Unfolding the pathogenesis of systemic sclerosis through epigenomics

  • Xiuzhi Jia Central Lab of Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, China
  • Hao Cheng Department of Dermatology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, China
  • Ying Xiao Central Lab of Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, China
Keywords: Epigenetics, Methylation, Histone modification, microRNA, Systemic sclerosis

Abstract

As a group of autoimmune diseases, systemic sclerosis (scleroderma, SSc) is characterized by immune dysregulation, micro-vessels dominant obliteration, and the final fibrosis of the skin and or internal organs. Although the precise mechanisms are still unknown, increasing data shows that epigenetic mechanisms, such as DNA methylation, histone modification, and microRNA (miRNA), are strictly related to the pathogenesis of scleroderma. Epigenetic mechanisms, which can link genetics and environmental stress, represents a promising field in systemic sclerosis investigation. The objective of this review is, to sum up the current information about epigenetic alteration.

Downloads

Download data is not yet available.

References

Rose NR, Mackay IR. CHAPTER 1 - The immune response in autoimmunity and autoimmune disease. In: Rose NR, Mackay IR, editors. The Autoimmune Diseases II. Boston: Academic Press; 1992. p. 1-26.

Firestein GS, Kelley WN. Kelley's textbook of rheumatology. Philadelphia, PA: Elsevier/Saunders; 2013.

Gourier G, Therene C, Mazeas M, Abasq-Thomas C, Brenaut E, Huet F, et al. Clinical characteristics of pruritus in systemic sclerosis vary according to the autoimmune subtype. Acta Derm Venereol. 2018;98(8):735-41.

Mayes MD. Scleroderma epidemiology. Rheum Dis Clin North Am. 2003;29(2):239-54.

Valesini G, Litta A, Bonavita MS, Luan FL, Purpura M, Mariani M, Balsano F. Geographical clustering of scleroderma in a rural area in the province of Rome. Clin Exp Rheumatol. 1993;11(1):41-7.

Sen D, Keung AJ. Designing epigenome editors: considerations of biochemical and locus specificities. In: Jeltsch A, Rots MG, editors. Epigenome Editing: Methods and Protocols. New York, NY: Springer New York; 2018. p. 65-87.

Dupont C, Armant DR, Brenner CA. Epigenetics: definition, mechanisms and clinical perspective. Semin Reprod Med. 2009;27(5):351-7.

Richardson B. Primer: epigenetics of autoimmunity. Nat Clin Pract Rheumatol. 2007;3(9):521-7.

Du J, Johnson LM, Jacobsen SE, Patel DJ. DNA methylation pathways and their crosstalk with histone methylation. Nat Rev Mol Cell Biol. 2015;16(9):519-32.

Altorok N, Tsou P-S, Coit P, Khanna D, Sawalha AH. Genome-wide DNA methylation analysis in dermal fibroblasts from patients with diffuse and limited systemic sclerosis reveals common and subset-specific DNA methylation aberrancies. Ann Rheum Dis. 2015;74(8):1612-20.

Wang Y, Fan P-S, Kahaleh B. Association between enhanced type I collagen expression and epigenetic repression of the FLI1 gene in scleroderma fibroblasts. Arthritis Rheum. 2006;54(7):2271-9.

Dees C, Tomcik M, Palumbo-Zerr K, Distler A, Beyer C, Lang V, et al. JAK-2 as a novel mediator of the profibrotic effects of transforming growth factor beta in systemic sclerosis. Arthritis Rheum. 2012;64(9):3006-15.

Bergmann C, Distler JHW. Canonical Wnt signaling in systemic sclerosis. Lab Invest. 2016;96(2):151-5.

Henderson WR, Jr., Chi EY, Ye X, Nguyen C, Tien Y-t, Zhou B, et al. Inhibition of Wnt/beta-catenin/CREB binding protein (CBP) signaling reverses pulmonary fibrosis. Proc Natl Acad Sci USA. 2010;107(32):14309-14.

Svegliati S, Marrone G, Pezone A, Spadoni T, Grieco A, Moroncini G, et al. Oxidative DNA damage induces the ATM-mediated transcriptional suppression of the Wnt inhibitor WIF-1 in systemic sclerosis and fibrosis. Sci Signal. 2014;7:ra84-ra.

Dees C, Schlottmann I, Funke R, Distler A, Palumbo-Zerr K, Zerr P, et al. The Wnt antagonists DKK1 and SFRP1 are downregulated by promoter hypermethylation in systemic sclerosis. Ann Rheum Dis. 2014;73:1232-9.

Wang Y, Kahaleh B. Epigenetic repression of bone morphogenetic protein receptor II expression in scleroderma. J Cell Mol Med. 2013;17(10):1291-9.

Romero L, Zhang D, Cooke J, Ho HK, Avalos E, Herrera R, et al. Differential expression of nitric oxide by dermal microvascular endothelial cells from patients with scleroderma. Vasc Med. 2000;5:147-58.

