Integrative Lncrna–mrna Co‐expression Network Analysis Identifies Novel Lncrna E2F3‐IT1 for Rheumatoid Arthritis
Long‐Fei Wu,Xingbo Mo,Jin-Bo He,Pei He,Xin Lu,Hong‐Wen Deng,Fei‐Yan Deng,Shu‐Feng Lei
DOI: https://doi.org/10.1002/ctm2.325
IF: 8.554
2021-01-01
Clinical and Translational Medicine
Abstract:Clinical and Translational MedicineVolume 11, Issue 2 e325 LETTER TO EDITOROpen Access Integrative lncRNA–mRNA co-expression network analysis identifies novel lncRNA E2F3-IT1 for rheumatoid arthritis Long-Fei Wu, Long-Fei Wu Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, P. R. China Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, P. R. China Long-Fei Wu, Xing-Bo Mo, and Jia-Hui He contributed equally to this work.Search for more papers by this authorXing-Bo Mo, Xing-Bo Mo orcid.org/0000-0001-8956-4918 Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, P. R. China Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, P. R. China Long-Fei Wu, Xing-Bo Mo, and Jia-Hui He contributed equally to this work.Search for more papers by this authorJia-Hui He, Jia-Hui He Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, P. R. China Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, P. R. China Long-Fei Wu, Xing-Bo Mo, and Jia-Hui He contributed equally to this work.Search for more papers by this authorPei He, Pei He Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, P. R. China Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, P. R. ChinaSearch for more papers by this authorXin Lu, Xin Lu Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, P. R. China Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, P. R. ChinaSearch for more papers by this authorHong-Wen Deng, Hong-Wen Deng Center of Bioinformatics and Genomics, Department of Global Biostatistics and Data Science, Tulane University, New Orleans, Louisiana, USASearch for more papers by this authorFei-Yan Deng, Corresponding Author Fei-Yan Deng fdeng@suda.edu.cn Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, P. R. China Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, P. R. China Correspondence Shu-Feng Lei and Fei-Yan Deng, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, 199 Renai Road, Suzhou, Jiangsu, P. R. China. Email: leisf@suda.edu.cn; fdeng@suda.edu.cnSearch for more papers by this authorShu-Feng Lei, Corresponding Author Shu-Feng Lei leisf@suda.edu.cn orcid.org/0000-0003-1453-8082 Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, P. R. China Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, P. R. China Correspondence Shu-Feng Lei and Fei-Yan Deng, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, 199 Renai Road, Suzhou, Jiangsu, P. R. China. Email: leisf@suda.edu.cn; fdeng@suda.edu.cnSearch for more papers by this author Long-Fei Wu, Long-Fei Wu Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, P. R. China Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, P. R. China Long-Fei Wu, Xing-Bo Mo, and Jia-Hui He contributed equally to this work.Search for more papers by this authorXing-Bo Mo, Xing-Bo Mo orcid.org/0000-0001-8956-4918 Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, P. R. China Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, P. R. China Long-Fei Wu, Xing-Bo Mo, and Jia-Hui He contributed equally to this work.Search for more papers by this authorJia-Hui He, Jia-Hui He Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, P. R. China Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, P. R. China Long-Fei Wu, Xing-Bo Mo, and Jia-Hui He contributed equally to this work.Search for more papers by this authorPei He, Pei He Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, P. R. China Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, P. R. ChinaSearch for more papers by this authorXin Lu, Xin Lu Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, P. R. China Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, P. R. ChinaSearch for more papers by this authorHong-Wen Deng, Hong-Wen Deng Center of Bioinformatics and Genomics, Department of Global Biostatistics and Data Science, Tulane University, New Orleans, Louisiana, USASearch for more papers by this authorFei-Yan Deng, Corresponding Author Fei-Yan Deng fdeng@suda.edu.cn Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, P. R. China Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, P. R. China Correspondence Shu-Feng Lei and Fei-Yan Deng, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, 199 Renai Road, Suzhou, Jiangsu, P. R. China. Email: leisf@suda.edu.cn; fdeng@suda.edu.cnSearch for more papers by this authorShu-Feng Lei, Corresponding Author Shu-Feng Lei leisf@suda.edu.cn orcid.org/0000-0003-1453-8082 Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, P. R. China Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, P. R. China Correspondence Shu-Feng Lei and Fei-Yan Deng, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, 199 Renai Road, Suzhou, Jiangsu, P. R. China. Email: leisf@suda.edu.cn; fdeng@suda.edu.cnSearch for more papers by this author First published: 24 February 2021 https://doi.org/10.1002/ctm2.325 AboutSectionsPDF ToolsRequest permissionExport citationAdd to favoritesTrack citation ShareShare Give accessShare full text accessShare full-text accessPlease review our Terms and Conditions of Use and check box below to share full-text version of article.I have read and accept the Wiley Online Library Terms and Conditions of UseShareable LinkUse the link below to share a full-text version of this article with your friends and colleagues. Learn more.Copy URL Share a linkShare onFacebookTwitterLinked InRedditWechat Dear Editor, Our previous study has reported that DNA methylation serves as an important epigenetic factor of gene–environment interaction, which contributes to pathogenesis of rheumatoid arthritis (RA).1 To investigate the role of another epigenetic factor (long noncoding RNA, lncRNA) in RA pathogenesis, we integrated lncRNA and mRNA transcriptomic information, constructed lncRNA→mRNA→RA regulatory network by performing co-expression networks analysis and causal inference test, and explored functional roles of the highlighted lncRNA E2F3-IT1 (E2F3 intronic transcript 1) in RA (Figure S1). This study first isolated peripheral blood mononuclear cells (PBMCs) from RA patients (N = 25)2 and age- and sex-matched controls (N = 18) (Table 1A), and tested 22,774 lncRNAs and 25,004 mRNAs expressions through human microarray (Figure 1A). Differential expression analyses identified a total of 402 lncRNAs and 832 mRNAs (fold-change > 2 and false discovery rate < 0.05) as potential targets for subsequent analyses (Table S1). Since the functions of lncRNAs are largely unknown, we performed the weighted gene co-expression network analysis (WGCNA)3 by simultaneously incorporating information of the above differential expressed genes, and two interesting co-expression modules were constructed, named yellow and magenta (Figure 1B). The yellow module has the highest module significance, indicating that the yellow module genes were more likely associated with RA. GO functional enrichment analysis showed that the genes in the yellow module were enriched in “regulation of innate immune response” and “regulation of gene expression” (Table S2). From the yellow module, a total of 31 hub lncRNAs and 30 hub mRNAs (correlation coefficient r2 ≥ 0.8) were selected (Table S2) to construct the regulatory chain of lncRNA (causal factor) →mRNA (mediator) →RA (outcome) through causal inference test (CIT) analysis.4 A total of 191 significant causative regulatory chains were identified including 20 lncRNAs and 21 mRNAs (Figure 1C and Table S3). TABLE 1. Basic characteristics of the study subjects and the expression levels of the selected mRNA and lncRNA in PBMCs in the validation sample (A) Basic characteristics of the study subjects Discovery group Validation group Variable RA patient (n = 25) Healthy control (n = 18) RA patient (n = 35) Healthy control (n = 35) Gender Female Female Female Female Age (year) 45.6 ± 9.84 47.11 ± 14.09 46.44 ± 10.99 47.57 ± 13.99 BMI (kg/m2) 22.07 ± 3.31 22.32 ± 2.79 22.24 ± 3.67 22.32 ± 2.79 DAS28 4.46 ± 0.99 n.d. 5.08 ± 1.32 n.d. CRP (mg/L) 