model could effectively predict the prognosis of SOC patients with different clinical characteristics and their responses to immunotherapy. Lastly, we validated the predictive reliability and sensitivity of the lncRNA-based model via a nomogram. This study explored the association between RNA modification writer-related lncRNAs and SOC prognosis, providing a potential complement for the clinical management of SOC patients.
Collapse
Affiliation(s)
- Lele Ye
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Kan Pan
- First Clinical College, Wenzhou Medical University, Wenzhou, China
| | - Su Fang
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Su-Ni Wu
- Department of Gynecologic Oncology, Wenzhou Central Hospital, Wenzhou, China
| | - Su Chen
- Department of Gynecologic Oncology, Wenzhou Central Hospital, Wenzhou, China
| | - Sangsang Tang
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Nan Wang
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Haoke Zhang
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xinya Tong
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xinyu Shi
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Shiyu Feng
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Dan Xiang
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Ruanmin Zou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yingying Hu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiangyang Xue
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Gangqiang Guo
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
23
|
Lu X, Tan Q, Ma J, Zhang J, Yu P. Emerging Role of LncRNA Regulation for NLRP3 Inflammasome in Diabetes Complications. Front Cell Dev Biol 2022; 9:792401. [PMID: 35087834 PMCID: PMC8789514 DOI: 10.3389/fcell.2021.792401] [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: 10/11/2021] [Accepted: 12/01/2021] [Indexed: 12/13/2022] Open
Abstract
Diabetes is a widespread metabolic disease with various complications, including diabetic nephropathy, retinopathy, cardiomyopathy, and other cardiovascular or cerebrovascular diseases. As the prevalence of diabetes increases in all age groups worldwide, diabetes and its complications cause an emerging public health burden. NLRP3 inflammasome is a complex of several proteins that play a critical role in inflammatory response and various diseases, including diabetes and its complications. Accumulating evidences indicate that NLRP3 inflammasome contributes to the development of diabetes and diabetic complications and that NLRP3 inflammation inactivation is beneficial in treating these illnesses. Emerging evidences suggest the critical role of long non-coding RNAs (lncRNAs) in regulating NLRP3 inflammasome activity in various diseases. LncRNAs are non-coding RNAs exceeding 200 nucleotides in length. Its dysregulation has been linked to the development of diseases, including diabetes. Recently, growing evidences hint that regulating lncRNAs on NLRP3 inflammasome is critical in developing and progressing diabetes and diabetic complications. Here, we discuss the role of lncRNAs in regulating NLRP3 inflammasome as well as its participation in diabetes and diabetic complications, providing novel insights into developing future therapeutic approaches for diabetes.
Collapse
Affiliation(s)
- Xiaolin Lu
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qihong Tan
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianyong Ma
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Jing Zhang
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Peng Yu
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
24
|
The Multifunctional Faces of T-Cell Intracellular Antigen 1 in Health and Disease. Int J Mol Sci 2022; 23:ijms23031400. [PMID: 35163320 PMCID: PMC8836218 DOI: 10.3390/ijms23031400] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/13/2022] [Accepted: 01/22/2022] [Indexed: 02/06/2023] Open
Abstract
T-cell intracellular antigen 1 (TIA1) is an RNA-binding protein that is expressed in many tissues and in the vast majority of species, although it was first discovered as a component of human cytotoxic T lymphocytes. TIA1 has a dual localization in the nucleus and cytoplasm, where it plays an important role as a regulator of gene-expression flux. As a multifunctional master modulator, TIA1 controls biological processes relevant to the physiological functioning of the organism and the development and/or progression of several human pathologies. This review summarizes our current knowledge of the molecular aspects and cellular processes involving TIA1, with relevance for human pathophysiology.
Collapse
|
25
|
Wu J, Wu Y, Guo Q, Wang S, Wu X. RNA-binding proteins in ovarian cancer: a novel avenue of their roles in diagnosis and treatment. J Transl Med 2022; 20:37. [PMID: 35062979 PMCID: PMC8783520 DOI: 10.1186/s12967-022-03245-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 01/11/2022] [Indexed: 12/18/2022] Open
Abstract
Ovarian cancer (OC), an important cause of cancer-related death in women worldwide, is one of the most malignant cancers and is characterized by a poor prognosis. RNA-binding proteins (RBPs), a class of endogenous proteins that can bind to mRNAs and modify (or even determine) the amount of protein they can generate, have attracted great attention in the context of various diseases, especially cancers. Compelling studies have suggested that RBPs are aberrantly expressed in different cancer tissues and cell types, including OC tissues and cells. More specifically, RBPs can regulate proliferation, apoptosis, invasion, metastasis, tumorigenesis and chemosensitivity and serve as potential therapeutic targets in OC. Herein, we summarize what is currently known about the biogenesis, molecular functions and potential roles of human RBPs in OC and their prospects for application in the clinical treatment of OC.
Collapse
Affiliation(s)
- Jiangchun Wu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, People's Republic of China
| | - Yong Wu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, People's Republic of China
| | - Qinhao Guo
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, People's Republic of China
| | - Simin Wang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, People's Republic of China
| | - Xiaohua Wu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
26
|
A diagnostic and prognostic value of blood-based circulating long non-coding RNAs in Thyroid, Pancreatic and Ovarian Cancer. Crit Rev Oncol Hematol 2022; 171:103598. [PMID: 35033662 DOI: 10.1016/j.critrevonc.2022.103598] [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] [Received: 07/09/2021] [Revised: 01/12/2022] [Accepted: 01/12/2022] [Indexed: 12/12/2022] Open
Abstract
Several studies have demonstrated the potential of circulating long non-coding RNAs (lncRNAs) as promising cancer biomarkers. Herein, we addressed the regulatory role of circulating lncRNAs and their potential value as diagnostic/prognostic markers for thyroid, pancreatic and ovarian cancers. Furthermore, we analyzed and measured the clinical implications and association of lncRNAs with sensitivity, specificity, and area under the ROC curve (AUC). Based on our meta-analysis, we found that GAS8-AS1 could discriminate thyroid cancer from non-cancer and other cancers with higher accuracy (AUC = 0.746; sensitivity = 61.70%, and specificity = 90.00%). Similarly, for ovarian cancer, lncRNA RP5-837J1.2 was found to have ideal diagnostic potential with critical clinical specifications of AUC = 0.996; sensitivity = 97.30% and specificity = 94.60%. Whereas we could not find any lncRNA having high diagnostic/prognostic efficiency in pancreatic cancer. We believe that lncRNAs mentioned above may explore clinical settings for the diagnosis and prognosis of cancer patients.
Collapse
|
27
|
Shen J, Yuan Z, Sheng J, Feng X, Wang H, Wang Y, Zhou Y. Long non-coding RNA NNT-AS1 positively regulates NPM1 expression to affect the proliferation of estrogen-mediated endometrial carcinoma by interacting. J Cancer 2022; 13:112-123. [PMID: 34976175 PMCID: PMC8692688 DOI: 10.7150/jca.62630] [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] [Received: 05/11/2021] [Accepted: 10/23/2021] [Indexed: 11/05/2022] Open
Abstract
Objective: This study aims to investigate the mechanism of long non-coding RNA NNT-AS1 in the proliferation of estrogen-mediated endometrial carcinoma (EC). Materials and methods: NNT-AS1, miR-30c, and Nucleophosmin 1 (NPM1) expressions were measured by quantitative real-time PCR and Western blotting. Cell Counting Kit-8 assay and 5-Ethynyl-2'-deoxyuridine (EdU) assay were used to detect the viability and proliferation of Ishikawa and HEC-1-A cells, respectively. RNA immunoprecipitation assay was used to confirm the interaction between NNT-AS1 and miR-30c. Luciferase reporter assay was performed to confirm the interaction between miR-30c and NPM1. Results: NNT-AS1 and NPM1 expressions in EC tissues and cell lines were higher than in benign endometrium and normal endometrial epithelial cells (EECs). miR-30c expression in EC tissues and cell lines was lower than in benign endometrium and normal EECs. NNT-AS1 interacted with miR-30c, and miR-30c negatively regulated NPM1 expression. Overexpression of NNT-AS1 increased NPM1 expression in EC cells, while overexpression of miR-30c reversed the effect. NNT-AS1 interference inhibited the mRNA level of NPM1, while the miR-30c inhibitor reversed the result. Estradiol (E2) promoted the proliferation of EC cells, small interfering RNA (siRNA) against NNT-AS1 inhibited EC cell proliferation, miR-30c inhibitor promoted cell proliferation, and NPM1 siRNA inhibited cell proliferation. E2 increased tumor volume, and NNT-AS1 interference reduced tumor volume in vivo. Conclusion: NNT-AS1 promoted the proliferation of estrogen-mediated EC by regulating miR-30c/NPM1.
