Generic placeholder image

Current Molecular Medicine

Editor-in-Chief

ISSN (Print): 1566-5240
ISSN (Online): 1875-5666

Systematic Review Article

Structural and Pharmacological Network Focused on MiRNAs Involved in Rheumatoid Arthritis: A Systematic Review

Author(s): Oscar Salvador Barrera-Vázquez and Olivia Hernández-González*

Volume 24, Issue 5, 2024

Published on: 09 June, 2023

Page: [599 - 609] Pages: 11

DOI: 10.2174/1566524023666230423144114

Price: $65

conference banner
Abstract

Background: Rheumatoid Arthritis (RA) is a chronic autoimmune disease that has a prevalence of over one percent of the world population, causing substantial pain, joint deformity, and functional disability in patients. The identification and measurement of miRNAs are relatively easy to perform. Future studies will corroborate if miRNAs can fulfill their roles as biomarkers with either predictive or diagnostic evaluation of treatment potential and provide actual clinical utility.

Methods: In the last decade, various advances have been made regarding the identification of the origin and exact functions of miRNAs, allowing us to have a potential use both in the research and clinical fields.

Objective: This systematic review aimed to collect, analyze, and improve the current understanding of RA-related miRNAs and their applicability in therapeutics. A bibliographic search of the miRNAs involved in RA was carried out, and through the use of databases, their target genes and small molecules that had some relationship with their expression were searched. The analysis of these data was done through structural network analysis.

Results: During the network analysis, miR-30a, miR-30c, let-7a, miR-144, miR-17-5p, miR-124, miR -23b, miR-23, miR-15a, miR-16 were the most connected, which could be used as possible biomarkers or be candidates for further analysis due to their interaction with other miRNAs and genes.

Conclusion: Additionally, this is the first systematic review, in which we proposed that small compounds like toxicants and drugs could have a potential role within RA because they regulate the expression of miRNAs involved in this pathology. Some of these compounds are commonly found as environmental contaminants, and others as drugs. These ideas open a new panorama of understanding RA, proposing possible causes or treatments against this pathology. Therefore, these small molecules would give us some indication of a relationship with RA, thereby helping in seeking causes, treatment, or prevention of this disease. Conclusion: This is the first time it is intended to use structural network analysis to determine possible biomarkers of AR for diagnosis and prognosis through the expression of these miRNAs and their relationship with compounds of daily life.

