Open access peer-reviewed chapter

Role of LncRNA in Rheumatoid Arthritis

Written By

Ayse Kocak

Submitted: 19 June 2021 Published: 19 January 2022

DOI: 10.5772/intechopen.99525

From the Edited Volume

Rheumatoid Arthritis

Edited by Hechmi Toumi

Chapter metrics overview

202 Chapter Downloads

View Full Metrics

Abstract

Long non-coding RNAs (lncRNAs) are a class of non-coding RNA (ncRNA) molecules that do not have protein coding. They are ubiquitous in the process of transcription and gene regulation. lncRNAs regulation is correlated with many diseases. Rheumatoid arthritis (RA) is a chronic inflammatory disorder and this disease can affect especially joints. Nevertheless, in some patients, RA and inflammation can damage body parts such as the eyes, lungs, skin, heart, and blood vessels. Lots of lncRNAs were confirmed to be correlated with rheumatoid arthritis (RA) pathogenesis. Particularly GAPLINC, ZFAS1, PTGS2, and HOTAIR lncRNAs play a role in RA. This chapter will be explained and summarized the relationship between IncRNAs and RA.

Keywords

  • lncRNA
  • rheumatoid arthritis

1. Introduction

Rheumatoid arthritis (RA) is a chronic and systemic autoimmune disease. It is associated with progressive joint destruction complications and decreased life expectancy [1]. RA’s main clinical features are typically symmetrical polyarthritis with swelling, redness, and pain in the distal joint, particularly the small joints of the hands and feet [2]. Advances in understanding the pathogenesis of the disease, RA treatment greatly improved with an emphasis in the early stage. To our best knowledge, lots of laboratory tests used for RA generally include rheumatoid factor (RF), c-reactive protein (CRP), erythrocyte sedimentation rate (ESR), and anti-cyclic peptide containing citrulline (anti-CCP) antibodies [3]. Nevertheless, RA pathogenesis is still unclear, but it is most likely related to the physiological structure of the joint and anatomy [4].

Long non-coding RNAs’ (lncRNAs) lengths are greater than >200 nucleotides, which are subclassified into five categories that include the natural antisense lncRNAs according to their positions relative to protein-coding genes, long intergenic ncRNAs (lincRNAs), intronic lncRNAs, bidirectional lncRNAs, sense-overlapping lncRNAs [5, 6]. Referring to the GENCODE database (version 31), 17,904 lncRNA genes were identified in the human genome. lncRNAs play a role as critical regulators of cellular processes and disease stage or progression. lncRNAs have been studied in cancer [7, 8], innate and adaptive immunity [9], and inflammation [10]. Recently, studies of the role of lncRNA in RA pathogenesis are increased. Sequencing or microarray analyses revealed the expression profiles of lncRNAs in RA. The lncRNA expression profile in RA is different in various immune cell types such as B cells, natural killer (NK) cells, and T cells, indicating immune cell-type specificity of lncRNA expression. Identification of aberrantly expressed lncRNAs in RA and investigation of the underlying molecular mechanisms.

Emerging evidence suggests that lncRNAs are involved in the development of RA. Although numerous aberrant-expressing lncRNAs (HOTAIR, MALAT1, GAPLINC, PVT-1, LERFS, GAS5, DILC, NEAT-1, Lnc-p21, THRIL, RMRP, NTT, MEG3, Lnc-IL7R, ZFAS1, UCA1, C5T1LncRNA) have been reported in RA [11, 12], only a few of them are functionally determined. In this chapter, we summarize here the current findings of lncRNAs that may be involved in the pathogenesis of RA, aiming to encourage future research on this topic.