Collapse
Affiliation(s)
- Jie Shen
- Department of Gynecology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhilin Yuan
- Department of Obstetrics and Gynecology, the Fourth Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jingjing Sheng
- Department of Obstetrics and Gynecology, the Fourth Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaoping Feng
- Department of Gynecology, Yiwu Central Hospital, Yiwu, Zhejiang, China
| | - Hao Wang
- Department of Gynecology, Yiwu Central Hospital, Yiwu, Zhejiang, China
| | - Yanli Wang
- Department of Pathology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yunxiao Zhou
- Department of Gynecology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
28
|
Metastatic EMT Phenotype Is Governed by MicroRNA-200-Mediated Competing Endogenous RNA Networks. Cells 2021; 11:cells11010073. [PMID: 35011635 PMCID: PMC8749983 DOI: 10.3390/cells11010073] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 12/12/2022] Open
Abstract
Epithelial–mesenchymal transition (EMT) is a fundamental physiologically relevant process that occurs during morphogenesis and organ development. In a pathological setting, the transition from epithelial toward mesenchymal cell phenotype is hijacked by cancer cells, allowing uncontrolled metastatic dissemination. The competing endogenous RNA (ceRNA) hypothesis proposes a competitive environment resembling a large-scale regulatory network of gene expression circuits where alterations in the expression of both protein-coding and non-coding genes can make relevant contributions to EMT progression in cancer. The complex regulatory diversity is exerted through an array of diverse epigenetic factors, reaching beyond the transcriptional control that was previously thought to single-handedly govern metastatic dissemination. The present review aims to unravel the competitive relationships between naturally occurring ceRNA transcripts for the shared pool of the miRNA-200 family, which play a pivotal role in EMT related to cancer dissemination. Upon acquiring more knowledge and clinical evidence on non-genetic factors affecting neoplasia, modulation of the expression levels of diverse ceRNAs may allow for the development of novel prognostic/diagnostic markers and reveal potential targets for the disruption of cancer-related EMT.
Collapse
|
29
|
Naz F, Tariq I, Ali S, Somaida A, Preis E, Bakowsky U. The Role of Long Non-Coding RNAs (lncRNAs) in Female Oriented Cancers. Cancers (Basel) 2021; 13:6102. [PMID: 34885213 PMCID: PMC8656502 DOI: 10.3390/cancers13236102] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/14/2021] [Accepted: 11/30/2021] [Indexed: 12/15/2022] Open
Abstract
Recent advances in molecular biology have discovered the mysterious role of long non-coding RNAs (lncRNAs) as potential biomarkers for cancer diagnosis and targets for advanced cancer therapy. Studies have shown that lncRNAs take part in the incidence and development of cancers in humans. However, previously they were considered as mere RNA noise or transcription byproducts lacking any biological function. In this article, we present a summary of the progress on ascertaining the biological functions of five lncRNAs (HOTAIR, NEAT1, H19, MALAT1, and MEG3) in female-oriented cancers, including breast and gynecological cancers, with the perspective of carcinogenesis, cancer proliferation, and metastasis. We provide the current state of knowledge from the past five years of the literature to discuss the clinical importance of such lncRNAs as therapeutic targets or early diagnostic biomarkers. We reviewed the consequences, either oncogenic or tumor-suppressing features, of their aberrant expression in female-oriented cancers. We tried to explain the established mechanism by which they regulate cancer proliferation and metastasis by competing with miRNAs and other mechanisms involved via regulating genes and signaling pathways. In addition, we revealed the association between stated lncRNAs and chemo-resistance or radio-resistance and their potential clinical applications and future perspectives.
Collapse
Affiliation(s)
- Faiza Naz
- Punjab University College of Pharmacy, Allama Iqbal Campus, University of the Punjab, Lahore 54000, Pakistan;
| | - Imran Tariq
- Punjab University College of Pharmacy, Allama Iqbal Campus, University of the Punjab, Lahore 54000, Pakistan;
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, 35037 Marburg, Germany or (S.A.); (A.S.); (E.P.)
| | - Sajid Ali
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, 35037 Marburg, Germany or (S.A.); (A.S.); (E.P.)
- Angström Laboratory, Department of Chemistry, Uppsala University, 75123 Uppsala, Sweden
| | - Ahmed Somaida
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, 35037 Marburg, Germany or (S.A.); (A.S.); (E.P.)
| | - Eduard Preis
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, 35037 Marburg, Germany or (S.A.); (A.S.); (E.P.)
| | - Udo Bakowsky
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, 35037 Marburg, Germany or (S.A.); (A.S.); (E.P.)
| |
Collapse
|
30
|
Pan L, Du M, Liu H, Cheng B, Zhu M, Jia B, Wang Y, He W, Li X, Liu C, Gu J, Li M, Zhang Y, Yao L, Zhang Y, Hao Q. LncRNA FTX promotes the malignant progression of colorectal cancer by regulating the miR-214-5p-JAG1 axis. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1369. [PMID: 34733921 PMCID: PMC8506562 DOI: 10.21037/atm-21-2755] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 07/19/2021] [Indexed: 12/22/2022]
Abstract
Background Long non-coding RNAs (lncRNAs) have recently been found to be vital regulators of various cancers, including colorectal cancer (CRC). It has been previously reported that the dysregulated expression of lncRNA Five prime to Xist (FTX) is involved in carcinogenesis. However, the role of lncRNA FTX in the progression of CRC is still unclear. Methods Fluorescence in situ hybridization (FISH) was used to detect the expression of lncRNA FTX and miR-214-5p in CRC tissues. Cell Counting Kit-8 assay, transwell assay, wound-healing assay, and proliferation assay were used to explore the function of lncRNA FTX in CRC cells. Quantitative real-time polymerase chain reaction (qRT-PCR), western blotting, and luciferase reporter assay were used to confirm the relationship between lncRNA FTX and miR-214-5p-jagged canonical Notch ligand 1 (JAG1). We further explored the role of lncRNA FTX in vivo using xenograft tumor assay. Results lncRNA FTX was found to be upregulated in CRC tissues by FISH. The downregulation of endogenous lncRNA FTX expression inhibited CRC cell proliferation, migration, and invasion. Mechanistically, lncRNA FTX sequestered miR-214-5p and thus released its repression on JAG1, driving the malignant progression of CRC. Conclusions These findings give rise to a new perspective, the lncRNA FTX-miR-214-5p-JAG1 regulatory axis, in exploring the cancer-promoting mechanism of lncRNA FTX in CRC.