13.51 + 16.74 n.d. 18.37 ± 31.17 n.d. ESR (mm/h) 42.61 ± 27.5 n.d. 48.29 ± 29.46 n.d. TJC 8.64 ± 6.49 n.d. 10 ± 7.7 n.d. SJC 5.48 ± 3.83 n.d. 6.79 ± 5.43 n.d. (B) The expression levels of the selected mRNA and lncRNA in PBMCs in the validation sample lncRNA Chromosome Fold change p-Value Class CYTOR 17q11.2 1.08 0.67 Intergenic DQ593252 11q12.3 1.21 0.05 Intergenic E2F3-IT1 6p22.3 1.73 0.03 Intronic uc.265 9q31.1 1.35 0.02 Antisense INE2 Xp22.2 1.43 0.049 Antisense mRNA DDX58 9p21.1 1.06 0.67 DExD/H-box helicase 58 IFI16 1q23.1 1.09 0.35 Interferon gamma inducible protein 16 LDLR 19p13.2 2.13 <0.001 Low-density lipoprotein receptor PLSCR1 3q24 -1.67 <0.001 Phospholipid scramblase 1 PARP9 3q21.1 1.39 0.015 Poly(ADP-ribose) polymerase family member 9 Abbreviations: BMI, body mass index; DAS28,28 joint Disease Activity Score; CRP, C reactive protein; ESR, equivalent series resistance; TJC, tender joint count; SJC, swollen joint count; n.d., not determined. Notes: Variables were expressed as the mean ± SD. p-Value represents the significance for the difference between RA patients(n = 35) and healthy controls(n = 35). FIGURE 1Open in figure viewerPowerPoint Integrative lncRNA and mRNA co-expression network analysis and causal inference test (CIT) analysis. (A) Volcano plot of lncRNA and mRNA expressed in RA patients and healthy controls. (B) Gene modules of differentially expressed mRNAs and lncRNAs in PBMCs identified by WGCNA. Left: Diagram of correlation of module's color and RA. The colored column indicates modules and the Y-axis represents gene significance. Right: Clustering dendrograms of PBMCs. System clustering tree was built based on PBMCs dataset. Three kinds of colors present three modules. (C) Significant lncRNA→mRNA→RA regulatory chains identified by CIT To verify hub lncRNAs and mRNAs in the causative regulatory chains, five lncRNAs (CYTOR, UC.265, DQ593252, E2F3-IT1, and INE2) and five mRNAs (DDX58, IFI16, LDLR, PLSCRI, and PARP9) were selected for validation in another sample including 35 RA patients and 35 healthy controls. The RT-qPCR results showed that three lncRNAs (UC.265, E2F3-IT1, and INE2) and three mRNAs (LDLR, PLSCRI, and PARP9) in PBMCs were also differentially expressed in the validation sample (Table 1B). The constructed lncRNA→mRNA→RA causative regulatory chains for the three validated lncRNAs and three validated mRNAs showed that LDLR serves as a significant mediator between lncRNAs (e.g., UC.265, E2F3-IT1, and INE2) and RA disease outcome (Table S4). Among the validated differentially expressed lncRNAs, E2F3-IT1 presented higher fold-change and smaller statistical p-values and hence was further assessed for its functional roles in the pathogenesis of RA. RA is characterized by the breakdown of immunological tolerance. Aberrant T-cell activation has been recognized as the central event in chronic inflammation and synovial hyperplasia.5 Next, lncRNA knock-down cells (E2F3-IT1-SH) were constructed by stably transfecting lentiviral vectors harboring E2F3-IT1 target sequence into Jurkat T cells. Comparing with negative control (E2F3-IT1-NC) cells, lncRNA E2F3-IT1-SH cells presented significantly decreased lncRNA E2F3-IT1 expression (Figure 2A and B), suggesting that E2F3-IT1 was successfully targeted and silenced in Jurkat T cells. To determine whether lncRNA E2F3-IT1 affects cell proliferation, cell number was examined by using the CCK8 assay. Compared with negative control cells, knockdown E2F3-IT1 significantly inhibits cell proliferation (Figure 2C). In addition, an increased percentage of apoptotic cells was observed in lncRNA E2F3-IT1 knockdown cells through flow cytometry analysis (Figure 2D). Furthermore, we found that knockdown E2F3-IT1 could lead to an increased percentage of S-phase cells and a reduced percentage of G2-phase cells through the cell cycle assay (Figure 2E). LncRNA E2F3-IT1 co-expression genes including LDLR, PLSCR1, and PARP9 were also successfully validated in vitro by using E2F3-IT1-SH cells (Figure 2F). We then examined the effect of lncRNA E2F3-IT1 on T-cell activation stimulated with immune activator phorbol-12-myristate-13-acetate (PMA). Under PMA stimulation, the inflammatory cytokines, such as IL-1, IFN-γ, and TNF-α, presented downregulated expression in E2F3-IT1-SH cells as compared with E2F3-IT1-NC cells (Figure 2G). Flow cytometry analysis showed that the expression of antigen CD69 (an early T cells activation biomarker) was increased by PMA induction in E2F3-IT1-NC cells, which was attenuated by knockdown of lncRNA E2F3-IT1 (Figure 2H). These results taken together indicated that lncRNA E2F3-IT1 may be involved in RA pathogenesis by affecting T-cell growth and activation. FIGURE 2Open in figure viewerPowerPoint The functional role of lncRNA E2F3-IT1 in Jurkat cells in vitro. (A) E2F3-IT1-SH Jurkat cells in bright field (left) and fluorescent field (right). (B) Relative expression of lncRNA E2F3-IT1 in E2F3-IT1-SH and E2F3-IT1-NC cells detected by RT-qPCR. (C) OD450 values of E2F3-IT1-SH and E2F3-IT1-NC Jurkat cells in cell proliferation assays. (D) Percentages of apoptotic Jurkat cells detected by annexin V/7AAD double staining through flow cytometry. (E) Percentages of stage-specific cells within the cell cycle for E2F3-IT1-SH and E2F3-IT1-NC. (F) Relative expression of LDLR, PLSCR1, and PARP9 genes in E2F3-IT1-SH and E2F3-IT1-NC cells examined by RT-qPCR. (G) Relative expression of IL-1β, IFN-γ, TNF-α cytokine in PMA-stimulated E2F3-IT1-SH and E2F3-IT1-NC cells. (H) Expression of cell activation marker CD69 in PMA-stimulated E2F3-IT1-SH and E2F3-IT1-NC Jurkat T cells were assessed by flow cytometry. Two-sided Student's t-test was used for intergroup comparisons. **p < 0.01. E2F3-IT1-SH: Jurkat cell transfected with short-hairpin (SH) RNA with knocked-down E2F3-IT1. E2F3-IT1-NC: Jurkat cell transfected with empty vector, serving as negative controls The lncRNA E2F3-IT1 is located at chromosome 6, an intronic transcript of transcriptional factor E2F3 (Figure S2A). Since the function of lncRNA is correlated with its subcellular localization, we carried out a cellular fractionation assay. The data indicated that the distribution of lncRNA E2F3-IT1 is similar to the nuclear-localized U6 snRNA and distinct from the cytoplasm-enriched protein-coding GAPDH mRNA (Figure S2B). We also carried out target prediction by using some bioinformatics tools, such as RNAInter.6 Of interest, lncRNA E2F3-IT1 mainly interacts with transcription factors, histone modification, and RNA binding proteins (Figure S2C). Based on lncRNA E2F3-IT1 subcellular localization and its predicted target moleculars, we inferred that the lncRNA E2F3-IT1 acts its regulation effect mainly through transcription regulatory in the nucleus. Interestingly, the other two E2F3-IT1 regulatory targets PLSCR1 and PARP9 were interferon response genes.7, 8 We proposed that E2F3-IT1 acts its regulation effects mainly on the three interferon response genes (LDLR, PLSCR1, and PARP9) at the transcription level in the nucleus, while the exact functional mechanisms underlying the associations are still needed to be further elucidated. In summary, the present study revealed a significant lncRNA–mRNA interaction network involved in RA and highlighted lncRNA E2F3-IT1 as a novel functional lncRNA associated with RA pathogenesis. These results further elucidate the important roles of lncRNA in RA and provided insights into the diagnosis, classification, and treatment for RA. To our knowledge, this study represents the first effort to explore the lncRNA role on RA by integrating the evidence from multi-omics data. ACKNOWLEDGMENTS The study was supported by the Natural Science Foundation of China (31401079, 81872681, and 81473046), the Science and Technology Project of Suzhou (SS202050 and SYS2019024), and a Project of the Priority Academic Program Development of Jiangsu Higher Education Institutions. CONFLICT OF INTEREST The authors declare no conflict of interest. AUTHOR CONTRIBUTIONS Long-Fei Wu, Xing-Bo Mo, Jia-Hui He, Pei He, and Xin Lu recruited the patients and conducted the experiments. Long-Fei Wu, Xing-Bo Mo, and Jia-Hui He wrote the manuscript and analyzed the