1
|
Wang Z, Liang X, Yi G, Wu T, Sun Y, Zhang Z, Fu M. Bioinformatics analysis proposes a possible role for long noncoding RNA MIR17HG in retinoblastoma. Cancer Rep (Hoboken) 2024; 7:e1933. [PMID: 38321787 PMCID: PMC10864729 DOI: 10.1002/cnr2.1933] [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: 04/14/2023] [Revised: 09/27/2023] [Accepted: 11/06/2023] [Indexed: 02/08/2024] Open
Abstract
BACKGROUND Retinoblastoma (RB) is the most common prevalent intraocular malignancy among infants and children, particularly in underdeveloped countries. With advancements in genomics and transcriptomics, noncoding RNAs have been increasingly utilized to investigate the molecular pathology of diverse diseases. AIMS This study aims to establish the competing endogenous RNAs network associated with RB, analyse the function of mRNAs and lncRNAs, and finds the relevant regulatory network. METHODS AND RESULTS This study establishes a network of competing endogenous RNAs by Spearman correlation analysis and prediction based on RB patients and healthy children. Enrichment analyzes based on Gene Ontology and the Kyoto Encyclopedia of Genes and Genomes are conducted to analyze the potential biological functions of lncRNA and mRNA networks. Weighted gene co-expression network analysis (WGCNA) is employed to identify gene cluster modules exhibiting the strongest correlation with RB. The results indicate a significant correlation between the lncRNA MIR17HG (R = .73, p = .02) and the RB phenotype. ceRNA networks reveal downstream miRNAs (hsa-mir-425-5p and hsa-mir455-5p) and mRNAs (MDM2, IPO11, and ITGA1) associated with MIR17Hg. As an inhibitor of the p53 signaling pathway, MDM2 can suppress the development of RB. CONCLUSION In conclusion, lncRNAs play a role in RB, and the MIR17HG/hsa-mir-425-5p/MDM2 pathway may contribute to RB development by inhibiting the p53 signaling pathway.
Collapse
Affiliation(s)
- Zijin Wang
- The Second Clinical Medicine SchoolSouthern Medical UniversityGuangzhouGuangdongChina
| | - Xiaotian Liang
- Department of Cardiovascular Medicine, Sun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Guoguo Yi
- Department of OphthalmologyThe Sixth Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Tong Wu
- The First Clinical Medicine SchoolSouthern Medical UniversityGuangzhouGuangdongChina
| | - Yuxin Sun
- The Second Clinical Medicine SchoolSouthern Medical UniversityGuangzhouGuangdongChina
| | - Ziran Zhang
- The Second Clinical Medicine SchoolSouthern Medical UniversityGuangzhouGuangdongChina
| | - Min Fu
- Department of Ophthalmology, Zhujiang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| |
Collapse
|
2
|
Jayarathna DK, Rentería ME, Batra J, Gandhi NS. A supervised machine learning approach identifies gene-regulating factor-mediated competing endogenous RNA networks in hormone-dependent cancers. J Cell Biochem 2022; 123:1394-1408. [PMID: 35757968 PMCID: PMC9542250 DOI: 10.1002/jcb.30300] [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: 04/21/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 11/17/2022]
Abstract
Competing endogenous RNAs (ceRNAs) have become an emerging topic in cancer research due to their role in gene regulatory networks. To date, traditional ceRNA bioinformatic studies have investigated microRNAs as the only factor regulating gene expression. Growing evidence suggests that genomic (e.g., copy number alteration [CNA]), transcriptomic (e.g., transcription factors [TFs]), and epigenomic (e.g., DNA methylation [DM]) factors can influence ceRNA regulatory networks. Herein, we used the Least absolute shrinkage and selection operator regression, a machine learning approach, to integrate DM, CNA, and TFs data with RNA expression to infer ceRNA networks in cancer risk. The gene‐regulating factors‐mediated ceRNA networks were identified in four hormone‐dependent (HD) cancer types: prostate, breast, colorectal, and endometrial. The shared ceRNAs across HD cancer types were further investigated using survival analysis, functional enrichment analysis, and protein–protein interaction network analysis. We found two (BUB1 and EXO1) and one (RRM2) survival‐significant ceRNA(s) shared across breast‐colorectal‐endometrial and prostate–colorectal–endometrial combinations, respectively. Both BUB1 and BUB1B genes were identified as shared ceRNAs across more than two HD cancers of interest. These genes play a critical role in cell division, spindle‐assembly checkpoint signalling, and correct chromosome alignment. Furthermore, shared ceRNAs across multiple HD cancers have been involved in essential cancer pathways such as cell cycle, p53 signalling, and chromosome segregation. Identifying ceRNAs' roles across multiple related cancers will improve our understanding of their shared disease biology. Moreover, it contributes to the knowledge of RNA‐mediated cancer pathogenesis.