Collapse
Affiliation(s)
- Luxiang Pan
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Mingrui Du
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, China.,School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Haixia Liu
- Department of Gynecology and Obstetrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Boyang Cheng
- School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Maorong Zhu
- School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Bo Jia
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yinwen Wang
- School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Wei He
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Xiaoju Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Chenlin Liu
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Jintao Gu
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Meng Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Yingqi Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Li Yao
- Department of Pathology, Xi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, China
| | - Yi Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Qiang Hao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
31
|
Sabol M, Calleja-Agius J, Di Fiore R, Suleiman S, Ozcan S, Ward MP, Ozretić P. (In)Distinctive Role of Long Non-Coding RNAs in Common and Rare Ovarian Cancers. Cancers (Basel) 2021; 13:5040. [PMID: 34680193 PMCID: PMC8534192 DOI: 10.3390/cancers13205040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/04/2021] [Accepted: 10/06/2021] [Indexed: 02/05/2023] Open
Abstract
Rare ovarian cancers (ROCs) are OCs with an annual incidence of fewer than 6 cases per 100,000 women. They affect women of all ages, but due to their low incidence and the potential clinical inexperience in management, there can be a delay in diagnosis, leading to a poor prognosis. The underlying causes for these tumors are varied, but generally, the tumors arise due to alterations in gene/protein expression in cellular processes that regulate normal proliferation and its checkpoints. Dysregulation of the cellular processes that lead to cancer includes gene mutations, epimutations, non-coding RNA (ncRNA) regulation, posttranscriptional and posttranslational modifications. Long non-coding RNA (lncRNA) are defined as transcribed RNA molecules, more than 200 nucleotides in length which are not translated into proteins. They regulate gene expression through several mechanisms and therefore add another level of complexity to the regulatory mechanisms affecting tumor development. Since few studies have been performed on ROCs, in this review we summarize the mechanisms of action of lncRNA in OC, with an emphasis on ROCs.
Collapse
Affiliation(s)
- Maja Sabol
- Laboratory for Hereditary Cancer, Division of Molecular Medicine, Ruđer Bošković Institute, HR-10000 Zagreb, Croatia;
| | - Jean Calleja-Agius
- Department of Anatomy, Faculty of Medicine and Surgery, University of Malta, MSD 2080 Msida, Malta; (J.C.-A.); (R.D.F.); (S.S.)
| | - Riccardo Di Fiore
- Department of Anatomy, Faculty of Medicine and Surgery, University of Malta, MSD 2080 Msida, Malta; (J.C.-A.); (R.D.F.); (S.S.)
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Sherif Suleiman
- Department of Anatomy, Faculty of Medicine and Surgery, University of Malta, MSD 2080 Msida, Malta; (J.C.-A.); (R.D.F.); (S.S.)
| | - Sureyya Ozcan
- Department of Chemistry, Middle East Technical University (METU), 06800 Ankara, Turkey;
- Cancer Systems Biology Laboratory (CanSyl), Middle East Technical University (METU), 06800 Ankara, Turkey
| | - Mark P. Ward
- Department of Histopathology, Trinity St James’s Cancer Institute, Emer Casey Molecular Pathology Laboratory, Trinity College Dublin and Coombe Women’s and Infants University Hospital, D08 RX0X Dublin, Ireland;
| | - Petar Ozretić
- Laboratory for Hereditary Cancer, Division of Molecular Medicine, Ruđer Bošković Institute, HR-10000 Zagreb, Croatia;
| |
Collapse
|
32
|
Xie W, Sun H, Li X, Lin F, Wang Z, Wang X. Ovarian cancer: epigenetics, drug resistance, and progression. Cancer Cell Int 2021; 21:434. [PMID: 34404407 PMCID: PMC8369623 DOI: 10.1186/s12935-021-02136-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 08/03/2021] [Indexed: 03/05/2023] Open
Abstract
Ovarian cancer (OC) is one of the most common malignant tumors in women. OC is associated with the activation of oncogenes, the inactivation of tumor suppressor genes, and the activation of abnormal cell signaling pathways. Moreover, epigenetic processes have been found to play an important role in OC tumorigenesis. Epigenetic processes do not change DNA sequences but regulate gene expression through DNA methylation, histone modification, and non-coding RNA. This review comprehensively considers the importance of epigenetics in OC, with a focus on microRNA and long non-coding RNA. These types of RNA are promising molecular markers and therapeutic targets that may support precision medicine in OC. DNA methylation inhibitors and histone deacetylase inhibitors may be useful for such targeting, with a possible novel approach combining these two therapies. Currently, the clinical application of such epigenetic approaches is limited by multiple obstacles, including the heterogeneity of OC, insufficient sample sizes in reported studies, and non-optimized methods for detecting potential tumor markers. Nonetheless, the application of epigenetic approaches to OC patient diagnosis, treatment, and prognosis is a promising area for future clinical investigation.
Collapse
Affiliation(s)
- Weiwei Xie
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University School of Medicine Xinhua Hospital, 1665 Kongjiang Road, Yangpu District, Shanghai, China
| | - Huizhen Sun
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University School of Medicine Xinhua Hospital, 1665 Kongjiang Road, Yangpu District, Shanghai, China
| | - Xiaoduan Li
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Feikai Lin
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University School of Medicine Xinhua Hospital, 1665 Kongjiang Road, Yangpu District, Shanghai, China
| | - Ziliang Wang
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University School of Medicine Xinhua Hospital, 1665 Kongjiang Road, Yangpu District, Shanghai, China.
| | - Xipeng Wang
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University School of Medicine Xinhua Hospital, 1665 Kongjiang Road, Yangpu District, Shanghai, China.
| |
Collapse
|
33
|
Cavaliere AF, Perelli F, Zaami S, Piergentili R, Mattei A, Vizzielli G, Scambia G, Straface G, Restaino S, Signore F. Towards Personalized Medicine: Non-Coding RNAs and Endometrial Cancer. Healthcare (Basel) 2021; 9:965. [PMID: 34442102 PMCID: PMC8393611 DOI: 10.3390/healthcare9080965] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 07/24/2021] [Accepted: 07/26/2021] [Indexed: 12/12/2022] Open
Abstract
Endometrial cancer (EC) is the most frequent female cancer associated with excellent prognosis if diagnosed at an early stage. The risk factors on which clinical staging is based are constantly updated and genetic and epigenetic characteristics have recently been emerging as prognostic markers. The evidence shows that non-coding RNAs (ncRNAs) play a fundamental role in various biological processes associated with the pathogenesis of EC and many of them also have a prognosis prediction function, of remarkable importance in defining the therapeutic and surveillance path of EC patients. Personalized medicine focuses on the continuous updating of risk factors that are identifiable early during the EC staging to tailor treatments to patients. This review aims to show a summary of the current classification systems and to encourage the integration of various risk factors, introducing the prognostic role of non-coding RNAs, to avoid aggressive therapies where not necessary and to treat and strictly monitor subjects at greater risk of relapse.
Collapse
Affiliation(s)
- Anna Franca Cavaliere
- Azienda USL Toscana Centro, Gynecology and Obstetric Department, Santo Stefano Hospital, 59100 Prato, Italy;
| | - Federica Perelli
- Azienda USL Toscana Centro, Gynecology and Obstetric Department, Santa Maria Annunziata Hospital, 50012 Florence, Italy;
| | - Simona Zaami
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00161 Roma, Italy;
| | - Roberto Piergentili
- Institute of Molecular Biology and Pathology, Italian National Research Council (CNR-IBPM), 00185 Rome, Italy;
| | - Alberto Mattei
- Azienda USL Toscana Centro, Gynecology and Obstetric Department, Santa Maria Annunziata Hospital, 50012 Florence, Italy;
| | - Giuseppe Vizzielli
- Gynecologic Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (G.V.); (G.S.)
