Skip to main content
  • Review
  • Published:

Dissecting complex epigenetic alterations in human lupus

Abstract

Systemic lupus erythematosus is a chronic relapsing autoimmune disease that primarilyafflicts women, and both a genetic predisposition and appropriate environmentalexposures are required for lupus to develop and flare. The genetic requirement isevidenced by an increased concordance in identical twins and by the validation of atleast 35 single-nucleotide polymorphisms predisposing patients to lupus. Genes alone,though, are not enough. The concordance of lupus in identical twins is oftenincomplete, and when concordant, the age of onset is usually different. Lupus is alsonot present at birth, but once the disease develops, it typically follows a chronicrelapsing course. Thus, genes alone are insufficient to cause human lupus, andadditional factors encountered in the environment and over time are required toinitiate the disease and subsequent flares. The nature of the environmentalcontribution, though, and the mechanisms by which environmental agents modify theimmune response to cause lupus onset and flares in genetically predisposed peoplehave been controversial. Reports that the lupus-inducing drugs procainamide andhydralazine are epigenetic modifiers, that epigenetically modified T cells aresufficient to cause lupus-like autoimmunity in animal models, and that patients withactive lupus have epigenetic changes similar to those caused by procainamide andhydralazine have prompted a growing interest in how epigenetic alterations contributeto this disease. Understanding how epigenetic mechanisms modify T cells to contributeto lupus requires an understanding of how epigenetic mechanisms regulate geneexpression. The roles of DNA methylation, histone modifications, and microRNAs inlupus pathogenesis will be reviewed here.

Epigenetics and gene expression

Epigenetics is defined as heritable changes in gene expression that do not involve achange in the DNA sequence, and the mechanisms include DNA methylation, a variety ofcovalent histone modifications, and microRNAs (miRNAs). DNA is packaged in the nucleusas chromatin. Chromatin consists of DNA wrapped twice around a histone core to form anucleosome, and the nucleosomes are stacked into higher-ordered structures to form thechromatin fiber that makes each chromosome. The DNA in chromatin is tightly packaged andinaccessible to the protein complexes that initiate RNA transcription. DNA methylationand histone modifications regulate gene expression by modifying chromatin structure topermit or prevent access of the transcription complexes to the DNA (Figure 1). In contrast, miRNAs target mRNAs for degradation. All threemechanisms - DNA methylation, histone modifications, and miRNAs - are being explored inhuman lupus.

Figure 1
figure 1

DNA methylation, histone acetylation, and chromatin structure. DNA ispackaged as chromatin, the basic subunit of which is the nucleosome. Eachnucleosome consists of two turns of DNA wrapped around a core histone of histoneproteins, the tails of which protrude. Transcriptionally active chromatin ischaracterized by unmethylated DNA and acetylated (green triangles) histone tails.(a) The DNA is exposed and accessible to transcription factor binding.(b) Methylation of cytosine bases in the DNA (red dots) attractsmethylcytosine-binding proteins which in turn attract and tether chromatininactivation complexes containing histone deacetylases and other proteins. (c) These complexes deacetylate the histones and promote condensation of thechromatin into a compressed structure inaccessible to the transcription initiationcomplexes. DNMT, DNA methyltransferase; HAT, histone acetylase; HDAC, histonedeacetylase; MBD, methyl-CpG-binding domain; RNA-PII, RNA polymerase II; TF,transcription factor. Figure reprinted with permission from Michigan Creative.

DNA methylation

DNA methylation refers to the methylation of cytosines in CpG pairs and silences genesby stabilizing chromatin in the tightly packaged, transcriptionally repressiveconfiguration. DNA methylation patterns are established during development and serve inpart to silence genes which would be inappropriate or detrimental to the function of anygiven cell but for which a cell might have transcription factors that would otherwisedrive their expression. Different cell types have different functions, determined by therepertoire of genes they express, so each cell type has a distinct pattern of methylatedand unmethylated genes.

Once established, the methylation patterns are replicated each time a cell divides byDNA methyltransferase 1 (Dnmt1). As cells enter S phase, Dnmt1 levels increase. Dnmt1binds the DNA replication fork and reads CpG pairs. Where deoxycytosine (dC) in theparent strand is methylated, Dnmt1 transfers the methyl group from S-adenosylmethionine(SAM) to the corresponding dC in the daughter strand to form deoxymethylcytosine,replicating the methylation patterns and producing S-adenosylhomocysteine (SAH), aninhibitor of transmethylation reactions [1]. Importantly, this reaction is sensitive to environmental agents and drugsthat decrease Dnmt1 activity, decrease SAM, or increase SAH, preventing methylation ofnewly synthesized DNA in the daughter cells and causing inappropriate gene expression [1–3]. Furthermore, the errors can be replicated during subsequent rounds of celldivision and accumulate over time, causing an age-dependent decrease in DNA methylationand increase in aberrant T-cell gene expression [4]. These age-dependent changes are evidenced by a report that lymphocyte DNAmethylation patterns are the same in identical twins at 3 years of age but different at50. DNA methylation patterns also diverged more when twins had different lifestyles orspent less of their lives together, compared to twins who had similar lifestyles orspent more of their lives together [5].

T-cell DNA methylation and gene expression

T lymphocytes are particularly dependent on DNA methylation to suppress inappropriategene expression. T cells differentiate into multiple subsets throughout life but haveoverlapping sets of transcription factors and use DNA methylation to silence genesinappropriate for specific subsets. Like other cells, resting T cells expressrelatively little Dnmt1, but as T cells enter mitosis, signals through theextracellular-signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK)pathways upregulate Dnmt1 to replicate the methylation patterns; and decreasing Dnmt1enzymatic activity with inhibitors like 5-azacytidine (5-azaC) or procainamide, orpreventing Dnmt1 upregulation with signaling inhibitors like hydralazine, preventsthe methylation of the newly synthesized DNA, activating normally silenced genes andaltering effector functions in the daughter cells [1]. For example, inhibiting DNA methylation induces interferon-gamma(IFNγ) in Th2 cells, FoxP3 in CD4+CD25− T cells [6], and others [7].