Advertisement

2. Long non-coding RNAs in RA

2.1 Long non-coding RNAs

LncRNAs play epigenetic regulation, cell cycle regulation, and cell differentiation and genetic roles [13] such as physiological and pathological and regulator process; also, lncRNAs are Central regulators of the immune response, but they are poorly conserved in species [14]. LncRNAs regulate the coding genes directly various molecular mechanisms [15]. LncRNAs expressed differentially and effects on immune cells in autoimmune diseases. The regulatory mechanism of lncRNAs is complex and needs to be investigated by more functional and mechanistic experiments. A group of lncRNAs is associated with clinical indicators such as CRP, ESR, serum proinflammatory cytokines, and DAS28, suggesting that lncRNAs may serve as biomarkers to monitor RA activity.

2.1.1 LncRNA HOTAIR

In RA patients, the expression of HOX transcript antisense RNA (HOTAIR), HOTAIR is decreased in fibroblast-like synoviocytes (FLSs), also HOTAIR suppresses the activation of MMP-2 and MMP-13. lncRNA HOTAIR, miR-138, and NF-kB axis have also been established in chondrocytes in RA, LncRNA HOTAIR may target miR-138 and inhibit the activation of NF-kB pathway [16], and the expression of HOTAIR increases in peripheral blood mononuclear cell and blood exosomes using lncRNA array analysis [17].

2.1.2 LncRNA MALAT1

In RA FLSs, MALAT1 plays a role regulation of cell proliferation and inflammation [18]. MALAT1 binds to the beta-catenin promoter in the WNT signaling pathway [19]. One group study suggests that MALAT1 plays a role in apoptosis proteins [19]. MALAT1 silencing suppressed Bax, and Bcl-2, caspase-3, caspase-9 in RA FLSs [19].

2.1.3 LncRNA GAPLINC

LncRNA long intergenic non-coding RNA (GAPLINC) may play act as a molecular sponge of miR-382-5p and miR-575. There is a negative correlation observed between the expression of GAPLINC and the miRNAs [20]. Also, GAPLINC may be a new therapeutic target for RA [21].

2.1.4 LncRNA PVT-1

Knockdown of plasmacytoma variant translocation 1 (PVT-1) in RA’s FLSs suppresses the TNF-α and IL-1β pro-inflammatory cytokines [22]. In the same study, PVT1 regulates inflammation and apoptosis in RA-FLSs through the Sirt6 demethylation. Furthermore, PVT-1 increase at synovial tissue of RA patients and RA model and PVT-1 bound to miR-543 positively regulated the expression of signal peptide-CUB-EGF-like containing protein 2 (SCUBE2) by inhibiting the miR-543, guide to FLSs inhibition of apoptosis and IL-1β secretion. Inhibition of PVT1 may be a new idea for the treatment of RA [23].

2.1.5 LncRNA LERFS

Lowly expressed in rheumatoid fibroblast-like synoviocytes (lncRNA LERFS) negatively regulated the invasion, proliferation, and migration of joint synovium by interacting with heterogeneous nuclear ribonucleoprotein Q (hnRNP Q) but in RA FLSs, LERFS is low expressed in RA FLSs and the reduced LERFS led to the reduction of LERFS-hnRNP Q complex [24]. In this study, LERFS regulates the expression and activity of CDC42, Rac1, RhoA and, probably by binding to the hnRNP Q complex.

2.1.6 LncRNA GAS5

In RA FLSs, LncRNA growth arrest-specific transcript 5 (GAS5) overexpression organizes cell apoptosis by activating cleaved caspase-9 and caspase-3 and inhibits PI3K/AKT signaling pathway [25, 26]. Also, GAS5 plays a role in the inflammatory response in RA. In synovial tissue and FLSs, GAS5 expression is decreased and expression of homeodomain-interacting protein kinase 2 (HIPK2) increases significantly and GAS5 reduced the level of TNF-α and IL-6 [27]. Also, overexpression of LncRNA GAS5 affects IL-18 levels, and IL-18 is downregulated by LncRNA GAS5 [28].

2.1.7 LncRNA DILC

DILC of plasma RA patients was downregulated, while IL-6 was upregulated and DILC level is negatively correlated with RA. DILC overexpression promoted the inhibition of IL-6 expression and FLSs apoptosis and in RA [29].