- Obstetrics, Gynecology and Pediatrics Department, Udine University Hospital, DAME, 33100 Udine, Italy;
| | - Giovanni Scambia
- Gynecologic Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (G.V.); (G.S.)
| | - Gianluca Straface
- Division of Perinatal Medicine, Policlinico Abano Terme, 35031 Abano Terme, Italy;
| | - Stefano Restaino
- Obstetrics, Gynecology and Pediatrics Department, Udine University Hospital, DAME, 33100 Udine, Italy;
| | - Fabrizio Signore
- Obstetrics and Gynecology Department, USL Roma2, Sant’Eugenio Hospital, 00144 Rome, Italy;
| |
Collapse
|
34
|
lncRNA OIP5-AS1 Suppresses Cell Proliferation and Invasion of Endometrial Cancer by Regulating PTEN/AKT via Sponging miR-200c-3p. J Immunol Res 2021; 2021:4861749. [PMID: 34368370 PMCID: PMC8342140 DOI: 10.1155/2021/4861749] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 07/05/2021] [Indexed: 11/17/2022] Open
Abstract
Background Endometrial carcinoma (EC) is one of the major gynecologic malignancy cancers affecting females with dismal prognosis and high mortality around the world. Numerous studies have proven that an aberrant level of long noncoding RNAs is present in many endometrial cancer patients, while the underlying molecular mechanism remains unclear. Method The expression levels of lncRNA OIP5-AS1, miR200c-3p, and PTEN were measured by a quantitative real-time polymerase chain reaction in endometrial cancer tissue and endometrial cancer cells. CCK8 assay, wound-healing assay, and cell colony formation were applied to evaluate cell proliferation, cell migration, and cell colony formation ability. Cell cycle and cell apoptosis were detected by flow cytometry. The interactions between OIP5-AS1, miR200c-3p, and PTEN were explored by luciferase activity. Results In the present study, we demonstrated that long noncoding RNA OIP5-AS1 was significantly reduced in EC tissue compared with normal tissue. The lower expression level of OIP5-AS1 was also confirmed in four kinds of EC cell lines compared with the normal endometrial cell line. Gain- and loss-of-function of experiments indicated that upregulation of OIP5-AS1 could inhibit the proliferation, migration, and invasion of EC cells in vitro. Meanwhile, overexpression of OIP5-AS1 could also suppress the growth of tumor in the xenograft model. Moreover, further study revealed that miR-200c-3p could bind to OIP5-AS1, and the loss function of miR-200c-3p could reverse the elevated OIP5-AS1's inhibitory effect on the progression of EC. Furthermore, we found that downregulation of miR-200c-3p was inversely correlated with PTEN expression in EC cells. Reduced OIP5-AS1 could lead to the accumulation of miR-200c-3p, which could induce the upregulation of PTEN indirectly. Conclusion Our study demonstrated a novel molecular mechanism that lncRNA OIP5-AS1 could modulate the progression of EC by combining competitively with miR-200c-3p to control the PTEN/AKT pathway in EC cells, which might supply important information for developing novel therapeutic strategies for EC patients.
Collapse
|
35
|
Tan X, Guo W, Peng Z, Gu C, Xiang P, Tu Y, Fei H, Liu X, Lu Y, Li M, Wang H, Luo Y, Yang J. LncRNA-Malat1 promoting inflammatory response aggravates cerebral ischemia-reperfusion neuronal injury in rats by targeting miR-211-5p. Biochem Pharmacol 2021; 192:114694. [PMID: 34324865 DOI: 10.1016/j.bcp.2021.114694] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 07/16/2021] [Accepted: 07/16/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Ischemic stroke is one kind of disorder of cerebral blood circulation, which poses a severe threaten to human health and quality of life, causing serious economic loss to families and society. LncRNA-Malat1(Metastasis-associated lung adenocarcinoma transcript 1) and miRNA-211-5p are abnormally expressed in stroke patients and in the middle cerebral ischemia-reperfusion injury (CIRI) model. However, the involvement between LncRNA-Malat1 and miR-211-5p in cerebral ischemia-reperfusion neuronal injury and its mechanism remain unclear. METHODS The middle cerebral ischemia-reperfusion injury (CIRI) model was established in rat, and the rat primary neuron injury model induced by oxygen-glucose deprivation/reoxygenation (OGD/R) was simulated in vitro. Level of LncRNA-Malat1, miR-211-5p and COX-2 mRNA were detected via RT-qPCR. The protein levels of COX-2, BAX and BCL-2 were measured by western blot. MTT assay was performed to detect cell viability. RESULTS In our study, LncRNA-Malat1 and COX-2 were up-regulated while miR-211-5p down-regulated in vitro and in vivo. Knockdown of LncRNA-Malat1 improved neurological deficit scores, reduced cerebral infarction volume in vivo, and significantly promoted cell viability, reduced apoptosis and proinflammatory factors PGE2 and IL-10 in vitro. More importantly, the level of miR-211-5p was negatively correlated with that of LncRNA-Malat1 and COX-2. Knockdown of LncRNA-Malat1 inhibited the expression of COX-2 by up-regulating miR-211-5p. CONCLUSION Higher expression of LncRNA-Malat1 was involved in neuronal injury, LncRNA-Malat1 knockdown may reduce inflammation and apoptosis by modulating miR-211-5p, which provided a new therapeutic strategy for clinical stroke intervention targets.
Collapse
Affiliation(s)
- Xiaodan Tan
- Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology, Chongqing Medical University, Chongqing 400016, China
| | - Wenjia Guo
- Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology, Chongqing Medical University, Chongqing 400016, China
| | - Zhe Peng
- Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology, Chongqing Medical University, Chongqing 400016, China
| | - Chao Gu
- Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology, Chongqing Medical University, Chongqing 400016, China
| | - Pu Xiang
- Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology, Chongqing Medical University, Chongqing 400016, China; Dianjiang People's Hospital of Chongqing, Dianjiang, Chongqing 408300, China
| | - Yunjun Tu
- Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology, Chongqing Medical University, Chongqing 400016, China
| | - Huizhi Fei
- Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology, Chongqing Medical University, Chongqing 400016, China
| | - Xia Liu
- Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology, Chongqing Medical University, Chongqing 400016, China
| | - Yi Lu
- Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology, Chongqing Medical University, Chongqing 400016, China
| | - Miaomiao Li
- Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology, Chongqing Medical University, Chongqing 400016, China
| | - Hong Wang
- Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology, Chongqing Medical University, Chongqing 400016, China
| | - Ying Luo
- Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology, Chongqing Medical University, Chongqing 400016, China
| | - Junqing Yang
- Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
36
|
Fu D, Zang L, Li Z, Fan C, Jiang H, Men T. Long non-coding RNA CRNDE regulates the growth and migration of prostate cancer cells by targeting microRNA-146a-5p. Bioengineered 2021; 12:2469-2479. [PMID: 34232111 PMCID: PMC8806644 DOI: 10.1080/21655979.2021.1935402] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The function of lncRNA CRNDE and its role in prostate cancer (PC) remains unclear. The aim of this study was to determine the expression level of lncRNA CRNDE in PC tissues and to elucidate its role in PC. The expression levels of lncRNA CRNDE were measured by quantitative reverse transcription polymerase chain reaction. The role of lncRNA CRNDE in PC cells was studied using loss-of-function assays in vitro. Cell proliferation, migration, invasion, and apoptosis were assessed via Cell Counting Kit-8, colony formation, flow cytometry, wound healing, and transwell chamber assays. A luciferase reporter assay was used to characterize the interaction between lncRNA CRNDE and miR-146a-5p. In PC tissues, the expression level of lncRNA CRNDE was upregulated. Moreover, knockdown of lncRNA CRNDE suppressed PC cell proliferation and migration and induced apoptosis in vitro. miR-146a-5p was verified as a direct target of lncRNA CRNDE. Moreover, the inhibition of miR-146a-5p partially counteracted the effects of lncRNA CRNDE on PC cell proliferation, migration, and invasion. In conclusion, lncRNA CRNDE may serve as a cancer promoter in PC by targeting miR-146a-5p. Therefore, lncRNA CRNDE could be a promising target for the clinical treatment of PC.