Collapse
Affiliation(s)
- Dulari K Jayarathna
- Centre for Genomics and Personalized Health, School of Chemistry and Physics, Queensland University of Technology, Brisbane, QLD, Australia.,Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Miguel E Rentería
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Jyotsna Batra
- Centre for Genomics and Personalized Health, School of Chemistry and Physics, Queensland University of Technology, Brisbane, QLD, Australia.,School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia.,Australian Prostate Cancer Research Centre-Queensland, Woolloongabba, QLD, Australia
| | - Neha S Gandhi
- Centre for Genomics and Personalized Health, School of Chemistry and Physics, Queensland University of Technology, Brisbane, QLD, Australia.,Cancer and Ageing Research Program, Translational Research Institute, Woolloongabba, QLD, Australia
| |
Collapse
|
3
|
Xue X, Liu M, Wang Y, Yang Y, Li Z, Shi R, Miao Y. MicroRNA-494-3p Exacerbates Renal Epithelial Cell Dysfunction by Targeting SOCS6 under High Glucose Treatment. Kidney Blood Press Res 2022; 47:247-255. [PMID: 35038704 DOI: 10.1159/000521647] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/21/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Diabetic nephropathy is a common complication of the kidneys induced by diabetes and is the main cause of end-stage renal disease. MicroRNA-494-3p was reported to be upregulated in renal tissues collected from db/db mice, but its specific role in diabetic nephropathy was still unclear. This study aimed to explore the effect of miR-494-3p on renal fibrosis using an in vitro cell model of diabetic nephropathy. METHODS After human renal tubular epithelial cells (HK-2) were treated with high glucose (HG), the viability and apoptosis of cells were examined by CCK-8 assays and flow cytometry analyses. Additionally, protein levels of fibronectin, collagen I, collagen III, collagen IV, and epithelial-mesenchymal transition (EMT) markers in HG-induced HK-2 cells were quantified by Western blotting. miR-494-3p expression in HK-2 cells was detected by reverse-transcription quantitative polymerase chain reaction. The binding relation between miR-494-3p and the messenger RNA suppressor of cytokine signaling 6 (SOCS6) was detected by luciferase reporter assays. RESULTS HG reduced cell viability and enhanced cell apoptosis in a time- or concentration-dependent manner. Additionally, HG induced collagen accumulation and triggered the EMT process. miR-494-3p was upregulated in HG-treated HK-2 cells. miR-494-3p inhibition alleviated HG-induced cell dysfunction. Mechanistically, miR-494-3p bound with SOCS6 and negatively regulated SOCS6 expression. Moreover, silencing SOCS6 rescued the suppressive effect of miR-499-5p inhibition on HG-induced cell dysfunction. CONCLUSION miR-494-3p aggravates renal fibrosis, EMT process, and cell apoptosis by targeting SOCS6, suggesting that the miR-494-3p/SOCS6 axis may become a potential strategy for the treatment of diabetic nephropathy.