Zhu H, Zhu C, Mi W, Chen T, Zhao H, Zuo X, et al. Integration of genome-wide DNA methylation and transcription uncovered aberrant methylation-regulated genes and pathways in the peripheral blood mononuclear cells of systemic sclerosis. Int J Rheumatol. 2018;2018:7342472-.

Ramos PS, Zimmerman KD, Haddad S, Langefeld CD, Medsger TA, Jr., Feghali-Bostwick CA. Integrative analysis of DNA methylation in discordant twins unveils distinct architectures of systemic sclerosis subsets. Clin Epigenetics. 2019;11(1):58-.

Sawalha AH, Jeffries M, Webb R, Lu Q, Gorelik G, Ray D, et al. Defective T-cell ERK signaling induces interferon-regulated gene expression and overexpression of methylation-sensitive genes similar to lupus patients. Genes & Immun. 2008;9(4):368-78.

Hellman A, Chess A. Gene body-specific methylation on the active X chromosome. Science. 2007;315:1141-3.

Lian X, Xiao R, Hu X, Kanekura T, Jiang H, Li Y, et al. DNA demethylation of CD40L in CD4+ T cells from women with systemic sclerosis: A possible explanation for female susceptibility. Arthritis Rheum. 2012;64(7):2338-45.

Fukasawa C, Kawaguchi Y, Harigai M, Sugiura T, Takagi K, Kawamoto M, et al. Increased CD40 expression in skin fibroblasts from patients with systemic sclerosis (SSc): role of CD40-CD154 in the phenotype of SSc fibroblasts. Eur J Immunol. 2003;33(10):2792-800.

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(5):1282-91.

Jiang H, Xiao R, Lian X, Kanekura T, Luo Y, Yin Y, et al. Demethylation of TNFSF7 contributes to CD70 overexpression in CD4+ T cells from patients with systemic sclerosis. Clin Immunol. 2012;143(1):39-44.

Yin Y, Liu W, Dai Y. SOCS3 and its role in associated diseases. Human Immunology. 2015;76(10):775-80.

Hill GR, Kuns RD, Raffelt NC, Don ALJ, Olver SD, Markey KA, et al. SOCS3 regulates graft-versus-host disease. Blood. 2010;116(2):287-96.

Kondo Y, Shen L, Cheng AS, Ahmed S, Boumber Y, Charo C, et al. Gene silencing in cancer by histone H3 lysine 27 trimethylation independent of promoter DNA methylation. Nat Genet. 2008;40(6):741-50.

Hemmatazad H, Rodrigues HM, Maurer B, Brentano F, Pileckyte M, Distler JHW, et al. Histone deacetylase 7, a potential target for the antifibrotic treatment of systemic sclerosis. Arthritis Rheum. 2009;60(5):1519-29.

Huber LC, Distler JHW, Moritz F, Hemmatazad H, Hauser T, Michel BA, et al. Trichostatin A prevents the accumulation of extracellular matrix in a mouse model of bleomycin-induced skin fibrosis. Arthritis Rheum. 2007;56(8):2755-64.

Matouk CC, Marsden PA. Epigenetic regulation of vascular endothelial gene expression. Circ Res. 2008;102(8):873-87.

Ciechomska M, O'Reilly S, Przyborski S, Oakley F, Bogunia-Kubik K, van Laar JM. Histone demethylation and Toll-like receptor 8-dependent cross-talk in monocytes promotes transdifferentiation of fibroblasts in systemic sclerosis via Fra-2. Arthritis Rheum. 2016;68(6):1493-504.

Maurer B, Distler JHW, Distler O. The Fra-2 transgenic mouse model of systemic sclerosis. Vascul Pharmacol. 2013;58(3):194-201.

Krämer M, Dees C, Huang J, Schlottmann I, Palumbo-Zerr K, Zerr P, et al. Inhibition of H3K27 histone trimethylation activates fibroblasts and induces fibrosis. Ann Rheum Dis. 2013;72:614-20.

Hong S, Cho Y-W, Yu L-R, Yu H, Veenstra TD, Ge K. Identification of JmjC domain-containing UTX and JMJD3 as histone H3 lysine 27 demethylases. Proc Natl Acad Sci USA. 2007;104(47):18439-44.

Agger K, Cloos PAC, Christensen J, Pasini D, Rose S, Rappsilber J, et al. UTX and JMJD3 are histone H3K27 demethylases involved in HOX gene regulation and development. Nature. 2007;449(7163):731-4.

Wang Y, Yang Y, Luo Y, Yin Y, Wang Q, Li Y, et al. Aberrant histone modification in peripheral blood B cells from patients with systemic sclerosis. Clin Immunol. 2013;149(1):46-54.

Wang Q, Xiao Y, Shi Y, Luo Y, Li Y, Zhao M, et al. Overexpression of JMJD3 may contribute to demethylation of H3K27me3 in CD4+ T cells from patients with systemic sclerosis. Clin Immunol. 2015;161(2):396-9.