data. Hong-Wen Deng, Fei-Yan Deng, and Shu-Feng Lei revised the manuscript. Fei-Yan Deng and Shu-Feng Lei designed and supervised the study. All authors read and approved the final manuscript. ETHICS APPROVAL AND CONSENT TO PARTICIPATE The study protocol was approved by the ethical committees of Soochow University. All study participants provided their written consent for participation in the study. DATA AVAILABILITY STATEMENT The data that support the findings of this study are available from the corresponding author upon reasonable request. Supporting Information Filename Description ctm2325-sup-0001-TableS1.xlsx109.1 KB Supporting Information ctm2325-sup-0002-TableS2.xlsx100.8 KB Supporting Information ctm2325-sup-0003-TableS3.xlsx19.2 KB Supporting Information ctm2325-sup-0004-TableS4.xlsx10.9 KB Supporting Information ctm2325-sup-0005-TableS5.docx16.2 KB Supporting Information ctm2325-sup-0006-SuppMat.docx546.5 KB Supporting Information Please note: The publisher is not responsible for the content or functionality of any supporting information supplied by the authors. Any queries (other than missing content) should be directed to the corresponding author for the article. REFERENCES 1Zhu H, Wu LF, Mo XB, et al. Rheumatoid arthritis-associated DNA methylation sites in peripheral blood mononuclear cells. Ann Rheum Dis. 2019; 78(1): 36- 42. Google Scholar 2Aletaha D, Neogi T, Silman AJ, et al. 2010 rheumatoid arthritis classification criteria: an American College of Rheumatology/European League Against Rheumatism collaborative initiative. Ann Rheum Dis. 2010; 69(9): 1580- 1588. Google Scholar 3Langfelder P, Horvath S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinform. 2008; 9: 559. Google Scholar 4Chen LS, Emmert-Streib F, Storey JD. Harnessing naturally randomized transcription to infer regulatory relationships among genes. Genome Biol. 2007; 8(10). https://doi.org/10.1186/gb-2007-8-10-r219Google Scholar 5Skapenko A, Leipe J, Lipsky PE, Schulze-Koops H. The role of the T cell in autoimmune inflammation. Arthritis Res Ther. 2005; 7(Suppl 2): S4- S14. Google Scholar 6Lin Y, Liu T, Cui T, et al. RNAInter in 2020: rNA interactome repository with increased coverage and annotation. Nucleic Acids Res. 2020; 48(D1): D189- D197. CrossrefGoogle Scholar 7Zhou QS, Zhao J, Al-Zoghaibi F, et al. Transcriptional control of the human plasma membrane phospholipid scramblase 1 gene is mediated by interferon-alpha. Blood. 2000; 95(8): 2593- 2599. Google Scholar 8Oriol-Tordera B, Berdasco M, Llano A, et al. Methylation regulation of Antiviral host factors, Interferon Stimulated Genes (ISGs) and T-cell responses associated with natural HIV control. PLoS Pathog. 2020; 16(8):e1008678. Google Scholar Volume11, Issue2February 2021e325 FiguresReferencesRelatedInformation
What problem does this paper attempt to address?
-
Identification of the key genes and long non-coding RNAs in ankylosing spondylitis using RNA sequencing (vol 43, pg 1179, 2019)
Zhengkuan Xu,Hao Li,Qixin Chen,Gang Chen
DOI: https://doi.org/10.3892/ijmm.2018.4038
IF: 5.314
2022-01-01
International Journal of Molecular Medicine
Abstract:Ankylosing spondylitis (AS) is an insidious and debilitating form of arthritis that involves the axial skeleton, and its etiology and pathogenesis remain unclear. In the present study, three patients with AS and three normal controls from our hospital were enrolled. RNA sequencing and bioinformatics analysis were performed in order to identify the differentially expressed (DE) mRNAs (DEmRNAs) and DE long non-coding RNAs (DElncRNAs) between the patients with AS and normal controls. Construction of an AS-specific protein-protein interaction network, a weighted DElncRNA-DEmRNA co-expression network and functional annotation of the DEmRNAs co-expressed with DElncRNAs was performed. Nearby cis-targeted DEmRNAs or DElncRNAs were identified by searching for DEmRNAs that were transcribed within 100-kb up- or downstream of DElncRNAs. Based on the Gene Expression Omnibus datasets GSE25101 and GSE73754, the expression of selected DEmRNAs and DElncRNAs were verified using published RNA sequencing data from blood samples, and receiver operating characteristic analysis of selected DEmRNAs was performed. Compared with the normal controls, 1,072 DEmRNAs and 372 DElncRNAs in the patients with AS were identified. Caspase recruitment domain family member 11 and DNA methyltransferase 1 have great diagnostic value for AS. MSTRG.8559 and LINC00987 were also identified as two hub DElncRNAs. The T-cell receptor signaling pathway was a significantly enriched pathway of the DEmRNAs co-expressed with DElncRNAs in patients with AS. In conclusion, the present study identified the key DEmRNAs and DElncRNAs in AS, which provides novel information for understanding the pathogenesis of AS and developing potential biomarkers for AS.
-
Novel Long Non-coding RNA Expression Profile of Peripheral Blood Mononuclear Cells Reveals Potential Biomarkers and Regulatory Mechanisms in Systemic Lupus Erythematosus
Qi Cheng,Mo Chen,Xin Chen,Xiaochan Chen,Huawei Jiang,Huaxiang Wu,Yan Du
DOI: https://doi.org/10.3389/fcell.2021.639321
IF: 27.973
2021-01-01
Annals of the Rheumatic Diseases
Abstract:ObjectiveThe multisystem involvement and high heterogeneity of systemic lupus erythematosus (SLE) lead to great challenges in its diagnosis and treatment. The purpose of this study was to find new lncRNAs in peripheral blood mononuclear cells of SLE patients by transcriptome sequencing and explore their potential as biomarkers and their correlation with clinical features.Materials and MethodsTranscriptome sequencing was used to screen differentially expressed lncRNAs (DELs) and mRNAs (DEMs). The expression of these selected lncRNAs and mRNAs in SLE patients and healthy controls was verified by qPCR. DAVID and WebGestalt were used to perform enrichment analysis. Cytoscape was used to construct a protein–protein network, a coexpression network, and a competitive endogenous RNA network to reveal the regulatory mechanisms of lncRNAs at the transcriptome level.ResultsA total of 1737 DELs and 4078 DEMs were identified between SLE patients and healthy controls. Ten lncRNAs and eight genes were verified by qPCR in a larger sample set. The lncRNA NONHSAT101022.2 was significantly downregulated in SLE patients and was also significantly related to the activity and severity of disease. The upregulated genes were enriched in defense and the immune response, while the downregulated genes were mainly enriched in SLE-related pathways. Topology network analysis revealed that the lncRNAs were involved in regulation at the transcriptome level, including acting directly on mRNA or indirectly affecting gene expression by acting on miRNA.ConclusionIn this work, we identified many mRNAs and novel lncRNAs by transcriptome sequencing. The functions and regulatory mechanisms of these lncRNAs were analyzed by bioinformatic methods. The novel lncRNA NONHSAT101022.2 is significantly downregulated in SLE patients and is significantly related to the activity and severity of disease. Additionally, we propose that NONHSAT101022.2 may enhance the signal transduction of β2-AR by cis regulating LMBRD2, inducing NK cells to produce high levels of IFN-γ and thereby exacerbating SLE.
-
LncRNA: an All-rounder in Rheumatoid Arthritis
Junyu Liang,Weiqian Chen,Jin Lin
DOI: https://doi.org/10.2478/jtim-2019-0002
2019-01-01
Journal of Translational Internal Medicine
Abstract:Rheumatoid arthritis (RA) is a chronic autoimmune disease and is supposed to have both genetic and environmental backgrounds. Plenty of studies have demonstrated the roles of long non-coding RNAs (lncRNAs) in the initiation and development of RA. Numerous lncRNAs have been found to be dysregulated in RA and to be correlated with disease activity of RA, which indicates potential diagnostic roles of lncRNAs. In addition to working as biomarkers for RA, lncRNAs participate in many specific pathological processes including inflammation, aberrant proliferation, migration, invasion and apoptosis. Further screenings and researches are required to validate the clinical potentials of lncRNAs as diagnostic and therapeutic targets in RA.