2.1.8 LncRNA NEAT-1

NEAT-1 is significantly upregulated in th17 cells differentiated CD44 cells from RA patients. Also, upregulation of NEAT-1 plays a role differentiation of CD4+ T cells into Th17 cells by regulating its downstream molecule STAT3 [30].

2.1.9 LncRNA Lnc-p21

In RA, Lnc-p21 expression is so low and can be renovated by the methotrexate treatment [31]. This LncRNA-p21 suppresses inflammation and is downregulated [31].

2.1.10 LncRNA THRIL

According to the information obtained from RA, THRIL is on the upward path [32]. This LncRNA may use as a biomarker for RA. THRIL expression in the blood of RA patients was positively correlated with TNF-α and erythrocyte sedimentation rate. THRIL inhibition is reversed the regulatory effect of TNF-α, and significantly reduced the activity of p-AKT and phosphoinositide 3-kinase (PI3K) signaling pathways. Also, expression of THRIL may promote the activating of the PI3K/AKT signaling pathway, and this leads to the result of inflammation and proliferation of FLSs [33, 34].

2.1.11 LncRNA RMRP

LncRNA RMRP expression is high in T cells from patients with RA [32]. Also, LncRNA RMRP has a positive correlation with RA progression [35]. This LncRNA may be a biomarker for RA.

2.1.12 LncRNA NTT

NTT expression is increased in a peripheral blood mononuclear cell (PBMC) from early patients with RA [36]. In the same study, the researchers found that in RA, lncRNA NTT/PBOV1 is capable of regulating monocyte differentiation.

2.1.13 LncRNA MEG3

The level of LncRNA maternally expressed gene 3 (MEG3) is significantly downregulated in FLSs of patients with RA [37]. Also, this study suggests that in lipopolysaccharide (LPS)-treated chondrocytes LncRNA MEG is downregulated. Overexpression of LncRNA MEG3 has an inhibitory effect on RA pathology can be achieved by increasing the rate of chondrocyte proliferation through negative regulation of miR-141 and AKT/mTOR signaling pathway [38, 39, 40]. In RA patients, low MEG3 expression correlated negatively with serum level of HIF-1α and vascular endothelial growth factor A (VEGF) and positively correlated with BAX. MEG3 gene rs941576(A/G) polymorphism has been confirmed to be associated with increased RA severity in the population [41]. We can say that LncRNA MEG3 promotes proliferation and it has an inhibitory effect.

2.1.14 LncRNA Lnc-IL7R

LncRNA long noncoding-interleukin-7 receptor (Lnc-IL7R) inhibits apoptosis and leads to proliferation [42]. Also, Lnc-IL7R interacts with the enhancer of zeste homolog 2 (EZH2) to assist the FLSs’ growth and it is necessary for PRC2-mediated inhibition of the cyclin-dependent kinase inhibitors 1A and 2A [42].

2.1.15 LncRNA ZFAS1

Ye et al. found that LncRNA ZFAS1 has abnormal activity in RA FLSs. Also, the knockout of LncRNA ZFAS1 suppresses the migration and invasion of FLSs and it takes miR-27 s as a target and increased the expression of miR-27a [43].

2.1.16 LncRNA UCA1

LncRNA UCA1 expression is low in RA FLSs [44]. It reduces caspase-3 and cell apoptosis via Wnt-6 [44].

2.1.17 C5T1LncRNA

LncRNA C5T1LncRNA is a new LncRNA and it inhibits the mRNA of C5 protein, this protein plays a role in inflammation in RA [45, 46].