Ambros V. The functions of animal microRNAs. Nature. 2004;431(7006):350-5.

Bartel DP. Metazoan MicroRNAs. Cell. 2018;173(1):20-51.

Bartel DP. MicroRNAs: Genomics, Biogenesis, Mechanism, and Function. Cell. 2004;116(2):281-97.

Maurer B, Stanczyk J, Jüngel A, Akhmetshina A, Trenkmann M, Brock M, et al. MicroRNA-29, a key regulator of collagen expression in systemic sclerosis. Arthritis Rheum. 2010;62(6):1733-43.

Zhu H, Li Y, Qu S, Luo H, Zhou Y, Wang Y, et al. MicroRNA expression abnormalities in limited cutaneous scleroderma and diffuse cutaneous scleroderma. J Clin Immunol. 2012;32(3):514-22.

Zhu H, Luo H, Li Y, Zhou Y, Jiang Y, Chai J, et al. MicroRNA-21 in scleroderma fibrosis and its function in TGF-β-regulated fibrosis-related genes expression. J Clin Immunol. 2013;33(6):1100-9.

Wermuth PJ, Piera-Velazquez S, Jimenez SA. Exosomes isolated from serum of systemic sclerosis patients display alterations in their content of profibrotic and antifibrotic microRNA and induce a profibrotic phenotype in cultured normal dermal fibroblasts. Clin Exp Rheumatol. 2017;35 Suppl 106(4):21-30.

Sing T, Jinnin M, Yamane K, Honda N, Makino K, Kajihara I, et al. microRNA-92a expression in the sera and dermal fibroblasts increases in patients with scleroderma. Rheumatology. 2012;51(9):1550-6.

Honda N, Jinnin M, Kajihara I, Makino T, Makino K, Masuguchi S, et al. TGF-β–mediated downregulation of microRNA-196a contributes to the constitutive upregulated type I collagen expression in scleroderma dermal fibroblasts. J Immunol. 2012;188:3323-31.

Honda N, Jinnin M, Kira-Etoh T, Makino K, Kajihara I, Makino T, et al. miR-150 down-regulation contributes to the constitutive type I collagen overexpression in scleroderma dermal fibroblasts via the induction of integrin β3. Am J Pathol. 2013;182(1):206-16.

Jiang X, Tsitsiou E, Herrick SE, Lindsay MA. MicroRNAs and the regulation of fibrosis. FEBS J. 2010;277(9):2015-21.

Makino K, Jinnin M, Kajihara I, Honda N, Sakai K, Masuguchi S, et al. Circulating miR-142-3p levels in patients with systemic sclerosis. Clin Exp Dermatol. 2012;37(1):34-9.

Christmann RB, Wooten A, Sampaio-Barros P, Borges CL, Carvalho CRR, Kairalla RA, et al. miR-155 in the progression of lung fibrosis in systemic sclerosis. Arthritis Res Ther. 2016;18(1):155-.

Wermuth PJ, Piera-Velazquez S, Rosenbloom J, Jimenez SA. Existing and novel biomarkers for precision medicine in systemic sclerosis. Nat Rev Rheumatol. 2018;14(7):421-32.

Vrba L, Garbe JC, Stampfer MR, Futscher BW. Epigenetic regulation of normal human mammary cell type-specific miRNAs. Genome Res. 2011;21(12):2026-37.

Messemaker TC, Chadli L, Cai G, Goelela VS, Boonstra M, Dorjée AL, et al. Antisense long non-coding RNAs are deregulated in skin tissue of patients with systemic sclerosis. J Invest Dermatol. 2018;138(4):826-35.

Wang Z, Jinnin M, Nakamura K, Harada M, Kudo H, Nakayama W, et al. Long non-coding RNA TSIX is upregulated in scleroderma dermal fibroblasts and controls collagen mRNA stabilization. Exp Dermatol. 2016;25(2):131-6.

Xu F, Jin L, Jin Y, Nie Z, Zheng H. Long noncoding RNAs in autoimmune diseases. J Biomed Mater Res A. 2019;107(2):468-75.

Wu G-C, Pan H-F, Leng R-X, Wang D-G, Li X-P, Li X-M, et al. Emerging role of long noncoding RNAs in autoimmune diseases. Autoimmun Rev. 2015;14(9):798-805.

Colletti M, Galardi A, De Santis M, Guidelli GM, Di Giannatale A, Di Luigi L, et al. Exosomes in systemic sclerosis: messengers between immune, vascular and fibrotic components? Int J Mol Sci. 2019;20(18):4337.

Published
2020-01-02
How to Cite
JiaX., ChengH., & XiaoY. (2020). Unfolding the pathogenesis of systemic sclerosis through epigenomics. STEMedicine, 1(1), e5. https://doi.org/10.37175/stemedicine.v1i1.5
Section
Review articles