Journal List > J Rheum Dis > v.20(3) > 1064033

Lee, Kim, Lee, and Kim: Epigenetic Modification in Systemic Rheumatic Diseases

Abstract

Epigenetics is defined as an inheritable effect that influen-ces gene activity, but does not involve a change in DNA sequence. Epigenetic gene regulation has an essential role in determining individual gene function and activity in each specific cell type. Epigenetics includes four predom-inant mechanisms: DNA methylation, histone modification, nucleosome positioning and microRNA (miRNA). These mechanisms influence gene expression, cell differentiation, proliferation, DNA repair and replication. Epigenetic modifications are far more sensitive to environmental stimuli than DNA sequence alterations. Candidate gene approaches have identified a small set of genes that undergo epigenetic changes, such as aberrant DNA demethylation, histone modification, as well as regulation by miRNA in rheumatic diseases. It is well known that T cells from patients with SLE or RA, as well as synovial fibroblasts from individuals with RA, have sequences undergoing DNA hypomethylation and/or histone modifications. In addition, miRNA regulates the gene expression by pairing with its target mRNAs and is often deregulated in systemic rheumatic diseases. High-throughput approaches are necessary for screening the epigenetic alterations, and it is essential to screen the specific tissue and cell types that are relevant to the disease pathogenesis. Identification of cell-specific targets of the epigenetic deregulation in rheumatic disorders will provide clinical markers for the diagnosis, disease progression and response to therapy. Our understanding of epigenetics is in its infancy. New generation of pharmaceuticals, which manipulate the epigenome to the switch targeted genes on or off are under investigation. The new field of repairing or optimizing the epigenome through epigenetic modifier and/or diet is wide open.