-
Comprehensive analysis of long non-coding RNA and mRNA expression profiles in rheumatoid arthritis.
Qing Luo,Chuxin Xu,Xue Li,Lulu Zeng,Jianqing Ye,Yang Guo,Zikun Huang,Junming Li
DOI: https://doi.org/10.3892/etm.2017.5284
IF: 2.7
2017-01-01
Experimental and Therapeutic Medicine
Abstract:Abnormal expression of long non-coding RNA (lncRNA) has been demonstrated to be involved in a variety of human diseases. However, the role of lncRNA remains largely unknown in rheumatoid arthritis (RA). The present study aimed to investigate whether lncRNA are differentially expressed in RA. Differentially expressed lncRNA and mRNA in peripheral blood mononuclear cells from individuals with RA and healthy controls were detected using a human lncRNA microarray containing 30,586 lncRNA and 26,109 coding transcripts. Several candidate lncRNA and mRNA in 24 paired samples were verified by reverse transcription-quantitative polymerase chain reaction analysis. Bioinformatics analyses (Gene Ontology and Kyoto Encyclopedia of Genes and Genomes) were used to evaluate signaling pathways and biological functions influenced by the differentially expressed mRNA. A total of 5,045 lncRNA (upregulated, 2,410; downregulated, 2,635) and 3,289 mRNA (upregulated, 1,403; downregulated, 1,886) were differentially expressed in patients with RA (fold-change > 2; P<0.05). The majority of abnormal lncRNA were from intergenic spacer regions (42%), natural antisense (19%) and intronic antisense (15%) to protein-coding loci. lncRNA target prediction indicated the presence of 135 potential lncRNA-mRNA target pairs for the 85 aberrant lncRNA and 109 aberrant mRNA. Significantly enriched (P< 0.05) signaling pathways based on deregulated mRNA were mostly implicated in bile secretion, T cell receptor signaling pathway and systemic lupus erythematosus. In summary, to the best of our knowledge, the present study executed global expression profiling of lncRNA and mRNA involved in RA for the first time. These results may provide important insights regarding lncRNA in RA pathogenesis and provide potential therapeutic targets.
-
Identification of the Potential Regulatory Interactions in Rheumatoid Arthritis Through a Comprehensive Analysis of Lncrna-Related Cerna Networks.
Mingyi Yang,Yani Su,Haishi Zheng,Ke Xu,Qiling Yuan,Yongsong Cai,Yirixiati Aihaiti,Peng Xu
DOI: https://doi.org/10.1186/s12891-023-06936-3
IF: 2.562
2023-01-01
BMC Musculoskeletal Disorders
Abstract:OBJECTIVE:This study aimed at constructing a network of competing endogenous RNA (ceRNA) in the synovial tissues of rheumatoid arthritis (RA). It seeks to discern potential biomarkers and explore the long non-coding RNA (lncRNA)-microRNA (miRNA)-messenger RNA (mRNA) axes that are intricately linked to the pathophysiological mechanisms underpinning RA, and providing a scientific basis for the pathogenesis and treatment of RA.METHODS:Microarray data pertaining to RA synovial tissue, GSE103578, GSE128813, and GSE83147, were acquired from the Gene Expression Omnibus (GEO) database ( http://www.ncbi.nlm.nih.gov/geo ). Conducted to discern both differentially expressed lncRNAs (DELncRNAs) and differentially expressed genes (DEGs). A ceRNA network was obtained through key lncRNAs, key miRNAs, and key genes. Further investigations involved co-expression analyses to uncover the lncRNA-miRNA-mRNA axes contributing to the pathogenesis of RA. To delineate the immune-relevant facets of this axis, we conducted an assessment of key genes, emphasizing those with the most substantial immunological correlations, employing the GeneCards database. Finally, gene set enrichment analysis (GSEA) was executed on the identified key lncRNAs to elucidate their functional implications in RA.RESULTS:The 2 key lncRNAs, 7 key miRNAs and 6 key genes related to the pathogenesis of RA were obtained, as well as 2 key lncRNA-miRNA-mRNA axes (KRTAP5-AS1-hsa-miR-30b-5p-PNN, XIST-hsa-miR-511-3p/hsa-miR-1277-5p-F2RL1). GSEA of two key lncRNAs obtained biological processes and signaling pathways related to RA synovial lesions.CONCLUSION:The findings of this investigation hold promise in furnishing a foundational framework and guiding future research endeavors aimed at comprehending the etiology and therapeutic interventions for RA.
-
Long Non-Coding RNA AL928768.3 Promotes Rheumatoid Arthritis Fibroblast-Like Synoviocytes Proliferation, Invasion and Inflammation, While Inhibits Apoptosis Via Activating Lymphotoxin Beta Mediated NF-κB Signaling Pathway.
Li Sun,Lingzhen Hu,Peirong Chen,Yongji Li,Jianxin Tu,Jianghua Chen
DOI: https://doi.org/10.1007/s10753-023-01927-x
2024-01-01
Inflammation
Abstract:Our previous study using RNA sequencing and reverse transcription quantitative polymerase chain reaction (RT-qPCR) validation identified a long non-coding RNA (lnc), lnc-AL928768.3, correlating with risk and disease activity of rheumatoid arthritis (RA), then the present study was conducted to further investigate the interaction of lnc-AL928768.3 with lymphotoxin beta (LTB) and their impact on proliferation, migration, invasion, and inflammation in RA-fibroblast-like synoviocytes (RA-FLS). Human RA-FLS was obtained and transfected with lnc-AL928768.3 overexpression, negative control overexpression, lnc-AL928768.3 short hairpin RNA (shRNA) and negative control shRNA plasmids. Then cell functions and inflammatory cytokine expressions were detected. Afterward, rescue experiments were conducted via transfecting lnc-AL928768.3 shRNA with or without LTB overexpression plasmids in RA-FLS. Lnc-AL928768.3 enhanced proliferation and invasion, inhibited apoptosis, while had little impact on migration in RA-FLS. In addition, lnc-AL928768.3 positively modulated interleukin-1β (IL-1β), IL-6 and IL-8 expressions in RA-FLS supernatant; moreover, it also positively regulated LTB mRNA expression, LTB protein expression, p-NF-κB protein expression, and p-IKB-α protein expression in RA-FLS. Furthermore, following experiment showed that lnc-AL928768.3 positively regulated LTB expression while LTB did not impact on lnc-AL928768.3 expression in RA-FLS. Furthermore, in rescue experiments, LTB overexpression curtailed the effect of lnc-AL928768.3 knock-down on regulating proliferation, invasion, apoptosis and inflammatory cytokine expressions in RA-FLS. Lnc-AL928768.3 promotes proliferation, invasion, and inflammation while inhibits apoptosis of RA-FLS via activating LTB mediated NF-κB signaling.
-
Differential long non-coding RNA expression profiles in the peripheral blood and CD4<SUP>+</SUP>T cells of patients with active rheumatoid arthritis
Ming Li,Kexun Ma,Zhe Feng,Jing Wang,Xueping Zhou,Lingling Zhou
DOI: https://doi.org/10.3892/etm.2020.8681
IF: 2.7
2020-01-01
Experimental and Therapeutic Medicine
Abstract:The human transcriptome is primarily composed of long non-coding RNAs (lncRNAs), which are key regulatory molecules of multiple biological processes. In the present study, the expression profiles of lncRNAs in the peripheral blood and CD4(+)T cells of patients with active rheumatoid arthritis (RA) were determined. Based on the expression profiles, 493 lncRNAs and 374 mRNAs were identified to be differentially expressed in the peripheral blood of active RA patients and healthy donors. Further verification of lncRNAs was performed using reverse transcription-quantitative (RT-q) PCR analysis of peripheral blood from 5 healthy donors and 5 patients with active RA and 14 additional differentially expressed genes were identified. CD4(+)T cells in peripheral blood from 12 patients with active RA and 8 healthy donors were isolated using magnetic beads and qPCR was used to assess differentially expressed lncRNAs. The results suggested that 7 lncRNAs were upregulated and 2 were downregulated. The results indicated that these 9 lncRNAs may be involved in the pathogenesis of RA. An increased ratio of Th17: T-regulatory (Treg) cells was also observed. It may be hypothesized that LncRNAs serve important roles in the differentiation of CD4(+)T cells. Receiver operating characteristic curve analysis suggested that these 9 lncRNAs are of potential clinical diagnostic value for RA. Pearson correlation analysis indicated that the correlation coefficient between Ensembl transcript (ENST)00000569543 and complement C4 was 0.623 (P<0.05), and that between ENST00000420096 and anti-cyclic citrullinated peptide antibody or disease activity evaluation score, the correlation coefficient was 0.662 and 0.605, respectively (P<0.05 for each). In conclusion, the results of the present study suggest a possible role of lncRNAs in the differentiation of CD4(+)T cells and the pathogenesis of RA, as well as the potential value as diagnostic biomarkers for active RA.