Collapse
Affiliation(s)
- Xianjun Xue
- Department of Nephrology, Puyang Oilfeld General Hospital, Puyang, China
| | - Minjie Liu
- Department of Nephrology, Puyang Oilfeld General Hospital, Puyang, China
| | - Yulu Wang
- Department of Nephrology, Puyang Oilfeld General Hospital, Puyang, China
| | - Yanlei Yang
- Department of Nephrology, Puyang Oilfeld General Hospital, Puyang, China
| | - Zhiping Li
- Department of Nephrology, Puyang Oilfeld General Hospital, Puyang, China
| | - Ruifang Shi
- Department of Nephrology, Puyang Oilfeld General Hospital, Puyang, China
| | - Yueting Miao
- Department of Nephrology, Puyang Oilfeld General Hospital, Puyang, China
| |
Collapse
|
4
|
Jayarathna DK, Rentería ME, Sauret E, Batra J, Gandhi NS. Identifying Complex lncRNA/Pseudogene-miRNA-mRNA Crosstalk in Hormone-Dependent Cancers. BIOLOGY 2021; 10:biology10101014. [PMID: 34681112 PMCID: PMC8533463 DOI: 10.3390/biology10101014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/02/2021] [Accepted: 10/04/2021] [Indexed: 12/13/2022]
Abstract
Simple Summary Competing endogenous RNAs (ceRNAs) have gained attention in cancer research owing to their involvement in microRNA-mediated gene regulation. Here, we identified a shared ceRNA network across five hormone-dependent (HD) cancers (prostate, breast, colon, rectal, and endometrial), that contain two long non-coding RNAs, nine mRNAs, and seventy-four microRNAs. Among them, two mRNAs and forty-one microRNAs were associated with at least one HD cancer survival. A similar analytical approach can be applied to identify shared ceRNAs across a group of related cancers, which will significantly contribute to understanding their shared disease biology. Abstract The discovery of microRNAs (miRNAs) has fundamentally transformed our understanding of gene regulation. The competing endogenous RNA (ceRNA) hypothesis postulates that messenger RNAs and other RNA transcripts, such as long non-coding RNAs and pseudogenes, can act as natural miRNA sponges. These RNAs influence each other’s expression levels by competing for the same pool of miRNAs through miRNA response elements on their target transcripts, thereby modulating gene expression and protein activity. In recent years, these ceRNA regulatory networks have gained considerable attention in cancer research. Several studies have identified cancer-specific ceRNA networks. Nevertheless, prior bioinformatic analyses have focused on long-non-coding RNA-associated ceRNA networks. Here, we identify an extended ceRNA network (including both long non-coding RNAs and pseudogenes) shared across a group of five hormone-dependent (HD) cancers, i.e., prostate, breast, colon, rectal, and endometrial cancers, using data from The Cancer Genome Atlas (TCGA). We performed a functional enrichment analysis for differentially expressed genes in the shared ceRNA network of HD cancers, followed by a survival analysis to determine their prognostic ability. We identified two long non-coding RNAs, nine genes, and seventy-four miRNAs in the shared ceRNA network across five HD cancers. Among them, two genes and forty-one miRNAs were associated with at least one HD cancer survival. This study is the first to investigate pseudogene-associated ceRNAs across a group of related cancers and highlights the value of this approach to understanding the shared molecular pathogenesis in a group of related diseases.
Collapse
Affiliation(s)
- Dulari K. Jayarathna
- Centre for Genomics and Personalised Health, School of Chemistry and Physics, Queensland University of Technology, Brisbane, QLD 4000, Australia; (D.K.J.); (J.B.)
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia;
| | - Miguel E. Rentería
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia;
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD 4059, Australia
| | - Emilie Sauret
- School of Mechanical, Medical & Process Engineering, Queensland University of Technology, Brisbane, QLD 4000, Australia;
| | - Jyotsna Batra
- Centre for Genomics and Personalised Health, School of Chemistry and Physics, Queensland University of Technology, Brisbane, QLD 4000, Australia; (D.K.J.); (J.B.)
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD 4059, Australia
- Translational Research Institute, Brisbane, QLD 4102, Australia
| | - Neha S. Gandhi
- Centre for Genomics and Personalised Health, School of Chemistry and Physics, Queensland University of Technology, Brisbane, QLD 4000, Australia; (D.K.J.); (J.B.)
- Translational Research Institute, Brisbane, QLD 4102, Australia
- Correspondence:
| |
Collapse
|
5
|
Ouyang D, Li R, Li Y, Zhu X. Erratum: Construction of a Competitive Endogenous RNA Network in Uterine Corpus Endometrial Carcinoma. Med Sci Monit 2021; 27:e934874. [PMID: 34584064 PMCID: PMC8489249 DOI: 10.12659/msm.934874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
There was an error in Figure 4. The correct Figure 4 is provided below. The picture error would not affect the conclusion of the paper. Reference: Dong Ouyang, Ruyi Li, Yaxian Li, Xueqiong Zhu. Construction of a Competitive Endogenous RNA Network in Uterine Corpus Endometrial Carcinoma. Med Sci Monit 2019; 25:7998-8010. DOI: 10.12659/MSM.915798.