Collapse
Affiliation(s)
- Dewang Fu
- Department of Urology Surgery, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Li'e Zang
- Department of Urology Surgery, The First Affiliate Hospital of Jinzhou Medical University, Jinzhou, China
| | - Zhaowei Li
- Department of Urology Surgery, The First Affiliate Hospital of Jinzhou Medical University, Jinzhou, China
| | - Chenghui Fan
- Department of Urology Surgery, The First Affiliate Hospital of Jinzhou Medical University, Jinzhou, China
| | - Huamao Jiang
- Department of Urology Surgery, The First Affiliate Hospital of Jinzhou Medical University, Jinzhou, China
| | - Tongyi Men
- Department of Urology Surgery, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
37
|
Klicka K, Grzywa TM, Klinke A, Mielniczuk A, Włodarski PK. The Role of miRNAs in the Regulation of Endometrial Cancer Invasiveness and Metastasis-A Systematic Review. Cancers (Basel) 2021; 13:3393. [PMID: 34298609 PMCID: PMC8304659 DOI: 10.3390/cancers13143393] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/18/2021] [Accepted: 07/02/2021] [Indexed: 12/17/2022] Open
Abstract
Endometrial cancer (EC) is the most common genital cancer in women with increasing death rates. MiRNAs are short non-coding RNAs that regulate gene expression on the post-transcriptional levels. Multiple studies demonstrated a fundamental role of miRNAs in the regulation of carcinogenesis. This systematic review is a comprehensive overview of the role of miRNAs in the regulation of cancer cell invasiveness and metastasis in EC. The literature was searched for studies investigating the role of miRNAs in the regulation of invasiveness and metastasis in EC. We explored PubMed, Embase, and Scopus using the following keywords: miRNA, metastasis, invasiveness, endometrial cancer. Data were collected from 163 articles that described the expression and role of 106 miRNAs in the regulation of EC invasiveness and metastasis out of which 63 were tumor suppressor miRNAs, and 38 were oncomiRNAs. Five miRNAs had a discordant role in different studies. Moreover, we identified 66 miRNAs whose expression in tumor tissue or concentration in serum correlated with at least one clinical parameter. These findings suggest a crucial role of miRNAs in the regulation of EC invasiveness and metastasis and present them as potential prognostic factors for patients with EC.
Collapse
Affiliation(s)
- Klaudia Klicka
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (K.K.); (T.M.G.); (A.K.); (A.M.)
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Tomasz M. Grzywa
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (K.K.); (T.M.G.); (A.K.); (A.M.)
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Alicja Klinke
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (K.K.); (T.M.G.); (A.K.); (A.M.)
| | - Aleksandra Mielniczuk
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (K.K.); (T.M.G.); (A.K.); (A.M.)
| | - Paweł K. Włodarski
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (K.K.); (T.M.G.); (A.K.); (A.M.)
| |
Collapse
|
38
|
Zhou X, Chen Z, Pei L, Sun J. MicroRNA miR-106a-5p targets forkhead box transcription factor FOXC1 to suppress the cell proliferation, migration, and invasion of ectopic endometrial stromal cells via the PI3K/Akt/mTOR signaling pathway. Bioengineered 2021; 12:2203-2213. [PMID: 34082653 PMCID: PMC8806537 DOI: 10.1080/21655979.2021.1933679] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Emerging evidence has exhibited an obvious decreased expression of miR-106a-5p in the ectopic endometrial tissue of endometriosis (EMS) patients. Thus far, the pathophysiological function of miR-106a-5p in EMS is unknown. A previous study showed an increased FOXC1 expression in the ectopic endometrial tissue of patients with EMS. Moreover, we found that there was a binding site of miR-106a-5p on the 3'UTR of FOXC1 through bioinformatics predictions. Hence, we speculated that miR-106a-5p might affect the development of EMS via targeting FOXC1. We first showed a decreased level of miR-106a-5p and an increased level of FOXC1 mRNA in ectopic endometrial tissues compared with normal tissues. Functionally, we transfected ectopic endometrial stromal cells (ESCs) with miR-106a-5p mimics or NC mimics and indicated an inhibitory role of miR-106a-5p on ESC proliferation, invasion, and migration. Mechanistically, FOXC1 was found to be a target gene of miR-106a-5p. To confirm whether miR-106a-5p exerted an inhibitory activity in ESCs via targeting FOXC1, miR-106a-5p mimic was co-transfected into ESCs with the FOXC1-plasmid or vector. We found that FOXC1 overexpression evidently reversed the results caused by a miR-106a-5p mimic in ESCs. Additionally, our results demonstrated that miR-106a-5p mimic inhibited the expression of p-Akt and p-PI3K. Collectively, these results revealed that miR-106a-5p inhibited the proliferative, migratory, and invasive ability of ESCs via directly binding to FOXC1, likely through the suppression of the PI3K and its downstream signaling pathway, which offered a potential and novel therapeutic strategy for EMS treatment.
Collapse
Affiliation(s)
- Xinyue Zhou
- Department of Obstetrics and Gynecology, The General Hospital of Northern Theater Command, Shenyang, People's Republic of China
| | - Zhenyu Chen
- Department of Obstetrics and Gynecology, The General Hospital of Northern Theater Command, Shenyang, People's Republic of China
| | - Lipeng Pei
- Department of Obstetrics and Gynecology, The General Hospital of Northern Theater Command, Shenyang, People's Republic of China
| | - Jingli Sun
- Department of Obstetrics and Gynecology, The General Hospital of Northern Theater Command, Shenyang, People's Republic of China
| |
Collapse
|
39
|
Zhang Z, Gu M, Gu Z, Lou YR. Role of Long Non-Coding RNA Polymorphisms in Cancer Chemotherapeutic Response. J Pers Med 2021; 11:jpm11060513. [PMID: 34199840 PMCID: PMC8228201 DOI: 10.3390/jpm11060513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 05/28/2021] [Accepted: 06/02/2021] [Indexed: 12/12/2022] Open
Abstract
Genetic polymorphisms are defined as the presence of two or more different alleles in the same locus, with a frequency higher than 1% in the population. Since the discovery of long non-coding RNAs (lncRNAs), which refer to a non-coding RNA with a length of more than 200 nucleotides, their biological roles have been increasingly revealed in recent years. They regulate many cellular processes, from pluripotency to cancer. Interestingly, abnormal expression or dysfunction of lncRNAs is closely related to the occurrence of human diseases, including cancer and degenerative neurological diseases. Particularly, their polymorphisms have been found to be associated with altered drug response and/or drug toxicity in cancer treatment. However, molecular mechanisms are not yet fully elucidated, which are expected to be discovered by detailed studies of RNA–protein, RNA–DNA, and RNA–lipid interactions. In conclusion, lncRNAs polymorphisms may become biomarkers for predicting the response to chemotherapy in cancer patients. Here we review and discuss how gene polymorphisms of lncRNAs affect cancer chemotherapeutic response. This knowledge may pave the way to personalized oncology treatments.
Collapse
Affiliation(s)
- Zheng Zhang
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China;
| | - Meng Gu
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai 201203, China;
| | - Zhongze Gu
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China;
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Correspondence: (Z.G.); (Y.-R.L.)
| | - Yan-Ru Lou
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai 201203, China;
- Correspondence: (Z.G.); (Y.-R.L.)