Inhibiting DNA methylation also makes CD4+ T cells autoreactive to selfclass II major histocompatibility complex (MHC) molecules. The autoreactivity is dueto LFA-1 (lymphocyte function-associated antigen 1) (CD11a/CD18) overexpression, andincreasing LFA-1 by transfection makes T cells similarly autoreactive [1]. Importantly, CD4+ T cells similarly responding to class II MHCmolecules cause a lupus-like disease in the chronic graft-versus-host disease model [8], suggesting that demethylated, autoreactive T cells may cause a similarlupus-like disease. This was confirmed by treating mouse CD4+ T cells with5-azaC and then injecting the cells into genetically identical mice. The recipientsdeveloped antidsDNA antibodies and an immune complex glomerulonephritis closelyresembling human lupus nephritis [9].

The observation that 5-azaC, a drug that inhibits DNA methylation, can cause alupus-like disease suggested that drugs which cause a lupus-like disease may be DNAmethylation inhibitors. Subsequent studies demonstrated that CD4+ T cellstreated with procainamide or hydralazine, which cause antinuclear antibodies (ANAs)in a majority of people and a lupus-like disease in genetically predisposed people,are also DNA methylation inhibitors [10] and that, when injected into genetically identical mice, mouse T cellstreated with these drugs also caused a lupus-like disease [9]. Procainamide was found to inhibit Dnmt1 enzymatic activity, whereashydralazine decreases Dnmt1 levels in dividing cells by inhibiting ERK pathwaysignaling [1].

Functional studies demonstrated that experimentally demethylated, autoreactiveCD4+ T cells are cytotoxic and kill autologous or syngeneic macrophages(Mφ), causing release of apoptotic nuclear material as well as impairing itsclearance, since Mφ clear apoptotic debris [1]. Others have reported that injecting apoptotic cells into mice orimpairing their clearance through genetic manipulation is sufficient to causeanti-DNA antibodies and a lupus-like disease [11], suggesting that the Mφ apoptosis caused by demethylated T cellscontributes to anti-DNA antibody development. The demethylated CD4+ Tcells also overstimulate B-cell antibody production through demethylation andoverexpression of B-cell co-stimulatory genes, including CD70,CD40L, IFNγ, and others [1, 12, 13]. When injected into mice, the demethylated cells accumulate in the spleen,where they kill Mφ and stimulate B cells, and removing the spleen prior toinjection prevents disease development [14].

T-cell DNA demethylates in patients with active lupus

These studies demonstrate that epigenetically altered CD4+ T cells aresufficient to cause a lupus-like disease in mice. Since the two drugs that mostcommonly cause drug-induced lupus are DNA methylation inhibitors, these studies alsosuggest that epigenetically altered CD4+ T cells may similarly causeidiopathic lupus. This was initially tested by comparing overall DNA methylationlevels in T cells from patients with active lupus, inactive lupus, or other rheumaticand inflammatory diseases and healthy controls. Patients with active lupus had lowermethylcytosine levels than the other groups [15]. Interestingly, T-cell DNA methylation levels also decline with age [16], suggesting an explanation for the development of ANAs with age inotherwise healthy older people [17].

The decrease in lupus T-cell Dnmt1 levels was traced to decreased ERK pathwaysignaling, which prevents Dnmt1 upregulation during mitosis [1]. A role for decreased signaling in lupus pathogenesis was confirmed byexperiments demonstrating that inhibiting ERK pathway signaling with MEK inhibitorsalso inhibited DNA methylation in mouse CD4+ T cells, and injecting thetreated cells into syngeneic mice also caused a lupus-like disease [1]. More recently, a double-transgenic mouse strain was generated in whichexpression of a dominant negative MEK (dnMEK) could be selectively induced in T cellsby adding doxycycline to their drinking water. Activating the dnMEK inhibited T-cellDNA methylation and caused anti-DNA antibodies and an 'interferon signature' in mice,similarly to what was observed in patients with lupus [18]. Interestingly, no kidney disease was seen in these transgenic mice on aBL6 background. However, crossing the double-transgenic BL6 strain with SJL mice,which have lupus genes, resulted in the development of an immune complexglomerulonephritis as well as anti-DNA antibodies when doxycycline was given [19]. Thus, impaired T-cell ERK pathway signaling is sufficient to causelupus-like autoantibodies in nonlupus-prone mice, but renal disease also requireslupus genes.

Evidence that hydralazine inhibits ERK pathway signaling, that ERK pathway signalingis impaired in T cells from patients with active lupus, and that inhibiting ERKpathway signaling causes a lupus-like disease in adoptive transfer and transgenicmouse models [20] prompted studies identifying the signaling molecule(s) inactivated byhydralazine and inactivated in CD4+ T cells from patients with activelupus. The ERK pathway defect was traced to PKCδ, which fails to respond todirect stimulation with phorbol myristate acetate in both the idiopathic lupus andhydralazine-induced models [3]. Importantly, PKCδ 'knockout' mice develop lupus [20], demonstrating a critical role for PKCδ in lupus-likeautoimmunity.

More recent studies demonstrate that PKCδ is inactivated by oxidative damage inlupus T cells. Lupus onset and flares are associated with environmental agents thatcause oxidative stress, such as ultraviolet light exposure, acute infections, silicaexposure, and smoking [21], and all cause oxidative stress [22]. Furthermore, lupus flares are characterized by biomarkers of oxidativestress such as protein nitration, caused by superoxide(O2−) combining with nitric oxide (NO), anintracellular signaling molecule, to form peroxynitrite (ONOO−) [23]. T-cell PKCδ is nitrated in patients with active lupus, and thenitrated fraction is catalytically inactive [3], providing a direct link between environmental agents associated withlupus and epigenetic changes in T cells.