References

1. Quintero-Ronderos P, Montoya-Ortiz G. Epigenetics and autoimmune diseases. Autoimmune Dis. 2012; 2012; 593720.
crossref
2. Deapen D, Escalante A, Weinrib L, Horwitz D, Bachman B, Roy-Burman P, et al. A revised estimate of twin concordance in systemic lupus erythematosus. Arthritis Rheum. 1992; 35:311–8.
3. Järvinen P, Kaprio J, Mäkitalo R, Koskenvuo M, Aho K. Systemic lupus erythematosus and related systemic diseases in a nationwide twin cohort: an increased prevalence of disease in MZ twins and concordance of disease features. J Intern Med. 1992; 231:67–72.
4. Aho K, Koskenvuo M, Tuominen J, Kaprio J. Occurrence of rheumatoid arthritis in a nationwide series of twins. J Rheumatol. 1986; 13:899–902.
5. Bellamy N, Duffy D, Martin N, Mathews J. Rheumatoid arthritis in twins: a study of aetiopathogenesis based on the Australian Twin Registry. Ann Rheum Dis. 1992; 51:588–93.
crossref
6. Silman AJ, MacGregor AJ, Thomson W, Holligan S, Carthy D, Farhan A, et al. Twin concordance rates for rheumatoid arthritis: results from a nationwide study. Br J Rheumatol. 1993; 32:903–7.
crossref
7. Fan S, Zhang X. CpG island methylation pattern in different human tissues and its correlation with gene expression. Biochem Biophys Res Commun. 2009; 383:421–5.
crossref
8. Ooi SK, Bestor TH. The colorful history of active DNA demethylation. Cell. 2008; 133:1145–8.
crossref
9. Kouzarides T. Chromatin modifications and their function. Cell. 2007; 128:693–705.
crossref
10. Gregory PD, Wagner K, Hörz W. Histone acetylation and chromatin remodeling. Exp Cell Res. 2001; 265:195–202.
crossref
11. Schones DE, Cui K, Cuddapah S, Roh TY, Barski A, Wang Z, et al. Dynamic regulation of nucleosome positioning in the human genome. Cell. 2008; 132:887–98.
crossref
12. Chodavarapu RK, Feng S, Bernatavichute YV, Chen PY, Stroud H, Yu Y, et al. Relationship between nucleosome positioning and DNA methylation. Nature. 2010; 466:388–92.
crossref
13. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004; 116:281–97.
14. Lu Q, Kaplan M, Ray D, Ray D, Zacharek S, Gutsch D, et al. Demethylation of ITGAL (CD11a) regulatory sequences in systemic lupus erythematosus. Arthritis Rheum. 2002; 46:1282–91.
15. Lu Q, Wu A, Tesmer L, Ray D, Yousif N, Richardson B. Demethylation of CD40LG on the inactive X in T cells from women with lupus. J Immunol. 2007; 179:6352–8.
crossref
16. Kaplan MJ, Lu Q, Wu A, Attwood J, Richardson B. Demethylation of promoter regulatory elements contributes to perforin overexpression in CD4+ lupus T cells. J Immunol. 2004; 172:3652–61.
crossref
17. Oelke K, Lu Q, Richardson D, Wu A, Deng C, Hanash S, et al. Overexpression of CD70 and overstimulation of IgG synthesis by lupus T cells and T cells treated with DNA methylation inhibitors. Arthritis Rheum. 2004; 50:1850–60.
crossref
18. Javierre BM, Fernandez AF, Richter J, Al-Shahrour F, Martin-Subero JI, Rodriguez-Ubreva J, et al. Changes in the pattern of DNA methylation associate with twin dis-cordance in systemic lupus erythematosus. Genome Res. 2010; 20:170–9.
crossref
19. Garaud S, Le Dantec C, Jousse-Joulin S, Hanrotel-Saliou C, Saraux A, Mageed RA, et al. IL-6 modulates CD5 expression in B cells from patients with lupus by regulating DNA methylation. J Immunol. 2009; 182:5623–32.
crossref
20. Neidhart M, Rethage J, Kuchen S, Künzler P, Crowl RM, Billingham ME, et al. Retrotransposable L1 elements expressed in rheumatoid arthritis synovial tissue: association with genomic DNA hypomethylation and influence on gene expression. Arthritis Rheum. 2000; 43:2634–47.
crossref
21. Nile CJ, Read RC, Akil M, Duff GW, Wilson AG. Methylation status of a single CpG site in the IL6 promoter is related to IL6 messenger RNA levels and rheumatoid arthritis. Arthritis Rheum. 2008; 58:2686–93.
22. Takami N, Osawa K, Miura Y, Komai K, Taniguchi M, Shiraishi M, et al. Hypermethylated promoter region of DR3, the death receptor 3 gene, in rheumatoid arthritis synovial cells. Arthritis Rheum. 2006; 54:779–87.
23. Kubo M, Czuwara-Ladykowska J, Moussa O, Markiewicz M, Smith E, Silver RM, et al. Persistent down-regulation of Fli1, a suppressor of collagen transcription, in fibrotic scleroderma skin. Am J Pathol. 2003; 163:571–81.
crossref
24. Dieker JW, Fransen JH, van Bavel CC, Briand JP, Jacobs CW, Muller S, et al. Apoptosis-induced acetylation of histones is pathogenic in systemic lupus erythematosus. Arthritis Rheum. 2007; 56:1921–33.
crossref
25. van Bavel CC, Dieker JW, Tamboer WP, van der Vlag J, Berden JH. Lupus-derived monoclonal autoantibodies against apoptotic chromatin recognize acetylated confor- mational epitopes. Mol Immunol. 2010; 48:248–56.
26. Hu N, Qiu X, Luo Y, Yuan J, Li Y, Lei W, et al. Abnormal histone modification patterns in lupus CD4+ T cells. J Rheumatol. 2008; 35:804–10.
27. Young DA, Lakey RL, Pennington CJ, Jones D, Kevor-kian L, Edwards DR, et al. Histone deacetylase inhibitors modulate metalloproteinase gene expression in chondrocytes and block cartilage resorption. Arthritis Res Ther. 2005; 7:R503–12.