-
Comprehensive analysis of serum exosome-derived lncRNAs and mRNAs from patients with rheumatoid arthritis
Li Xue,Biao Wang,Xueyi Li,Jianhong Zhu,Wei Wang,Fang Huang,Xiaofei Wang,Yaofeng Jin,Chaoliang Xiong,Li Tao,Ke Xu,Jing Wang,Ying Guo,Jing Xu,Xin Yang,Na Wang,Ning Gao,Yan Wang,Ke Li,Ming Li,Yan Geng
DOI: https://doi.org/10.1186/s13075-023-03174-9
2023-10-16
Abstract:Background: Serum exosomes play important roles in intercellular communication and are promising biomarkers of several autoimmune diseases. However, the biological functions and potential clinical importance of long non-coding RNAs (lncRNAs) and mRNAs from serum exosomes in rheumatoid arthritis (RA) have not yet been studied. Methods: Serum exosomal lncRNAs and mRNAs were isolated from patients with RA and osteoarthritis (OA) and healthy controls. The differentially expressed lncRNAs (DE-lncRNAs) and mRNA profiles in the serum exosomes of patients with RA were analysed using high-throughput sequencing, and their functions were predicted using Gene Ontologyenrichment, Kyoto Encyclopedia of Genes and Genomes pathway, and gene set enrichment analysis. We constructed a DE-lncRNA-mRNA network and a protein-protein interaction network of differentially expressed mRNAs (DE-mRNAs) in RA using the Cytoscape software. The expression of several candidate a DE-lncRNAs and DE-mRNAs in the serum of patients with RA, patients with OA, and healthy controls was confirmed by qRT-PCR. We assessed the diagnostic ability of DE-lncRNAs and DE-mRNAs in patients with RA using receiver operating characteristic analysis. Furthermore, we analysed the characteristics of immune cell infiltration in RA by digital cytometry using the CIBERSORT algorithm and determined the correlation between immune cells and several DE-lncRNAs or DE-mRNAs in RA. Results: The profiles of serum exosomal lncRNAs and mRNAs in patients with RA were different from those in healthy controls and patients with OA. The functions of both DE-lncRNAs and DE-mRNAs in RA are associated with the immune response and cellular metabolic processes. The RT-PCR results show that NONHSAT193357.1, CCL5, and MPIG6B were downregulated in patients with RA. The combination of three DE-mRNAs, CCL5, MPIG6B, and PFKP, had an area under the curve of 0.845 for differentiating RA from OA. Digital cytometry using the CIBERSORT algorithm showed that the neutrophil counts were higher in patients with RA than those in healthy controls and patients with OA. Conclusions: These findings help to elucidate the role of serum exosomal lncRNAs and mRNAs in the specific mechanisms underlying RA.
-
Long noncoding RNA expression profile in fibroblast-like synoviocytes from patients with rheumatoid arthritis
Yu Zhang,Yu-Zhong Xu,Ning Sun,Jian-Hong Liu,Fang-Fang Chen,Xiao-Long Guan,Ang Li,Fei Wang,Qin-Fei Zhao,Hai-Yong Wang,Shu-Sheng Song,Wei Yu,Jian-Ning Zhao,Xiao-Jun Li
DOI: https://doi.org/10.1186/s13075-016-1129-4
2016-01-01
Arthritis Research & Therapy
Abstract:Background Long noncoding RNAs (lncRNAs) have recently received wide attention as key molecules that mediate a variety of physiological and pathological processes by regulating gene expression; however, knowledge of lncRNAs in rheumatoid arthritis (RA) is limited. Thus, we investigated the lncRNA expression profile in fibroblast-like synoviocytes (FLSs) from patients with RA and explored the function of abundantly expressed lncRNAs. Methods LncRNA and mRNA microarrays were performed to identify differentially expressed lncRNAs in RA FLSs compared with normal FLSs. Quantitative polymerase chain reaction (qPCR) was used to validate the results, and correlation analysis was used to analyze the relationship between these aberrantly expressed lncRNAs and clinical characteristics. A receiver operating characteristic (ROC) curve was constructed to evaluate the diagnostic value of the lncRNAs identified. Results According to the gene expression profiles, 135 lncRNAs were differentially expressed between RA and normal FLSs. Furthermore, qPCR data showed that lncRNA ENST00000483588 was up-regulated and that three lncRNAs (ENST00000438399, uc004afb.1, and ENST00000452247) were down-regulated in RA FLSs. The expression level of ENST00000483588 was positively correlated with the level of C-reactive protein and the Simplified Disease Activity Index score. Moreover, the areas under the ROC curve were 0.85, 0.92, 0.97, and 0.92 for ENST00000483588, ENST00000438399, uc004afb.1, and ENST00000452247, respectively. Conclusions The results indicate that the dysregulation of ENST00000483588, ENST00000438399, uc004afb.1, and ENST00000452247 may be involved in the pathological processes of RA and that these lncRNAs may have potential value for the diagnosis and assessment of the disease activity of RA.
-
Screening and analysis for autophagy - related lncRNA in fibroblast - like synoviocytes from patients with rheumatoid arthritis
Hua Shang,Qing Peng,Jiajun Liu,Yan Liu,Li Long
DOI: https://doi.org/10.11817/j.issn.1672-7347.2021.210374
2021-10-28
Abstract:Objectives: Long non-coding RNA (lncRNA) has become a key epigenetic regulator that regulates gene expression and affects a variety of biological processes. LncRNA plays an important role in the occurrence and development of rheumatoid arthritis (RA). The study on lncRNA in peripheral blood cells of RA patients has been reported. However, there is no study on autophagy regulation by lncRNA in RA patients. This study aims to provide a new direction for the diagnosis and treatment of RA via screening the changes of lncRNAs in RA fibroblast-like synoviocytes (RA-FLSs) before and after autophagy and finding the key lncRNAs targeting RA-FLSs autophagy. Methods: Synovial tissues of 6 RA patients after knee and hip joint surgery were obtained, and RA-FLSs were cultured to the 5th generation for further experiments (tissue culture method). After treatment with mTOR inhibitor PP242, the expression of LC3-II was detected by Western blotting. Total RNAs of 3 cases of RA-FLSs before and after treatment with mTOR inhibitor PP242 were extracted by TRIzol and screened by Agilent Human ceRNA Microarray 2019 (4×180 K, design ID: 086188) chip. The lncRNAs with significantly changed expression levels were selected (difference multiple≥2.0, P<0.05). Bioinformatics technology was used to analyze the gene ontology (GO) function and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway of the differentially expressed lncRNAs, and to explore the possible role of differentially expressed lncRNAs in the pathogenesis of RA. Subsequently, ENST00000584721.1 and ENST00000615939.1 were identified in all the 6 samples of RA-FLSs using real-time RT-PCR, which were selected by previous chip screen combined with GO enrichment analysis and KEGG analysis. Results: RA-FLSs were successfully isolated and cultured from the synovial tissues of the patient's knee or hip joint. After 6 RA-FLSs were treated with PP242, the expression level of autophagy marker protein LC3-II was increased (P<0.05). PP242 induced autophagy in RA-FLSs. LncRNA sequencing analysis showed that a total of 591 lncRNAs were significantly changed, of which 428 were up-regulated and 163 were down-regulated.Go analysis showed that these differentially expressed lncRNAs were associated with negative regulation of Th17 function, T cell related cytokines production, and TGF-β receptor signaling pathway, as well as IL-6 receptor complex formation and type I TGF-β receptor binding. KEGG analysis showed that autophagy pathway, TGF-β, TNF-α, IL-17 signaling pathway, and Th17, Th1, Th2, osteoclast differentiation pathway were the most abundant signal pathways of differentially expressed lncRNAs during autophagy. The expression of ENST00000584721.1 was up-regulated (P<0.05) and the expression ofENST00000615939.1 was down-regulated (P<0.05) in RA-FLSs undergoing autophagy, by using real-time RT-PCR validation which was in consistent with the microarray results. The reliability of microarray screening differential genes was confirmed. GO analysis showed that ENST00000584721.1 and ENST00000615939.1 were related to ribosome transport and autophagy assembly, respectively. KEGG analysis showed that ENST00000584721.1 was related to mitogen-activated protein kinase (MAPK) signaling pathway, and ENST00000615939.1 was related to FoxO signaling pathway. Conclusions: Differentially expressed lncRNAs in RA-FLSs have been identified with microarray analysis. In RA, differential expression of lncRNAs is involved in the autophagy of RA-FLSs. The underlying mechanisms based on bioinformatics analysis include regulating the secretion of cytokines, such as IL-6, TGF-β, TNF-α and IL-17, participating in the immune cell differentiation, such as Th17, Th1, Th2 cells and osteoclasts, as well as regulating the autophagy pathway, MAPK, FoxO, and other signaling pathways. It has been verified that the expression of ENST0000584721.1 is up-regulated and ENST0000615939.1 is down-regulated after autophagy of RA FLSs, which provides a good experimental basis for further study on the mechanism of lncRNA in RA-FLSs autophagy.