Advertisement

3. LncRNA as novel RA biomarkers

As with other pathological and chronic diseases, RA affects patients’ life and status. Many pieces of evidence have confirmed the role of lncRNAs in the pathogenesis of RA [47]. Luo et al. found 5.045 irregular lncRNAs in PBMCs (2.635 downregulated and 2.410 upregulated) of RA patients compared to controls [48], 135 potential lncRNA-mRNA target pairs and RP11-498C9.15 targeted RA-related genes and pathways. Lots of LncRNAs such as PVT-1, MEG3, HOTAIR suggest that LncRNAs may serve as novel biomarkers to monitor RA pathogenesis.

Advertisement

4. Discussion

LncRNAs are of great importance in gene regulation and various RA biological processes. Expression profiles of lncRNAs vary in PBMCs, serum exosomes, osteoclasts, FLS, synovial tissues, plasma, synovium in RA. Some of these are differentially expressed, and LncRNAs are related to RA activity.

Advertisement

5. Conclusion

Emerging evidence shows us that lncRNAs are important regulators in RA. Continuing to explore the functions of lncRNAs in RA, their aberrant expression profile, and determining their role and mode of action will help us understand the underlying causes of the disease. Also, the identified lncRNAs related to the pathogenesis of RA may be potential diagnostic markers or target molecules that regulate RA progression. In the future, LncRNA-based therapeutic tools will likely lead to treatment insights into RA.