28. Nishida K, Komiyama T, Miyazawa S, Shen ZN, Furumatsu T, Doi H, et al. Histone deacetylase inhibitor sup-pression of autoantibody-mediated arthritis in mice via regulation of p16INK4a and p21(WAF1/Cip1) expression. Arthritis Rheum. 2004; 50:3365–76.
crossref
29. Tang Y, Luo X, Cui H, Ni X, Yuan M, Guo Y, et al. MicroRNA-146A contributes to abnormal activation of the type I interferon pathway in human lupus by targeting the key signaling proteins. Arthritis Rheum. 2009; 60:1065–75.
crossref
30. Zhao X, Tang Y, Qu B, Cui H, Wang S, Wang L, et al. MicroRNA-125a contributes to elevated inflammatory chemokine RANTES levels via targeting KLF13 in systemic lupus erythematosus. Arthritis Rheum. 2010; 62:3425–35.
crossref
31. Pan W, Zhu S, Yuan M, Cui H, Wang L, Luo X, et al. MicroRNA-21 and microRNA-148a contribute to DNA hypomethylation in lupus CD4+ T cells by directly and indirectly targeting DNA methyltransferase 1. J Immunol. 2010; 184:6773–81.
32. Nakamachi Y, Kawano S, Takenokuchi M, Nishimura K, Sakai Y, Chin T, et al. MicroRNA-124a is a key regulator of proliferation and monocyte chemoattractant protein 1 secretion in fibroblast-like synoviocytes from patients with rheumatoid arthritis. Arthritis Rheum. 2009; 60:1294–304.
crossref
33. Kawano S, Nakamachi Y. miR-124a as a key regulator of proliferation and MCP-1 secretion in synoviocytes from patients with rheumatoid arthritis. Ann Rheum Dis. 2011; 70(Suppl 1):i88–91.
crossref
34. Stanczyk J, Ospelt C, Karouzakis E, Filer A, Raza K, Kolling C, et al. Altered expression of microRNA-203 in rheumatoid arthritis synovial fibroblasts and its role in fi-broblast activation. Arthritis Rheum. 2011; 63:373–81.
crossref
35. Stanczyk J, Pedrioli DM, Brentano F, Sanchez-Pernaute O, Kolling C, Gay RE, et al. Altered expression of Micro-RNA in synovial fibroblasts and synovial tissue in rheumatoid arthritis. Arthritis Rheum. 2008; 58:1001–9.
crossref
36. Alevizos I, Illei GG. MicroRNAs in Sjögren's syndrome as a prototypic autoimmune disease. Autoimmun Rev. 2010; 9:618–21.
crossref
37. Barreto G, Schäfer A, Marhold J, Stach D, Swaminathan SK, Handa V, et al. Gadd45a promotes epigenetic gene activation by repair-mediated DNA demethylation. Nature. 2007; 445:671–5.
crossref
38. Fritz EL, Papavasiliou FN. Cytidine deaminases: AIDing DNA demethylation? Genes Dev. 2010; 24:2107–14.
crossref
39. Lee BH, Yegnasubramanian S, Lin X, Nelson WG. Procainamide is a specific inhibitor of DNA methyltransferase 1. J Biol Chem. 2005; 280:40749–56.
crossref
40. Deng C, Lu Q, Zhang Z, Rao T, Attwood J, Yung R, et al. Hydralazine may induce autoimmunity by inhibiting extracellular signal-regulated kinase pathway signaling. Arthritis Rheum. 2003; 48:746–56.
crossref
41. Yung R, Chang S, Hemati N, Johnson K, Richardson B. Mechanisms of drug-induced lupus. IV. Comparison of procainamide and hydralazine with analogs in vitro and in vivo. Arthritis Rheum. 1997; 40:1436–43.
crossref
42. Fu LH, Ma CL, Cong B, Li SJ, Chen HY, Zhang JG. Hypomethylation of proximal CpG motif of interleukin-10 promoter regulates its expression in human rheumatoid arthritis. Acta Pharmacol Sin. 2011; 32:1373–80.
crossref
43. Bruns A, Bläss S, Hausdorf G, Burmester GR, Hiepe F. Nucleosomes are major T and B cell autoantigens in systemic lupus erythematosus. Arthritis Rheum. 2000; 43:2307–15.
crossref
44. Dai Y, Zhang L, Hu C, Zhang Y. Genome-wide analysis of histone H3 lysine 4 trimethylation by ChIP-chip in peripheral blood mononuclear cells of systemic lupus erythematosus patients. Clin Exp Rheumatol. 2010; 28:158–68.
45. Zhang Z, Song L, Maurer K, Petri MA, Sullivan KE. Global H4 acetylation analysis by ChIP-chip in systemic lupus erythematosus monocytes. Genes Immun. 2010; 11:124–33.
crossref
46. Manabe H, Nasu Y, Komiyama T, Furumatsu T, Kitamura A, Miyazawa S, et al. Inhibition of histone deacetylase down-regulates the expression of hypoxia-induced vascular endothelial growth factor by rheumatoid synovial fibroblasts. Inflamm Res. 2008; 57:4–10.
crossref
47. Nasu Y, Nishida K, Miyazawa S, Komiyama T, Kadota Y, Abe N, et al. Trichostatin A, a histone deacetylase inhibitor, suppresses synovial inflammation and subsequent cartilage destruction in a collagen antibody-induced arthritis mouse model. Osteoarthritis Cartilage. 2008; 16:723–32.
crossref
48. Grabiec AM, Korchynskyi O, Tak PP, Reedquist KA. Histone deacetylase inhibitors suppress rheumatoid arthritis fibroblast-like synoviocyte and macrophage IL-6 production by accelerating mRNA decay. Ann Rheum Dis. 2012; 71:424–31.
crossref
49. Angelov D, Molla A, Perche PY, Hans F, Côté J, Khochbin S, et al. The histone variant macroH2A inter-feres with transcription factor binding and SWI/SNF nucleosome remodeling. Mol Cell. 2003; 11:1033–41.
crossref
50. Li J, Wan Y, Guo Q, Zou L, Zhang J, Fang Y, et al. Altered microRNA expression profile with miR-146a upregulation in CD4+ T cells from patients with rheumatoid arthritis. Arthritis Res Ther. 2010; 12:R81.
crossref
51. Niimoto T, Nakasa T, Ishikawa M, Okuhara A, Izumi B, Deie M, et al. MicroRNA-146a expresses in interleukin- 17 producing T cells in rheumatoid arthritis patients. BMC Musculoskelet Disord. 2010; 11:209.
crossref