| |
Collapse
|
40
|
Zhang D, Fang C, Li H, Lu C, Huang J, Pan J, Yang Z, Liang E, Liu Z, Zhou X, Xin Z, Chen Y, Cai Q. Long ncRNA MALAT1 promotes cell proliferation, migration, and invasion in prostate cancer via sponging miR-145. Transl Androl Urol 2021; 10:2307-2319. [PMID: 34295718 PMCID: PMC8261405 DOI: 10.21037/tau-20-1526] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 04/12/2021] [Indexed: 12/17/2022] Open
Abstract
Background The long non-coding (lncRNA) RNA MALAT1 (metastasis-associated lung adenocarcinoma transcript 1) is known to promote tumorigenesis, whereas microRNA-145 (miR-145) plays an antitumor role in several cancers. In this study, we aimed to elucidate the role of MALAT1 and miR-145 in prostate cancer cells and investigate the effect of MALAT1 downregulation on prostate cancer (PCa) cells in vitro in vivo. Methods The Cancer Genome Atlas (TCGA) datasets were used to carry out the initial bioinformatics analysis; the findings were then tested in LNCaP and CWR22Rv1 cell lines. Western blot and quantitative reverse transcription-polymerase chain reaction (qRT-PCR) was used to evaluate the levels of MALAT1 and miR-145 along with related biomarkers. Furthermore, wound-healing and Transwell assays were performed to test the migratory and invasive abilities of PCa cells. Luciferase reporter assays were used to validate the relationship between MALAT1 and miR-145; their down-stream target genes were also studied. To further substantiate these findings in an animal model, tumor studies including immunofluorescence staining of tissues were carried in nude mice. Results The expression of MALAT1 was upregulated in both LNCaP cell lines and CWR22Rv1 cell lines (F=2.882, t=13.370, P<0.001; F=2.268, t=15.859, P<0.001). Knockdown of MALAT1 reduced the migratory and invasive capabilities of PCa cells (F=0.017, t=12.212, P<0.001; F=10.723, t=6.016, P=0.002). Using direct binding, MALAT1 suppressed the antitumor function of miR-145, which in turn upregulated transforming growth factor-β1 (TGF-β1)-induced epithelial-mesenchymal transition (EMT) via SMAD3 and TGFBR2 (F=2.097, t=5.389, P=0.006; F=1.306, t=4.155, P=0.014). Conclusions We confirmed that MALAT1 acts as a competing endogenous RNA (ceRNA) of miR-145. The MALAT1 based regulation of MiR-145-5p-SMAD3/TGFBR2 interactions could be an intriguing molecular pathway for the progression of PCa.
Collapse
Affiliation(s)
- Dingrong Zhang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Cheng Fang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Haibo Li
- Department of Urology, Tianjin Baodi Hospital, Baodi Clinical College of Tianjin Medical University, Tianjin, China
| | - Chunyuan Lu
- Department of Anesthesia, the Second hospital of Tianjin Medical University, Tianjin, China
| | - Jiaohong Huang
- Department of Geriatric, the Second Hospital of Tianjin Medical University, Tianjin, China
| | - Jiancheng Pan
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhizhao Yang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Enli Liang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhifei Liu
- Department of Urology, Tangshan People's Hospital, Tangshan, China
| | - Xiaodong Zhou
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhongcheng Xin
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China.,Andrology Center, Peking University First Hospital, Peking University, Beijing, China
| | - Yegang Chen
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Qiliang Cai
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
41
|
Zhu X, Pan H, Liu L. Long noncoding RNA network: Novel insight into hepatocellular carcinoma metastasis (Review). Int J Mol Med 2021; 48:134. [PMID: 34013360 PMCID: PMC8148093 DOI: 10.3892/ijmm.2021.4967] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 04/16/2021] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common, aggressive malignancies with poor prognosis and high mortality. Although great progress has been made in recent decades, overall survival of HCC patients remains unsatisfactory due to high recurrence and metastasis. Accordingly, understanding and clarifying the underlying molecular mechanisms of metastasis has become increasingly important. Recently, accumulated reports have supported that long noncoding RNAs (lncRNAs) are dysregulated in HCC and are involved in various pivotal biological processes, including metastasis. The aim of this review was to investigate the dysregulation of lncRNAs in HCC and their function as oncogenes or tumour suppressors. Furthermore, reciprocal regulatory networks between lncRNAs and various molecules that were identified in HCC metastasis, including regulating epithelial-mesenchymal transition (EMT), controlling metastasis-associated genes, and regulating tumour angiogenesis were examined. Numerous reports and information on lncRNAs may help identify lncRNAs that are potential novel diagnostic markers, prognostic markers and therapeutic targets.
Collapse
Affiliation(s)
- Xiuming Zhu
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | - Lili Liu
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| |
Collapse
|
42
|
Xu M, Chen Y, Lu W, Kong L, Fang J, Li Z, Zhang L, Pian C. SPMLMI: predicting lncRNA-miRNA interactions in humans using a structural perturbation method. PeerJ 2021; 9:e11426. [PMID: 34055486 PMCID: PMC8140594 DOI: 10.7717/peerj.11426] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/18/2021] [Indexed: 01/06/2023] Open
Abstract
Long non-coding RNA (lncRNA)-microRNA (miRNA) interactions are quickly emerging as important mechanisms underlying the functions of non-coding RNAs. Accordingly, predicting lncRNA-miRNA interactions provides an important basis for understanding the mechanisms of action of ncRNAs. However, the accuracy of the established prediction methods is still limited. In this study, we used structural consistency to measure the predictability of interactive links based on a bilayer network by integrating information for known lncRNA-miRNA interactions, an lncRNA similarity network, and an miRNA similarity network. In particular, by using the structural perturbation method, we proposed a framework called SPMLMI to predict potential lncRNA-miRNA interactions based on the bilayer network. We found that the structural consistency of the bilayer network was higher than that of any single network, supporting the utility of bilayer network construction for the prediction of lncRNA-miRNA interactions. Applying SPMLMI to three real datasets, we obtained areas under the curves of 0.9512 ± 0.0034, 0.8767 ± 0.0033, and 0.8653 ± 0.0021 based on 5-fold cross-validation, suggesting good model performance. In addition, the generalizability of SPMLMI was better than that of the previously established methods. Case studies of two lncRNAs (i.e., SNHG14 and MALAT1) further demonstrated the feasibility and effectiveness of the method. Therefore, SPMLMI is a feasible approach to identify novel lncRNA-miRNA interactions underlying complex biological processes.
Collapse
Affiliation(s)
- Mingmin Xu
- College of Agriculture, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Yuanyuan Chen
- College of Sciences, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Wei Lu
- College of Life Sciences, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Lingpeng Kong
- College of Agriculture, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Jingya Fang
- College of Agriculture, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Zutan Li
- College of Agriculture, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Liangyun Zhang
- College of Sciences, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Cong Pian
- College of Sciences, Nanjing Agricultural University, Nanjing, Jiangsu, China
| |
Collapse
|
43
|
Jian F, Che X, Zhang J, Liu C, Liu G, Tang Y, Feng W. The long-noncoding RNA SOCS2-AS1 suppresses endometrial cancer progression by regulating AURKA degradation. Cell Death Dis 2021; 12:351. [PMID: 33824269 PMCID: PMC8024384 DOI: 10.1038/s41419-021-03595-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 02/02/2021] [Accepted: 02/19/2021] [Indexed: 12/12/2022]
Abstract
Aberrant long-noncoding RNA (lncRNA) expression has been shown to be involved in the pathogenesis of endometrial cancer (EC). Herein, we report a novel tumor suppressor lncRNA SOCS2-AS1 in EC. Quantitative real-time PCR was performed to detect RNA expression. In situ hybridization and nuclear/cytoplasmic fractionation assays were used to detect the subcellular location. We found that SOCS2-AS1 was downregulated in EC tissues. Its reduced expression was correlated with advanced clinical stage and poor prognosis. Forced expression of SOCS2-AS1 suppressed EC cell proliferation and induced cell-cycle arrest and apoptosis. SOCS2-AS1-binding proteins were detected using RNA pull-down assay and mass spectrometry. Mechanistically, SOCS2-AS1 bound to Aurora kinase A (AURKA) and increased its degradation through the ubiquitin-proteasome pathway. In conclusion, SOCS2-AS1 may thus serve as a prognostic predictor and a biomarker for AURKA-inhibitor treatment in EC patients.
