Skip to main content
Journal of Scleroderma and Related Disorders logoLink to Journal of Scleroderma and Related Disorders
. 2019 Jul 3;4(3):238–250. doi: 10.1177/2397198319855872

Epigenetics of scleroderma: Integrating genetic, ethnic, age, and environmental effects

Paula S Ramos 1,
PMCID: PMC8922566  NIHMSID: NIHMS1744821  PMID: 35382507

Abstract

Scleroderma or systemic sclerosis is thought to result from the interplay between environmental or non-genetic factors in a genetically susceptible individual. Epigenetic modifications are influenced by genetic variation and environmental exposures, and change with chronological age and between populations. Despite progress in identifying genetic, epigenetic, and environmental risk factors, the underlying mechanism of systemic sclerosis remains unclear. Since epigenetics provides the regulatory mechanism linking genetic and non-genetic factors to gene expression, understanding the role of epigenetic regulation in systemic sclerosis will elucidate how these factors interact to cause systemic sclerosis. Among the cell types under tight epigenetic control and susceptible to epigenetic dysregulation, immune cells are critically involved in early pathogenic events in the progression of fibrosis and systemic sclerosis. This review starts by summarizing the changes in DNA methylation, histone modification, and non-coding RNAs associated with systemic sclerosis. It then discusses the role of genetic, ethnic, age, and environmental effects on epigenetic regulation, with a focus on immune system dysregulation. Given the potential of epigenome editing technologies for cell reprogramming and as a therapeutic approach for durable gene regulation, this review concludes with a prospect on epigenetic editing. Although epigenomics in systemic sclerosis is in its infancy, future studies will help elucidate the regulatory mechanisms underpinning systemic sclerosis and inform the design of targeted epigenetic therapies to control its dysregulation.

Keywords: Systemic sclerosis, scleroderma, epigenetics, epigenomics, environment, DNA methylation, histone modification, non-coding RNA, gene regulation, epigenome editing

Introduction

Systemic sclerosis (SSc, scleroderma) is a multisystem autoimmune disease characterized by cutaneous and visceral fibrosis, immune dysregulation, and vasculopathy. It is often considered a fibrosing disease preceded and accompanied by autoimmunity and vasculopathy, 1 as immune system activation is an early and key event in SSc pathogenesis. 2

As reviewed elsewhere, SSc shows great variation in reported prevalence estimates (varying between 31 and 659 cases per million), a strong sex bias (between three and 14 affected women for one man), and marked ethnic disparities.3,4 While twin and family studies support a high genetic contribution to the development of autoimmunity in SSc, multiple lines of evidence support a modest genetic etiology in SSc, 3 suggesting a substantial role for epigenetic or environmental factors in SSc susceptibility.

Epigenetics refers to chromatin modifications that influence gene function but not involving changes in the DNA sequence that can be propagated through cell division. By allowing or preventing molecules’ access to regulatory sequences, the chromatin’s structure provides gene regulation. Commonly studied epigenetic mechanisms include DNA methylation, histone modification, non-coding RNA, and chromatin accessibility representing transcription factor-binding sites. These epigenetic marks can change the spatial conformation of chromatin to modulate or regulate gene expression. The epigenetic architecture is influenced by genetic variation and environmental exposures, and it changes with chronological age and between populations. Given that epigenetic changes reflect influences of both genetic and environmental factors, and epigenetics thus provides a molecular mechanism linking genetic background and environmental exposures to disease, there is much interest in understanding the role of epigenetic regulation in disease.

Comprehensive reviews of epigenetic research in SSc have been recently published.58 In the first part, this review summarizes the changes in DNA methylation, histone modification, and non-coding RNAs associated with SSc. It then discusses the role of genetic, ethnic, age, and environmental effects on epigenetic regulation, followed by a prospect on epigenetic editing. Since immune system dysregulation critically contributes to early pathogenic events in the progression of fibrosis and SSc, and epigenetic regulation is critically involved in immune cell differentiation and response to stimuli,9,10 the second part of this review focuses on epigenetic regulation of immune cells.

DNA methylation

DNA methylation consists of the addition of a CH3 methyl group to cytosine phospho-guanine dinucleotides (CpG). CpG-rich sequences, called CpG islands, often locate within gene promoter regions. CpG methylation is essential for multiple biological processes, including gene regulation, genomic imprinting, X chromosome inactivation or repression of transposable elements. Methylation is catalyzed by a family of DNA methyltransferases (DNMTs). Although generally associated with transcriptional silencing, 11 the precise relationships between DNA methylation, genetic factors, and gene expression are complex and poorly understood.1214 The correlation between DNA methylation and gene expression can be positive or negative, is tissue-specific, and context-specific, in that the local DNA sequence and genomic features largely account for local patterns of methylation.11,1517

In 2006, Wang and colleagues 18 first implicated DNA methylation mechanisms as mediators of the fibrotic manifestations of SSc by unveiling hypermethylation and histone deacetylation at the friend leukemia integration 1 (FLI1) promoter. As thoroughly reviewed elsewhere,5,6 a total of 18 studies have documented changes in enzyme (including methyl-CpG-binding domain proteins (MBD) and DNMTs), global methylation, and candidate gene methylation levels across multiple cell types (blood, CD4+ T cells, fibroblasts, endothelial cells). Among these, three genome-wide DNA methylation studies1921 on skin, lung fibroblasts, CD4+ T cells, and CD8+ T cells, further confirm the role of aberrant DNA methylation on SSc pathogenesis.

DNA methylation changes associated with SSc are summarized in Table 1. In CD4+ T cells there is global DNA hypomethylation and concomitant underexpression of methylation-regulating genes (e.g. DNMT1, MBD3, and MBD). 22 Overexpression of CD40L, CD70, and ITGAL (aka CD11a) has been attributed to hypomethylation of the genes’ promoters.2830 Demethylation of the costimulatory CD70 (aka TNFSF7) and X-chromosome-encoded CD40L genes leads to their overexpression on activated CD4+ T cells, and ultimately B-cell autoantibody production. In the case of the integrin subunit alpha L (ITGAL), the gene’s overexpression contributes to increased CD4+ T cell proliferation, IgG production, and collagen production, suggesting a role for epigenetic dysregulation in immune and fibrotic dysfunction is SSc. 30 Forkhead box P3 (FOXP3), a transcription factor required for generation of regulatory T (Treg) cells that is hypomethylated in Treg cells and hypermethylated in conventional T cells, 36 shows hypermethylation in CD4+ T cells of patients with SSc, which has effect in the number of Tregs. 31

Table 1.

Summary of DNA methylation changes in SSc.

Modification Cell type/tissue Refs
GlobalZ
 DNMTs levels CD4+ T cells 22
Fibroblasts 18,23
 MBDs levels CD4+ T cells 22
Fibroblasts 18
 TET levels Fibroblasts 24,25
 MECP2 levels Fibroblasts 18,25
 Global methylation levels Whole blood 26
CD4+ T cells 22
Fibroblasts 24
Gene-specific
 RORC1, RORC2 PBMCs 27
 CD40L CD4+ T cells 28
 CD70 CD4+ T cells 29
 CD11a CD4+ T cells 30
 FOXP3 CD4+ T cells 31
 BMPR2 Endothelial cells, skin 32
 FLI1 Fibroblasts 18
 KLF5 Fibroblasts 33
 C8orf4/TCIM Lung fibroblasts 21
 DKK1, SFRP1 PBMCs, fibroblasts 34
 Several X chromosome genes PBMCs 35
 ITGAL, CD11a, CD70, FOXP3, and other Whole blood 26
 ITGA9, COL4A2, TNXB, CDH11, and other Fibroblasts 19
 Type I interferon genes CD4+ and CD8+ T cells 20

SSc: scleroderma or systemic sclerosis; Refs: references; PBMCs: peripheral blood mononuclear cells; DNMTs: DNA methyltransferases; MBDs: methyl-CpG-binding domain; TET: ten-eleven translocation; MECP2: methyl-CpG binding protein 2; RORC: RAR-related orphan receptor C; FOXP3: forkhead box P3; BMPR2: bone morphogenetic protein receptor type 2; FLI1: Fli-1 proto-oncogene, ETS transcription factor; KLF5: Kruppel like factor 5; TCIM: transcriptional and immune response regulator; DKK1: dickkopf WNT signaling pathway inhibitor 1; SFRP1: secreted frizzled related protein 1; ITGAL: integrin subunit alpha L; ITGA9: integrin subunit alpha 9; COL4A2: collagen type IV alpha 2 chain; TNBX: tenascin XB; CDH11: cadherin 11.