Collapse
Affiliation(s)
- Dong Ouyang
- Department of Obstetrics and Gynecology, Akesu Hospital of Traditional Chinese Medicine, Akesu, Xinjiang, China (mainland)
| | - Ruyi Li
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China (mainland)
| | - Yaxian Li
- Department of Obstetrics and Gynecology, Akesu Hospital of Traditional Chinese Medicine, Akesu, Xinjiang, China (mainland)
| | - Xueqiong Zhu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China (mainland)
| |
Collapse
|
6
|
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: 46] [Impact Index Per Article: 11.5] [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
|
7
|
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
|
8
|
Liu S, Xin W, Lu Q, Tang X, Wang F, Shao W, Zhang Y, Qiu J, Hua K. Knockdown of lncRNA H19 suppresses endometriosis in vivo. ACTA ACUST UNITED AC 2021; 54:e10117. [PMID: 33656053 PMCID: PMC7917710 DOI: 10.1590/1414-431x202010117] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 11/27/2020] [Indexed: 12/12/2022]
Abstract
The long noncoding RNA (lncRNA) H19 is involved in the pathogenesis of endometriosis by modulating the proliferation and invasion of ectopic endometrial cells in vitro, but related in vivo studies are rare. This study aimed to investigate the role of lncRNA H19 in a nude mouse model of endometriosis. Ectopic endometrial stromal cells (ecESCs) were isolated from ectopic endometrium of patients with endometriosis and infected with lentiviruses expressing short hairpin RNA (shRNA) negative control (LV-NC-shRNA) or lncRNA-H19 shRNA (LV-H19-shRNA). The ecESCs infected with LV-NC-shRNA and LV-H19-shRNA were subcutaneously implanted into forty 6- to 8-week-old female nude mice. The size and weight of the endometriotic implants were measured at 1, 2, 3, and 4 weeks after implantation and compared, and lncRNA H19 levels in endometriotic implants were evaluated using real-time polymerase chain reaction (RT-PCR). All nude mice survived the experimental period, and no significant differences in body weight were observed between the experimental group and the control group. All nude mice developed histologically confirmed subcutaneous endometriotic lesions with glandular structures and stroma after 1 week of implantation. The subcutaneous lesions in the LV-NC-shRNA group after 1, 2, 3, and 4 weeks of implantation were larger than those in the LV-H19-shRNA group, and lncRNA H19 levels in subcutaneous lesions in the LV-NC-shRNA group were significantly higher than those in the LV-H19-shRNA group. Knockdown of lncRNA H19 suppresses endometriosis in vivo. Further study is required to explore the underlying mechanism in the future.
Collapse
Affiliation(s)
- Songping Liu
- Department of Obstetrics and Gynecology, Jinshan Hospital of Fudan University, Shanghai, China.,Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China.,Department of Obstetrics and Gynecology, Zhenjiang Maternal and Child Health Hospital, Zhenjiang, Jiangsu, China
| | - Weijuan Xin
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Qi Lu
- Department of Obstetrics and Gynecology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Xiaoyan Tang
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Fengqin Wang
- Department of Obstetrics and Gynecology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Wei Shao
- Department of Obstetrics and Gynecology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Yajiao Zhang
- Department of Obstetrics and Gynecology, Zhenjiang Maternal and Child Health Hospital, Zhenjiang, Jiangsu, China
| | - Junjun Qiu
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Keqin Hua
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| |
Collapse
|
9
|
Gao L, Zhao Y, Ma X, Zhang L. Integrated analysis of lncRNA-miRNA-mRNA ceRNA network and the potential prognosis indicators in sarcomas. BMC Med Genomics 2021; 14:67. [PMID: 33653335 PMCID: PMC7927383 DOI: 10.1186/s12920-021-00918-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 02/22/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Competitive endogenous RNA (ceRNA) networks have revealed a new mechanism of interaction between RNAs, and play crucial roles in multiple biological processes and development of neoplasms. They might serve as diagnostic and prognosis markers as well as therapeutic targets. METHODS In this work, we identified differentially expressed mRNAs (DEGs), lncRNAs (DELs) and miRNAs (DEMs) in sarcomas by comparing the gene expression profiles between sarcoma and normal muscle samples in Gene Expression Omnibus (GEO) datasets. Gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) pathway enrichment analyses were applied to investigate the primary functions of the overlapped DEGs. Then, lncRNA-miRNA and miRNA-mRNA interactions were predicted, and the ceRNA regulatory network was constructed using Cytoscape software. In addition, the protein-protein interaction (PPI) network and survival analysis were performed. RESULTS A total of 1296 DEGs were identified in sarcoma samples by combining the GO and KEGG enrichment analyses, 338 DELs were discovered after the probes were reannotated, and 36 DEMs were ascertained through intersecting two different expression miRNAs sets. Further, through target gene prediction, a lncRNA-miRNA-mRNA ceRNA network that contained 113 mRNAs, 69 lncRNAs and 29 miRNAs was constructed. The PPI network identified the six most significant hub proteins. Survival analysis revealed that seven mRNAs, four miRNAs and one lncRNA were associated with overall survival of sarcoma patients. CONCLUSIONS Overall, we constructed a ceRNA network in sarcomas, which might provide insights for further research on the molecular mechanism and potential prognosis biomarkers.