Collapse
Affiliation(s)
- Fangfang Jian
- Department of obstetrics and gynecology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaoxia Che
- Department of obstetrics and gynecology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Obstetrics and gynecology hospital, Fudan University, Shanghai, 200011, China
| | - Jingjing Zhang
- Department of obstetrics and gynecology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chang Liu
- Department of obstetrics and gynecology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Gedan Liu
- Department of obstetrics and gynecology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yujing Tang
- Department of obstetrics and gynecology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Weiwei Feng
- Department of obstetrics and gynecology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
44
|
Goyal B, Yadav SRM, Awasthee N, Gupta S, Kunnumakkara AB, Gupta SC. Diagnostic, prognostic, and therapeutic significance of long non-coding RNA MALAT1 in cancer. Biochim Biophys Acta Rev Cancer 2021; 1875:188502. [PMID: 33428963 DOI: 10.1016/j.bbcan.2021.188502] [Citation(s) in RCA: 202] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/30/2020] [Accepted: 01/02/2021] [Indexed: 12/20/2022]
Abstract
Metastasis Associated Lung Adenocarcinoma Transcript 1 (MALAT1) is a widely studied lncRNA in cancer. Although dispensable for normal physiology, MALAT1 is important for cancer-related pathways regulation. It is localized in the nuclear speckles periphery along with centrally located pre-RNA splicing factors. MALAT1 associated cancer signaling pathways include MAPK/ERK, PI3K/AKT, β-catenin/Wnt, Hippo, VEGF, YAP, etc. Molecular tools such as immunoprecipitation, RNA pull-down, reporter assay, Northern blotting, microarray, and q-RT-PCR has been used to elucidate MALAT1's function in cancer pathogenesis. MALAT1 can regulate multiple steps in the development of tumours. The diagnostic and prognostic significance of MALAT1 has been demonstrated in cancers of the breast, cervix, colorectum, gallbladder, lung, ovary, pancreas, prostate, glioma, hepatocellular carcinoma, and multiple myeloma. MALAT1 has also emerged as a novel therapeutic target for solid as well as hematological malignancies. In experimental models, siRNA and antisense oligonucleotide (ASO) based strategy has been used for targeting MALAT1. The lncRNA has also been targeted for the chemosensitization and radiosensitization of cancer cells. However, most studies have been performed in preclinical models. How the cross-talk of MALAT1 with other signaling pathways affect cancer pathogenesis is the focus of this article. The diagnostic, prognostic, and therapeutic significance of MALAT1 in multiple cancer types are discussed.
Collapse
Affiliation(s)
- Bela Goyal
- Department of Biochemistry, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, India
| | - Shashi Ranjan Mani Yadav
- Department of Biochemistry, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, India
| | - Nikee Awasthee
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Sweety Gupta
- Department of Radiation Oncology, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, India
| | - Ajaikumar B Kunnumakkara
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, 781039, India
| | - Subash Chandra Gupta
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
45
|
Piergentili R, Zaami S, Cavaliere AF, Signore F, Scambia G, Mattei A, Marinelli E, Gulia C, Perelli F. Non-Coding RNAs as Prognostic Markers for Endometrial Cancer. Int J Mol Sci 2021; 22:3151. [PMID: 33808791 PMCID: PMC8003471 DOI: 10.3390/ijms22063151] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/12/2021] [Accepted: 03/12/2021] [Indexed: 02/06/2023] Open
Abstract
Endometrial cancer (EC) has been classified over the years, for prognostic and therapeutic purposes. In recent years, classification systems have been emerging not only based on EC clinical and pathological characteristics but also on its genetic and epigenetic features. Noncoding RNAs (ncRNAs) are emerging as promising markers in several cancer types, including EC, for which their prognostic value is currently under investigation and will likely integrate the present prognostic tools based on protein coding genes. This review aims to underline the importance of the genetic and epigenetic events in the EC tumorigenesis, by expounding upon the prognostic role of ncRNAs.
Collapse
Affiliation(s)
- Roberto Piergentili
- Institute of Molecular Biology and Pathology, Italian National Research Council (CNR-IBPM), 00185 Rome, Italy;
| | - Simona Zaami
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, “Sapienza” University of Rome, Viale Regina Elena 336, 00161 Rome, Italy
| | - Anna Franca Cavaliere
- Gynecology and Obstetric Department, Azienda USL Toscana Centro, Santo Stefano Hospital, 59100 Prato, Italy;
| | - Fabrizio Signore
- Obstetrics and Gynecology Department, USL Roma2, Sant’Eugenio Hospital, 00144 Rome, Italy;
| | - Giovanni Scambia
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Gynecologic Oncology Unit, 00168 Rome, Italy;
- Universita’ Cattolica Del Sacro Cuore, 00168 Rome, Italy
| | - Alberto Mattei
- Gynecology and Obstetric Department, Azienda USL Toscana Centro, Santa Maria Annunziata Hospital, 50012 Florence, Italy; (A.M.); (F.P.)
| | - Enrico Marinelli
- Unit of Forensic Toxicology (UoFT), Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University, 00161 Rome, Italy;
| | - Caterina Gulia
- Department of Urology, Misericordia Hospital, 58100 Grosseto, Italy;
| | - Federica Perelli
- Gynecology and Obstetric Department, Azienda USL Toscana Centro, Santa Maria Annunziata Hospital, 50012 Florence, Italy; (A.M.); (F.P.)
| |
Collapse
|
46
|
Qiao FH, Tu M, Liu HY. Role of MALAT1 in gynecological cancers: Pathologic and therapeutic aspects. Oncol Lett 2021; 21:333. [PMID: 33692865 DOI: 10.3892/ol.2021.12594] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 02/02/2021] [Indexed: 12/15/2022] Open
Abstract
Gynecological cancers, including breast, ovarian, uterine, vaginal, cervical and vulvar cancers are among the major threats to modern life, particularly to female health. Long non-coding RNAs (lncRNAs) play critical roles in normal development of organisms, as well as the tumorigenesis process, and metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is a large infrequently spliced lncRNA, which have been implicated in different gynecological cancers. MALAT1 is overexpressed in breast, ovarian, cervical and endometrial cancers, which initiates cancer progression by inducing changes in the expression of several anti-apoptotic and epithelial-to-mesenchymal transition-related genes. Targeting MALAT1 is an important strategy to combat gynecological cancers, and application of RNA-interference technology and chemotherapeutic process are crucial to target and minimize MALAT1 activity. The present review discusses the role of MALAT1 in gynecological cancers, and potential strategies to target this lncRNA to develop cancer therapeutics. However, further clinical studies are required to determine the prognostic potential of MALAT1 in gynecological cancers.
Collapse
Affiliation(s)
- Feng-Hua Qiao
- Department of Gynecology, Second People's Hospital of Jingmen, Jingmen, Hubei 448000, P.R. China
| | - Min Tu
- Department of Orthopedics, Second People's Hospital of Jingmen, Jingmen, Hubei 448000, P.R. China
| | - Hong-Yan Liu
- Department of Gynecology, Maternal and Child Health Hospital of Jingmen, Jingmen, Hubei 448000, P.R. China
| |
Collapse
|
47
|
Zuo Y, Chen L, He X, Ye Z, Li L, Liu Z, Zhou S. Atorvastatin Regulates MALAT1/miR-200c/NRF2 Activity to Protect Against Podocyte Pyroptosis Induced by High Glucose. Diabetes Metab Syndr Obes 2021; 14:1631-1645. [PMID: 33880049 PMCID: PMC8053520 DOI: 10.2147/dmso.s298950] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 03/18/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Diabetic nephropathy (DN) is one of the main complications of diabetes mellitus (DM), which leads to the long-term loss of kidney functions. Long noncoding RNAs (LncRNAs) can alleviate DN by interacting with microRNAs (miRNAs). In this work, we aimed to explore the effects of the MALAT1/miR-200c/NRF2 regulatory axis on the pyroptosis and oxidative stress (Oxidative stress, OS) of renal podocytes in high glucose (HG) environment and whether the lipid-lowering drug atorvastatin (AT) can relieve renal OS through this approach. METHODS MPC-5, a mouse podocyte cell line, was induced by HG as a cell model. The protein expressions of caspase-1, GSDMD, NLRP3, NRF2, etc. were detected by Western blotting and immunofluorescence, and the mRNA level of caspase-1, GSDMD, NLRP3, NRF2, MALAT1, miR-200c was tested by qRT-PCR. The cell pyroptosis of podocytes treated with AT was verified by CCK-8 or flow cytometry. The levels of Malondialdehyde (MDA), superoxide dismutase (SOD), and glutathione (GSH) were measured by spectrophotometer, respectively. RESULTS The caspase-1 was upregulated in time-dependent manner and got the peak at 48 h and 30 mmol/L respectively in MPC-5 cells treated with HG. Further, the expression of GSDMD, MALAT1 and miR-200c were increased, while the level of NRF2, HO-1, OS-related indicators, were decreased simultaneously. Knockdown the MALAT1 protected MPC-5 cells from pyroptosis and OS induced by HG. However, overexpressing miR-200c in control-group cells increased pyroptosis and upregulated the OS level with HG culture medium. Further, atorvastatin protected MPC-5 cells from cell pyroptosis and downregulated the level of renal OS via attenuating the expression of MALAT1 and miR-200c. CONCLUSION Atorvastatin protects podocyte cells via MALAT1/miR-200c/NRF2 signal pathway from pyroptosis and OS induced by HG.