Figure 1.
Environmental factors affecting epigenetic modification.
jrd-20-140f1.tif
Figure 2.
Mechanism by which epigenetic changes are inherited.
jrd-20-140f2.tif
Figure 3.
DNA methylation and histone modification.
jrd-20-140f3.tif
Table 1.
Concordance rate of autoimmune diseases between monozygotic twins
Disease Concordance rate References
Systmeic lupus erythematosus 11∼25% (2, 3)
Rheumatoid arthritis 12∼22% (4–6)
Table 2.
Epigenetic modification in autoimmune diseases
Mechanisms Diseases Details References
DNA methylation Systemic lupus erythematosus Global hypomethylation of promoter regions of genes:  
      ITGAL (14)
      CD40LG (15)
      PRF1 (16)
      CD70 (17)
      IFGNR2 (18)
      MMP14 (18)
      LCN2 (18)
    Ribosomal RNA gene promoter (18)
      e1B promoter of CD5 in resting B cells (19)
  Rheumatoid arthritis Hypomethylation:  
      CpG islands upstream of an L1 open-reading frame (20)
      IL-6 promoter gene in monocytes (21)
    Hypermethylation:  
      Promoter of death receptor 3 (DR-3) (22)
Systemic sclerosis Hypermethylation:  
      CpG islands in Fli1 promoter (23)
Histone modification Systemic lupus erythematosus Predisposition to apoptotic nucleosomes:  
      H3K4me3  
      H4K8 triacetylation (24)
      H3K27me3 (25)
      H2BK12ac  
      Global acetylation:  
      Histone H3 and H4 in active CD4+ T cells (26)
  Rheumatoid arthritis HDAC inhibitors:  
      Block induction of MMPs (27)
      Repress ADAMTs enzymes (27)
  Hyperacetylation of histones induces p16 and p21 (28)
microRNA Systemic lupus erythematosus Decreased expression:  
      miR-146a (29)
      miR-125a (30)
    Upregulation:  
      miR-21 and miR-148a (31)
  Rheumatoid arthritis Decreased expression:  
      miR-124 (32, 33)
    Overexpression:  
      miR-203 (34)
      miR-146 (35)
      miR-155 (35)
  Sjögren's syndrome Overexpression:  
      miR-547-3p and miR-168-3p (36)
      miR-150 and miR-149 (36)

H: histone, K: lysine, HDAC: histone deacetylase, miR: microRNA, MMP: matrix metalloproteinase

Table 3.
Hypomethylated genes in autoimmune diseases
Genetic element Disease Cell type Product and/or Function References
CD70 SLE CD4+ T cell T cell proliferation (17)
CD40LG SLE CD4+ T cell IgG overproduction of B cell (15)
CD5 SLE CD19+ B cell Cytokine production and BCR signaling regulation of T cell (19)
il6 RA PBMC IL-6 (21)
miRNA-203 RA SFMC MMP, IL-6 (34)
Ribosomal DNA (18S, 28S) SLE PBMC Part of ribosomal particles (18)

BCR: B cell receptor, MMP: matrix metalloproteinase, PBMC: peripheral blood mononuclear cell, RA: rheumatoid arthritis, SFMC: synovial fluid mononuclear cell, SLE: systemic lupus erythematosus

TOOLS
Similar articles