Lupus T-cell epigenomics

The observation that experimentally demethylated CD4+ T cells overexpressLFA-1 due to ITGAL (CD11a) demethylation, making them autoreactive [1], raised the possibility that other genes may similarly demethylate and beinappropriately overexpressed by T cells from patients with active lupus. Additionalgenes were sought by treating normal human CD4+ T cells with 5-azaC andcomparing gene expression with mRNA expression arrays. These experiments identifiedCD70 (TNFSF7), perforin (PRF1), and the KIR gene family asgenes primarily regulated by DNA methylation in CD4+ T cells. CD70 isexpressed on some but not all CD4+ T cells and promotes B-cell antibodyproduction [1]. Perforin is a cytotoxic molecule expressed in killer cells and lysestarget cells by forming a pore in their cytoplasmic membrane [1]. The KIR genes normally are expressed by natural killer (NK) cells but notT cells and encode proteins that recognize I MHC molecules. Stimulatory Kir proteinsmediate cytotoxic and inflammatory responses. Aberrant expression of KIR genes bydemethylated T cells makes them likely candidates for contributing to autoimmuneresponses [24].

The DNA sequences demethylated to activate these genes were identified by bisulfitesequencing. Bisulfite converts cytosine to uracil but does not affect methylcytosine,and the change can be detected by DNA sequencing. Bisulfite sequencing revealed thatregulatory sequences upstream of the CD11a, CD70, KIR, and perforin genes alldemethylate in 5-azaC- treated CD4+ T cells, and methylation of theseregions in transfection experiments silenced these genes [1, 25].

Subsequent experiments compared expression and methylation of the same genes inCD4+ T cells from patients with either inactive or active lupus. Aspredicted, CD11a, perforin, and the KIR genes were overexpressed in patients withactive but not inactive lupus, and the same sequences demethylated in proportion todisease activity and gene overexpression in these patients [1, 24]. The exception was CD70, which remains demethylated once itdemethylates [1]. Others have reported that protein phosphatase 2A, a signaling molecule,is also demethylated and overexpressed in T cells from patients with active lupus [26]. Very recently, Jeffries and colleagues [27] used microarrays to survey hypomethylated and hypermethylated genes inCD4+ T cells from patients active lupus and identified 236hypomethylated and 105 hypermethylated sites, confirming widespread methylationchanges throughout the genome.

Together, these results demonstrate that inhibiting T-cell DNA methylation, eitherwith drugs in vitro or environmental agents in patients with lupus, causesthe same epigenetic changes in DNA methylation patterns and overexpression of thesame genes. Since similar demethylated T cells cause lupus-like autoimmunity in mice,these studies indicate that T-cell DNA demethylation is likely fundamental to lupusonset and flares.

Histone modifications

Histone modifications also regulate gene expression. Histone acetyltransferases (HATs)and histone deacetylases (HDACs) regulate gene expression by adding or removing acetylgroups on lysine residues in the histone proteins [28]. Acetylation neutralizes the positive charge of lysines, weakeningelectrostatic DNA-histone interactions and increasing DNA accessibility for geneexpression [29]. Conversely, deacetylation strengthens DNA-histone interactions, decreasingDNA accessibility and subsequent gene expression [30]. Promoters of actively transcribed genes are also characterized bymethylation of lysine 4 on histone H3 (H3K4) and of lysine 36 on H3 (H3K36) [31]. In contrast, inactive genes are methylated at H3K27 and permanently silencedgenes are frequently methylated at H3K9 [32]. A large number of other histone modifications serve a number of regulatoryand other functions (reviewed in [33]).

Lupus T-cell histone modifications

Histone acetylation may also contribute to lupus pathogenesis. Hu and colleagues [34] showed that SLE CD4+ T cells have decreased overall acetylationof histones H3 and H4, and the degree of H3 deacetylation correlated inversely withSLE disease activity. This raises the possibility that decreased histone acetylationcontributes to lupus pathogenesis by promoting silencing of some genes. However, thegenes affected are unclear.

Histone methylation patterns are also altered in CD4+ lupus T cells [35]. Hu and colleagues [34] also reported hypomethylation at lysine 9 of histone H3 (H3K9) in SLECD4+ T cells, compared with healthy controls. mRNA levels of thehistone methyltransferases SUV39H2 and EZH2 were also decreased in CD4+ SLE T cells [34]. Zhao and colleagues [36] subsequently reported that a different histone methyltransferase, SUV39H1,is recruited to the CD11a and CD70 promoters. This is mediated by RFX1, and RFX1 isdownregulated in lupus CD4+ T cells [36]. RFX1 recruitment to SUV39H1 causes both increased H3K9 methylation anddecreased CD11a and CD70 expression [35]. In a second report, Zhou and colleagues [37] showed increased H3K4 methylation at the CD70 promoter in lupus CD4+ T cells. Zhang and colleagues [38] showed that hematopoietic progenitor kinase 1 (HPK1) levels are decreasedin lupus CD4+ T cells. Furthermore, blocking HPK1 expression increasedT-cell proliferation, cytokine secretion, and B-cell co-stimulatory functions. Thesechanges reversed when HPK1 was overexpressed. The decreased HPK1 expression in lupusCD4+ T cells is due to decreased H3K27 methylation, and the decreasedmethylation is due to decreased binding of the enzyme jumonji domain containing 3(JMJD3), which methylates H3K27 [38].

MicroRNAs

MiRNAs are 18- to 22-nucleotide non-coding RNA molecules that regulate gene expressionby degrading mRNA or blocking protein translation. An miRNA binds to a target sequencein the 3' untranslated region of its target mRNA, leading to either degradation ortranslational silencing by other mechanisms. Multiple miRNAs can bind to and blockexpression of a target mRNA. Additionally, a single miRNA can bind to and block theexpression of multiple target mRNAs. These events are possible because the sequencesthat mediate binding between an miRNA and mRNA are short and thus recognizable by manymiRNAs [39].