Collapse
Affiliation(s)
- Yi Zuo
- Department of Endocrinology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, People’s Republic of China
| | - Li Chen
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin, Guangxi, 541004, People’s Republic of China
| | - Xiaoyun He
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
| | - Zhen Ye
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin, Guangxi, 541004, People’s Republic of China
| | - Ling Li
- Department of Endocrinology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, People’s Republic of China
| | - Zhanhong Liu
- Department of Endocrinology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, People’s Republic of China
| | - Suxian Zhou
- Department of Endocrinology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, People’s Republic of China
- Correspondence: Suxian Zhou Department of Endocrinology, Affiliated Hospital of Guilin Medical University, 15 Lequn Road, Guilin, Guangxi, 541001, People’s Republic of China Email
| |
Collapse
|
48
|
Ni J, Tian W, Liang S, Wang H, Ren Y. Promoter Methylation-mediated Silencing of the MiR-192-5p Promotes Endometrial Cancer Progression by Targeting ALX1. Int J Med Sci 2021; 18:2510-2520. [PMID: 34104082 PMCID: PMC8176185 DOI: 10.7150/ijms.58954] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/16/2021] [Indexed: 12/24/2022] Open
Abstract
Background: Epigenetic regulation by promoter methylation-mediated silencing of cancer-related microRNAs plays vital roles in tumorigenesis. MiR-192-5p promotes tumor progression in various human cancers with conflicting biological effects. However, its expression levels and biological functions in endometrial carcinoma (EC) have not been reported. Methods: The methylation status of miR-192-5p in tissue samples and cell lines, was examined using bisulfite sequencing PCR. miR-192-5p expression was also measured. EC cell lines transfected with specifically designed vectors overexpressing miR-192-5p, its target gene ALX1 or both, were constructed. Tumorigenicity of these cell lines were examined by in vitro and in vivo experiments. Dual-luciferase reporter assay were employed to verify the target of miR-192-5p. Results: The promoter region of miR-192-5p gene was highly methylated and its expression significantly repressed in EC samples. Moreover, a higher level of promoter methylation as well as a lower expression of miR-192-5p, was significantly associated with advanced Federation of Gynecology and Obstetrics stage and shorter disease-free survival in patients with curatively resected EC. Functional studies demonstrated that miR-192-5p overexpression inhibited in vitro tumor progression, in vivo tumorigenicity and the expression of several oncoproteins that was highly related to epithelial-to-mesenchymal transition. ALX1 was verified as a direct target of miR-192-5p and demonstrated to mediate the tumor-suppressive function of miR-192-5p. Conclusion: miR-192-5p is a tumor suppressor miRNA that is epigenetically silenced by promoter methylation and may serve as a potential prognostic biomarker in EC.
Collapse
Affiliation(s)
- Jianjiao Ni
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenjuan Tian
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shanhui Liang
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Huaying Wang
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yulan Ren
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
49
|
Liao H, Chen Q, Xiao J. Reflections on the Role of Malat1 in Gynecological Cancer. Cancer Manag Res 2020; 12:13489-13500. [PMID: 33408521 PMCID: PMC7779295 DOI: 10.2147/cmar.s286804] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 12/01/2020] [Indexed: 12/26/2022] Open
Abstract
Non-coding RNAs (ncRNAs) have received significant attention over the last few years. Malat1, as one of the most extensively studied ncRNAs, is believed to be not only a potential biomarker for disease diagnosis and prognosis, but also a candidate drug target for gynecological cancers. This potential is supported by a growing body of experimental evidence demonstrating that Malat1 participates in the occurrence, progression, and metastasis of tumors. Research has also shown that Malat1 can influence patient survival by regulating a range of target genes and signaling pathways. However, previous review articles have generally failed to consider the role of Malat1 in gynecological cancer in detail. In the present review, we summarize recent progress in research relating to the clinical relevance of Malat1 and the molecular mechanisms underlying the action of this ncRNA. Besides, we put forward some action points for further research after taking into consideration the sub-location and other essential properties of Malat1, which might enable us to have a better understanding of the potential of this molecule regarding clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Huiyan Liao
- The 2nd Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province 510006, People's Republic of China
| | - Qi Chen
- The 6th Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province 510006, People's Republic of China
| | - Jing Xiao
- Department of Gynecology, the University Town Branch, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province 510120, People's Republic of China
| |
Collapse
|
50
|
Fu S, Wang Y, Li H, Chen L, Liu Q. Regulatory Networks of LncRNA MALAT-1 in Cancer. Cancer Manag Res 2020; 12:10181-10198. [PMID: 33116873 PMCID: PMC7575067 DOI: 10.2147/cmar.s276022] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/03/2020] [Indexed: 12/18/2022] Open
Abstract
Long noncoding (lnc)RNAs are a group of RNAs with a length greater than 200 nt that do not encode a protein but play an essential role in regulating the expression of target genes in normal biological contexts as well as pathologic processes including tumorigenesis. The lncRNA metastasis-associated lung adenocarcinoma transcript (MALAT)-1 has been widely studied in cancer. In this review, we describe the known functions of MALAT-1; its mechanisms of action; and associated signaling pathways and their clinical significance in different cancers. In most malignancies, including lung, colorectal, thyroid, and other cancers, MALAT-1 functions as an oncogene and is upregulated in tumors and tumor cell lines. MALAT-1 has a distinct mechanism of action in each cancer type and is thus at the center of large gene regulatory networks. Dysregulation of MALAT-1 affects cellular processes such as alternative splicing, epithelial–mesenchymal transition, apoptosis, and autophagy, which ultimately results in the abnormal cell proliferation, invasion, and migration that characterize cancers. In other malignancies, such as glioma and endometrial carcinoma, MALAT-1 functions as a tumor suppressor and thus forms additional regulatory networks. The current evidence indicates that MALAT-1 and its associated signaling pathways can serve as diagnostic or prognostic biomarker or therapeutic target in the treatment of many cancers.
Collapse
Affiliation(s)
- Shijian Fu
- The First Affiliated Hospital of Harbin Medical University, Harbin 150081, People's Republic of China
| | - Yanhong Wang
- Department of Laboratory Medicine, Yuebei People's Hospital of Shaoguan, The Affiliated Hospital of Shantou University, Shaoguan 512025, People's Republic of China
| | - Hang Li
- The First Affiliated Hospital of Harbin Medical University, Harbin 150081, People's Republic of China
| | - Leilei Chen
- Department of Cardiology, Beijing Anzhen Hospital, Beijing Institute of Heart Lung and Blood Vessel Disease, Capital Medical University, Beijing 100029, People's Republic of China
| | - Quanzhong Liu
- Department of Medical Genetics, Harbin Medical University, Harbin 150081, People's Republic of China
| |
Collapse
|