[1]
Guo Q, Wang Y, Xu D, Nossent J, Pavlos NJ, Xu J. Rheumatoid arthritis: Pathological mechanisms and modern pharmacologic therapies. Bone Res 2018; 6(1): 15.
[http://dx.doi.org/10.1038/s41413-018-0016-9] [PMID: 29736302]
[2]
Shams S, Martinez JM, Dawson JRD, et al. The therapeutic landscape of rheumatoid arthritis: Current state and future directions. Front Pharmacol 2021; 12680043.
[http://dx.doi.org/10.3389/fphar.2021.680043] [PMID: 34122106]
[3]
Zhang R, Zhou X, Jin Y, et al. Identification of differential key biomarkers in the synovial tissue between rheumatoid arthritis and osteoarthritis using bioinformatics analysis. Clin Rheumatol 2021; 40(12): 5103-10.
[http://dx.doi.org/10.1007/s10067-021-05825-1] [PMID: 34224029]
[4]
Wei ST, Sun YH, Zong SH. Serum levels of IL-6 and TNF-α may correlate with activity and severity of rheumatoid arthritis. Med Sci Monit 2015; 21: 4030.
[5]
Koutrouli M, Karatzas E, Paez-Espino D, Pavlopoulos GA. A guide to conquer the biological network era using graph theory. Front Bioeng Biotechnol 2020; 8: 34.
[http://dx.doi.org/10.3389/fbioe.2020.00034] [PMID: 32083072]
[6]
Sonawane AR, Weiss ST, Glass K, Sharma A. Network medicine in the age of biomedical big data. Front Genet 2019; 10: 294.
[http://dx.doi.org/10.3389/fgene.2019.00294] [PMID: 31031797]
[7]
Vocaturo E, Veltri P. On the use of networks in biomedicine. Procedia Comput Sci 2017; 110: 498-503.
[http://dx.doi.org/10.1016/j.procs.2017.06.132]
[8]
Sobhy N, Ghoniem SA, Eissa BM, Kamal A, Medhat A, Elsaid NYJTER. Disease characteristics in high versus low titers of rheumatoid factor or anti-citrullinated peptide antibody in rheumatoid arthritis patients. Egypt Rheumatol 2022; 44(4): 325-8.
[http://dx.doi.org/10.1016/j.ejr.2022.04.004]
[9]
Ormseth MJ, Solus JF, Sheng Q, et al. Development and validation of a MicroRNA panel to differentiate between patients with rheumatoid arthritis or systemic lupus erythematosus and controls. J Rheumatol 2020; 47(2): 188-96.
[http://dx.doi.org/10.3899/jrheum.181029] [PMID: 31092710]
[10]
Paradowska-Gorycka A, Stypinska B. MicroRNAs in rheumatoid arthritis: From pathogenesis to clinical utility. In: New Developments in the Pathogenesis of Rheumatoid Arthritis. InTech 2017.
[11]
Moher D, Liberati A, Tetzlaff J, Altman DG. Preferred reporting items for systematic reviews and meta-analyses: The PRISMA statement. PLoS Med 2009; 6(7): e1000097.
[http://dx.doi.org/10.1371/journal.pmed.1000097] [PMID: 19621072]
[12]
Shannon P, Markiel A, Ozier O, et al. Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res 2003; 13(11): 2498-504.
[http://dx.doi.org/10.1101/gr.1239303] [PMID: 14597658]
[13]
Chin CH, Chen SH, Wu HH, Ho CW, Ko MT, Lin CY. cytoHubba: Identifying hub objects and sub-networks from complex interactome. BMC Syst Biol 2014; 8(S4): S11.
[http://dx.doi.org/10.1186/1752-0509-8-S4-S11] [PMID: 25521941]
[14]
Winterhalter C, Widera P, Krasnogor N. JEPETTO: A Cytoscape plugin for gene set enrichment and topological analysis based on interaction networks. Bioinformatics 2014; 30(7): 1029-30.
[http://dx.doi.org/10.1093/bioinformatics/btt732] [PMID: 24363376]
[15]
Sui S, Wang X, Zheng H, Guo H, Chen T, Ji DM. Gene set enrichment and topological analyses based on interaction networks in pediatric acute lymphoblastic leukemia. Oncol Lett 2015; 10(6): 3354-62.
[http://dx.doi.org/10.3892/ol.2015.3761] [PMID: 26788135]
[16]
Glaab E, Baudot A, Krasnogor N, Schneider R, Valencia A. EnrichNet: Network-based gene set enrichment analysis. Bioinformatics 2012; 28(18): i451-7.
[http://dx.doi.org/10.1093/bioinformatics/bts389] [PMID: 22962466]
[17]
Kanehisa M, Furumichi M, Tanabe M, Sato Y, Morishima K. KEGG: New perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res 2017; 45(D1): D353-61.
[http://dx.doi.org/10.1093/nar/gkw1092] [PMID: 27899662]
[18]
Lenert A, Fardo DW. Detecting novel micro RNAs in rheumatoid arthritis with gene-based association testing. Clin Exp Rheumatol 2017; 35(4): 586-92.