In contrast to CD4+ T cells, fibroblasts from SSc patients show high levels of methylation-regulating genes such as DNMT1, MBD1, and MECP2. There is hypermethylation in the promoter region of FLI1, an inhibitor of collagen expression. 18 In lung fibroblasts, hypermethylation of the transcriptional and immune response regulator (TCIM, aka C8orf4) gene, which functions as a positive regulator of the Wnt/β-catenin signaling pathway, leads to its underexpression, resulting in a decreased capacity of the fibrotic lung fibroblasts to up-regulate cyclooxygenase-2 (COX2) and COX-2-derived prostaglandin E2 (PGE2) synthesis. 21 It is important to note that many of these epigenetic modifications (e.g. epigenetic silencing of FLI1 or TCIM) might drive fibroblast activation and transition to myofibroblasts. Recent reviews summarize the role of DNA methylation, histone modifications, and non-coding RNAs involved in myofibroblast activation.37,38 For example, MeCP2 promotes myofibroblast differentiation in mice and human,39,40 and hypomethylation of genes such as integrin subunit alpha (ITGA9) and cadherin 11 (CDH11) in SSc skin fibroblasts 19 might contribute to myofibroblast differentiation.

In microvascular endothelial cells, hypermethylation of the promoter of the bone morphogenetic protein type II receptor (BMPR2) gene leads to reduced gene expression, resulting in endothelial cell enhanced apoptosis. 32

Histone modification

Chromatin is formed by DNA wrapped around histone octamers formed by two copies of the core histones (H2A, H2B, H3, and H4) and linker histones (H2 and H5). Covalent post-translational modifications at specific amino acid residues in the side chains of histone tails can affect chromatin structure and influence its accessibility. Histone tail modifications include methylation, phosphorylation, acetylation, ubiquitylation, sumoylation, and ADP ribosylation. Histone acetyltransferases (HATs) (e.g. EP300/p300, KAT/MYST) and histone deacetylases (HDACs) (e.g. HDAC1-11, SIRT1-7) regulate histone acetylation required for biological processes such as DNA replication and transcription.

Histone acetylation and histone methylation are the most common modifications. Histone acetylation typically occurs at lysine residues on histone H3 or H4 and can present as mono-, di-, or tri-acetylation. Because it leads to an open chromatin conformation accessible to transcription factors, histone acetylation marks are associated with transcriptional activation. Histone methylation can be associated with either transcriptional repression or activation depending on the number of methyl groups that are added and the location of the amino acid being methylated. For example, enhancers marked by acetylation of histone H3 at lysine 27 (H3K27ac) and methylation of H3 at lysine 4 (H3K4me1) promote transcription, 41 while H3K27 trimethylation (H3K27me3) is a repressive epigenetic mark.

The first evidence for histone modification involvement in SSc came from a study investigating the role of the transcriptional coactivator histone acetyltransferase p300 in the profibrotic responses elicited by TGF-beta in fibroblasts. 42 The authors found that elevated p300 expression contributes to the intensity of profibrotic responses, establishing the role of the HAT p300 is mediating stimulation of collagen synthesis in fibroblasts. 42 Soon after another study in fibroblasts reported increased levels of epigenetic mediators, as well as normalized collagen expression upon addition of HDAC inhibitors. 18

Table 2 summarizes the histone modification changes associated with SSc. Most studies of histone modification in SSc have focused on fibroblasts. For example, the histone deacetylase sirtuin 1 (SIRT1) is reduced in SSc skin and fibroblasts. 43 The histone demethylase lysine demethylase 6B (KDM6B, aka Jumonji domain-containing protein 3 (JMJD3)) is increased in fibroblasts, 46 resulting in accumulation of H3K27me3 at the promoter of the profibrotic FOS like 2, AP-1 transcription factor (FOSL2, aka FRA2). 49 Thus, JMJD3 and H3K27me3 modulate fibroblast activation by regulating FOSL2 expression. As reviewed elsewhere,37,38 histone modifications are known contributors to myofibroblast activation. For example, JMJD3 regulates myofibroblast activation, 52 p300 regulates fibrosis through myofibroblast transformation, 53 and elevated levels of the histone methyltransferase enhancer of zeste homolog 2 (EZH2) contribute to myofibroblast transdifferentiation in SSc. 54

Table 2.

Summary of histone modification changes in SSc.

Modification Cell type/tissue Gene Refs
Global
 HDACs levels Fibroblasts NA 18,43,44
B cells NA 45
Endothelial cells NA 46
 HATs levels Fibroblasts NA 42
B cells NA 47
 HDMs levels CD4+ T cells NA 48
B cells NA 45
 HMTs levels B cells NA 45
 H4 acetylation levels B cells NA 45
 H3K9 methylation levels B cells NA 45
 H3K27me3 Fibroblasts NA 49
CD4+ T cells NA 48
Gene-specific
 H3, H4 hypoacetylation Fibroblasts FLI1 18
 H3, H4 hypoacetylation Fibroblasts KLF5 33
 H3K27me3 Fibroblasts FRA2/FOSL2 49
 H4 hyperacetylation Fibroblasts COL1A2 47
 H3, H4 hyperacetylation Fibroblasts NR4A1 50
 Histone hypoacetylation Fibroblasts WIF1 51
 HDAC5 levels Endothelial cells CCN1, NECTIN2, FSTL1 46

SSc: Scleroderma or systemic sclerosis; Refs: references; HDACs: histone deacetylases; HATs: histone acetyltransferases; HDMs: histone demethylases; HMT: histone methyltransferase; H3: histone 3; H4: histone 4; H3K9: histone 3 lysine 9; H3K27me3: histone 3 lysine 27 trimethylation; FLI1: Fli-1 proto-oncogene, ETS transcription factor; KLF5: Kruppel like factor 5; FOSL2: FOS like 2, AP-1 transcription factor subunit; COL1A2: collagen type I alpha 2 chain; NR4A1: receptor subfamily 4 group A member 1; WIF1: WNT inhibitory factor 1; CCN1: cellular communication network factor 1; NECTIN2: nectin cell adhesion molecule 2; FSTL1: follistatin like 1.

In B cells from SSc patients, global hyperacetylation of histone H4, and hypomethylation of H3K9 were correlated with levels of histone enzymes like HDAC2 and HDAC7, disease activity, and skin thickness. 45 Similarly to what was observed in fibroblasts, KDM6B (aka JMJD3) overexpression in CD4+ T cells of SSc patients was associated with lower levels of the repressive H3K27me3 mark. 48

Endothelial cells from SSc patients show increased expression of HDAC5, which alters the regulation of key proangiogenic and profibrotic genes, resulting in impaired angiogenesis. 46

Non-coding RNAs

Non-coding RNAs (ncRNAs) are functional RNA molecules that mediate various intracellular processes. 55 Classes of ncRNAs include microRNAs (miRNAs), long non-coding RNAs (lncRNAs), small nucleolar RNAs (snoRNAs), and circular RNAs (circRNAs), all with different regulatory functions. The best characterized ncRNAs are miRNAs and lncRNAs. miRNAs (19–24 nucleotides in length) regulate gene silencing of more than 60% of human protein-coding genes at the transcriptional and/or translational level. lncRNAs (over 200 nucleotides in length) are found in sense or antisense orientation to protein-coding genes, or within intergenic regions. lncRNAs function as chromatin remodelers, transcriptional regulators, and post-transcriptional regulators.

Comprehensive lists of ncRNAs dysregulated in SSc patient tissues have been recently reviewed.5,6 Table 3 summarizes the ncRNAs with target genes that have been reported in SSc; several studies that did not report target genes for the ncRNAs are not included, including all studies evaluating panels of candidate circulating RNAs in serum, plasma, or exosomes (recently reviewed elsewhere 5 ). The vast majority of studies in SSc has focused on the role of miRNAs, especially in fibroblasts.

Table 3.

Summary of non-coding RNA with reported target genes in SSc.

Modification Cell type/tissue Target genes Refs
miRNAs
 miR-618 pDCs IRF8 56
 miR-7 Fibroblasts, skin, serum COL1A1, COL2A1 57,58
 miR-21 Fibroblasts, skin SMAD7 59,60
 miR-27a-3p Fibroblasts ACTA2, SMAD2, SMAD4 61
 miR-29a Fibroblasts, hair COL1A1, COL3A1 59,6265
 miR-30a-3p Fibroblasts TNFSF13B 66
 miR-30b Skin, serum PDGFRB 67
 miR-92a Fibroblasts, serum MMP1 68
 miR-129-5p Fibroblasts COL1A1 69
 miR-130b Fibroblasts, skin PPARG 70
 miR-135b Fibroblasts, serum, monocytes STAT6 25
 miR-145 Fibroblasts, skin SMAD3 59
 miR-150 Fibroblasts, skin, serum ITGB3 71
 miR-155 Fibroblasts, skin CSNK1A1, INPP5D 72
 mir-193b Fibroblasts, skin PLAU 73
 miR-196a Fibroblasts, skin, serum, hair COL1A1, COL2A1 7477
 miR-202-3p Fibroblasts, skin MMP1 78
 miR-5196 Monocytes, serum FOSL2 79
 let-7a Fibroblasts, skin, serum COL1A1, COL2A1 80
lncRNAs
 TSIX Fibroblasts, serum COL1A1, COL1A2 81

SSc: scleroderma or systemic sclerosis; Refs: references; pDCs: plasmacytoid dendritic cells; IRF8: interferon regulatory factor 8; COL1A1: collagen type I alpha 1 chain; COL2A1: collagen type II alpha 1 chain; SMAD7: SMAD family member 7; ACTA2: actin alpha 2, smooth muscle; COL3A1: collagen type III alpha 1 chain; TNFSF13B: TNF superfamily member 13b; PDGFRB: platelet-derived growth factor receptor beta; MMP1: matrix metallopeptidase 1; PPARG: peroxisome proliferator activated receptor gamma; STAT6: signal transducer and activator of transcription 6; ITGB3: integrin subunit beta 3; CSNK1A1: casein kinase 1 alpha 1; INPP5D: inositol polyphosphate-5-phosphatase D; PLAU: plasminogen activator, urokinase; FOSL2: FOS like 2, AP-1 transcription factor subunit; TSIX: TSIX transcript, XIST antisense RNA; COL1A2: collagen type I alpha 2 chain.

miRNAs were first implicated in SSc in 2010, when miR-29a was shown to be downregulated in SSc fibroblasts and skin, leading to an increase in type I and type III collagen in fibroblasts. 62 Further studies in dermal fibroblasts have suggested that miR-29a is an inducer of apoptosis and an attenuator of extracellular matrix (ECM) production, 65 that it reduces tissue inhibitor of metalloproteinases 1 (TIMP1) secretion, a key player in ECM deposition, via targeting TGF-beta activated kinase 1 binding protein 1 (TAB1) repression, 63 and that it increases functional matrix metallopeptidase 1 (MMP1) production resulting in collagen degradation. 63 As summarized in Table 3, expression of miR-7, miR-129-5p, miR-196a, and let-7a in fibroblasts also affect the expression of collagen. It should be noted that the antifibrotic miR-7 was upregulated in SSc 57 but downregulated in patients with limited cutaneous SSc, 58 suggesting different mechanistic roles for miR-7 depending on specific disease manifestations. Other downregulated, antifibrotic miRNAs include miR-30b, miR-135b, and miR-150. Overexpressed, profibrotic miRNAs include miR-21, miR-92a, miR-130b, miR-155, and miR-202-3p. Among these studies focused on fibroblasts, two miRNAs were shown to target immune genes, and one to target a vascular gene. miR-30a-3p, which was downregulated in SSc fibroblasts, is a basal repressor of the TNF superfamily member 13b gene (TNFSF13B, aka BAFF), thus leading to decreased B cell survival. 66 miR-135b, which was downregulated in SSc fibroblasts, targets the transcription factor signal transducer and activator of transcription 6 gene (STAT6), which in turn regulates IL-13-mediated collagen production by fibroblasts. 25 miR-135b is thus important to reduce collagen induction independent from TGFβ1. Interestingly, the authors also showed that methylation regulates miR-135b, underscoring the joint role of multiple epigenetic factors in gene regulation. Finally, the downregulation of miR-193b in SSc skin and fibroblasts promoted the expression of the plasminogen activator, urokinase gene (PLAU, aka u-PA), resulting in proliferation of vascular smooth muscle cells, and thus contributing to the proliferative vasculopathy with intimal hyperplasia characteristic for SSc. 73 The effects of ncRNAs in myofibroblast activation in SSc have been reviewed in detail elsewhere. 37

Analyses of the role of miRNAs in immune cells are emerging. In monocytes, miRNA-5196 was increased and positively correlated with C-reactive protein (CRP) level in SSc patients, which led to inhibition of the FOSL2 gene (aka FRA2) and TIMP1 expression 79 . As mentioned above, accumulation of H3K27me3 at the promoter of FOSL2 in fibroblasts also regulates FOSL2 expression, 49 once again highlighting the orchestrated control of gene regulation involving multiple epigenetic mechanisms. miRNA expression profiling of plasmacytoid dendritic cells (pDCs) revealed that the observed overexpression of miR-618 suppressed the development of pDCs and increased their ability to secrete interferon α (IFNα), potentially contributing to the type I IFN signature observed in SSc patients. 56

A large number of studies have investigated a broad range of circulating miRNAs in SSc.5,6,82 The resulting profiles of miRNAs can help discriminate patients from controls, patients with different disease subtypes, and with different clinical criteria or disease manifestations. Circulating miRNAs can be found in body fluids such as serum or plasma, which given the easy tissue accessibility, makes them good candidates for disease biomarkers. Circulating miRNAs can also be found in extracellular vesicles such as exosomes, which mediate intercellular communication by releasing their macromolecular content into target cells. Interestingly, serum exosomes from SSc patients contain miRNA displaying a markedly profibrotic profile and induce a profibrotic phenotype in target normal fibroblasts. 83 This ability to alter the phenotype of distant target cells could explain the extension of the SSc-associated fibrotic process to unaffected cells and tissues. 83

Very little is known about the involvement of lncRNA in SSc. In 2016, TSIX (TSIX transcript, XIST antisense RNA) was reported as upregulated in the serum and dermal fibroblasts of SSc patients. 81 More recently, RNA-seq of skin tissue uncovered 676 lncRNAs differentially expressed between patients and controls. 84

Genetic, ethnic, and age effects on the immune epigenome

The epigenome refers to the collection of chemical modifications of chromatin, alterations in chromatin constituents, and changes in the spatial chromatin organization that regulate gene expression. Dynamic regulation of these epigenetic marks underlies cell function and plasticity and provides a heritable response to developmental and environmental cues. This epigenomic regulation is critical for immune cell development and differentiation, immune function in host defense mechanisms, and immune dysregulation exhibited during autoimmune disease. 85 Given the need of the immune system to respond prompt and efficiently to stimuli by eliciting rapid immune gene expression, immune cells may be more susceptible to epigenetic reprogramming that might poise them to a sustained, activated phenotype. Evidence shows that many immune gene promoters exist in an epigenetically primed, or accessible state prior to activation, which permits robust gene transcription upon activation by transcription factors. 86 As recently reviewed elsewhere, 87 this primed epigenetic state is also observed in innate immune genes; interestingly, monocytes in a resting state can be epigenetically primed, and many tumor necrosis factor (TNF)-responsive innate immune genes are engaged in preformed enhancer-promoter chromosomal looping interactions required for gene expression. Since immune system dysregulation critically contributes to early pathogenic events in the progression of fibrosis and SSc, and epigenetic phenomena may explain this early immune system dysregulation, there has been increasing interest in identifying differential epigenetic patterns in autoimmune diseases. 85 In SSc, for example, the hypomethylation and overexpression of CD40L in CD4+ T cells of SSc patients might be contributing to the overproduction of autoantibodies that drive disease pathogenesis. 28

Multiple genome-wide association (GWA) and other large-scale studies identified genetic variants associated with SSc. 5 These results, coupled with the regions shared with other autoimmune diseases, emphasize the role of dysregulation of immune pathways as a key etiologic factor in SSc pathogenesis. 3 However, the molecular mechanisms linking each variant to the disease phenotype are largely unknown. This is because most variants, including 90% of causal autoimmune disease variants, lie in non-coding regions of the genome and are concentrated in regulatory DNA whose mechanisms are not yet well characterized.88,89 Since the effects of environmental exposures on phenotypes might be mediated by epigenetic changes, there has been great interest in understanding the role of the epigenome in gene regulation, cellular differentiation, and human disease. Several large-scale initiatives such as the ENCODE, 90 Epigenomics Roadmap, 91 and BLUEPRINT 92 projects have mapped gene regulatory elements across a wide range of tissues and cell types using a variety of assays.

The DNA sequence variation can exert effects of gene expression though epigenetic modifications. The influence of genetic factors on methylation levels is well established, as multiple studies have mapped DNA sequence variants associated with DNA methylation variation, that is, methylation quantitative trait loci (meQTLs), across a variety of tissues.13,9397 About ~ 20% of the inter-individual variation in DNA methylation has been attributed to genetics.98,99 In addition, genetic variants associated with complex traits by genome-wide association studies (GWAS) often overlap both expression QTLs and meQTLs, suggesting that disease risk can be mediated, directly or indirectly, by variation in DNA methylation.100105

Despite the disproportionate burden on African Americans, the majority of SSc studies have been conducted in populations of European ancestry. This underrepresentation in research continues to exacerbate the health disparities gap, as epigenetic changes have been shown to vary between populations,106113 and might be involved in the heightened autoimmune dysregulation in African ancestry relative to European ancestry SSc patients.114,115 Although differential methylation between ethnic groups is partially explained by their distinct genetic ancestry, environmental factors not captured by ancestry are significant contributors to variation in methylation. 108 Evidence for the causal impact of ancestry-related differences in DNA methylation on immune gene regulation is emerging. Husquin et al. 109 unveiled extensive population differences in DNA methylation between individuals of African and European descent, detected in primary monocytes. They report higher levels of DNA methylation in genes involved in the activation and regulation of immune responses in individuals of European ancestry, mostly owing to genetic control. Since up to 16% of immune-related genes that are hypermethylated in Europeans are also differentially expressed between populations, 110 the ancestry-related differences in transcriptional responses to bacteria reported in macrophages, where European ancestry is associated with lower inflammatory responses, 116 might be partially due to these ancestry-related epigenetic differences. 109 At the time of writing, only one epigenetic study has been conducted in SSc patients of African ancestry: Matatiele and colleagues 26 assessed DNA methylation levels of selected genes in black South African SSc patients, revealing patterns similar to those observed in studies of other ancestries (Table 1).

In addition to genetic background and ancestry, aging can also influence epigenetic patterns. Specifically, thousands of CpG sites whose tissue-specific methylation levels are strongly correlated with chronological age have been previously identified,117,118 and many of these CpG sites are shared and replicate across a broad range of human populations. 111 In SSc, premature activation of aging-associated molecular mechanisms is emerging as an important contributor to the autoimmune, vascular, and fibrotic disease pathogenesis. 119 Notably, the HDACs sirtuins (SIRTs), which are key regulators of the aging process and decline during normal aging, show decreased levels in SSc, 119 supporting the notion that epigenomic dysregulation contributes to disease pathogenesis.

Environmental effects on the epigenome

Multiple chemical and physical environmental stressors, diet, life habits, and pharmacological treatments that can alter the epigenome and determine disease phenotypes have been thoroughly reviewed elsewhere. 120 Briefly, environmental pollutants with epigenetic effects include metals (arsenic, mercury, nickel, lead, and cadmium), air pollutants, asbestos, benzene, and electromagnetic radiation (ultraviolet (UV) light). Endocrine disruptors include pesticides (DDT and methoxychlor), fungicides (vinclozolin), herbicides (atrazine), industrial chemicals (PCBs, dioxins), plant hormones (phytoestrogens), and plastics (bisphenol A (BPA) and phthalates). Diet is one of the most studied and better understood environmental epigenetic factors, and well-known examples of diet affecting epigenetic marks include folate (vitamin B9), caloric restriction, and bioactive dietary compounds such as polyphenols. Lifestyle and environmental conditions of the mother during pregnancy (including diet, chemical stressors, endocrine disruptors, smoking or alcohol consumption) affect the epigenome of the offspring,120,121 and can have long-term effects on the health of the offspring.122124 There is evidence that certain medications can also induce genome-wide epigenetic changes (sodium valproate (VPA), diethylstilbestrol (DES), procaine, pyrazinamide, doxorubicin, gemcitabine, cisplatin, and temozolomide). 120 Infectious agents (Leishmania and mycobacterium tuberculosis),14,125 tobacco smoke,126,127 diesel exhaust particles, 128 and other indoor and outdoor pollutants 129 have been shown to affect methylation levels. Psychosocial factors, including measures of traumatic experiences,130132 socioeconomic status,133,134 and general perceived stress 135 also affect methylation levels.

Notably, exposures during sensitive periods of immune development can have lasting effects on inflammation, and recent empirical work has implicated environments in infancy and childhood as important determinants of inflammatory phenotypes. For example, individuals born at lower birth weight and infants breastfed for shorter durations have higher concentrations of CRP, and major psychosocial stressors and socioeconomic adversity in childhood are associated with proinflammatory activity in adulthood.136138 Several studies have documented associations between early life psychosocial exposures and DNA methylation levels of inflammatory genes.139141 Collectively, these data support the notion that early environmental exposures might shape adult inflammatory phenotypes through epigenetic mechanisms.

Several environmental exposures have been implicated in the development of SSc; these include silica, organic solvents, epoxy resins, welding fumes, pesticides, silicone breast implants, viruses, and drugs.142,143 Despite the paucity of studies investigating the effects of many of these exposures on the epigenome, there is growing evidence that environmental factors have a crucial impact on both alterations and modulation of epigenetic determinants, resulting in SSc onset and progression. 143 It has been well established that there is a marked correlation between SSc onset and occupational exposure to crystalline silica. 143 Although the interactions between silica exposure and the epigenome have not been assessed in SSc, an investigation of associations between silica particles and SSc-associated genetic variants in a fibroblast model unveiled a genetic variant in TNF alpha induced protein 3 (TNFAIP3) to be associated with silica-induced profibrotic response of fibroblasts. 144 Interestingly, the variant is thought to affect the binding activities of the transcription factors for TNFAIP3, 144 underscoring the role of regulatory variation on mediating environmental effects on phenotypes.

Organic solvents also have established associations with SSc onset. These are compounds commonly used in dry cleaning (tetrachloroethylene), paint thinners (toluene), nail polish removers and glue solvents (acetone, methyl acetate, ethyl acetate), and spot removers. 143 The effects of organic solvents on the epigenome remains unclear. One study reported that exposure to trichloroethylene resulted in DNA hypermethylation on rat cardiac myoblasts. 143

Exposure to heavy metals is also associated with SSc, including antimony, cadmium, lead, and mercury. Cadmium, lead, and mercury exposure have all been associated with DNA methylation changes, and cadmium and mercury with changes in miRNA profiles. 120 Interestingly, collagen type I alpha 2 chain (COL1A2) promoter DNA hypomethylation has been associated with high exposure to lead. 145

Viral infections (Epstein-Barr virus (EBV), hepatitis B virus (HBV)) are known to induce epigenetic changes that involve DNA methylation and histone modifications, reprogramming the infected host cells in ways that persist from one generation to the next and leave long-lasting phenotypes.146,147 Clearly, the deregulation of gene expression is a phenotypic consequence of epigenome plasticity modulated by the virus. Infections have long been speculated to play a role in SSc onset, as several mechanisms, such as endothelial cell damage, molecular mimicry, and self-reactive antibodies, could explain disease etiology. 143 It is not known if the pathogens implicated in SSc (parvovirus B19, cytomegalovirus, Helicobacter pylori ((H. pylori)) cause epigenetically driven changes in gene expression that affect the cellular dysfunction seen in SSc. 143

Pesticides and BPA, a carbon-based synthetic compound employed to make, among other plastics, the epoxy resins that line metal food and drink cans, are well documented endocrine disruptors that alter DNA methylation levels. 120 Future studies should investigate if the apparently increased risk of SSc after exposure to epoxy resins and pesticides is mediated by epigenetic changes. Similarly, although the extent to which particulate air pollution increases susceptibility to SSc requires further assessment, 143 the effects of air pollutants on the epigenome of SSc patients ought to be evaluated.

Several case reports have described the development of SSc or SSc-like manifestations after exposure to several drugs, namely some chemotherapy agents (bleomycin, taxanes, gemcitabin, tegafur-uracil, IFNα, and aldesleukin), appetite suppressants, and tryptophan, but there is insufficient evidence to establish a causative role on the development of SSc. 148 Nevertheless, chemotherapy drugs such as bleomycin or paclitaxel are considered to be associated with the induction of SSc, 149 although their roles as epigenetic modifiers have not been investigated. Doxorubicin, gemcitabine, and temozolomide have been found to induce epigenetic changes. 120 Furthermore, aberrant DNA methylation patterns have been found in the sperm of patients treated with temozolomide, raising caution about the potential transmission of epigenetic alterations to offspring of individuals treated with chemotherapeutic agents. 120

Prospects for epigenetic editing

Since the epigenome exerts a pivotal role during immune cell development and modulates functional immune programs during autoimmune disease, and epigenetic modifications can be reversed or modified, they offer an opportunity for the development of targeted therapies. 85 Several Food and Drug Administration (FDA)-approved drugs alter DNA methylation and histone acetylation. The epigenetic compounds investigated in the context of SSc have been recently reviewed. 5 However, a limitation of the current epigenetic-targeting approaches (e.g. DNMT, HDAC, or HAT inhibitors, or miRNA mimics) is their lack of specificity, as they affect epigenetic marks at many genomic sites indiscriminately. The most commonly used compounds are the DNMT inhibitors 5-azacytidine and 5-aza-2’-deoxycytidine (collectively known as 5-aza). As reviewed,5,6 treatment of fibroblasts with 5-aza reduces the expression of alpha-smooth muscle actin, collagen genes, and has potent antifibrotic effects.

An alternative to these global pharmacological compounds is the recently developed CRISPR (clustered regulatory interspaced short palindromic repeat)/Cas9-based RNA-guided DNA endonuclease technology. This gene editing technology has been retooled for epigenome editing, including the potential to modulate epigenetic marks and gene expression, manipulate nuclear architecture and chromatin loops, and visualize chromosome organization and dynamics via chromosome imaging. 150 Use of epigenome editing technologies for cell programming holds unprecedented potential as a therapeutic approach for durable regulation of disease-related genes and in cellular reprogramming. 151 Notably, Farhang et al. 152 used CRISPR-based epigenome editing to repress inflammatory cytokine cell receptors, specifically TNFR1 and IL1R1, which mitigated the downstream activation of NF-κB in the presence of TNF-α or IL-1β. The authors thus demonstrated that modulating cell response to inflammatory signaling can be used in engineering cells delivered to inflammatory environments, and as a direct gene therapy to protect endogenous cells. This indicates that deregulated loci identified in SSc might become the targets of epigenome editing to inhibit cell-specific dysregulation in SSc. Targeted regulation of deregulated loci using epigenome editing can thus be a potential therapeutic avenue to control fibrosis and autoimmunity in SSc.151,153

Despite the role of genetic factors and environmental exposures in SSc, an understanding of their effects on gene regulation is lacking. To characterize genetic and environmental risk factors for SSc, it is critical to investigate gene regulation in a relevant cell type under controlled treatment conditions. A tractable way to study genetic and environmental effects on gene regulation and cellular phenotypes is to integrate genetic, epigenetic, and expression data in cells exposed to environmental perturbations, such as the addition of a drug. In SSc, one study has investigated the interactions between genetic and environmental factors (silica particles) in SSc fibroblasts, revealing a genetic variant associated with the silica-induced profibrotic response of fibroblasts. 144 Epigenome editing might also be used to dissect the regulatory mechanisms involved in disease etiology, as genetic and epigenetic variants can be incorporated into human induced pluripotent stem cells (iPSCs) via genome editing to characterize phenotypic differences after differentiation, under different environmental conditions. Future studies aimed at identifying active regulatory elements for gene expression response to specific environmental perturbations are needed to advance our understanding of SSc risk. Results from these mechanistic studies integrating genetic, epigenetic, transcriptomic, and exposure data are likely to inform the design of epigenetic therapies to mitigate autoimmunity and fibrosis in SSc.

Conclusion

Multiple epigenetic modifications continue to be implicated in SSc pathogenesis. Similarly, genetic risk loci continue to be identified, but molecular mechanisms linking each variant to the disease phenotype are largely unknown. Multiple environmental exposures are associated with SSc, but only a few have been associated with epigenetic changes. Epigenetic modifications provide a mechanistic link between a cell’s genotype and their functional responses to environmental exposures. However, the lack of experimentally validated regulatory mechanisms is hampering the understanding of the regulatory mechanisms underlying SSc. Studies integrating genetic, epigenetic, and gene expression data, in relevant cell subsets, under different exposures, coupled with new epigenetic editing technologies, will provide a better understanding of the regulatory mechanisms underpinning SSc. This knowledge will ultimately inform the design of targeted epigenetic therapies for SSc.

Footnotes

Declaration of conflicting interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Funding: The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This work was supported by the US National Institute of Arthritis and Musculoskeletal and Skin Diseases of the National Institutes of Health (NIH) (grant number K01 AR067280). The content is solely the responsibility of the author and does not necessarily represent the official views of the NIH.

ORCID iD: Paula S. Ramos Inline graphic https://orcid.org/0000-0002-9433-6634

References

  • 1. Varga J, Abraham D. Systemic sclerosis: a prototypic multisystem fibrotic disorder. J Clin Invest 2007; 117(3): 557–567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Mahoney JM, Taroni J, Martyanov V, et al. Systems level analysis of systemic sclerosis shows a network of immune and profibrotic pathways connected with genetic polymorphisms. PLoS Comput Biol 2015; 11: e1004005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Ramos PS, Silver RM, Feghali-Bostwick CA. Genetics of systemic sclerosis: recent advances. Curr Opin Rheumatol 2015; 27: 521–529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Morgan ND, Shah AA, Mayes MD, et al. Clinical and serological features of systemic sclerosis in a multicenter African American cohort: analysis of the genome research in African American scleroderma patients clinical database. Medicine 2017; 96(51): e8980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Angiolilli C, Marut W, van der Kroef M, et al. New insights into the genetics and epigenetics of systemic sclerosis. Nat Rev Rheumatol 2018; 14(11): 657–673. [DOI] [PubMed] [Google Scholar]
  • 6. Tsou PS, Sawalha AH. Unfolding the pathogenesis of scleroderma through genomics and epigenomics. J Autoimmun 2017; 83: 73–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. O’Reilly S. Epigenetic modulation as a therapy in systemic sclerosis. Rheumatology (Oxford) 2019; 58: 191–196. [DOI] [PubMed] [Google Scholar]
  • 8. Aslani S, Sobhani S, Gharibdoost F, et al. Epigenetics and pathogenesis of systemic sclerosis; the ins and outs. Hum Immunol 2018; 79: 178–187. [DOI] [PubMed] [Google Scholar]
  • 9. Ballestar E, Li T. New insights into the epigenetics of inflammatory rheumatic diseases. Nat Rev Rheumatol 2017; 13(10): 593–605. [DOI] [PubMed] [Google Scholar]
  • 10. Kondilis-Mangum HD, Wade PA. Epigenetics and the adaptive immune response. Mol Aspects Med 2013; 34(4): 813–825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Schubeler D. Function and information content of DNA methylation. Nature 2015; 517(7534): 321–326. [DOI] [PubMed] [Google Scholar]
  • 12. Pai AA, Pritchard JK, Gilad Y. The genetic and mechanistic basis for variation in gene regulation. PLoS Genet 2015; 11(1): e1004857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Gutierrez-Arcelus M, Lappalainen T, Montgomery SB, et al. Passive and active DNA methylation and the interplay with genetic variation in gene regulation. eLife 2013; 2: e00523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Pacis A, Tailleux L, Morin AM, et al. Bacterial infection remodels the DNA methylation landscape of human dendritic cells. Genome Res 2015; 25(12): 1801–1811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Gutierrez-Arcelus M, Ongen H, Lappalainen T, et al. Tissue-specific effects of genetic and epigenetic variation on gene regulation and splicing. PLoS Genet 2015; 11: e1004958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Ambrosi C, Manzo M, Baubec T. Dynamics and Context-Dependent Roles of DNA Methylation. J Mol Biol 2017; 429(10): 1459–1475. [DOI] [PubMed] [Google Scholar]
  • 17. Teschendorff AE, Relton CL. Statistical and integrative system-level analysis of DNA methylation data. Nat Rev Genet 2018; 19(3): 129–147. [DOI] [PubMed] [Google Scholar]
  • 18. Wang Y, Fan PS, Kahaleh B. Association between enhanced type I collagen expression and epigenetic repression of the FLI1 gene in scleroderma fibroblasts. Arthritis Rheum 2006; 54(7): 2271–2279. [DOI] [PubMed] [Google Scholar]
  • 19. Altorok N, Almeshal N, Wang Y, et al. Epigenetics, the holy grail in the pathogenesis of systemic sclerosis. Rheumatology (Oxford, England) 2015; 54: 1759–1770. [DOI] [PubMed] [Google Scholar]
  • 20. Ding W, Pu W, Wang L, et al. Genome-wide DNA methylation analysis in systemic sclerosis reveals hypomethylation of IFN-associated genes in CD4(+) and CD8(+) T Cells. J Invest Dermatol 2018; 138(5): 1069–1077. [DOI] [PubMed] [Google Scholar]
  • 21. Evans IC, Barnes JL, Garner IM, et al. Epigenetic regulation of cyclooxygenase-2 by methylation of c8orf4 in pulmonary fibrosis. Clin Sci (Lond) 2016; 130(8): 575–586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Lei W, Luo Y, Lei W, et al. Abnormal DNA methylation in CD4+ T cells from patients with systemic lupus erythematosus, systemic sclerosis, and dermatomyositis. Scand J Rheumatol 2009; 38(5): 369–374. [DOI] [PubMed] [Google Scholar]
  • 23. Qi Q, Guo Q, Tan G, et al. Predictors of the scleroderma phenotype in fibroblasts from systemic sclerosis patients. Eur Acad Dermatol Venereol 2009; 23(2): 160–168. [DOI] [PubMed] [Google Scholar]
  • 24. Hattori M, Yokoyama Y, Hattori T, et al. Global DNA hypomethylation and hypoxia-induced expression of the ten eleven translocation (TET) family, TET1, in scleroderma fibroblasts. Exp Dermatol 2015; 24: 841–846. [DOI] [PubMed] [Google Scholar]
  • 25. O’Reilly S, Ciechomska M, Fullard N, et al. IL-13 mediates collagen deposition via STAT6 and microRNA-135b: a role for epigenetics. Scientific Reports 2016; 6: 25066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Matatiele P, Tikly M, Tarr G, et al. DNA methylation similarities in genes of black South Africans with systemic lupus erythematosus and systemic sclerosis. J Biomed Sci 2015; 22: 34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Almanzar G, Klein M, Schmalzing M, et al. Disease manifestation and inflammatory activity as modulators of Th17/Treg balance and RORC/FoxP3 methylation in systemic sclerosis. Int Arch Allergy Immunol 2016; 171: 141–154. [DOI] [PubMed] [Google Scholar]
  • 28. Lian X, Xiao R, Hu X, 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): 2338–2345. [DOI] [PubMed] [Google Scholar]
  • 29. Jiang H, Xiao R, Lian X, 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. [DOI] [PubMed] [Google Scholar]
  • 30. Wang Y, Shu Y, Xiao Y, et al. Hypomethylation and overexpression of ITGAL (CD11a) in CD4(+) T cells in systemic sclerosis. Clin Epigenetics 2014; 6(1): 25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Wang YY, Wang Q, Sun XH, et al. DNA hypermethylation of the forkhead box protein 3 (FOXP3) promoter in CD4+ T cells of patients with systemic sclerosis. Br J Dermatol 2014; 171(1): 39–47. [DOI] [PubMed] [Google Scholar]
  • 32. Wang Y, Kahaleh B. Epigenetic repression of bone morphogenetic protein receptor II expression in scleroderma. J Cell Mol Med 2013; 17(10): 1291–1299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Noda S, Asano Y, Nishimura S, et al. Simultaneous downregulation of KLF5 and Fli1 is a key feature underlying systemic sclerosis. Nat Commun 2014; 5: 5797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Dees C, Schlottmann I, Funke R, et al. The Wnt antagonists DKK1 and SFRP1 are downregulated by promoter hypermethylation in systemic sclerosis. Ann Rheum Dis 2014; 73(6): 1232–1239. [DOI] [PubMed] [Google Scholar]
  • 35. Selmi C, Feghali-Bostwick CA, Lleo A, et al. X chromosome gene methylation in peripheral lymphocytes from monozygotic twins discordant for scleroderma. Clin Exp Immunol 2012; 169: 253–262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Kim HP, Leonard WJ. CREB/ATF-dependent T cell receptor-induced FoxP3 gene expression: a role for DNA methylation. J Exp Med 2007; 204(7): 1543–1551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Bergmann C, Distler JH. Epigenetic factors as drivers of fibrosis in systemic sclerosis. Epigenomics 2017; 9(4): 463–477. [DOI] [PubMed] [Google Scholar]
  • 38. Gyftaki-Venieri DA, Abraham DJ, Ponticos M. Insights into myofibroblasts and their activation in scleroderma: opportunities for therapy? Curr Opin Rheumatol 2018; 30(6): 581–587. [DOI] [PubMed] [Google Scholar]
  • 39. Mann J, Chu DC, Maxwell A, et al. MeCP2 controls an epigenetic pathway that promotes myofibroblast transdifferentiation and fibrosis. Gastroenterology 2010; 138(2): 705–714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. He Y, Tsou PS, Khanna D, et al. Methyl-CpG-binding protein 2 mediates antifibrotic effects in scleroderma fibroblasts. Ann Rheum Dis 2018; 77(8): 1208–1218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Ostuni R, Piccolo V, Barozzi I, et al. Latent enhancers activated by stimulation in differentiated cells. Cell 2013; 152(1–2): 157–171. [DOI] [PubMed] [Google Scholar]
  • 42. Bhattacharyya S, Ghosh AK, Pannu J, et al. Fibroblast expression of the coactivator p300 governs the intensity of profibrotic response to transforming growth factor beta. Arthritis Rheum 2005; 52(4): 1248–1258. [DOI] [PubMed] [Google Scholar]
  • 43. Wei J, Ghosh AK, Chu H, et al. The histone deacetylase sirtuin 1 Is reduced in systemic sclerosis and abrogates fibrotic responses by targeting transforming growth factor beta signaling. Arthritis Rheumatol 2015; 67(5): 1323–1334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Zerr P, Palumbo-Zerr K, Huang J, et al. Sirt1 regulates canonical TGF-beta signalling to control fibroblast activation and tissue fibrosis. Ann Rheum Dis 2016; 75(1): 226–233. [DOI] [PubMed] [Google Scholar]
  • 45. Wang Y, Yang Y, Luo Y, et al. Aberrant histone modification in peripheral blood B cells from patients with systemic sclerosis. Clin Immunol 2013; 149(1): 46–54. [DOI] [PubMed] [Google Scholar]
  • 46. Tsou PS, Wren JD, Amin MA, et al. Histone Deacetylase 5 Is Overexpressed in Scleroderma Endothelial Cells and Impairs Angiogenesis via Repression of Proangiogenic Factors. Arthritis & Rheumatology 2016; 68(12): 2975–2985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Ghosh AK, Bhattacharyya S, Lafyatis R, et al. p300 is elevated in systemic sclerosis and its expression is positively regulated by TGF-beta: epigenetic feed-forward amplification of fibrosis. J Invest Dermatol 2013; 133(5): 1302–1310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Wang Q, Xiao Y, Shi Y, 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): 396–399. [DOI] [PubMed] [Google Scholar]
  • 49. Kramer M, Dees C, Huang J, et al. Inhibition of H3K27 histone trimethylation activates fibroblasts and induces fibrosis. Ann Rheum Dis 2013; 72(4): 614–620. [DOI] [PubMed] [Google Scholar]
  • 50. Palumbo-Zerr K, Zerr P, Distler A, et al. Orphan nuclear receptor NR4A1 regulates transforming growth factor-beta signaling and fibrosis. Nat Med 2015; 21(2): 150–158. [DOI] [PubMed] [Google Scholar]
  • 51. Svegliati S, Marrone G, Pezone A, 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(341): ra84. [DOI] [PubMed] [Google Scholar]
  • 52. Bergmann C, Brandt A, Merlevede B, et al. The histone demethylase Jumonji domain-containing protein 3 (JMJD3) regulates fibroblast activation in systemic sclerosis. Ann Rheum Dis 2018; 77(1): 150–158. [DOI] [PubMed] [Google Scholar]
  • 53. Ghosh AK, Varga J. The transcriptional coactivator and acetyltransferase p300 in fibroblast biology and fibrosis. Journal of Cellular Physiology 2007; 213(3): 663–671. [DOI] [PubMed] [Google Scholar]
  • 54. Tsou PS, Campbell P, Amin MA, et al. Inhibition of EZH2 prevents fibrosis and restores normal angiogenesis in scleroderma. Proc Natl Acad Sci U S A 2019; 116(9): 3695–3702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Huang B, Jiang C, Zhang R. Epigenetics: the language of the cell? Epigenomics 2014; 6(1): 73–88. [DOI] [PubMed] [Google Scholar]
  • 56. Rossato M, Affandi AJ, Thordardottir S, et al. Association of MicroRNA-618 expression with altered frequency and activation of plasmacytoid dendritic cells in patients with systemic sclerosis. Arthritis Rheumatol 2017; 69(9): 1891–1902. [DOI] [PubMed] [Google Scholar]
  • 57. Kajihara I, Jinnin M, Yamane K, et al. Increased accumulation of extracellular thrombospondin-2 due to low degradation activity stimulates type I collagen expression in scleroderma fibroblasts. Am J Pathol 2012; 180: 703–714. [DOI] [PubMed] [Google Scholar]
  • 58. Etoh M, Jinnin M, Makino K, et al. microRNA-7 down-regulation mediates excessive collagen expression in localized scleroderma. Arch Dermatol Res 2013; 305(1): 9–15. [DOI] [PubMed] [Google Scholar]
  • 59. Zhu H, Li Y, Qu S, et al. MicroRNA expression abnormalities in limited cutaneous scleroderma and diffuse cutaneous scleroderma. J Clin Immunol 2012; 32(3): 514–522. [DOI] [PubMed] [Google Scholar]
  • 60. Zhu H, Luo H, Li Y, et al. MicroRNA-21 in scleroderma fibrosis and its function in TGF-beta-regulated fibrosis-related genes expression. J Clin Immunol 2013; 33(6): 1100–1109. [DOI] [PubMed] [Google Scholar]
  • 61. Cui H, Banerjee S, Xie N, et al. MicroRNA-27a-3p is a negative regulator of lung fibrosis by targeting myofibroblast differentiation. Am J Respir Cell Mol Biol 2016; 54(6): 843–852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Maurer B, Stanczyk J, Jungel A, et al. MicroRNA-29, a key regulator of collagen expression in systemic sclerosis. Arthritis Rheum 2010; 62(6): 1733–1743. [DOI] [PubMed] [Google Scholar]
  • 63. Ciechomska M, O’Reilly S, Suwara M, et al. MiR-29a reduces TIMP-1 production by dermal fibroblasts via targeting TGF-beta activated kinase 1 binding protein 1, implications for systemic sclerosis. PLoS One 2014; 9(12): e115596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Takemoto R, Jinnin M, Wang Z, et al. Hair miR-29a levels are decreased in patients with scleroderma. Exp Dermatol 2013; 22(12): 832–833. [DOI] [PubMed] [Google Scholar]
  • 65. Jafarinejad-Farsangi S, Farazmand A, Mahmoudi M, et al. MicroRNA-29a induces apoptosis via increasing the Bax:Bcl-2 ratio in dermal fibroblasts of patients with systemic sclerosis. Autoimmunity 2015; 48(6): 369–378. [DOI] [PubMed] [Google Scholar]
  • 66. Alsaleh G, Francois A, Philippe L, et al. MiR-30a-3p negatively regulates BAFF synthesis in systemic sclerosis and rheumatoid arthritis fibroblasts. PLoS One 2014; 9(10): e111266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Tanaka S, Suto A, Ikeda K, et al. Alteration of circulating miRNAs in SSc: miR-30b regulates the expression of PDGF receptor beta. Rheumatology (Oxford) 2013; 52(11): 1963–1972. [DOI] [PubMed] [Google Scholar]
  • 68. Sing T, Jinnin M, Yamane K, et al. microRNA-92a expression in the sera and dermal fibroblasts increases in patients with scleroderma. Rheumatology (Oxford) 2012; 51(9): 1550–1556. [DOI] [PubMed] [Google Scholar]
  • 69. Nakashima T, Jinnin M, Yamane K, et al. Impaired IL-17 signaling pathway contributes to the increased collagen expression in scleroderma fibroblasts. J Immunol 2012; 188: 3573–3583. [DOI] [PubMed] [Google Scholar]
  • 70. Luo H, Zhu H, Zhou B, et al. MicroRNA-130b regulates scleroderma fibrosis by targeting peroxisome proliferator-activated receptor gamma. Mod Rheumatol 2015; 25(4): 595–602. [DOI] [PubMed] [Google Scholar]
  • 71. Honda N, Jinnin M, Kira-Etoh T, et al. miR-150 down-regulation contributes to the constitutive type I collagen overexpression in scleroderma dermal fibroblasts via the induction of integrin beta3. Am J Pathol 2013; 182(1): 206–216. [DOI] [PubMed] [Google Scholar]
  • 72. Yan Q, Chen J, Li W, et al. Targeting miR-155 to treat experimental scleroderma. Scientific Reports 2016; 6: 20314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Iwamoto N, Vettori S, Maurer B, et al. Downregulation of miR-193b in systemic sclerosis regulates the proliferative vasculopathy by urokinase-type plasminogen activator expression. Ann Rheum Dis 2016; 75(1): 303–310. [DOI] [PubMed] [Google Scholar]
  • 74. Honda N, Jinnin M, Kajihara I, et al. TGF-beta-mediated downregulation of microRNA-196a contributes to the constitutive upregulated type I collagen expression in scleroderma dermal fibroblasts. J Immunol 2012; 188: 3323–3331. [DOI] [PubMed] [Google Scholar]
  • 75. Wang Z, Jinnin M, Kudo H, et al. Detection of hair-microRNAs as the novel potent biomarker: evaluation of the usefulness for the diagnosis of scleroderma. J Dermatol Sci 2013; 72(2): 134–141. [DOI] [PubMed] [Google Scholar]
  • 76. Makino K, Jinnin M, Aoi J, et al. Discoidin domain receptor 2-microRNA 196a-mediated negative feedback against excess type I collagen expression is impaired in scleroderma dermal fibroblasts. J Invest Dermatol 2013; 133(1): 110–119. [DOI] [PubMed] [Google Scholar]
  • 77. Makino T, Jinnin M, Etoh M, et al. Down-regulation of microRNA-196a in the sera and involved skin of localized scleroderma patients. Eur J Dermatol 2014; 24(4): 470–476. [DOI] [PubMed] [Google Scholar]
  • 78. Zhou B, Zhu H, Luo H, et al. MicroRNA-202-3p regulates scleroderma fibrosis by targeting matrix metalloproteinase 1. Biomed Pharmacother 2017; 87: 412–418. [DOI] [PubMed] [Google Scholar]
  • 79. Ciechomska M, Zarecki P, Merdas M, et al. The role of microRNA-5196 in the pathogenesis of systemic sclerosis. Eur J Clin Invest 2017; 47(8): 555–564. [DOI] [PubMed] [Google Scholar]
  • 80. Makino K, Jinnin M, Hirano A, et al. The downregulation of microRNA let-7a contributes to the excessive expression of type I collagen in systemic and localized scleroderma. J Immunol 2013; 190(8): 3905–3915. [DOI] [PubMed] [Google Scholar]
  • 81. Wang Z, Jinnin M, Nakamura K, et al. Long non-coding RNA TSIX is upregulated in scleroderma dermal fibroblasts and controls collagen mRNA stabilization. Exp Dermatol 2016; 25(2): 131–136. [DOI] [PubMed] [Google Scholar]
  • 82. Dolcino M, Pelosi A, Fiore PF, et al. Gene profiling in patients with systemic sclerosis reveals the presence of oncogenic gene signatures. Front Immunol 2018; 9: 449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. 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): 21–30. [PMC free article] [PubMed] [Google Scholar]
  • 84. Messemaker TC, Chadli L, Cai G, et al. Antisense long non-coding RNAs are deregulated in skin tissue of patients with systemic sclerosis. J Invest Dermatol 2018; 138(4): 826–835. [DOI] [PubMed] [Google Scholar]
  • 85. Morales-Nebreda L, McLafferty FS, Singer BD. DNA methylation as a transcriptional regulator of the immune system. Transl Res 2019; 204: 1–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Bhatt DM, Pandya-Jones A, Tong AJ, et al. Transcript dynamics of proinflammatory genes revealed by sequence analysis of subcellular RNA fractions. Cell 2012; 150(2): 279–290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Fanucchi S, Mhlanga MM. Lnc-ing trained immunity to chromatin architecture. Front Cell Dev Biol 2019; 7: 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Maurano MT, Humbert R, Rynes E, et al. Systematic localization of common disease-associated variation in regulatory DNA. Science 2012; 337(6099): 1190–1195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Farh KK, Marson A, Zhu J, et al. Genetic and epigenetic fine mapping of causal autoimmune disease variants. Nature 2015; 518: 337–343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Consortium EP. An integrated encyclopedia of DNA elements in the human genome. Nature 2012; 489(7414): 57–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Roadmap Epigenomics C, Kundaje A, Meuleman W, et al. Integrative analysis of 111 reference human epigenomes. Nature 2015; 518(7539): 317–330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Adams D, Altucci L, Antonarakis SE, et al. BLUEPRINT to decode the epigenetic signature written in blood. Nature Biotechnol 2012; 30(3): 224–226. [DOI] [PubMed] [Google Scholar]
  • 93. Wagner JR, Busche S, Ge B, et al. The relationship between DNA methylation, genetic and expression inter-individual variation in untransformed human fibroblasts. Genome Biol 2014; 15(2): R37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Hannon E, Spiers H, Viana J, et al. Methylation QTLs in the developing brain and their enrichment in schizophrenia risk loci. Nat Neurosci 2016; 19(1): 48–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Olsson AH, Volkov P, Bacos K, et al. Genome-wide associations between genetic and epigenetic variation influence mRNA expression and insulin secretion in human pancreatic islets. PLoS Genet 2014; 10(11): e1004735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Banovich NE, Lan X, McVicker G, et al. Methylation QTLs are associated with coordinated changes in transcription factor binding, histone modifications, and gene expression levels. PLoS Genet 2014; 10(9): e1004663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Bell JT, Pai AA, Pickrell JK, et al. DNA methylation patterns associate with genetic and gene expression variation in HapMap cell lines. Genome Biol 2011; 12(1): R10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. van Dongen J, Nivard MG, Willemsen G, et al. Genetic and environmental influences interact with age and sex in shaping the human methylome. Nat Commun 2016; 7: 11115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. McClay JL, Shabalin AA, Dozmorov MG, et al. High density methylation QTL analysis in human blood via next-generation sequencing of the methylated genomic DNA fraction. Genome Biol 2015; 16: 291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Richardson TG, Zheng J, Davey Smith G, et al. Mendelian randomization analysis identifies CpG sites as putative mediators for genetic influences on cardiovascular disease risk. Am J Hum Genet 2017; 101(4): 590–602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Hannon E, Weedon M, Bray N, et al. Pleiotropic effects of trait-associated genetic variation on DNA methylation: utility for refining GWAS Loci. Am J Hum Genet 2017; 100(6): 954–959. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Bell CG, Gao F, Yuan W, et al. Obligatory and facilitative allelic variation in the DNA methylome within common disease-associated loci. Nat Commun 2018; 9(1): 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Bonder MJ, Luijk R, Zhernakova DV, et al. Disease variants alter transcription factor levels and methylation of their binding sites. Nat Genet 2017; 49(1): 131–138. [DOI] [PubMed] [Google Scholar]
  • 104. Pickrell JK. Joint analysis of functional genomic data and genome-wide association studies of 18 human traits. Am J Hum Genet 2014; 94(4): 559–573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Schaub MA, Boyle AP, Kundaje A, et al. Linking disease associations with regulatory information in the human genome. Genome Res 2012; 22(9): 1748–1759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Michels KB, Binder AM, Dedeurwaerder S, et al. Recommendations for the design and analysis of epigenome-wide association studies. Nat Methods 2013; 10(10): 949–955. [DOI] [PubMed] [Google Scholar]
  • 107. Barfield RT, Almli LM, Kilaru V, et al. Accounting for population stratification in DNA methylation studies. Genet Epidemiol 2014; 38(3): 231–241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Galanter JM, Gignoux CR, Oh SS, et al. Differential methylation between ethnic sub-groups reflects the effect of genetic ancestry and environmental exposures. eLife 2017; 6: e20532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Husquin LT, Rotival M, Fagny M, et al. Exploring the genetic basis of human population differences in DNA methylation and their causal impact on immune gene regulation. Genome Biol 2018; 19(1): 222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Quach H, Rotival M, Pothlichet J, et al. Genetic adaptation and neandertal admixture shaped the immune system of human populations. Cell 2016; 167(3): 643–656.e617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Gopalan S, Carja O, Fagny M, et al. Trends in DNA methylation with age replicate across diverse human populations. Genetics 2017; 206(3): 1659–1674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Fagny M, Patin E, MacIsaac JL, et al. The epigenomic landscape of African rainforest hunter-gatherers and farmers. Nat Commun 2015; 6: 10047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Heyn H, Moran S, Hernando-Herraez I, et al. DNA methylation contributes to natural human variation. Genome Res 2013; 23(9): 1363–1372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Lee R, Reese C, Perry B, et al. Enhanced chemokine-receptor expression, function, and signaling in healthy African American and scleroderma-patient monocytes are regulated by caveolin-1. Fibrogenesis Tissue Repair 2015; 8: 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Reese C, Perry B, Heywood J, et al. Caveolin-1 deficiency may predispose African Americans to systemic sclerosis-related interstitial lung disease. Arthritis Rheumatol 2014; 66(7): 1909–1919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Nedelec Y, Sanz J, Baharian G, et al. Genetic ancestry and natural selection drive population differences in Immune Responses to Pathogens. Cell 2016; 167(3): 657–669. [DOI] [PubMed] [Google Scholar]
  • 117. Horvath S. DNA methylation age of human tissues and cell types. Genome Biology 2013; 14: R115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Weidner CI, Lin Q, Koch CM, et al. Aging of blood can be tracked by DNA methylation changes at just three CpG sites. Genome Biol 2014; 15(2): R24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Wyman AE, Atamas SP. Sirtuins and accelerated aging in scleroderma. Curr Rheumatol Rep 2018; 20(4): 16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Torano EG, Garcia MG, Fernandez-Morera JL, et al. The impact of external factors on the epigenome: in utero and over Lifetime. Biomed Res Int 2016; 2016: 2568635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Heijmans BT, Tobi EW, Stein AD, et al. Persistent epigenetic differences associated with prenatal exposure to famine in humans. Proc Natl Acad Sci U S A 2008; 105(44): 17046–17049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Zacharasiewicz A. Maternal smoking in pregnancy and its influence on childhood asthma. ERJ Open Res 2016; 2(3): 00042–2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Banik A, Kandilya D, Ramya S, et al. Maternal factors that induce epigenetic changes contribute to neurological disorders in offspring. Genes (Basel) 2017; 8(6): 150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Kereliuk SM, Brawerman GM, Dolinsky VW. Maternal macronutrient consumption and the developmental origins of metabolic disease in the offspring. Int J Mol Sci 2017; 18(7): E1451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Marr AK, MacIsaac JL, Jiang R, et al. Leishmania donovani infection causes distinct epigenetic DNA methylation changes in host macrophages. PLoS Pathog 2014; 10(10): e1004419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Joubert BR, Haberg SE, Nilsen RM, et al. 450K epigenome-wide scan identifies differential DNA methylation in newborns related to maternal smoking during pregnancy. Environ Health Perspect 2012; 120(10):1425–1431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Joubert BR, Felix JF, Yousefi P, et al. DNA Methylation in newborns and maternal smoking in pregnancy: genome-wide consortium Meta-analysis. Am J Hum Genet 2016; 98: 680–696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Jiang R, Jones MJ, Sava F, et al. Short-term diesel exhaust inhalation in a controlled human crossover study is associated with changes in DNA methylation of circulating mononuclear cells in asthmatics. Part Fibre Toxicol 2014; 11: 71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Ho SM, Johnson A, Tarapore P, et al. Environmental epigenetics and its implication on disease risk and health outcomes. ILAR J 2012; 53(3–4): 289–305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. van der Knaap LJ, Riese H, Hudziak JJ, et al. Glucocorticoid receptor gene (NR3C1) methylation following stressful events between birth and adolescence. The TRAILS study. Transl Psychiatry 2014; 4: e381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Ressler KJ, Mercer KB, Bradley B, et al. Post-traumatic stress disorder is associated with PACAP and the PAC1 receptor. Nature 2011; 470(7335): 492–497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Chen W, Boutaoui N, Brehm JM, et al. ADCYAP1R1 and asthma in Puerto Rican children. Am J Respir Crit Care Med 2013; 187: 584–588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Lam LL, Emberly E, Fraser HB, et al. Factors underlying variable DNA methylation in a human community cohort. Proc Natl Acad Sci U S A 2012; 109(Suppl 2): 17253–17260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Borghol N, Suderman M, McArdle W, et al. Associations with early-life socio-economic position in adult DNA methylation. Int J Epidemiol 2012; 41: 62–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Vidal AC, Benjamin Neelon SE, Liu Y, et al. Maternal stress, preterm birth, and DNA methylation at imprint regulatory sequences in humans. Genet Epigenet 2014; 6: 37–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Danese A, Pariante CM, Caspi A, et al. Childhood maltreatment predicts adult inflammation in a life-course study. Proc Natl Acad Sci U S A 2007; 104(4): 1319–1324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. McDade TW, Hoke M, Borja JB, et al. Do environments in infancy moderate the association between stress and inflammation in adulthood? Initial evidence from a birth cohort in the Philippines. Brain Behav Immun 2013; 31: 23–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Taylor SE, Lehman BJ, Kiefe CI, et al. Relationship of early life stress and psychological functioning to adult C-reactive protein in the coronary artery risk development in young adults study. Biol Psychiatry 2006; 60(8): 819–824. [DOI] [PubMed] [Google Scholar]
  • 139. Needham BL, Smith JA, Zhao W, et al. Life course socioeconomic status and DNA methylation in genes related to stress reactivity and inflammation: The multi-ethnic study of atherosclerosis. Epigenetics 2015; 10(10): 958–969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Esposito EA, Jones MJ, Doom JR, et al. Differential DNA methylation in peripheral blood mononuclear cells in adolescents exposed to significant early but not later childhood adversity. Dev Psychopathol 2016; 28: 1385–1399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. McDade TW, Ryan C, Jones MJ, et al. Social and physical environments early in development predict DNA methylation of inflammatory genes in young adulthood. Proc Natl Acad Sci U S A 2017; 114(29): 7611–7616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Rubio-Rivas M, Moreno R, Corbella X. Occupational and environmental scleroderma. Systematic review and meta-analysis. Clin Rheumatol 2017; 36: 569–582. [DOI] [PubMed] [Google Scholar]
  • 143. Marie I. Systemic sclerosis and exposure to heavy metals. Autoimmun Rev 2019; 18: 62–72. [DOI] [PubMed] [Google Scholar]
  • 144. Wei P, Yang Y, Guo X, et al. Identification of an association of TNFAIP3 polymorphisms with matrix metalloproteinase expression in fibroblasts in an integrative study of systemic sclerosis-associated genetic and environmental factors. Arthritis Rheumatol 2016; 68(3): 749–760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Hanna CW, Bloom MS, Robinson WP, et al. DNA methylation changes in whole blood is associated with exposure to the environmental contaminants, mercury, lead, cadmium and bisphenol A, in women undergoing ovarian stimulation for IVF. Hum Reprod 2012; 27(5): 1401–1410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Scott RS. Epstein-Barr virus: a master epigenetic manipulator. Curr Opin Virol 2017; 26: 74–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Hensel KO, Rendon JC, Navas MC, et al. Virus-host interplay in hepatitis B virus infection and epigenetic treatment strategies. FEBS J 2017; 284(21): 3550–3572. [DOI] [PubMed] [Google Scholar]
  • 148. Marie I, Gehanno JF. Environmental risk factors of systemic sclerosis. Seminars in Immunopathology 2015; 37: 463–473. [DOI] [PubMed] [Google Scholar]
  • 149. Niklas K, Niklas AA, Majewski D, et al. Rheumatic diseases induced by drugs and environmental factors: the state-of-the-art - part two. Reumatologia 2016; 54(4): 165–169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Lau CH, Suh Y. In vivo epigenome editing and transcriptional modulation using CRISPR technology. Transgenic Res 2018; 27: 489–509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Thakore PI, Black JB, Hilton IB, et al. Editing the epigenome: technologies for programmable transcription and epigenetic modulation. Nat Methods 2016; 13(2): 127–137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Farhang N, Brunger JM, Stover JD, et al. (*) CRISPR-based epigenome editing of cytokine receptors for the promotion of cell survival and tissue deposition in inflammatory environments. Tissue Eng Part A 2017; 23(15–16): 738–749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Koeferle A, Stricker SH, Beck S. Brave new epigenomes: the dawn of epigenetic engineering. Genome Medicine 2015; 7: 59. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of Scleroderma and Related Disorders are provided here courtesy of World Scleroderma Foundation, EUSTAR, and SAGE Publications

RESOURCES