References

  1. 1. Firestein GS. Evolving concepts of rheumatoid arthritis. Nature. 2003;423:356-361. DOI: 10.1038/nature01661
  2. 2. Davis JM III, Matteson EL. My treatment approach to rheumatoid arthritis. Mayo Clinic Proceedings. 2012;87:659-673. DOI: 10.1016/j.mayocp.2012.03.011
  3. 3. Tavasolian F, Abdollahi E, Rezaei R, Momtazi-Borojeni AA, Henrotin Y, Sahebkar A. Altered expression of microRNAs in rheumatoid arthritis. Journal of Cellular Biochemistry. 2018;119:478-487. DOI: 10.1002/jcb.26205
  4. 4. Alpizar-Rodriguez D, Finckh A. Is the prevention of rheumatoid arthritis possible? Clinical Rheumatology. 2020;39(5):1383-1389. DOI: 10.1007/s10067-020-04927-6
  5. 5. Schonrock N, Harvey RP, Mattick JS. Long noncoding RNAs in cardiac development and pathophysiology. Circulation Research. 2012;111:1349-1362. DOI: 10.1161/CIRCRESAHA.112.268953
  6. 6. Shi X, Sun M, Liu H, Yao Y, Song Y. Long non-coding RNAs: A new frontier in the study of human diseases. Cancer Letters. 2013;339:159-166. DOI: 10.1016/j.canlet.2013.06
  7. 7. Silva A, Bullock M, Calin G. The clinical relevance of long non-coding RNAs in cancer. Cancers. 2015;7(4):2169-2182
  8. 8. Yang G, Lu X, Yuan L. LncRNA: A link between RNA and cancer. Biochimica et Biophysica Acta (BBA)—Gene Regulatory Mechanisms. 2014;1839(11):1097-1109
  9. 9. Fitzgerald KA, Caffrey DR. Long noncoding RNAs in innate and adaptive immunity. Current Opinion in Immunology. 2014;26C:140-146
  10. 10. Chew CL. Noncoding RNAs: Master regulators of inflammatory signaling. Trends in Molecular Medicine. 2018;24(1):66-84
  11. 11. Zhang Y, Xu YZ, Sun N, Liu JH, Chen FF, Guan XL, et al. Long noncoding RNA expression profile in fibroblast-like synoviocytes from patients with rheumatoid arthritis. Arthritis Research & Therapy. 2016;18:227. DOI: 10.1186/s13075-016-1129-4
  12. 12. Yuan M, Wang S, Yu L, Qu B, Xu L, Liu L, et al. Long noncoding RNA profiling revealed differentially expressed lncRNAs associated with disease activity in PBMCs from patients with rheumatoid arthritis. PLoS One. 2017;12:e0186795. DOI: 10.1371/journal. pone.0186795
  13. 13. Wei JW, Huang K, Yang C, Kang CS. Non-coding RNAs as regulators in epigenetics. Oncology Reports. 2017;37(1):3-9. DOI: 10.3892/or.2016.5236
  14. 14. Mathy NW, Chen XM. Long non-coding RNAs (lncRNAs) and their transcriptional control of inflammatory responses. The Journal of Biological Chemistry. 2017;292(30):12375-12382. DOI: 10.1074/jbc.R116.760884
  15. 15. Hombach S, Kretz M. Non-coding RNAs: Classification, biology and functioning. Advances in Experimental Medicine and Biology. 2016;937:3-17. DOI: 10.1007/978-3-319-42059-2_1
  16. 16. Zhang HJ, Wei QF, Wang SJ, Zhang HJ, Zhang XY, Geng Q , et al. LncRNA HOTAIR alleviates rheumatoid arthritis by targeting miR-138 and inactivating NF-κB pathway. International Immunopharmacology. 2017;50:283-290. DOI: 10.1007/s10238-013-0271-4
  17. 17. Song J, Kim D, Han J, Kim Y, Lee M, Jin EJ. PBMC and exosomederived Hotair is a critical regulator and potent marker for rheumatoid arthritis. Clinical and Experimental Medicine. 2015;15:121-126. DOI: 10.1007/s10238-013-0271-4
  18. 18. Li GQ , Fang YX, Liu Y, Meng FR, Wu X, Zhang CW, et al. MALAT1-driven inhibition of Wnt signal impedes proliferation and inflammation in fibroblast-like synoviocytes through CTNNB1 promoter methylation in rheumatoid arthritis. Human Gene Therapy. 2019;30:1008-1022. DOI: 10.1089/hum.2018.212
  19. 19. Pan F, Zhu L, Lv H, Pei C. Quercetin promotes the apoptosis of fibroblast-like synoviocytes in rheumatoid arthritis by upregulating lncRNA MALAT1. International Journal of Molecular Medicine. 2016;38:1507-1514. DOI: 10.3892/ijmm.2016.2755