Collapse
Affiliation(s)
- Lu Gao
- College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China
- Department of Oncology, The General Hospital of Western Theater Command, Chengdu, 610083, Sichuan, China
| | - Yu Zhao
- Department of Oncology, The General Hospital of Western Theater Command, Chengdu, 610083, Sichuan, China
| | - Xuelei Ma
- Department of Biotherapy, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ling Zhang
- Department of Oncology, The General Hospital of Western Theater Command, Chengdu, 610083, Sichuan, China.
| |
Collapse
|
10
|
Geng R, Zheng Y, Zhou D, Li Q, Li R, Guo X. ZBTB7A, a potential biomarker for prognosis and immune infiltrates, inhibits progression of endometrial cancer based on bioinformatics analysis and experiments. Cancer Cell Int 2020; 20:542. [PMID: 33292231 PMCID: PMC7654049 DOI: 10.1186/s12935-020-01600-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 10/06/2020] [Indexed: 12/24/2022] Open
Abstract
Backgroud ZBTB protein is an important member of the C2H2 zinc finger protein family. As a transcription factor, it is widely involved in the transcriptional regulation of genes, cell proliferation, differentiation, and apoptosis. The ZBTB7A has been largely linked to different kinds of tumors due to its diverse function. However, the value for ZBTB7A in uterine corpus endometrial carcinoma (UCEC) is unclear. Methods In our work, we assessed the importance of ZBTB7A in UCEC. Firstly, Using Oncomine and Tumor Immunoassay Resource (TIMER) databases to evaluate the expression of ZBTB7A. Secondly, we explored the co-expression network of ZBTB7A through the cBioPortal online tool, Metascape, and LinkedOmics. TIMER was also used to explore the relationship between ZBTB7A and tumor immune invasion, and to detect the correlation between the ZBTB7A and the marker genes related to immune infiltration. Finally, CCK8, migration, ChIP assays were introduced to partly validate ZBTB7A function in endometrial cancer cells. Results We found the ZBTB7A expression in TIMER was associated with various cancers, especially UCEC. The decreased expression of ZBTB7A was markedly related to the stage and prognosis of UCEC. Furthermore, ZBTB7A was also related to the expression of various immune markers such as Neutrophils, Dendritic cell, T cell (general), Th1, Th2, and Treg. Finally, we verified that ZBTB7A repressed E2F4 transcription and inhibited cells proliferation and migration. These results indicate that ZBTB7A may play a vital role in regulating immune cell infiltration in UCEC, and is a valuable prognostic marker. Conclusions In summary, we demonstrate that ZBTB7A is notably downregulated in UCEC, plays a vital role in regulating immune cell infiltration, possesses diagnostic and prognostic values and attenuates E2F4 transcription and cell proliferation, migration in vitro.