[19]
Lefèvre S, Knedla A, Tennie C, et al. Synovial fibroblasts spread rheumatoid arthritis to unaffected joints. Nat Med 2009; 15(12): 1414-20.
[http://dx.doi.org/10.1038/nm.2050] [PMID: 19898488]
[20]
Wang L, Wang C, Jia X, Yu J. Circulating exosomal miR-17 inhibits the induction of regulatory T cells via suppressing TGFBR II expression in rheumatoid arthritis. Cell Physiol Biochem 2018; 50(5): 1754-63.
[21]
Evangelatos G, Fragoulis GE, Koulouri V, Lambrou GI. MicroRNAs in rheumatoid arthritis: From pathogenesis to clinical impact. Autoimmun Rev 2019; 18(11): 102391.
[http://dx.doi.org/10.1016/j.autrev.2019.102391] [PMID: 31520804]
[22]
Murata K, Furu M, Yoshitomi H, et al. Comprehensive microRNA analysis identifies miR-24 and miR-125a-5p as plasma biomarkers for rheumatoid arthritis. PLoS One 2013; 8(7): e69118.
[http://dx.doi.org/10.1371/journal.pone.0069118] [PMID: 23874885]
[23]
Mao L, Liu S, Hu L, Jia L, Wang H, Guo M. miR-30 family: A promising regulator in development and disease. Biomed Res Int 2018; 2018: 9623412.
[24]
Roush S. The let-7 family of microRNAs. Trends Cell Biol 2008; 18(10): 505-16.
[25]
Alexandri C, Stamatopoulos B, Rothé F, Bareche Y, Devos M. MicroRNA profiling and identification of let-7a as a target to prevent chemotherapy-induced primordial follicles apoptosis in mouse ovaries. Sci Rep 2019; 9(1): 1-10.
[26]
Stoen MJ, Andersen S, Rakaee M, Pedersen MI, Ingebriktsen LM, Bremnes RM. High expression of miR-17-5p in tumor epithelium is a predictor for poor prognosis for prostate cancer patients. Sci Rep 2021; 11(1): 13864.
[http://dx.doi.org/10.1038/s41598-021-93208-6]
[27]
Wang Z, Zhang J, Zhang Z, et al. Prognostic value of miR-17-5 p in gastrointestinal cancers: A systematic review and meta-analysis. OncoTargets Ther 2018; 11: 5991-9.
[http://dx.doi.org/10.2147/OTT.S157670] [PMID: 30275704]
[28]
Kooshkaki O, Rezaei Z, Rahmati M, Vahedi P, Derakhshani A, Brunetti O. MiR-144: A new possible therapeutic target and diagnostic/prognostic tool in cancers. Int J Mol Sci 2020; 21(7): 2578.
[29]
Zhu S, Pan W, Song X, Liu Y, Shao X, Tang Y. The microRNA miR-23b suppresses IL-17-associated autoimmune inflammation by targeting TAB2, TAB3 and IKK-α. Nat Med 2012; 18(7): 1077-86.
[30]
Liu X, Ni S, Li C, Xu N, Chen W, Wu M. Circulating microRNA-23b as a new biomarker for rheumatoid arthritis. Gene 2019; 712: 143911.
[http://dx.doi.org/10.1016/j.gene.2019.06.001]
[31]
Guo Y-X, Wang N, Wu W-C, Li C-Q, Chen R-H, Zhang Y. The role of miR-23b in cancer and autoimmune disease. J Oncol 2021; 2021: 6473038.
[http://dx.doi.org/10.1155/2021/6473038]
[32]
Wang W, Wang Y, Liu W, van Wijnen AJJG. Regulation and biological roles of the multifaceted miRNA-23b (MIR23B). Gene 2018; 642.
[33]
Han D, Dong X, Zheng D. MiR-124 and the underlying therapeutic promise of neurodegenerative disorders. Front Pharmacol 2020; 10: 1555.
[34]
Kmiołek T, Paradowska-Gorycka AJC. miRNAs as biomarkers and possible therapeutic strategies in rheumatoid arthritis. Cells 2022; 11(3): 452.
[35]
Filková M, Aradi B, Šenolt L, Ospelt C, Vettori S, Mann H. Association of circulating miR-223 and miR-16 with disease activity in patients with early rheumatoid arthritis. Ann Rheum Dis 2014; 73(10): 1898-904.
[http://dx.doi.org/10.1136/annrheumdis-2012-202815]
[36]
Moran-Moguel MC, Petarra-del Rio S, Mayorquin-Galvan EE, Zavala-Cerna MG. Rheumatoid arthritis and miRNAs: A critical review through a functional view. J Immunol Res 2018; 2018: 1-16.
[http://dx.doi.org/10.1155/2018/2474529] [PMID: 29785401]
[37]
Pauley KM, Satoh M, Chan AL, Bubb MR, Reeves WH, Chan EKL. Upregulated miR-146a expression in peripheral blood mononuclear cells from rheumatoid arthritis patients. Arthritis Res Ther 2008; 10(4): R101.
[http://dx.doi.org/10.1186/ar2493] [PMID: 18759964]
[38]