  20. 20. Mo BY, Guo XH, Yang MR, Liu F, Bi X, Liu Y, et al. Long non-coding RNA GAPLINC promotes tumor-like biologic behaviors of fibroblast-like synoviocytes as microRNA sponging in rheumatoid arthritis patients. Frontiers in Immunology. 2018;10(9):702. DOI: 10.3389/fimmu.2018.00702
  21. 21. Liao S, Zhou S, Wang C. GAPLINC is a predictor of poor prognosis and regulates cell migration and invasion in osteosarcoma. Bioscience Reports. 2018;38(5):BSR20181171. DOI: 10.1042/BSR20181171
  22. 22. Zhang CW, Wu X, Liu D, Zhou W, Tan W, Fang YX, et al. Long non-coding RNA PVT1 knockdown suppresses fibroblast-like synoviocyte inflammation and induces apoptosis in rheumatoid arthritis through demethylation of sirt6. Journal of Biological Engineering. 2019;13:60. DOI: 10.1186/s13036-019-0184-1
  23. 23. Wang J, Kong X, Hu H, Shi S. Knockdown of long non-coding RNA PVT1 induces apoptosis of fibroblast-like synoviocytes through modulating miR-543-dependent SCUBE2 in rheumatoid arthritis. Journal of Orthopaedic Surgery and Research. 2020;15(1):142-151. DOI: 10.1186/s13018-020-01641-6
  24. 24. Zou Y, Xu S, Xiao Y, Qiu Q , Shi M, Wang J, et al. Long noncoding RNA LERFS negatively regulates rheumatoid synovial aggression and proliferation. The Journal of Clinical Investigation. 2018;128(10):4510-4524. DOI: 10.1172/JCI97965
  25. 25. Zhang HJ, Wei QF, Wang SJ, Zhang XY, Geng Q , Cui YH, et al. LncRNA HOTAIR alleviates rheumatoid arthritis by targeting miR-138 and inactivating NF-kB pathway. International Immunopharmacology. 2017;50:283-290. DOI: 10.1016/j.intimp.2017.06.021
  26. 26. Jiang H, Ma R, Zou S, Wang Y, Li Z, Li W. Reconstruction and analysis of the lncRNA-miRNA-mRNA network based on competitive endogenous RNA reveal functional lncRNAs in rheumatoid arthritis. Molecular BioSystems. 2017;13:1182-1192. DOI: 10.1039/C7MB00094D
  27. 27. Li M, Wang N, Shen Z, Yan J. Long non-coding RNA growth arrest-specific transcript 5 regulates rheumatoid arthritis by targeting homeodomain-interacting protein kinase 2. Clinical and Experimental Rheumatology. 2020;38(6):1145-1154
  28. 28. Ma C, Wang W, Li P. LncRNA GAS5 overexpression downregulates IL-18 and induces the apoptosis of fibroblast-like synoviocytes. Clinical Rheumatology. 2019;38(11):3275-3280. DOI: 10.1007/s10067-019-04691-2
  29. 29. Wang G, Tang L, Zhang X, Li Y. LncRNA DILC participates in rheumatoid arthritis by inducing apoptosis of fibroblast-like synoviocytes and down-regulating IL-6. Bioscience Reports. 2019;39:pii:BSR20182374. DOI: 10.1042/BSR20182374
  30. 30. Shui X, Chen S, Lin J, Kong J, Zhou C, Wu J. Knockdown of lncRNA NEAT1 inhibits Th17/CD4+ T cell differentiation through reducing the STAT3 protein level. Journal of Cellular Physiology. 2019;234:22477-22484. DOI: 10.1002/jcp.28811
  31. 31. Spurlock CF 3rd, Tossberg JT, Matlock BK, Olsen NJ, Aune TM. Methotrexate inhibits NF-kB activity via long intergenic (noncoding) RNA-p21 induction. Arthritis & Rhematology. 2014;66:2947-2957. DOI: 10.1002/art.38805
  32. 32. Moharamoghli M, Hassan-Zadeh V, Dolatshahi E, Alizadeh Z, Farazmand A. The expression of GAS5, THRIL, and RMRP lncRNAs is increased in T cells of patients with rheumatoid arthritis. Clinical Rheumatology. 2019;38:3073-3080. DOI: 10.1007/s10067-019-04694-z
  33. 33. Zhu LJ, Yang TC, Wu Q , Yuan LP, Chen ZW, Luo MH, et al. Tumor necrosis factor receptor-associated factor (TRAF) 6 inhibition mitigates the pro-inflammatory roles and proliferation of rheumatoid arthritis fibroblast-like synoviocytes. Cytokine. 2017;93:26-33. DOI: 10.1016/j.cyto.2017.05.001