Collapse
Affiliation(s)
- Rong Geng
- Department of Gynecology, Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, 52800, China.,Department of Gynecology and Obstetrics, The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China.,Foshan Maternal and Children Healthy Research Institute, Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, China
| | - Yuhua Zheng
- Department of Gynecology, Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, 52800, China
| | - Donghua Zhou
- Department of Pathology, Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, China
| | - Qingdong Li
- Department of Gynecology, Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, 52800, China
| | - Ruiman Li
- Department of Gynecology and Obstetrics, The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China.
| | - Xiaoling Guo
- Department of Gynecology, Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, 52800, China.
| |
Collapse
|
11
|
Dorn A, Glaß M, Neu CT, Heydel B, Hüttelmaier S, Gutschner T, Haemmerle M. LINC00261 Is Differentially Expressed in Pancreatic Cancer Subtypes and Regulates a Pro-Epithelial Cell Identity. Cancers (Basel) 2020; 12:cancers12051227. [PMID: 32414223 PMCID: PMC7281485 DOI: 10.3390/cancers12051227] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/30/2020] [Accepted: 05/11/2020] [Indexed: 12/11/2022] Open
Abstract
Pancreatic adenocarcinoma (PDAC) is one of the major causes of cancer-associated deaths worldwide, with a dismal prognosis that has not significantly changed over the last decades. Transcriptional analysis has provided valuable insights into pancreatic tumorigenesis. Specifically, pancreatic cancer subtypes were identified, characterized by specific mutations and gene expression changes associated with differences in patient survival. In addition to differentially regulated mRNAs, non-coding RNAs, including long non-coding RNAs (lncRNAs), were shown to have subtype-specific expression patterns. Hence, we aimed to characterize prognostic lncRNAs with deregulated expression in the squamous subtype of PDAC, which has the worst prognosis. Extensive in silico analyses followed by in vitro experiments identified long intergenic non-coding RNA 261 (LINC00261) as a downregulated lncRNA in the squamous subtype of PDAC, which is generally associated with transforming growth factor β (TGFβ) signaling in human cancer cells. Its genomic neighbor, the transcription factor forkhead box protein A2 (FOXA2), regulated LINC00261 expression by direct binding of the LINC00261 promoter. CRISPR-mediated knockdown and promoter knockout validated the importance of LINC00261 in TGFβ-mediated epithelial–mesenchymal transition (EMT) and established the epithelial marker E-cadherin, an important cell adhesion protein, as a downstream target of LINC00261. Consequently, depletion of LINC00261 enhanced motility and invasiveness of PANC-1 cells in vitro. Altogether, our data suggest that LINC00261 is an important tumor-suppressive lncRNA in PDAC that is involved in maintaining a pro-epithelial state associated with favorable disease outcome.
Collapse
Affiliation(s)
- Agnes Dorn
- Institute of Pathology, Section for Experimental Pathology, Medical Faculty, Martin-Luther University Halle-Wittenberg, 06120 Halle/Saale, Germany; (A.D.); (C.T.N.); (B.H.)
| | - Markus Glaß
- Institute of Molecular Medicine, Section for Cell Biology, Medical Faculty, Martin-Luther University Halle-Wittenberg, 06120 Halle/Saale, Germany; (M.G.); (S.H.)
| | - Carolin T. Neu
- Institute of Pathology, Section for Experimental Pathology, Medical Faculty, Martin-Luther University Halle-Wittenberg, 06120 Halle/Saale, Germany; (A.D.); (C.T.N.); (B.H.)
| | - Beate Heydel
- Institute of Pathology, Section for Experimental Pathology, Medical Faculty, Martin-Luther University Halle-Wittenberg, 06120 Halle/Saale, Germany; (A.D.); (C.T.N.); (B.H.)
| | - Stefan Hüttelmaier
- Institute of Molecular Medicine, Section for Cell Biology, Medical Faculty, Martin-Luther University Halle-Wittenberg, 06120 Halle/Saale, Germany; (M.G.); (S.H.)
| | - Tony Gutschner
- Junior Research Group ‘RNA biology and Pathogenesis’, Medical Faculty, Martin-Luther University Halle-Wittenberg, 06120 Halle/Saale, Germany
- Correspondence: (T.G.); (M.H.); Tel.: +49-345-5573945 (T.G.); +49-345-5573964 (M.H.)
| | - Monika Haemmerle
- Institute of Pathology, Section for Experimental Pathology, Medical Faculty, Martin-Luther University Halle-Wittenberg, 06120 Halle/Saale, Germany; (A.D.); (C.T.N.); (B.H.)
- Correspondence: (T.G.); (M.H.); Tel.: +49-345-5573945 (T.G.); +49-345-5573964 (M.H.)
| |
Collapse
|