-
Association of gene polymorphisms and the decreased expression of long non-coding RNA LOC553103 with rheumatoid arthritis
Sha-Sha Tao,Xi Fang,Liang-Zi Xu,Ruo-Di Zhang,Qing-Qing Luo,Jian Tang,Xiao-Fan Dai,Shu-Zhen Xu,Xiao-Ke Yang,Hai-Feng Pana
DOI: https://doi.org/10.1093/postmj/qgae055
2024-04-24
Postgraduate Medical Journal
Abstract:Abstract Background Long non-coding RNAs (lncRNAs) are involved in many key bioprocesses, including the occurrence and development of rheumatoid arthritis (RA). We aimed to analyze the association of genetic variants of long non-coding RNA LOC553103 and its peripheral blood mononuclear cells (PBMC) expression with RA. Methods We enrolled 457 RA patients and 551 healthy controls and conducted a case–control study to analyze the relationship between LOC553103 gene rs272879 and the susceptibility of RA by TaqMan single nucleotide polymorphism genotyping. Among them, we sampled 92 cases and 92 controls, respectively, to detect the PBMC level of LOC553103 using quantitative real-time polymerase chain reaction technology. We explored the association between LOC553103 rs272879 and its PBMC expression levels in 71 RA patients. Mann–Whitney, Chi-square, and Spearman correlation analysis were used for statistical analysis and P-value <.05 was considered statistically significant. Results The genotype frequency of LOC553103 rs272879 CC was increased, and CG was decreased in RA patients compared to the control group (χ2 = 6.772, P = .034). The LOC553103 expression level in PBMC of RA patients was downregulated compared to healthy control (Z = −4.497, P < .001). Moreover, negative correlations were observed between the PBMC level of LOC553103 and erythrocyte sedimentation rate (rs = −0.262, P = .018), white blood cell count (rs = −0.382, P = .004), platelet (rs = −0.293, P = .030), and disease activity score in 28 joints (rs = −0.271, P = .016) in RA patients. Conclusions This study provides the first evidence supporting an association between LOC553103 gene polymorphisms and susceptibility of RA and a relationship of PBMC level of LOC553103 with clinical manifestations and laboratory indicators of RA patients.
medicine, general & internal
-
Identification of serum exosomal lncRNAs and their potential regulation of characteristic genes of fibroblast-like synoviocytes in rheumatoid arthritis
Tong-Sheng Zhou,Chun-Lan Yang,Jie-Quan Wang,Ling Fang,Quan Xia,Ya-Ru Liu
DOI: https://doi.org/10.1016/j.intimp.2024.113382
2024-12-25
Abstract:Rheumatoid arthritis (RA) is a common autoimmune disease whose pathogenesis is poorly understand. Gaps in laboratory biomarkers cause a lack of clinically available strategies for the early diagnosis and treatment of RA. This study aims to identify serum exosomal lncRNAs as promising biomarkers and to unravel potential mechanisms by which they affect characteristic genes of fibroblast-like synoviocytes (FLSs) to induce RA malignant properties. RNA sequencing datasets of serum exosomes (GSE271161 and PRJNA911001) and FLSs (GSE103578, GSE122616, GSE128813, GSE181614 and GSE83147) were purposively mined. Visualization and functional enrichment of differentially expressed (DE) lncRNAs/protein-coding genes, screening of significant lncRNAs, and construction of competing endogenous RNAs (ceRNAs) and protein-protein interaction (PPI) network were carried out. Quantitative real-time PCR, receiver operating characteristic curve (ROC) and correlation analysis were conducted on the validation cohort. As a result, we screened a total of 131 serum exosomal DElncRNAs and 125 FLSs DEmRNAs, which were predominantly enriched in the proliferative, inflammatory and metabolic pathways. In-depth learning of DElncRNAs expression profiles was performed to identify models with better performance and lncRNAs with higher importance scores using 4 machine learning algorithms (SVM, KNN, RF, Logit), which led to the establishment of ceRNAs network linking serum exosomal lncRNAs and characteristic genes of FLSs. In short, we proposed that 4 RA-representative serum exosomal lncRNAs (DLEU2, FAM13A-AS1, MEG3 and SNHG15) may be applied as valuable indicators for laboratory tests, and their-mediated intercellular communication and ceRNAs network may regulate the characteristic genes of FLSs, thereby generating malignant phenotypes and adaptive synovial microenvironment in RA.
-
Epigenetically Regulated Co-Expression Network of Genes Significant for Rheumatoid Arthritis.
Pei He,Xing-Bo Mo,Shu-Feng Lei,Fei-Yan Deng
DOI: https://doi.org/10.2217/epi-2019-0028
2019-01-01
Epigenomics
Abstract:Aim: To identify epigenetically regulated network of genes in peripheral blood mononuclear cells significant for rheumatoid arthritis (RA). Methods: Differentially expressed genes (DEGs) and their associated differentially expressed miRNAs and differentially methylated positions (DMPs) were identified. Causal inference test (CIT) identified the causal regulation chains. The analyses, for example, weighted gene co-expression network (WGCNA), protein-protein interaction and functional enrichment, evaluated interaction patterns among the DEGs and the associated epigenetic factors. Results: A total of 181 DEGs were identified. The DEGs were significantly regulated by DMPs and/or differentially expressed miRNAs. Causal inference test analyses identified 18 causal chains of DMP-DEG-RA and 16 intermediate DEGs enriched in 'protein kinase inhibitor activity'. BTN2A1 was co-expressed with other 9 intermediate genes and 11 known RA-associated genes and played a pivotal role in the co-expression network. Conclusion: Epigenetically regulated network of genes in peripheral blood mononuclear cells (PBMC) contributed to RA. The causal DMPs and key intermediate genes may serve as potential biomarkers for RA.
-
The Study of m6A, m5C, m1A, m7G, Adenosine-to-Inosine and Pseudouridine RNA Modification-related Molecules in Peripheral Blood of Rheumatoid Arthritis
Ziqing Yu,Jifeng Tang,Jinfang Xia,Yinfeng Xiao,Yujue He,Renquan Jiang,Shuhui Chen,Jinpiao Lin
DOI: https://doi.org/10.21203/rs.3.rs-964198/v1
2021-10-26
Abstract:Abstract Background The overall situation of RNA epigenetics in rheumatoid arthritis (RA) is still unclear. This study aims to investigate the expression level of m6A, m5C, m1A, m7G, adenosine-to-inosine, pseudouridine(ψ) modification-related molecules in peripheral blood of RA. Methods 33 RA patients and 36 healthy donors (HD) were included in this study. Gene expression levels were detected by real time-polymerase chain reaction (RT-PCR). Correlation analyses were performed by Pearson correlation test. Least absolute shrinkage and selection operator regression (LASSO) regression and Logistic regression were used for seeking risk factors. Results The expression levels of METTL3 , TET2 , ADAR1 increased while ALKBH5 , TRMT10C TRMT61B ALKBH3 ALYREF METTL1 decreased in RA patients. Analysis shown 47 weak correlations, 43 moderate correlations and 2 strong correlations existed among these molecules. Area under curve (AUC) of ALYREF and TET2 combined ROC was 0.976 which indicated them were the most valuable risk factors in RA. The expression of ALKBH3 and TRMT10C was higher in ESR-negative RA patients, while the expression of METTL1 and METTL3 was higher in RF-negative group or CRP-negative group respectively. Besides, the level of METTL3 and TRMT10C was increased in DAS-28-ESR. Furthermore, METTL3 level was negative related with the levels of RF and DAS28-ESR. The level of TRMT10C was negative with ESR and DAS-28-ESR levels, and ALKBH5 expression was positive related with CRP level. The differential expression genes were mostly correlated with PLT and MCHC of blood routine examination. Conclusions Our study constructed a network of six kinds of mRNA modifications in RA. We found multiple mRNA-modification-related genes were different expression between RA patients and HD. Complicated mutual relations existed among these genes. ALYREF and TET2 were the most valuable risk factors in RA. This work hopes to support a new perspective for clarifying pathogenic mechanism of RA.
-
Differential expressions and potential clinical values of lncRNAs in the plasma exosomes of rheumatoid arthritis
Zi-Qiang Shuai,Zhi-Xin Wang,Jia-Le Ren,Xiao-Ke Yang,Bin Xu
DOI: https://doi.org/10.1016/j.intimp.2024.111511
2024-02-15
Abstract:Background: Rheumatoid arthritis (RA) is a common autoimmune disease with unclear pathogenesis. Progress in its clinical diagnosis and treatment mainly depends on the elucidation of its pathogenesis and the exploration of new biomarkers. Exosomes contain various biomolecules, including long non-coding ribonucleic acids (lncRNAs). lncRNAs may participate in the regulation of autoimmune and inflammatory processes during RA pathogenesis by transmitting these biomolecules via exosomes among different cells. Therefore, the investigation of lncRNAs in RA exosomes may be a feasible pathway to elucidate RA pathogenesis, identify new diagnostic biomarkers, and identify potential therapeutic targets. Methods: In the first phase of exosomal non-coding RNAs screening, exosomes were isolated from the peripheral blood of six patients with RA and healthy controls (HC). High-throughput RNA sequencing was performed to obtain lncRNA expression profiles, and 15 lncRNAs with the highest differential expression were selected as candidate lncRNAs. In the second phase of validation using real-time quantitative polymerase chain reaction (qRT-PCR), differential expression of the 15 candidate lncRNAs was verified in 42 patients with RA and their matched HC. Their potential value as RA diagnostic biomarkers was assessed using receiver operating characteristic (ROC) curve analysis. Their relationships with common clinical indices of RA were explored using Spearman's rank correlation and linear regression analyses. Result: Compared to HC, patients with RA had 206 upregulated and 2,332 downregulated lncRNAs. Fifteen candidate lncRNAs were validated by qRT-PCR, of which 12 (SNHG6, RPS18P9, RPL21P28, EBLN3P, FAM153CP, RPL23P8, SNHG31, NORAD, H3P6, DLEU2, TUG1, and OIP5-AS1) were upregulated, and three (CXXC4-AS1, OLMALINC, and NPHP3-AS1) were downregulated. In the ROC analysis of the 15 candidate lncRNAs, the area under the curve (AUC) ranged from 0.847 (0.767, 0.927) for OLMALINC to 0.994 (0.984, 1.000) for CXXC4-AS1. Spearman rank correlation analysis revealed erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), and disease activity score of 28 (DAS28) were correlated with seven, six, and five lncRNAs, respectively. Further linear regression analysis revealed a negative relationship between exosomal SNHG6 and ESR (B = -0.384, P = 0.006), and a positive relationship between SNHG31 and ESR (B = 0.381, P = 0.007). Exosomal SNHG6 also showed a negative relationship with CRP (B = -0.361, P = 0.019). Moreover, exosomal RPS18P9 and SNGH31 had a negative effect and a positive effect on DAS28, respectively (B = -0.463, P < 0.001; B = 0.586, P < 0.001), implying novel exosomal lncRNAs were the independent influencing factors of the main RA-related clinical indices. Conclusions: lncRNAs in RA plasma exosomes have characteristic expression profiles, including some lncRNAs with potential as diagnostic biomarkers and therapeutic targets for RA.
-
Analysis of lncRNA expression profiles by sequencing reveals that lnc-AL928768.3 and lnc-AC091493.1 are novel biomarkers for disease risk and activity of rheumatoid arthritis
Li Sun,Jianxin Tu,Cailong Liu,Axiao Pan,Xiaoru Xia,Xiaowei Chen
DOI: https://doi.org/10.1007/s10787-019-00666-6
2019-01-01
Inflammopharmacology
Abstract:Background This study aimed to analyze long non-coding RNA (lncRNA) expression profiles in synovium tissue of patients with RA using RNA sequencing, and to further assess the clinical values of dysregulated lncRNAs in RA diagnosis and monitoring. Methods Thirty patients with RA who underwent knee arthroscopy and 30 controls with knee trauma who underwent surgery were consecutively enrolled and synovium tissue samples of both groups were obtained during surgery. In the exploration stage, lncRNA and mRNA expression profiles in three RA samples and three control samples were detected by RNA sequencing and bioinformatic analyses were then performed. In the validation stage, quantitative polymerase chain reaction (qPCR) was subsequently used to detect expression of five candidate lncRNAs in 30 patients with RA and 30 control patients. Results A total of 349 lncRNAs and 1582 mRNAs were upregulated and 806 lncRNAs and 1295 mRNAs were downregulated in patients with RA compared with controls. Enrichment analyses revealed that these dysregulated lncRNAs and mRNAs were mainly involved in regulating immune response, leukocyte migration, complement activation, and B cell receptor signaling pathway. Subsequent qPCR validation discovered that lnc-AL928768.3 ( P < 0.001) and lnc-AC091493.1 ( P < 0.001) were elevated in patients with RA compared with controls and afford good predictive values for RA risk by receiver operating characteristic (ROC) curve analysis. Additionally, the two lncRNAs were positively associated with C-reactive protein level and disease activity score in 28 joints (ESR) (all P < 0.05). Conclusion Analysis of lncRNA expression profiles by sequencing reveals that lnc-AL928768.3 and lnc-AC091493.1 are novel biomarkers for RA risk and activity.
-
Epigenetically-regulated RPN2 gene influences lymphocyte activation and is involved in pathogenesis of rheumatoid arthritis
Pei He,Fei-Yan Deng,Bing-Hua Wang,Long-Fei Wu,Xu Zhou,Shu-Feng Lei
DOI: https://doi.org/10.1016/j.gene.2021.146059
IF: 3.913
2022-02-01
Gene
Abstract:BACKGROUND: To identify RA-associated genes and to ascertain epigenetic factors and functional mechanisms underlying RA pathogenesis.METHODS: Peripheral blood mononuclear cells (PBMC) transcriptome- and proteome- wide gene expressions were profiled in a case-control study sample. Differentially expressed genes (DEGs) were discovered and validated independently. In-house PBMC genome-wide SNP genotyping data, miRNA expression data and DNA methylation data in the same sample were utilized to identify SNPs [expression quantitative trait locus (eQTLs) and protein quantitative trait locus (pQTLs)], miRNAs, and DNA methylation positions (DMPs) regulating key DEG of interest. Lentivirus transfection was conducted to study the effects of RPN2 on T lymphocyte activation, proliferation, apoptosis, and inflammatory cytokine expression. Rpn2 protein level in plasma was quantitated by ELISA to assess its performance in discriminating RA cases and controls.RESULTS: Twenty-two DEGs were discovered in PBMCs. The most significant DEG, i.e., RPN2, was validated to be up-regulated with RA in PBMCs. A complex regulatory network for RPN2 gene expression in PBMCs was constructed, which consists of 38 eQTL and 53 pQTL SNPs, 3 miRNAs and 2 DMPs. Besides, RPN2 expression was significantly up-regulated with RA in primary T lymphocytes, as well as in PHA-activated T lymphocytes. RPN2 over-expression in T lymphocytes significantly inhibited apoptosis and IL-4 expression and promoted proliferation and activation. PBMCs-expressed RPN2 mRNA and plasma Rpn2 protein demonstrated superior and modest performances in discriminating RA cases and controls, respectively.CONCLUSIONS: RPN2 gene influences T lymphocyte growth and activation and is involved in the pathogenesis of RA. Rpn2 may serve as a novel protein biomarker for RA diagnosis.
genetics & heredity
-
Implication of LncRNAs MEG3 and LINC00305 in Pathophysiological Mechanisms associated with Rheumatoid Arthritis
Alaa Samir Wahba,Maha Emad Ibrahim,Noha Mesbah,Samy Saleh,Dina Abo-El-Matty,Eman T. Mehanna,Alaa Wahba,Maha Ibrahim,Eman Mehanna
DOI: https://doi.org/10.21608/rpbs.2020.32723.1068
2021-01-01
Records of Pharmaceutical and Biomedical Sciences
Abstract:Rheumatoid arthritis (RA) is a joint destructive disorder with great morbidity. Early diagnosis and early and effective therapy may prevent joint damage and lead to better long-term results. Therefore, reliable biomarkers and outcome measures are needed. Complex interplay between multiple risk factors contributes to RA. These risk factors include environmental factors, genetic factors and epigenetic modifications. Long non-coding RNAs (lncRNAs) are involved in these epigenetic mechanisms. Accumulating evidence has shown that lncRNAs participate in the processes of inflammation, aberrant proliferation, apoptosis and angiogenesis. They also play roles in autoimmune diseases, such as SLE, Sjogren syndrome, RA and multiple sclerosis. Maternally expressed gene 3 (MEG3) is a maternally expressed lncRNA of the imprinted DLK1–MEG3 locus located on human 14q32 chromosome. It functions as a tumor suppressor. Decreased MEG3 expression has been observed in various human cancers, both type 1 and type 2 diabetes, osteoarthritis and in RA. Decreased serum levels of lncRNA MEG3 in RA could be attributed to MEG3 promoter hypermethylation induced by hypoxia. This downregulation was associated with increased inflammation, cell proliferation and cell invasion and decreased apoptosis. Long intergenic non-Protein coding RNA 00305 (LINC00305) was identified as a pro-inflammatory atherosclerosis-associated lncRNA. RA was associated with increased LINC00305 which served as a regulator of inflammatory, hypoxic, invasive, apoptotic and proliferative mechanisms associated with RA.
-
A Novel Long Non-Coding RNA, Lnc-Rnu12, Influences the T-cell Cycle Via C-Jun and CCNL2 in Rheumatoid Arthritis
Xing-Bo Mo,Yang-Hua Sun,Long-Fei Wu,Pei He,Rong-Rong Cao,Xin Lu,Yong-Hong Zhang,Fei-Yan Deng,Shu-Feng Lei
DOI: https://doi.org/10.1093/rheumatology/keac553
2023-01-01
Rheumatology
Abstract:OBJECTIVES:Long non-coding RNAs (lncRNAs) play important roles in RA pathogenesis. However, specific lncRNAs that regulate gene expression in RA pathogenesis are poorly known. This study was undertaken to characterize a novel lncRNA (lnc-RNU12) that has a lower-than-normal expression level in RA patients.METHODS:We performed initial genome-wide lncRNA microarray screening in peripheral blood mononuclear cells from 28 RA cases and 18 controls. Multiple methods were used to validate the detected associations between lncRNAs and RA. Furthermore, we identified the source and characteristics of the highlighted lncRNAs, detected the target genes, and determined the functional effect on immune cells through lncRNA knock-down in Jurkat T cell lines.RESULTS:lnc-RNU12 was downregulated in peripheral blood mononuclear cells and T cell subtypes of RA patients and was genetically associated with RA risk. lnc-RNU12 mediates the effect of microbiome alterations on RA risk. Activation of T cells caused low expression of lnc-RNU12. Knock-down of lnc-RNU12 in Jurkat T cells caused cell cycle S-phase arrest and altered the expression of protein-coding genes related to the cell cycle and apoptosis (e.g. c-JUN, CCNL2, CDK6, MYC, RNF40, PKM, VPS35, DNAJB6 and FLCN). Finally, c-JUN and CCNL2 were identified as target genes of lnc-RNU12 at the mRNA and protein expression levels. RNA-binding protein immunoprecipitation assays verified the interaction between lnc-RNU12 and the two proteins (c-Jun and cyclin L2) in Jurkat cells.CONCLUSIONS:Our study suggested that lnc-RNU12 was involved in the pathogenesis of RA by influencing the T cell cycle by targeting c-JUN and CCNL2.
-
P053 LncRNA HOTAIR Promotes Proliferation and Invasion of Fibroblast-Like Synoviocytes As Microrna Sponging in RA Patients
X. Bi,Y. F. Pan,X. Q. Luo,M. L. Wang,Y. X. Chen
DOI: https://doi.org/10.1136/annrheumdis-2018-ewrr2019.45
IF: 27.973
2019-01-01
Annals of the Rheumatic Diseases
Abstract:Career situation of first and presenting author: Student for a master or a PhD. Introduction Long non-coding RNAs (lncRNAs) have drawn increasing attention because of the pivotal roles which they play in various types of autoimmune diseases, including rheumatoid arthritis (RA). LncRNA HOTAIR is a crucial lncRNA function as an oncogene in multiple cancers. Fibroblast-like synoviocytes (FLSs), a prominent component of hyperplastic synovial pannus tissue, are critical to synovial aggression and joint destruction in RA. However, the functions of lncRNA and the potential mechanisms remain to be further elucidated in FLSs of RA patients. Objectives Our present study aimed to investigate the expression and roles of lncRNA HOTAIR in RA-FLSs and explore its possible mechanism. Methods FLSs were cultured from synovial tissues of join. LncRNA and mircoRNA expression profiles in FLSs were screened by microarrays, and then we validated the results by Real-time Quantitative polymerase chain reaction (qRT-PCR). Small interfering RNA (siRNA) was then used to knock down the expression of HOTAIR in order to determine its role in RA FLSs. Cell viability was evaluated using the CCK-8 assay and flow cytometry. Cell invasion was analyzed by transwell chamber methodology. Bioinformatics analysis were performed to predict the possible competitive endogenous RNA (ceRNA) mechanisms via miRanda, PITA, RNAhybrid, as well as KEGG and Gene Ontology(GO) analysis. Results Both microarray analysis and qRT-PCR showed the expressions of lncRNA HOTAIR were up-regulated in RA FLSs compared with healthy controls (HCs). Transfection of HOTAIR-siRNA significantly decreased the expression of lncRNA HOTAIR in RA FLSs. HOTAIR knockdown largely inhibited cell proliferation and invasion of RA FLSs. Furthermore, the bioinformatics analysis predicted that some of microRNAs and mRNAs may be the downstream molecules of lncRNA HOTAIR. Considering the mircoRNA expression profiles detected by microarrays and the results from qRT-PCR, we designated miR-138 and miR-17–5 p as potential ceRNAs which lncRNA HOTAIR could directly bind to. In addition, the expressions of miR-138 and miR-17–5 p were markedly downregulated in RA FLSs, whereas the knockdown of lncRNA HOTAIR upregulated the expressions compared with the negative control group (NC-siRNA). Conclusions Our study illuminated that elevated lncRNA HOTAIR expression promoted the proliferation and invasion of RA FLSs. Meanwhile, it may function as a novel microRNAs sponging agent and regulate RA FLSs pathological behaviors via miR-138 or miR-17–5 p associated ceRNA network. In summary, the regulation of lncRNA HOTAIR may be a promising therapeutic strategy for RA in the future. Acknowledgements This work was supported by grants provided from Province Natural Science Fund of Guangdong, China (No.2014A030313080) and National Natural Science Foundation of China (No.81771750). Disclosure of Interest None declared