  34. 34. Liang Y, Li H, Gong X, Ding C. Long non-coding RNA THRIL mediates cell growth and inflammatory response of fibroblast-like synoviocytes by activating PI3K/AKT signals in rheumatoid arthritis. Inflammation. 2020;43(3):1044-1053. DOI: 10.1007/s10753-020-01189-x
  35. 35. Wu GC, Hu Y, Guan SY, Ye DQ , Pan HF. Differential plasma expression profiles of long non-coding RNAs reveal potential biomarkers for systemic lupus erythematosus. Biomolecules. 2019;9(6):206-215. DOI: 10.3390/biom9060206
  36. 36. Yang CA, Li JP, Yen JC, Lai IL, Ho YC, Chen YC, et al. LncRNA NTT/PBOV1 axis promotes monocyte differentiation and is elevated in rheumatoid arthritis. International Journal of Molecular Sciences. 2018;19:pii: E2806. DOI: 10.3390/ijms19092806
  37. 37. Wang A, Hu N, Zhang Y, Chen Y, Su C, Lv Y, et al. MEG3 promotes proliferation and inhibits apoptosis in osteoarthritis chondrocytes by miR-361-5p/FOXO1 axis. BMC Medical Genomics. 2019;12(1):201-212. DOI: 10.1186/s12920-019-0649-6
  38. 38. Li G, Liu Y, Meng F, Xia Z, Wu X, Fang Y, et al. LncRNA MEG3 inhibits rheumatoid arthritis through miR-141 and inactivation of AKT/mTOR signalling pathway. Journal of Cellular and Molecular Medicine. 2019;23(10):7116-7120. DOI: 10.1111/jcmm.14591
  39. 39. Chen K, Zhu H, Zheng MQ , Dong QR. LncRNA MEG3 inhibits the degradation of the extracellular matrix of chondrocytes in osteoarthritis via targeting miR-93/TGFBR2 axis. Cartilage. 2019;28:194760351985575. DOI: 10.1177/1947603519855759
  40. 40. Lu X, Qian J. Downregulated MEG3 participates in rheumatoid arthritis via promoting proliferation of fibroblast-like synoviocytes. Experimental and Therapeutic Medicine. 2019;17(3):1637-1642. DOI: 10.3892/etm.2018.7100
  41. 41. Wahba AS, Ibrahim ME, Mesbah NM, Saleh SM, Abo-Elmatty DM, Mehanna ET. Long non-coding RNA MEG3 and its genetic variant rs941576 are associated with rheumatoid arthritis pathogenesis in Egyptian patients. Archives of Physiology and Biochemistry. 2020;1:1-8. DOI: 10.1080/13813455.2020.1784951
  42. 42. Ye Z, Xu J, Li S, Cai C, Li T, Sun L, et al. Lnc-IL7R promotes the growth of fibroblast-like synoviocytes through interaction with enhancer of zeste homolog 2 in rheumatoid arthritis. Molecular Medicine Reports. 2017;15:1412-1418. DOI: 10.3892/mmr.2017.6150
  43. 43. Ye Y, Gao X, Yang N. LncRNA ZFAS1 promotes cell migration and invasion of fibroblast-like synoviocytes by suppression of miR-27a in rheumatoid arthritis. Human Cell. 2018;31(1):14-21. DOI: 10.1007/s13577-017-0179-5
  44. 44. Yan ZF, Zhao XY, Liu W, Liu XP. UCA1 impacts progress of rheumatoid arthritis by inducing the apoptosis of fibroblast-like synoviocyte. European Review for Medical and Pharmacological Sciences. 2018;22:914-920. DOI: 10.26355/eurrev_201802_14370
  45. 45. Cooke TD, Hurd ER, Jasin HE, Bienenstock J, Ziff M. Identification of immunoglobulins and complement in rheumatoid articular collagenous tissues. Arthritis and Rheumatism. 1975;18:541-551. DOI: 10.1002/art.1780180603
  46. 46. Wang Y, Kristan J, Hao L, Lenkoski CS, Shen Y, Matis LA. A role for complement in antibody-mediated inflammation: C5-deficient DBA/1 mice are resistant to collagen-induced arthritis. Journal of Immunology. 2000;164:4340-4347. DOI: 10.4049/jimmunol.164.8.4340
  47. 47. Dolcino M, Tinazzi E, Puccetti A, Lunardi C. Long non-coding RNAs target pathogenetically relevant genes and pathways in rheumatoid arthritis. Cells. 2019;8(8):816
  48. 48. Luo Q , Xu C, Li X, Zeng L, Ye J, Guo Y, et al. Comprehensive analysis of long non-coding RNA and mRNA expression profiles in rheumatoid arthritis. Experimental and Therapeutic Medicine. 2017;14(6):5965-5973

Written By

Ayse Kocak

Submitted: 19 June 2021 Published: 19 January 2022