Interestingly, histone modifications and DNA methylation also affect miRNAs in lupusCD4+ T cells. Ding and colleagues [40] showed that miR-142-3p and -5p are decreased in lupus CD4+ Tcells. This causes overexpression of SLAM-associated protein, IL-10, and CD84. H3K27methylation levels were increased in the putative miR-142 regulatory regions. The threeCpG pairs closest to the miR-142 transcription start site were also hypermethylated inlupus CD4+ T cells, but average methylation of 11 CpG pairs across themiR-142 regulatory region was not different in lupus CD4+ T cells [40].

Lupus T-cell miRNAs

Yu and colleagues [41] provided the first report of a possible role for miRNAs in autoimmunediseases. Sanroque mice, which have a defect in Roquin, develop a lupus-like syndromeattributed to increased ICOS (inducible co-stimulatory molecule) expression on Tcells. A sequence in the ICOS 3' untranslated region is required for Roquin to blockICOS expression, and this sequence is recognized by miR-101 [41]. MiR-101 does not completely explain Roquin's ability to block ICOSexpression, although other miRNAs could also be responsible.

Multiple miRNAs are differentially expressed in CD4+ T cells from patientswith lupus compared with healthy donors, and two recent reports link miRNAs and DNAmethylation. MiRNAs-21, -126, and -148a are upregulated in CD4+ lupus Tcells and decrease Dnmt1 expression [42, 43]. MiRNAs-126 and -148a decrease Dnmt1 directly. MiR-21 decreases RASGRP1,in the JNK signaling pathway, which leads to decreased Dnmt1 expression. BlockingmiR-21, 126, or 148a in lupus CD4+ T cells increases Dnmt1 levels anddecreases CD70 and CD11a levels. MiR-21 is also increased in CD4+ lupus Tcells [44], and blocking miR-21 expression decreases levels of themethylation-sensitive gene CD40L. Blocking miR-21 also decreases proliferation, IL-10expression, and B-cell maturation. Overexpressing miR-21 has opposing effects,including increased CD40L expression. A therapeutic potential for miRNAs is shown inthese cases since blocking their expression reverses lupus-like phenotypes.

Effects of miRNAs in total CD3+ T cells and PBMCs from patients with lupushave also been studied. For example, miR-31 and IL-2 levels are decreased in lupus Tcells. This effect is mediated by RhoA, which blocks IL-2 expression. MiR-31decreases RhoA, which increases IL-2 [45]. MiR-125a is also decreased, and RANTES (regulated and normal T cellexpressed and secreted) increased, in lupus T cells, and these effects are mediatedby interactions between miR-125a and KLF13, a regulator of RANTES expression [46]. Again, suppressing these miRNAs in lupus T cells reversed the defects inIL-2 and RANTES expression. MiR-146a is also downregulated in lupus PBMCs. MiR-146anegatively regulates IFN-α/β expression by targeting STAT-1 and IRF-5,suggesting a link between miR-146a and lupus pathogenesis [47].

Mouse models corroborate these findings. MiR-21 is increased in CD3+ Tcells from lupus-prone B6.Sle123 mice, and blocking miR-21 expression decreasessplenomegaly [48]. Suppressing miR-21 also increases PDCD4 expression. PDCD4 is decreased inCD4+ T cells from patients with lupus, and miR-21 suppresses PDCD4 whentransfected into normal CD4+ T cells [44].

Genetic/epigenetic interactions in lupus

As discussed above, lupus develops when genetically predisposed people encounterenvironmental agents that initiate disease onset and flares, and the environment appearsto trigger lupus onset and flares at least in part by modifying T-cell DNA methylation.The relationship between T-cell DNA demethylation and genetic predisposition is complexbecause both can vary. T-cell DNA demethylates in lupus, and the degree of DNAdemethylation is directly related to lupus flare severity [1]. Genetic predisposition to lupus approximates a continuous variable. So far,38 lupus single-nucleotide polymorphisms (SNPs), each with its own relative risk or oddsratio for lupus, have been identified [49]. These genes assort largely independently, so any given person can inherit 0to 38 distinct lupus genes, with 0, 1, or 2 copies of each. It is reasonable to proposethat those with a higher total genetic risk for lupus may have more problems with lupusthan those with a lower genetic risk.

Genetic/epigenetic interactions and age of lupus onset

The variable nature of T-cell DNA demethylation and genetic risk suggests that thedegree of T-cell DNA demethylation may interact with total genetic predisposition toinitiate lupus onset and flares in any given person. One example of this interactionis suggested by a study relating age of lupus onset to genetic risk. T-cell DNAdemethylates with age [4], and T-cell DNA demethylation contributes to lupus flares [1]. The age of lupus onset is also variable [50]. Typing for 19 risk alleles, this study confirmed that those developinglupus early in life had a greater genetic risk than those developing lupus later, andsome differences between ethnic groups were observed [50]. Typing for 22 lupus SNPs and adjusting each for their relative risk forlupus, Criswell and colleagues [51] found a similar relationship between age of lupus onset and total lupusgenetic risk in Caucasians. Since young people have higher T-cell DNA methylationlevels than older individuals, these studies suggest that young people with high DNAmethylation levels may require more lupus genes to develop lupus but that those witha lesser genetic risk require a greater environmental impact on their T-cellepigenome. This also suggests that those with an even lower total genetic risk mayonly develop an ANA with age.

Genetic/epigenetic interactions in women and men with lupus

The strongest genetic factor predisposing patients to lupus is female sex. Women havetwo × chromosomes whereas men have just one, and the second × in women isinactivated by mechanisms that include DNA methylation. This raises the possibilitythat the second × chromosome may demethylate in women with active lupus,allowing overexpression of X-linked immune genes in women but not in men. CD40L(CD40LG) is an X-linked B-cell co-stimulatory molecule previously reported to beoverexpressed on lupus lymphocytes, and murine lymphocytes overexpressing CD40Linduce lupus [52, 53]. Treating CD4+ T cells from healthy men and women with 5-azaCcaused CD40L overexpression on the female but not the male T cells, and bisulfitesequencing confirmed that women have one methylated and one unmethylated CD40LG geneand that 5-azaC demethylated the methylated gene. In contrast, men had only one,unmethylated gene, and 5-azaC had no further effect on CD40L gene methylation orexpression [12].

Similar experiments compared CD40L on CD4+ T cells from men and women withactive lupus. CD40L levels increased with disease activity on CD4+ T cellsfrom the women, and the degree of overexpression correlated with demethylation oftheir methylated CD40LG gene [12], similar to CD11a, CD70, perforin, and KIR [1, 24]. In contrast, no change in CD40L expression levels was seen in men matchedwith the women for disease activity, consistent with their one unmethylated gene.Controls included demonstrating that the men had an increase in CD70 (encoded onchromosome 19) expression equivalent to that in women with active lupus [12]. These results indicate that genes on the female inactive × cande-methylate and be overexpressed in women with lupus, potentially contributing tothe female predisposition to this disease. This is supported by a report that menwith Klinefelter's syndrome (XXY) develop lupus at approximately the same rate aswomen but that women with Turner's syndrome (XO) do not develop lupus [54].

Since the genetic and environmental contributions to lupus are variable and women arepredisposed to lupus because their second × chromosome can demethylate, it isreasonable to propose that men with only one × chromosome might require agreater degree of T-cell DNA demethylation or a greater total genetic risk (or both)to develop a flare equal in severity to that of women. This was tested by comparingthe interaction between the degree of DNA methylation in KIR and perforin genes,total genetic risk, and SLEDAI (Systemic Lupus Erythematosus Disease Activity Index)in men and women with lupus. Interestingly, the men had a slightly higher totalgenetic risk than the women (P = 0.05). Comparing the level of KIR orperforin methylation with SLEDAI scores showed no significant differences between themen and women. However, when the level of DNA methylation was adjusted by the totalgenetic risk and plotted against the SLEDAI (SLEDAI = risk/methylation) for eachsubject, the men required a stronger genetic/epigenetic interaction to achieve aflare equal in severity to that in women for both KIR (P = 0.01) andperforin (P = 0.005) [55].

Conclusions

Epigenetic mechanisms, including DNA methylation, histone modifications, and miRNAs,play an essential role in regulating gene expression. Recent evidence indicates thatdysregulation of these mechanisms alters gene expression in immune cells, contributingto the development of lupus in genetically predisposed people. Characterizing theepigenetic alterations and the mechanisms causing them is likely to provide importantnew insights into mechanisms causing human lupus and suggest new approaches to thetreatment of this disease.

Note

This article is part of the series on Epigenetics and rheumatic diseases,edited by Nan Shen. Other articles in this series can be found athttp://arthritis-research.com/series/epigenetic

Abbreviations

5-azaC:

5-azacytidine

ANA:

antinuclear antibody

dC:

deoxycytosine

dnMEK:

dominantnegative MEK

Dnmt1:

DNA methyltransferase 1

ERK:

extracellular-signal-regulatedkinase

HPK1:

hematopoietic progenitor kinase 1

ICOS:

inducible co-stimulatorymolecule

IFNγ:

interferon-gamma

JNK:

c-Jun N-terminal kinase

LFA-1:

lymphocytefunction-associated antigen 1

Mφ:

macrophage

MHC:

major histocompatibilitycomplex

miRNA:

microRNA

RANTES:

regulated and normal T cell expressed and secreted

SAH:

S-adenosylhomocysteine

SAM:

S-adenosylmethionine

SLE:

systemic lupuserythematosus

SLEDAI:

Systemic Lupus Erythematosus Disease Activity Index

SNP:

single-nucleotide polymorphism.

References

  1. Richardson B: Primer: epigenetics of autoimmunity. Nat Clin Pract Rheumatol. 2007, 3: 521-527.

    Article  CAS  PubMed  Google Scholar 

  2. Li Y, Liu Y, Strickland FM, Richardson B: Age-dependent decreases in DNA methyltransferase levels and low transmethylationmicronutrient levels synergize to promote overexpression of genes implicated inautoimmunity and acute coronary syndromes. Exp Gerontol. 2010, 45: 312-322. 10.1016/j.exger.2009.12.008.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  3. Gorelik GJ, Yarlagadda S, Richardson BC: Protein kinase Cdelta oxidation contributes to ERK inactivation in lupus Tcells. Arthritis Rheum. 2012, 64: 2964-2974. 10.1002/art.34503.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  4. Richardson B: Impact of aging on DNA methylation. Ageing Res Rev. 2003, 2: 245-261. 10.1016/S1568-1637(03)00010-2.

    Article  CAS  PubMed  Google Scholar 

  5. Fraga MF, Ballestar E, Paz MF , Ropero S, Setien F, Ballestar ML, Heine-Suner D, Cigudosa JC, Urioste M, Benitez J, Boix-Chornet M, Sanchez-Aguilera A, Ling C, Carlsson E, Poulsen P, Vaag A, Stephan Z, Spector TD, Wu YZ, Plass C, Esteller M: Epigenetic differences arise during the lifetime of monozygotic twins. Proc Natl Acad Sci USA. 2005, 102: 10604-10609. 10.1073/pnas.0500398102.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  6. Kim HP, Leonard WJ: CREB/ATF-dependent T cell receptor-induced FoxP3 gene expression: a role for DNAmethylation. J Exp Med. 2007, 204: 1543-1551.

    PubMed Central  CAS  PubMed  Google Scholar 

  7. Wilson CB, Rowell E, Sekimata M : Epigenetic control of T-helper-cell differentiation. Nat Rev Immunol. 2009, 9: 91-105. 10.1038/nri2487.

    Article  CAS  PubMed  Google Scholar 

  8. Rozendaal L, Pals ST, Gleichmann E, Melief CJ: Persistence of allospecific helper T cells is required for maintainingautoantibody formation in lupuslike graft-versus-host disease. Clin Exp Immunol. 1990, 82: 527-532.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  9. Quddus J, Johnson KJ, Gavalchin J, Amento EP, Chrisp CE, Yung RL, Richardson BC: Treating activated CD4+ T cells with either of two distinct DNA methyltransferaseinhibitors, 5-azacytidine or procainamide, is sufficient to cause a lupus-likedisease in syngeneic mice. J Clin Invest. 1993, 92: 38-53. 10.1172/JCI116576.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  10. Cornacchia E, Golbus J, Maybaum J, Strahler J, Hanash S, Richardson B: Hydralazine and procainamide inhibit T cell DNA methylation and induceautoreactivity. J Immunol. 1988, 140: 2197-2200.

    CAS  PubMed  Google Scholar 

  11. Walport MJ: Lupus, DNase and defective disposal of cellular debris. Nat Genet. 2000, 25: 135-136. 10.1038/75963.

    Article  CAS  PubMed  Google Scholar 

  12. Lu Q, Wu A, Tesmer L, Ray D, Yousif N, Richardson B: Demethylation of CD40LG on the inactive × in T cells from women withlupus. J Immunol. 2007, 179: 6352-6358.

    Article  CAS  PubMed  Google Scholar 

  13. Young HA: Regulation of interferon-gamma gene expression. J Interferon Cytokine Res. 1996, 16: 563-568. 10.1089/jir.1996.16.563.

    Article  CAS  PubMed  Google Scholar 

  14. Yung R, Williams R, Johnson K, Phillips C, Stoolman L, Chang S, Richardson B: Mechanisms of drug-induced lupus. III. Sex-specific differences in T cell homingmay explain increased disease severity in female mice. Arthritis Rheum. 1997, 40: 1334-1343.

    CAS  PubMed  Google Scholar 

  15. Richardson B, Scheinbart L, Strahler J, Gross L, Hanash S, Johnson M: Evidence for impaired T cell DNA methylation in systemic lupus erythematosus andrheumatoid arthritis. Arthritis Rheum. 1990, 33: 1665-1673. 10.1002/art.1780331109.

    Article  CAS  PubMed  Google Scholar 

  16. Golbus J, Palella TD, Richardson BC: Quantitative changes in T cell DNA methylation occur during differentiation andageing. Eur J Immunol. 1990, 20: 1869-1872. 10.1002/eji.1830200836.

    Article  CAS  PubMed  Google Scholar 

  17. Tan EM, Smolen JS, McDougal JS, Butcher BT, Conn D, Dawkins R, Fritzler MJ, Gordon T, Hardin JA, Kalden JR, Lahita RG, Maini RN, Rothfield NF, Smeenk R, Takasaki Y, van Venrooij WJ, Wiik A, Wilson M, Koziol JA: A critical evaluation of enzyme immunoassays for detection of antinuclearautoantibodies of defined specificities. I. Precision, sensitivity, andspecificity. Arthritis Rheum. 1999, 42: 455-464. 10.1002/1529-0131(199904)42:3<455::AID-ANR10>3.0.CO;2-3.

    Article  CAS  PubMed  Google Scholar 

  18. Sawalha AH, Jeffries M, Webb R, Lu Q, Gorelik G, Ray D, Osban J, Knowlton N, Johnson K, Richardson B: Defective T-cell ERK signaling induces interferonregulated gene expression andoverexpression of methylation-sensitive genes similar to lupus patients. Genes Immun. 2008, 9: 368-378. 10.1038/gene.2008.29.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  19. Strickland FM, Hewagama A, Lu Q, Wu A, Hinderer R, Webb R, Johnson K, Sawalha AH, Delaney C, Yung R, Richardson BC: Environmental exposure, estrogen and two × chromosomes are required fordisease development in an epigenetic model of lupus. J Autoimmun. 2012, 38: J135-143. 10.1016/j.jaut.2011.11.001.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  20. Gorelik G, Richardson B: Key role of ERK pathway signaling in lupus. Autoimmunity. 2010, 43: 17-22. 10.3109/08916930903374832.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  21. Zandman-Goddard G, Solomon M, Rosman Z, Peeva E, Shoenfeld Y: Environment and lupus-related diseases. Lupus. 2012, 21: 241-250. 10.1177/0961203311426568.

    Article  CAS  PubMed  Google Scholar 

  22. Chiurchiu V, Maccarrone M: Chronic inflammatory disorders and their redox control: from molecular mechanismsto therapeutic opportunities. Antioxid Redox Signal. 2011, 15: 2605-2641. 10.1089/ars.2010.3547.

    Article  CAS  PubMed  Google Scholar 

  23. Oates JC, Gilkeson GS: The biology of nitric oxide and other reactive intermediates in systemic lupuserythematosus. Clin Immunol. 2006, 121: 243-250. 10.1016/j.clim.2006.06.001.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  24. Basu D, Liu Y, Wu A, Yarlagadda S, Gorelik GJ, Kaplan MJ, Hewagama A, Hinderer RC, Strickland FM, Richardson BC: Stimulatory and inhibitory killer Ig-like receptor molecules are expressed andfunctional on lupus T cells. J Immunol. 2009, 183: 3481-3487. 10.4049/jimmunol.0900034.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  25. Liu Y, Kuick R, Hanash S, Richardson B: DNA methylation inhibition increases T cell KIR expression through effects on bothpromoter methylation and transcription factors. Clin Immunol. 2009, 130: 213-224. 10.1016/j.clim.2008.08.009.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  26. Sunahori K, Juang YT, Kyttaris VC, Tsokos GC: Promoter hypomethylation results in increased expression of protein phosphatase 2Ain T cells from patients with systemic lupus erythematosus. J Immunol. 2011, 186: 4508-4517. 10.4049/jimmunol.1000340.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  27. Jeffries MA, Dozmorov M, Tang Y, Merrill JT, Wren JD, Sawalha AH: Genomewide DNA methylation patterns in CD4+ T cells from patients with systemiclupus erythematosus. Epigenetics. 2011, 6: 593-601. 10.4161/epi.6.5.15374.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  28. Kornberg RD, Lorch Y: Chromatin-modifying and -remodeling complexes. Curr Opin Genet Dev. 1999, 9: 148-151. 10.1016/S0959-437X(99)80022-7.

    Article  CAS  PubMed  Google Scholar 

  29. Lee DY, Hayes JJ, Pruss D, Wolffe AP: A positive role for histone acetylation in transcription factor access tonucleosomal DNA. Cell. 1993, 72: 73-84. 10.1016/0092-8674(93)90051-Q.

    Article  CAS  PubMed  Google Scholar 

  30. Ura K, Kurumizaka H, Dimitrov S, Almouzni G, Wolffe AP: Histone acetylation: influence on transcription, nucleosome mobility andpositioning, and linker histone-dependent transcriptional repression. EMBO J. 1997, 16: 2096-2107. 10.1093/emboj/16.8.2096.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  31. Barski A, Cuddapah S, Cui K, Roh TY, Schones DE, Wang Z, Wei G, Chepelev I, Zhao K: High-resolution profiling of histone methylations in the human genome. Cell. 2007, 129: 823-837. 10.1016/j.cell.2007.05.009.

    Article  CAS  PubMed  Google Scholar 

  32. Lachner M, O'Sullivan RJ, Jenuwein T: An epigenetic road map for histone lysine methylation. J Cell Sci. 2003, 116 (Pt 11): 2117-2124.

    Article  CAS  PubMed  Google Scholar 

  33. Murr R: Interplay between different epigenetic modifications and mechanisms. Adv Genet. 2010, 70: 101-141.

    Article  CAS  PubMed  Google Scholar 

  34. Hu N, Qiu X, Luo Y, Yuan J, Zhang G, Zhou Y, Su Y, Lu Q: Abnormal histone modification patterns in lupus CD4+ T cells. J Rheumatol. 2008, 35: 804-810.

    CAS  PubMed  Google Scholar 

  35. Zhao M, Wu X, Zhang Q, Luo S, Liang G, Su Y, Tan Y, Lu Q: RFX1 regulates CD70 and CD11a expression in lupus T cells by recruiting thehistone methyltransferase SUV39H1. Arthritis Res Ther. 2010, 12: R227-10.1186/ar3214.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  36. Zhao M, Sun Y, Gao F, Wu X, Tang J, Yin H, Luo Y, Richardson B, Lu Q: Epigenetics and SLE: RFX1 downregulation causes CD11a and CD70 overexpression byaltering epigenetic modifications in lupus CD4+ T cells. J Autoimmun. 2010, 35: 58-69. 10.1016/j.jaut.2010.02.002.

    Article  PubMed  Google Scholar 

  37. Zhou Y, Qiu X, Luo Y, Yuan J, Zhong Q, Zhao M, Lu Q: Histone modifications and methyl-CpG-binding domain protein levels at the TNFSF7(CD70) promoter in SLE CD4+ T cells. Lupus. 2011, 20: 1365-1371. 10.1177/0961203311413412.

    Article  CAS  PubMed  Google Scholar 

  38. Zhang Q, Long H, Liao J, Zhao M, Liang G, Wu X, Zhang P, Ding S, Luo S, Lu Q: Inhibited expression of hematopoietic progenitor kinase 1 associated with loss ofjumonji domain containing 3 promoter binding contributes to autoimmunity insystemic lupus erythematosus. J Autoimmun. 2011, 37: 180-189. 10.1016/j.jaut.2011.09.006.

    Article  CAS  PubMed  Google Scholar 

  39. Bartel DP: MicroRNAs: target recognition and regulatory functions. Cell. 2009, 136: 215-233. 10.1016/j.cell.2009.01.002.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  40. Ding S, Liang Y, Zhao M, Liang G, Long H, Zhao S, Wang Y, Yin H, Zhang P, Zhang Q, Lu Q: Decreased microRNA-142-3p/5p expression causes CD4+ T cell activation and B cellhyperstimulation in systemic lupus erythematosus. Arthritis Rheum. 2012, 64: 2953-2963. 10.1002/art.34505.

    Article  CAS  PubMed  Google Scholar 

  41. Yu D, Tan AH, Hu X, Athanasopoulos V, Simpson N, Silva DG, Hutloff A, Giles KM, Leedman PJ, Lam KP, Goodnow CC, Vinuesa CG: Roquin represses autoimmunity by limiting inducible T-cell co-stimulator messengerRNA. Nature. 2007, 450: 299-303. 10.1038/nature06253.

    Article  CAS  PubMed  Google Scholar 

  42. Pan W, Zhu S, Yuan M, Cui H, Wang L, Luo X, Li J, Zhou H, Tang Y, Shen N: MicroRNA-21 and microRNA-148a contribute to DNA hypomethylation in lupus CD4+Tcells by directly and indirectly targeting DNA methyltransferase 1. J Immunol. 2010, 184: 6773-6781. 10.4049/jimmunol.0904060.

    Article  CAS  PubMed  Google Scholar 

  43. Zhao S, Wang Y, Liang Y, Zhao M, Long H, Ding S , Yin H, Lu Q: MicroRNA-126 regulates DNA methylation in CD4+ T cells and contributes to systemiclupus erythematosus by targeting DNA methyltransferase 1. Arthritis Rheum. 2011, 63: 1376-1386. 10.1002/art.30196.

    Article  CAS  PubMed  Google Scholar 

  44. Stagakis E, Bertsias G, Verginis P, Nakou M, Hatziapostolou M, Kritikos H, Iliopoulos D, Boumpas DT: Identification of novel microRNA signatures linked to human lupus disease activityand pathogenesis: miR-21 regulates aberrant T cell responses through regulation ofPDCD4 expression. Ann Rheum Dis. 2011, 70: 1496-1506. 10.1136/ard.2010.139857.

    Article  CAS  PubMed  Google Scholar 

  45. Fan W, Liang D, Tang Y, Qu B, Cui H, Luo X , Huang X, Chen S, Higgs BW, Jallal B, Yao Y, Harley JB, Shen N: Identification of microRNA-31 as a novel regulator contributing to impaired IL-2production in T cells from patients with systemic lupus erythematosus. Arthritis Rheum. 2012, 64: 3715-3725. 10.1002/art.34596.

    Article  CAS  PubMed  Google Scholar 

  46. Zhao X, Tang Y, Qu B, Cui H, Wang S, Wang L , Luo X, Huang X, Li J, Chen S, Shen N: MicroRNA-125a contributes to elevated inflammatory chemokine RANTES levels viatargeting KLF13 in systemic lupus erythematosus. Arthritis Rheum. 2010, 62: 3425-3435. 10.1002/art.27632.

    Article  CAS  PubMed  Google Scholar 

  47. Tang Y, Luo X, Cui H, Ni X, Yuan M, Guo Y, Huang X, Zhou H, de Vries N, Tak PP, Chen S, Shen N: MicroRNA-146A contributes to abnormal activation of the type I interferon pathwayin human lupus by targeting the key signalling proteins. Arthritis Rheum. 2009, 60: 1065-1075. 10.1002/art.24436.

    Article  CAS  PubMed  Google Scholar 

  48. Garchow BG, Bartulos Encinas O, Leung YT, Tsao PY, Eisenberg RA, Caricchio R, Obad S, Petri A, Kauppinen S, Kiriakidou M: Silencing of microRNA-21 in vivo ameliorates autoimmune splenomegaly inlupus mice. EMBO Mol Med. 2011, 3: 605-615. 10.1002/emmm.201100171.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  49. Lessard CJ, Adrianto I, Ice JA, Wiley GB, Kelly JA, Glenn SB, Adler AJ, Li H, Rasmussen A, Williams AH, Ziegler J, Comeau ME, Marion M, Wakeland BE, Liang C, Ramos PS, Grundahl KM, Gallant CJ, Alarcon-Riquelme ME, Alarcon GS, Anaya JM, Bae SC, Boackle SA, Brown EE, Chang DM, Cho SK, Criswell LA, Edberg JC, Freedman BI, Gilkeson GS, et al: Identification of IRF8, TMEM39A, and IKZF3-ZPBP2 as susceptibility loci forsystemic lupus erythematosus in a large-scale multiracial replication study. Am J Hum Genet. 2012, 90: 648-660. 10.1016/j.ajhg.2012.02.023.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  50. Webb R, Kelly JA, Somers EC, Hughes T, Kaufman KM, Sanchez E, Nath SK, Bruner G, Alarcon-Riquelme ME, Gilkeson GS, Kamen DL, Richardson BC, Harley JB, Sawalha AH: Early disease onset is predicted by a higher genetic risk for lupus and isassociated with a more severe phenotype in lupus patients. Ann Rheum Dis. 2011, 70: 151-156. 10.1136/ard.2010.141697.

    Article  PubMed Central  PubMed  Google Scholar 

  51. Taylor KE, Chung SA, Graham RR, Ortmann WA, Lee AT, Langefeld CD, Jacob CO, Kamboh MI, Alarcon-Riquelme ME, Tsao BP, Moser KL, Gaffney PM, Harley JB, Petri M, Manzi S, Gregersen PK, Behrens TW, Criswell LA: Risk alleles for systemic lupus erythematosus in a large case-control collectionand associations with clinical subphenotypes. PLoS Genet. 2011, 7: e1001311-10.1371/journal.pgen.1001311.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  52. Crow MK, Kirou KA: Regulation of CD40 ligand expression in systemic lupus erythematosus. Curr Opin Rheumatol. 2001, 13: 361-369. 10.1097/00002281-200109000-00004.

    Article  CAS  PubMed  Google Scholar 

  53. Higuchi T, Aiba Y, Nomura T, Matsuda J, Mochida K, Suzuki M, Kikutani H, Honjo T, Nishioka K, Tsubata T: Cutting edge: ectopic expression of CD40 ligand on B cells induces lupus-likeautoimmune disease. J Immunol. 2002, 168: 9-12.

    Article  CAS  PubMed  Google Scholar 

  54. Scofield RH, Bruner GR, Namjou B, Kimberly RP, Ramsey-Goldman R, Petri M, Reveille JD, Alarcon GS, Vila LM, Reid J, Harris B, Li S, Kelly JA, Harley JB: Klinefelter's syndrome (47,XXY) in male systemic lupus erythematosus patients:support for the notion of a gene-dose effect from the × chromosome. Arthritis Rheum. 2008, 58: 2511-2517. 10.1002/art.23701.

    Article  PubMed Central  PubMed  Google Scholar 

  55. Sawalha AH, Wang L, Nadig A, Somers EC, McCune WJ, Hughes T, Merrill JT, Scofield RH, Strickland FM, Richardson B: Sex-specific differences in the relationship between genetic susceptibility, Tcell DNA demethylation and lupus flare severity. J Autoimmun. 2012, 38: J216-222. 10.1016/j.jaut.2011.11.008.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank Julie Olivero for assistance in preparing the manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Bruce C Richardson.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.

Authors’ original file for figure 1

Rights and permissions

Reprints and permissions

About this article

Cite this article

Patel, D.R., Richardson, B.C. Dissecting complex epigenetic alterations in human lupus. Arthritis Res Ther 15, 201 (2013). https://doi.org/10.1186/ar4125

Download citation

  • Published:

  • DOI: https://doi.org/10.1186/ar4125

Keywords