Wakabayashi I, Sourij H, Sotoda Y, Daimon T, Groschner K. Ethnic differences in serum levels of micrornas potentially regulating alcohol dehydrogenase 1B and aldehyde dehydrogenase 2. J Clin Med 2021; 10(16): 3678.
[39]
Lazzerini PE, Capecchi PL, El Sherif N, Laghi Pasini F. Emerging arrhythmic risk of autoimmune and inflammatory cardiac channelopathies. J Am Heart Assoc 2018; 7(22): e010595.
[40]
Craparo A, Freund R. 14-3-3 (ϵ) interacts with the insulin-like growth factor I receptor and insulin receptor substrate I in a phosphoserine-dependent manner. J Biol Chem 1997; 272(17): 11663-9.
[41]
Jin T. The Wnt signaling pathway effector TCF7L2 and type 2 diabetes mellitus. Mol Endocrinol 2008; 22(11): 2383-92.
[42]
Vallée A, Vallée J-N, Guillevin R, Lecarpentier YJC. Interactions between the canonical WNT/beta-catenin pathway and PPAR gamma on neuroinflammation, demyelination, and remyelination in multiple sclerosis. Cell Mol Neurobiol 2018; 38(4): 783-95.
[43]
Diaz MB. Krones Herzig A, Metzger D, Ziegler A, Vegiopoulos A, Klingenspor M. Nuclear receptor cofactor receptor interacting protein 140 controls hepatic triglyceride metabolism during wasting in mice. Hepatology 2008; 48(3): 782-91.
[http://dx.doi.org/10.1002/hep.22383]
[44]
Arias de la Rosa I, Escudero-Contreras A, Ruiz-Ponce M, et al. Molecular changes in the adipose tissue induced by rheumatoid arthritis: Effects of disease-modifying anti-rheumatic drugs. Front Immunol 2021; 12744022.
[http://dx.doi.org/10.3389/fimmu.2021.744022] [PMID: 34721412]
[45]
Harris RBS. Direct and indirect effects of leptin on adipocyte metabolism. Biochim Biophys Acta Mol Basis Dis 2014; 1842(3): 414-23.
[http://dx.doi.org/10.1016/j.bbadis.2013.05.009] [PMID: 23685313]
[46]
Gerhards R, Pfeffer LK, Lorenz J, et al. Oligodendrocyte myelin glycoprotein as a novel target for pathogenic autoimmunity in the CNS. Acta Neuropathol Commun 2020; 8(1): 207.
[http://dx.doi.org/10.1186/s40478-020-01086-2] [PMID: 33256847]
[47]
Sağ S, Sağ MS, Tekeoğlu I, Kamanlı A, Nas K, Acar BA. Central nervous system involvement in rheumatoid arthritis: Possible role of chronic inflammation and tnf blocker therapy. Acta Neurol Belg 2020; 120(1): 25-31.
[http://dx.doi.org/10.1007/s13760-017-0879-3] [PMID: 29288410]
[48]
Gasparyan AY, Stavropoulos-Kalinoglou A, Mikhailidis DP, Douglas KMJ, Kitas GD. Platelet function in rheumatoid arthritis: Arthritic and cardiovascular implications. Rheumatol Int 2011; 31(2): 153-64.
[http://dx.doi.org/10.1007/s00296-010-1446-x] [PMID: 20390282]
[49]
Vasilopoulos Y, Gkretsi V, Armaka M, Aidinis V, Kollias G. Actin cytoskeleton dynamics linked to synovial fibroblast activation as a novel pathogenic principle in TNF-driven arthritis. Annals of the rheumatic diseases. Ann Rheum Dis 2007; 66(S3): iii23-8.
[50]
Mewar D, Wilson AG. Treatment of rheumatoid arthritis with tumour necrosis factor inhibitors. Br J Pharmacol 2011; 162(4): 785-91.
[http://dx.doi.org/10.1111/j.1476-5381.2010.01099.x] [PMID: 21039421]
[51]
McInnes IB, Buckley CD, Isaacs JD. Cytokines in rheumatoid arthritis — shaping the immunological landscape. Nat Rev Rheumatol 2016; 12(1): 63-8.
[http://dx.doi.org/10.1038/nrrheum.2015.171] [PMID: 26656659]
[52]
Gerosa M, De Angelis V, Riboldi P, Meroni PL. Rheumatoid arthritis: A female challenge. Womens Health 2008; 4(2): 195-201.
[http://dx.doi.org/10.2217/17455057.4.2.195] [PMID: 19072521]
[53]
Ge C, Tong D, Lönnblom E, et al. Antibodies to cartilage oligomeric matrix protein are pathogenic in mice and may be clinically relevant in rheumatoid arthritis. Arthritis Rheumatol 2022; 74(6): 961-71.
[http://dx.doi.org/10.1002/art.42072] [PMID: 35080151]
[54]
Castro-Villegas C, Pérez-Sánchez C, Escudero A, Filipescu I, Verdu M, Ruiz-Limón P. Circulating miRNAs as potential biomarkers of therapy effectiveness in rheumatoid arthritis patients treated with anti-TNFα. Arthritis Res Ther 2015; 17(1): 1-15.
[http://dx.doi.org/10.1186/s13075-015-0555-z]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy