Epigenetics in autoimmune diseases: pathogenesis and prospects for therapy Zimu Zhang, Rongxin Zhang PII: DOI: Reference:
S1568-9972(15)00116-0 doi: 10.1016/j.autrev.2015.05.008 AUTREV 1722
To appear in:
Autoimmunity Reviews
Received date: Accepted date:
15 May 2015 20 May 2015
Please cite this article as: Zhang Zimu, Zhang Rongxin, Epigenetics in autoimmune diseases: pathogenesis and prospects for therapy, Autoimmunity Reviews (2015), doi: 10.1016/j.autrev.2015.05.008
This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT Epigenetics in autoimmune diseases: pathogenesis and prospects for therapy Zimu Zhang1 & Rongxin Zhang1,* 1
IP
T
Laboratory of Immunology and Inflammation, Department of Immunology and Research Center of Basic Medical Science; Basic Medical College; Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironments and Diseases of Educational Ministry of China, Tianjin Medical University, Tianjin 300070, China;
SC R
* Correspondence: Rongxin Zhang, Research Center of Basic Medical Science, Tianjin Medical University, Tianjin 300070, China, Email:
[email protected] or
[email protected], Telephone and Fax: 86-22-83336563
TE
D
MA
NU
Abstract Epigenetics is the study of heritable changes in genome function without underlying modifications in their nucleotide sequence. Disorders of epigenetic processes, which involve DNA methylation, histone modification non-coding RNA and nucleosome remodeling, may influence chromosomal stability and gene expression, resulting in complicated syndromes. In the past few years, it has been disclosed that identified epigenetic alterations give rise to several typical human autoimmune diseases such as systemic lupus erythematosus (SLE), rheumatoid arthritis (RA) and multiple sclerosis (MS). These emerging epigenetic studies provide new insights into autoimmune diseases. The identification of specific epigenetic dysregulation may inspire more discoveries of other uncharacterized mechanisms. Further elucidation of the biological functions and clinical significance of these epigenetic alternations may be exploited for diagnostic biomarkers and therapeutic benefits.
CE P
Keywords: autoimmune disease, DNA methylation, epigenetics, epigenetic therapy, histone modification, ncRNA
AC
1. Introduction Autoimmune diseases, such as systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), multiple sclerosis (MS) and type 1 diabetes (T1DM), are characterized by the presence of autoreactive immune cells or the development of specific autoantibodies [1]. Autoimmune diseases possess diverse epidemiology or symptoms, however, they have a common origin and it is clear that genetic predisposition is involved in the etiopathology of autoimmune disorders[2]. Genetic studies revealed that the extended major histocompatibility complex (MHC), particularly human leukocyte antigen (HLA) genes, has a strong association with autoimmune diseases[3]. In recent decades, more susceptibility genes shared by most autoimmune diseases have been uncovered, such as PTPN22, IRF5, STAT4, BANK1 and ICAM3. These associated genes point to some common pathways, expanding our understanding of autoimmune diseases[1]. Whereas the high discordance in the onset rate of autoimmune diseases was subsequently observed in detailed investigations into monozygotic twins whose genetic information and susceptibility variants are virtually identical. The fact that different phenotypes arise from the same genotype suggests that environmental factors, other than genetics, also have an essential role in the onset and progression of diseases[4]. Epigenetics refers to the study of heritable alterations in genome function without underlying modifications in their nucleotide sequence[5]. Via epigenetic mechanisms, a number of internal and external environmental risk factors, including smoking, nutrition, viral infection and the exposure to chemicals, could exert their influence on the pathogenesis of autoimmune diseases. Such factors could impact the epigenetic mechanisms, which, in turn, build relationship with the regulation of gene expression, and eventually triggering immunologic events that result in instability of immune system[6]. For example, smoking, the best-known environmental risk factor, plays a 1
ACCEPTED MANUSCRIPT
AC
CE P
TE
D
MA
NU
SC R
IP
T
critical role in autoimmune diseases. Epidemiologic studies of RA and MS showed that heavy-smokers had a significantly increased risk of getting diseases compared with never-smokers [7, 8]. In addition, increasing levels of serum Vitamin D, a steroid hormone, could decrease the risk of MS[9]. Epstein-Barr virus (EBV) exposure was hypothesized to be related to the increased risk of RA and SLE as well[10]. To bridge the gap between environmental and genetic factors, over the past few years, great progress has been made in identifying detailed epigenetic mechanisms for autoimmune diseases. Furthermore, with rapid advances in technological development, high-throughput screening approaches and other novel technologies support the systematic investigations and facilitate the epigenetic identification. These emerging epigenetic studies provide new insights into autoimmune diseases, raising great expectations among researchers and clinicians. They might reveal not only new clinical biomarkers for diagnosis and disease progression but also novel targets for potential epigenetic therapeutic treatment. In this review, we summarize and discuss the epigenetic alterations in autoimmune diseases, with a focus on prospective candidates for ‘epigenetic therapy’, and outline the future steps to be taken in the field. 2. Roles of epigenetic alterations in the pathogenesis of autoimmune diseases Epigenetic regulations plays a critical role in determining gene function and activities, meanwhile they remain highly sensitive and could be even reversed by environmental influences. Disorders of epigenetic processes, which involve DNA methylation, histone modification and non-coding RNA expression, were found associated with the pathogenesis of autoimmune diseases. Although research progress varies in different kinds of autoimmune diseases, they could provide useful references for each other. 2.1 DNA methylation DNA methylation, a dynamic process involving methylation and demethylation events, occurs in different regions of the genome and is crucially important for embryogenesis, cellular proliferation and differentiation [11, 12]. Methylation is established and maintained by the DNA methyltransferase (DNMT) family which is composed of a group of enzymes, including DNMT1, DNMT3a, DNMT3b, and DNMT3L. Among them, DNMT1 contributes to maintaining methylation status during replication. DNMT3a and DNMT3b are de novo DNMTs that are responsible for methylation during embryonic development. DNMT3L is reported to be a cofactor for the de novo DNMT stimulating DNMT3a/3b activity. Methyl-CpG-binding domain (MBD) proteins could also regulate methylation together with DNMTs[13]. Demethylation could be passive or active. Passive demethylation is induced by inhibition of DNMTs, providing the basis for treatments with the aim of erasing abnormal hypermethylation [14, 15]. Active demethylation might depend on the activity of cytosine deaminases occurring predominantly in cell differentiation and associated with the activation of immune cells. For instance, activation-induced cytidine deaminase (AICDA) plays a critical role in activating B cells as well as generating IgG anti-nucleosomal antibodies of murine SLE[16, 17]. In mammals, DNMTs catalyze the donation of a methyl group to CpG dinucleotides, particularly CpG islands located within promoter regions, leading to the structural changes of chromatin. Methylated regions block the accessibility to transcriptional activators and thereby inhibit the gene transcription.[13] In contrast, an unmethylated state will permit the transcription [14]. 2.1.1 DNA methylation alterations in lymphocytes The analysis of epigenetic alterations in CD4+ T cells from patients has revealed that DNA methylation pattern is profoundly disrupted (Table 1). A range of autoimmune diseases, including SLE, RA and systemic sclerosis (SSc), display global hypomethylation associated with a concurrent decreased expression of methylation-related genes, such as DNMT1 and MBD4. [18, 19]. CD4+ T cells from patients with SLE, RA and SSc show a substantial reduction in the levels of DNA methylation on the promoter of CD40LG (which encodes the B cell costimulatory molecule CD40L; also known as CD154)[20-22], which is thought to be important in disease pathogenesis given that the CD40-CD40L interaction is critically associated with Th17 cells differentiation and IL-17 production [23]. It is noteworthy that 2
ACCEPTED MANUSCRIPT
AC
CE P
TE
D
MA
NU
SC R
IP
T
the fact that demethylation of CD40L regulatory elements on the female inactive X chromosome upregulates CD40L expression in CD4+ T cells may contribute to the susceptibility of women to these autoimmune diseases [20-22]. In addition, the hypomethylation of CD70 (also known as TNFSF7 or CD27L) is observed and probably contributes to the overexpression of CD70 by CD4+ T cells from patients with SLE, SSc and primary Sjögren's syndrome (pSS)[24-26]. Moreover, with the development of the genome-wide DNA methylation analysis approach, hypomethylation patterns were observed at interferon-regulated genes, such as IRF5, IFIT2, STAT1 and USP18, in CD4+ T cells in the context of SLE and pSS. This hypomethylation exists in T cells before activation and differentiation, as well as maintains in the immune system throughout and even beyond active stages of the disease. It might provide an explanation for the mechanism of type I interferon pathway playing an important role in the pathogenesis of SLE and pSS [27-29]. Despite of the importance of B cells in the pathogenesis of autoimmune diseases, few studies have addressed methylation changes in B cells. Garaud et al. reported that the E1B promoter of CD5 is hypomethylated in SLE B cells. High levels of CD5-E1B could block the inhibitory effects of CD5 on B cell receptor signaling and antibody production, leading to the activation of B cells. Moreover, in a recent study, HRES-1, the prototype of Human Endogenous Retroviruses (HERV), is demonstrated hypomethylated in SLE B cells. This hypomethylation might provide explanations for increasing detection of HERV in patients with autoimmune diseases including SLE, pSS and MS. On the other hand, in these two studies, high level of interleukin-6 (IL-6) is also showed in SLE B cells, which could reduce the expression of DNMT1 by arresting the cell cycle at late G1 phase and thereby affect the methylation of CD5 and HRES-1, contributing to disease activity [30, 31]. However, it is noteworthy that not all overexpressed genes in autoimmune diseases are accompanied by a concurrent epigenetic alteration. It has been demonstrated that IRF5 mRNA expression and CD40L on CD4+ T cells are increased in patients with pSS, but the aberrant DNA methylation profiles are absent from corresponding putative regulatory sequences[32, 33]. Further mechanisms leading to their overexpression need to be investigated. Moreover, not all epigenetic altered genes in lymphocytes are subject to hypomethylation. It has been reported that regulatory regions of FOXP3 in peripheral blood CD4+ T cells from patients with SSc, RA and T1DM is hypermethylated, affecting the expression of key transcription factor which is required for the generation of regulatory T cells[34-36]. These findings might indicate an epigenetic alteration responsible for the loss of immune homeostasis in development of SSc, T1DM and RA. 2.1.2 DNA methylation alterations in other cell types In addition to lymphocytes, a number of evidences support that DNA methylation pattern in other cell types could be altered (Table 1). Fibroblasts from patients with SSc showed increased DNA methylation in the promoter region of FLI1 that encodes an inhibitor of collagen expression [37]. Hypermethylation also silences the expression of the DKK1 and SFRP1, which are antagonists of Wnt signaling, permitting the profibrotic pathway in RA and SSc [38, 39]. These findings suggest that epigenetic mechanisms might mediate the fibrotic manifestations of RA and SSc. Peripheral blood mononuclear cells (PBMCs) from patients with RA showed hypomethylation of the promoter of IL6 encoding IL-6, which is instrumental in B cell response and hyperactivation of the inflammation circuit[40]. In addition, specifically hypermethylation is observed in synovial cell, which is a major pathology contributing to development of RA, silencing the expression of DR3 whose encoded protein is associated with cell apoptosis[41]. In the context of MS, discordance in CD4+ T cells of monozygotic twins has been studied but found no evidences for epigenetic differences[42]. Whereas Mastronardi et al. demonstrated hypomethylation at the promoter of PAD2 in normal-appearing white matter (NAWM) from patients [43]. MS is a chronic inflammatory disease characterized by myelin destruction followed by progressive neurodegeneration. Overexpression of PAD2 3
ACCEPTED MANUSCRIPT
AC
CE P
TE
D
MA
NU
SC R
IP
T
plays a critical role in citrullination process of myelin basic protein (MBP) followed by increased production of immunodominant peptides, promoting the protein autocleavage responsible for proteolytic digestion, myelin instability and chronic inflammation response[44]. The different conclusions between two studies mentioned above suggest that perhaps microglia, oligodendrocyte or other cell types should be studied for MS, and emphasize the importance of selecting appropriate cell types or tissues for study. 2.2 Histone modifications Histones, involved in packaging and organizing nucleosomes, could be grouped into core histones (H2A, H2B, H3, and H4) and linker histones (H1 and H5). As another major source of epigenetic alteration, histone post-translational modifications, including acetylation and methylation, could modulate the chromatin structure, influencing its accessibility to transcription factors at gene promoters and enhancers. The balance of histone modifications are established and maintained by a range of enzymes including, but not limited to histone methyltransferases (HMT) and demethylases (HDM), and histone acetyltransferases (HAT) and deacetylases (HDAC)[45]. In some cases, the correlation of specific histone modifications and gene expression associated with autoimmune diseases has been well investigated (Table 2). 2.2.1 Histone acetylation Histone acetylation is catalyzed by HAT, adding an acetyl group on lysine residues in the N-terminal tail and promoting a more open chromatin structure. Generally, H3 and H4 hyperacetylation (H3Ac, H4Ac) are characteristic of active genes. Whereas histone deacetylation catalyzed by HDAC removes the acetyl groups, causing the DNA wrapped around nucleosome more tightly, thereby repressing gene expression[45]. SLE, as a systemic disease characterized by autoantibodies against nuclear or cytoplasmic self-antigens resulting in inflammation-mediated multiorgan damage, displays global H3 and H4 hypoacetylation in the CD4+ T cells of patients[46]. However, gene-specific histone modification patterns might differ from those of genome-wide ones. By investigating the alterations of histone modification on CD70 gene transcription, Zhou et al. showed that H3 acetylation was significantly increased in patients with active SLE and was correlated positively with disease activity[47]. Furthermore, histone modification patterns could vary in different cell types. Owing to the decreased HDAC2 and HDAC7, histone H4 hyperacetylation was demonstrated in B cells from patients with SSc[48], while in fibroblast, increased H3 and H4 deacetylation downregulate FLI1 gene expression, contributing to excessive synthesis of collagen and other extracellular matrix components[37]. In addition, in context of MS, histone H3 deacetylation could inhibit oligodendrocyte differentiation[49]. As for T1DM, it has been investigated that the increased H3K9 acetylation (H3K9Ac) could upregulate HLA-DRB1, HLA-DQB1 gene expression[50]. These aforesaid altered histone acetylation patterns might provide potential therapeutic targets for the treatment. 2.2.2 Histone methylation Histone methylation, which adds a methyl group to the histone subunit, is catalyzed by HMT, while demethylation occurs as a result of interaction of HDM. In most cases, histone hypermethylations, such as histone H3 lysine9 methylation (H3K9me) or trimethyl-lysine27 (H3K27me3), is present in repressed genes[45]. H3K9 hypomethylation has been detected in B cells from patients with SSc, which is correlated with downregulated histone methyltransferase SUV39H2 and upregulated histone demethylase JHDM2A[48]. Moreover, H3K27me3 is inhibited in fibroblast from patients with SSc, upregulating FOSL2 gene expression involved in fibroblast activation and fibrosis [51]. Similarly, CLTA4, a susceptibility gene of T1DM, displays increased promoter histone H3 lysine 9 dimethylation (H3K9me2), which is associated with T cells activation[52]. Synovial fibroblasts from patients with RA show increased histone methyltransferase EZH2 which could silence SFRP1 gene expression and thereby abrogate its inhibitory role on aberrant collagen deposition by such fibroblasts [39]. However, not all histone methylation are markers for inactive regions of chromatin. Typically, methylation of 4
ACCEPTED MANUSCRIPT
AC
CE P
TE
D
MA
NU
SC R
IP
T
lysine 4 of histone H3 denotes an activation mark and is predominantly found at promoters of active genes[45]. For instance, CD4+ T cells from patients with SLE show a significantly elevated level of dimethylated H3 lysine 4 (H3K4me2) upregulating CD70 gene expression associated with excessive autoimmune responses[47]. 2.3 Non-coding RNA Only around 2% of mammalian transcripts could be translated into functional proteins. The rest of the remaining transcripts were defined as non-coding RNAs (ncRNAs) which serve as epigenetic modifiers and play a critical role on gene regulation. These ncRNAs could be further classified into several groups including microRNAs and long non-coding RNAs [53]. 2.3.1 MicroRNAs MicroRNAs (miRNAs) are approximately 22 nucleotides short, single-stranded ncRNAs involved in regulating the gene expression in a posttranscriptional manner. Via binding to complementary sequences on target mRNA, the miRNA-induced silencing complex (miRISC) could lead to translational repression, mRNA destabilization and degradation. It has been revealed that miRNA expression could control dynamic aspects of immune responses which are important for maintaining homeostasis and defending the host against antigens [54]. Functional studies subsequently demonstrated that a common set of aberrantly expressed miRNAs exert a significant role in pathogenesis of most autoimmune diseases by modulating multiple autoimmunity-related genes and other epigenetic processes (Table 3). Aberrant miR-21 expression has been reported to correlate with the progress of a range of autoimmune diseases. The expression of miR-21 is increased in CD4+ T cell from patients with SLE and has been revealed to target an important autoimmune gene RASGRP1, which mediates the Ras-MAPK pathway, downregulating DNMT1 expression and thereby contributing to the DNA hypomethylation[55]. Similarly, it has been demonstrated that increased levels of miR-21 contribute to the B cell hyperresponsiveness and aberrant T cell response in SLE by targeting the gene PTEN and PDCD4 respectively[56, 57]. Overexpression of miR-21 has been also found to negatively regulate SMAD7, enhancing TGF-β signaling and upregulating fibrosis-related genes, such as ACTA2, COL1A1, COL1A2 and FN1, in fibroblasts from patients with SSc [58]. Similarly, by depleting SMAD7 and therefore enhancing TGF-β signaling, miR-21 could limit the autocrine inhibitory effects of IL-2, and thereby promotes Th17 differentiation and mediates MS [59]. By contrast, CD4+ T cells show a significant reduction in levels of miR-21 in the context of RA, accompanied by increased STAT3 expression, decreased STAT5/pSTAT5 protein and Foxp3 mRNA levels which might contribute to the imbalance of Th17 and Treg cells[60]. Given that decreased Foxp3 expression has been also found in CD4+ T cell from patients with T1DM[35] and furthermore miR-21 has been reported to prevent T1DM[61], the association between its abnormal differentiation of regulatory T cells and miR-21 expression could be further investigated. The miR-29 family, including miR-29a, miR-29b and miR-29c, play a critical role in the pathogenesis of autoimmune diseases. It has been demonstrated that miR-29b targets directly DNMT3A and DNMT3B, inducing DNA hypomethylation, while CD4+ T cells in patients with SLE show that miR-29b downregulates DNMT1 indirectly by targeting Sp1 which is a transactivator of DNMT1 gene [62]. In the context of MS, miR-29b is upregulated in CD4+ T cells where it targets T-bet and IFN-γ, controlling Th1 differentiation [63]. Furthermore, in fibroblasts from patients with SSc, miR-29a serves as an antifibrotic miRNA that opposes the profibrotic activity of miR-21[64]. As well as prominent roles of miR-21 and miR-29 in regulating the autoimmune responses, aberrant expressions of other miRNAs, such as miR-30, miR-146a and miR-155, could also dysregulate epigenetic processes and, consequently, contribute to the pathogenesis of autoimmune diseases (Table 3). In addition to well-established roles of these miRNAs, the detection of specific circulating miRNAs that correlate with disease severity could provide reliable and accurate biomarkers of autoimmune diseases. For instance, altered circulating miR-223 expression has been found in T cells or other samples, such as plasma and PBMCs, from patients with SLE, RA and MS [65-67], however, further studies on their induced epigenetic alterations and molecular 5
ACCEPTED MANUSCRIPT
AC
CE P
TE
D
MA
NU
SC R
IP
T
mechanisms are clearly warranted. 2.3.2 Long non-coding RNAs Long non-coding RNAs (lncRNAs), which are larger than 200 nucleotides and lack of protein-coding function, have been found to be pervasively transcribed but highly diverse in sequence, structure and function. Presently, according to their position relative to protein-coding mRNAs, lncRNAs are classified into the long intergenic ncRNA (lincRNA), intronic lncRNA, antisense lncRNA, transcribed pseudogene lncRNAs and enhancer RNA (eRNA)[68]. Compared to widely-investigated miRNAs, however, studies of autoimmune diseases in search of lncRNA expression are just beginning. Understanding of lncRNAs involving immunity, inflammation and degeneration might provide useful references for investigating this type of defect in autoimmune diseases. The largest group of lncRNAs is the lincRNAs which are located between protein-coding genes. Pin et al. has demonstrated that lnc-DC regulates differentiation of human and mouse monocytes into dendritic cells. Lnc-DC knockdown results in altered expression of 664 protein-coding genes, affecting the antigen uptaking, allogenic CD4+ T cells production and cytokine release which might have potential relevance to autoimmune diseases involving monocytes and DC dysfunction[69]. Moreover, lincRNA-Cox2 mediates the induction or repression of immune response-related genes, including Il6, Stat1 and Irf7, in mouse macrophages [70]. LncRNA-ATB, which is activated by TGF-β involved in hepatocellular invasion-metastasis cascade as well as profibrotic activity, could promote autocrine induction of IL-11 and trigger STAT3 signaling [71]. NRON, an intronic lncRNA transcribed from intronic regions in the sense or antisense orientation, serves as the non-coding repressor of nuclear factor of activated T cells[72]. Lethe, a transcribed pseudogene lncRNA arising from a gene losing its intrinsic protein-coding ability through mutation or inaccurate duplication, blocks NF-κB-driven inflammatory responses in mouse fibroblasts[73]. Whether these lncRNAs are associated with the pathogenesis of autoimmune diseases deserves further investigations. Antisense lncRNAs, which are transcribed across the exons of protein-coding genes from the opposite strand, could form sense-antisense pairs by pairing with the protein-coding strand to regulate epigenetic silencing, transcription or mRNA stability. BDNF-AS has been found to normally repress brain-derived neurotrophic factor (BDNF). Inhibition of this antisense lncRNA leads to increased BDNF level, promoting neuronal outgrowth and differentiation [74]. Carrieri et al. identified a nuclear-enriched lncRNA antisense to mouse ubiquitin carboxy-terminal hydrolase L1 (Uchl1) involved in brain function and neurodegeneration given that oxidative inactivation of UCHL1 protein has been reported in Parkinson’s and Alzheimer’s disease brains. Via a 5' overlapping sequence and an embedded inverted SINEB2 element-dependent manner, antisense Uchl1 could upregulate mouse ubiquitin carboxy-terminal hydrolase L1 (UCHL1) protein synthesis, furthermore, this lncRNA is under the control of stress signaling pathways, particularly the mTORC1 pathway[75]. These data reveal novel mechanisms of gene expression control at post-transcriptional level and might expand the potential roles of lncRNA for neuroprotection in MS. Of note, it is relatively easy to extrapolate miRNA functions in humans from experiment results in mouse given that miRNAs and their targets are generally well conserved throughout evolution. In contrast, lncRNAs demonstrate low evolutionary sequence conservation, for less mouse lncRNAs have homologues in humans[76]. Non-coding RNAs open up new avenues for epigenetic research, however, multiple challenges still need to be appropriately addressed. 2.4 Cross-talk between multiple epigenetic mechanisms It is noteworthy that all these aforementioned epigenetic mechanisms act together at the same time rather than regulate gene expression separately. DNA methylation status is established and maintained by DNMTs, whereas methyl-CpG-binding domain (MBD) proteins associated with methyl-cytosine could also recruit silencing complexes participating in transcriptional repression. These silencing complexes which contain histone deacetylases (HDACs) and histone methyltransferases (HMTs) contribute to a cross-talk among the DNA methylation, histone modifications and 6
ACCEPTED MANUSCRIPT
AC
CE P
TE
D
MA
NU
SC R
IP
T
nucleosome remodeling[12]. Meanwhile, cross-talks among various ncRNAs or between ncRNAs and other epigenetic mechanisms exist extensively as well[77]. Typically, a recent study by Hedrich et al. showed an epigenetic cross-talk contributing the pathogenesis of SLE. Reduced DNA methylation of the IL10 gene promoter leads to Stat3 and Stat5 recruitment to and trans-activation of the IL10 promoter and an intronic enhancer which is referred to as intronic Stat-responsive element (I-SRE). The effects of Stat3 on the I-SRE were found higher compared with Stat5. In T cells from patients with SLE, increased Stat3 phosphorylation enhances its recruitment to the IL10 promoter and the replacement of Stat5 at the I-SRE, accompanied by the recruitment of histone acetyltransferase p300 to those regions, resulting in trans-activation and nucleosome remodeling which thereby elevates IL-10 level inducing autoantibody production and tissue damage in SLE[78]. 3. Prospects for epigenetic therapy Rapid progress in understanding epigenetic dysregulation in autoimmune diseases has enabled us to determine the general mechanisms of epigenetic alterations and attract intensive investigation aimed at identifying specific clinical markers for early diagnosis, furthermore, embark on the development of novel therapeutic drugs and other therapeutic approaches. 3.1 Drugs with epigenetic effects Many epigenetic aetiology-related human diseases, including cancer, encourage the development of a new therapeutic approach termed ‘epigenetic therapy’. Drugs with the ability to alter methylation patterns on DNA or the modification of histones are a means of inducing large-scale recovery in the epigenome. Repurposing drugs approved for cancer could be a promising orientation, as these drugs might have properties that allow them to be beneficial for multiple diseases. Among them, HDAC inhibitors and DNMT inhibitors are currently the most widely studied epigenetic therapeutics for autoimmune diseases (Table 4). Trichostatin A (TSA), which is an HDAC inhibitor inducing growth arrest and apoptosis in tumors, has demonstrated its inhibitory effect on fibroblast activation in fibrosis-related autoimmune diseases such as RA and SSc. TSA could restore the expression of FLI1 which is the negative regulator of collagen expression but usually repressed in SSc by promoter hypoacetylation[37]. Furthermore, the inhibition of HDAC-7 by TSA downregulates the expression of fibrosis-related genes COL1A1 and COL3A1[79]. In the context of RA, TSA significantly impairs the stability of IL-6 mRNA and thereby disrupts inflammatory cytokine production in synovial cells [80]. Apart from its anti-fibrosis effects, TSA reverses the aberrant increased CD40L and IL10 expression as well as the decreased IFN-γ expression in T cells from patients with SLE[81]. As for T1DM, it is suggested that TSA could increase the expression of IFN-γ and its transcription factor Tbx21 in T lymphocytes, reducing the cellular infiltration of islets [82]. In clinical trials, orally active histone deacetylase inhibitor givinostat (also termed ITF2357) was performed on 17 patients with systemic juvenile idiopathic arthritis. Although 6 adverse events exhibited in 3 patients, two thirds of the patients achieved significant therapeutic benefit after 12 weeks, particularly in terms of the arthritic component of the disease[83]. Decreased production of IL-6 by givinostat might contribute to this therapeutic benefit [80]. With regard to DNMT inhibitors, 5-aza-2'-deoxycytidine reverses the aberrant hypermethylation of DKK1, SFRP1 and FLI1 in fibroblasts from patients with SSc, normalizing the Wnt signalling and type I collagen expression respectively[37, 38]. In the mice with experimental autoimmune encephalomyelitis (EAE) which resemble the human demyelinating disease MS in both clinical course and histopathology, 5-aza-2'-deoxycytidine treatment increases the population and immunosuppressive function of Foxp3+ regulatory T cells as well as inhibits the effector cells in the periphery, preventing EAE development and suppressing CNS inflammation[84]. Of note, because of the large number of genes targeted by each HDAC, even a very enzyme-specific HDAC inhibitor is still unavoidable to alter expression of many genes which is likely to lead to unacceptable toxicity[85]. Therefore, HDAC inhibitors might act as a double edged sword in epigenetic therapy and the same is true for the 7
ACCEPTED MANUSCRIPT
AC
CE P
TE
D
MA
NU
SC R
IP
T
risk of DNMT inhibitors in clinical applications. Further investigations into targets downstream of HDACs and DNMTs, such as proteins that bind to modified histones, could be carried out to identify safer targets for therapy. Moreover, in hypomethylation-related autoimmune disease, including SLE and RA, DNMT inhibitors would not be appropriate to restore their DNA methylation patterns, thus drugs should be further designed to specifically increase methylation without triggering hypermethylation-related tumorigenesis. 3.2 MicroRNAs and lncRNAs-targeting therapeutics MicroRNAs have been demonstrated closely associated with autoimmune diseases in terms of diagnosis marks as well as therapeutic targets. The modulation of miRNA expression by using miRNA mimics or inhibitors might, therefore, represent attractive future opportunities for autoimmune disease treatment. A study by Tang et al. has demonstrated that reduced level of miR-146a, which directly targets IRF5 and STAT1, increases the induction of type I interferon in PBMCs from patients with SLE. In clinical trials, 5 patients with SLE who had high interferon scores were transfected with miR-146a expression plasmid. After 24 hours, PBMCs from the patients were tested for the mRNA levels of the selected interferon-inducible genes. As expected, the expressions of selected genes were notably reduced by miR-146a, indicating that manipulation of miR-146a levels could potentially provide a therapeutic benefit to SLE patients, even though the alleviation of related symptoms need to be further determined[86]. As for strategies of miRNA inhibition, miRNA sponges, small-molecule inhibitors and antisense oligonucleotides are main approaches that have been taken (Figure 1). Termed miRNA sponges, also known as vector-based strategies, are constructed based on mRNA sequences containing multiple artificial miRNA-binding sites which could act as decoy or ‘sponges’. When vectors encoding these sponges are transfected into cultured cells, sponges could selectively deplete endogenous miRNAs and thereby allow translation of the target mRNAs[87]. A study by Du et al. has shown that in vivo silencing of miR-326 by miRNA sponges caused a decrease in the number of Th17 cells and disease severity and vice versa, indicating that miR-326 plays an important role in Th17 polarization and the pathogenesis of MS[88]. Interestingly, it seems that some lncRNAs could serve as natural sponges to abrogate miRNA availability and lead to upregulation of downstream target genes. For instance, lnc-ATB was found to be competing endogenous RNAs (ceRNA) binding the miR-200 family, which might provide inspiration for further investigations [71]. Small-molecule inhibitors might target multiple steps of miRNA assembly and function, including transcription of primary miRNAs, the formation of miRNA-induced silencing complex (miRISC) and interactions between miRISC and target mRNA. It has been reported that azobenzene could affect miR-21 expression and several compounds exhibit inhibitory effect on miR-122 expression [89, 90]. Small-molecule inhibitors targeting miRNAs are less used in research on autoimmune diseases but provide alternative experimental methods. The miRNA-targeting antisense oligonucleotides (anti-miRs) are designed to be high complementary to the target miRNA. When delivered into cultured cells, the anti-miRs could bind to and specifically suppress target miRNAs function, blocking their inhibitory effect on the expression of endogenous target genes. Moreover, many anti-miRs have been demonstrated to induce degradation of targeted miRNAs. Recently, anti-miRs delivered by lentiviral vectors have been widely used to investigate miRNA function. For instance, recombinant lentiviral vectors silencing miR-223 was constructed and intraperitoneally injected into mice with collagen-induced arthritis. The anti-miR-223-treated mice showed the amelioration of score and incidence of arthritis, histopathologic examination, as well as the attenuation of osteoclastogenesis and bone erosion in joints[91]. In clinical trials, miR-122-targeted therapy in chronic hepatitis C patients has been reported to be long-term safe and effective [92, 93]. Furthermore, miRNA-masking (miR-mask) technology is another sort of antisense oligonucleotides approaches. Unlike anti-miRs directly binding to the target miRNAs, a miR-mask covers up the sites on mRNAs where are fully complementary to target miRNA, derepressing its target mRNAs [94]. In summary, antisense oligonucleotides technology provides valuable experimental platforms and opens up new avenues of clinical treatments of autoimmune diseases. 8
ACCEPTED MANUSCRIPT
TE
D
MA
NU
SC R
IP
T
4. Conclusion & future perspective Ranging from candidate-gene to genome-wide association studies, a large number of susceptibility genes have been identified in the pathogenesis of autoimmune diseases. However, genetic factors are unable to fully account for the risk of autoimmune diseases. A growing series of evidences suggest that gene function depends on not only DNA sequences, but also the epigenetic modifications, including DNA methylation, histone modification, non-coding RNA and their cross-talks, which are thought to be crossroads between environment and genetics. The endeavor to completely understand molecular mechanisms governing epigenetic alterations will provide more research orientations. It is noteworthy that epigenetic alterations vary among different tissue and cell types. Each cell type could be characterized by a particular epigenome that are associated with a specific gene expression profile, for instance, epigenome-wide profile analysis showed DNA methylation patterns varying dramatically between T and B lymphocytes[95]. Furthermore, different environment exposed and roles in pathogenesis could also contribute to their distinction in epigenetic alterations, so that appropriate selection of cell or tissue types for analysis is particularly required. Autoimmune diseases share many features, and a multitude of studies have revealed the existence of many common susceptibility genes and epigenetic alterations. It is viable that different kinds of autoimmune diseases could acquire useful references from each other when it comes to researching into pathogenesis. Meanwhile, the discovery of shared aspects of disease pathogenesis could offer the opportunity to use known drugs or therapeutic strategies across multiple diseases. Drugs with epigenetic effects, such as HDAC inhibitors and DNMT inhibitors, have been applied on animal models and even in clinical trials. Furthermore, with the rapid development of basic and clinical studies, microRNAs-targeting therapeutics by mimics or inhibitors has become a valuable and promising way for the modulation of multiple aspects of human diseases. We are optimistic that taking individual epigenetic variation into account will lead to a more personalized application of treatment for autoimmune diseases in the coming years.
CE P
Conflict of interest The authors declare that they have no competing financial interests.
AC
Take-home messages ·Aberrant epigenetic alterations, including DNA methylation, histone modification and non-coding RNAs expression which could lead to direct effects on expression of susceptibility genes as well as cross-talks with each other, occur in lymphocytes and other cell types from patients, contributing to the pathogenesis of autoimmune diseases. ·Drugs with epigenetic effects, such as HDAC inhibitors and DNMT inhibitors, have been applied on animal models and even in clinical trials, displaying positive therapeutic effect. ·The microRNAs-targeting therapeutics by mimics or inhibitors provides a valuable and promising way for the modulation of multiple aspects of human diseases. A range of miRNA-targeting therapeutic is in now at preclinical stage or even in clinical trials. ·With the rapid development of microRNAs-targeting therapeutics, taking individual epigenetic variation into account will lead to a more personalized application of treatment for autoimmune diseases in the coming years. Acknowledgment This work is supported by the Ministry of Science and Technology of China through Grants No 2012CB932503 and 2011CB933100; the National Natural Science Foundation of China through Grants No 91029705, 81172864 and 81272317. Grant TD12-5025 and 14JCTPJC00487 from Tianjin city. References 9
ACCEPTED MANUSCRIPT
[8] [9] [10] [11] [12] [13] [14] [15] [16]
[17] [18]
[19]
[20] [21]
T
IP
SC R
NU
[7]
MA
[5] [6]
D
[4]
TE
[3]
CE P
[2]
A. Zhernakova, S. Withoff and C. Wijmenga. Clinical implications of shared genetics and pathogenesis in autoimmune diseases. Nat Rev Endocrinol 2013; 9: 646-59. J.M. Anaya, L. Gomez and J. Castiblanco. Is there a common genetic basis for autoimmune diseases? Clin Dev Immunol 2006; 13: 185-95. B.A. Lie and E. Thorsby. Several genes in the extended human MHC contribute to predisposition to autoimmune diseases. Curr Opin Immunol 2005; 17: 526-31. E. Ballestar. Epigenetics lessons from twins: prospects for autoimmune disease. Clin Rev Allergy Immunol 2010; 39: 30-41. A. Bird. Perceptions of epigenetics. Nature 2007; 447: 396-8. K.H. Costenbader, S. Gay, M.E. Alarcon-Riquelme, L. Iaccarino and A. Doria. Genes, epigenetic regulation and environmental factors: which is the most relevant in developing autoimmune diseases? Autoimmun Rev 2012; 11: 604-9. E.W. Karlson, S.C. Chang, J. Cui, L.B. Chibnik, P.A. Fraser, I. De Vivo, et al. Gene-environment interaction between HLA-DRB1 shared epitope and heavy cigarette smoking in predicting incident rheumatoid arthritis. Ann Rheum Dis 2010; 69: 54-60. M.A. Hernan, M.J. Olek and A. Ascherio. Cigarette smoking and incidence of multiple sclerosis. Am J Epidemiol 2001; 154: 69-74. K.L. Munger, L.I. Levin, B.W. Hollis, N.S. Howard and A. Ascherio. Serum 25-hydroxyvitamin D levels and risk of multiple sclerosis. JAMA 2006; 296: 2832-8. K.D. Deane and H. El-Gabalawy. Pathogenesis and prevention of rheumatic disease: focus on preclinical RA and SLE. Nat Rev Rheumatol 2014; 10: 212-28. H. Guo, P. Zhu, L. Yan, R. Li, B. Hu, Y. Lian, et al. The DNA methylation landscape of human early embryos. Nature 2014; 511: 606-10. M. Ehrlich and M. Lacey. DNA methylation and differentiation: silencing, upregulation and modulation of gene expression. Epigenomics 2013; 5: 553-68. J. Arand, D. Spieler, T. Karius, M.R. Branco, D. Meilinger, A. Meissner, et al. In vivo control of CpG and non-CpG DNA methylation by DNA methyltransferases. PLoS Genet 2012; 8: e1002750. S.K. Ooi and T.H. Bestor. The colorful history of active DNA demethylation. Cell 2008; 133: 1145-8. E.L. Fritz and F.N. Papavasiliou. Cytidine deaminases: AIDing DNA demethylation? Genes Dev 2010; 24: 2107-14. E.L. Fritz, B.R. Rosenberg, K. Lay, A. Mihailovic, T. Tuschl and F.N. Papavasiliou. A comprehensive analysis of the effects of the deaminase AID on the transcriptome and methylome of activated B cells. Nat Immunol 2013; 14: 749-55. T. Detanico, W. Guo and L.J. Wysocki. Predominant role for activation-induced cytidine deaminase in generating IgG anti-nucleosomal antibodies of murine SLE. J Autoimmun 2015; 58: 67-77. B. Richardson, L. Scheinbart, J. Strahler, L. Gross, S. Hanash and M. Johnson. Evidence for impaired T cell DNA methylation in systemic lupus erythematosus and rheumatoid arthritis. Arthritis Rheum 1990; 33: 1665-73. W. Lei, Y. Luo, K. Yan, S. Zhao, Y. Li, X. Qiu, et al. Abnormal DNA methylation in CD4+ T cells from patients with systemic lupus erythematosus, systemic sclerosis, and dermatomyositis. Scand J Rheumatol 2009; 38: 369-74. Q. Lu, A. Wu, L. Tesmer, D. Ray, N. Yousif and B. Richardson. Demethylation of CD40LG on the inactive X in T cells from women with lupus. J Immunol 2007; 179: 6352-8. X. Lian, R. Xiao, X. Hu, T. Kanekura, H. Jiang, Y. Li, 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: 2338-45.
AC
[1]
10
ACCEPTED MANUSCRIPT [22] [23]
[29]
[30]
[31] [32]
[33] [34]
[35] [36] [37] [38]
[39]
SC R
NU
MA
D
[28]
TE
[27]
CE P
[26]
AC
[25]
IP
T
[24]
J. Liao, G. Liang, S. Xie, H. Zhao, X. Zuo, F. Li, et al. CD40L demethylation in CD4(+) T cells from women with rheumatoid arthritis. Clin Immunol 2012; 145: 13-8. G. Iezzi, I. Sonderegger, F. Ampenberger, N. Schmitz, B.J. Marsland and M. Kopf. CD40-CD40L cross-talk integrates strong antigenic signals and microbial stimuli to induce development of IL-17-producing CD4+ T cells. Proc Natl Acad Sci U S A 2009; 106: 876-81. K. Oelke, Q. Lu, D. Richardson, A. Wu, C. Deng, S. Hanash, et al. Overexpression of CD70 and overstimulation of IgG synthesis by lupus T cells and T cells treated with DNA methylation inhibitors. Arthritis Rheum 2004; 50: 1850-60. H. Jiang, R. Xiao, X. Lian, T. Kanekura, Y. Luo, Y. Yin, et al. Demethylation of TNFSF7 contributes to CD70 overexpression in CD4+ T cells from patients with systemic sclerosis. Clin Immunol 2012; 143: 39-44. H. Yin, M. Zhao, X. Wu, F. Gao, Y. Luo, L. Ma, et al. Hypomethylation and overexpression of CD70 (TNFSF7) in CD4+ T cells of patients with primary Sjogren's syndrome. J Dermatol Sci 2010; 59: 198-203. D.M. Absher, X. Li, L.L. Waite, A. Gibson, K. Roberts, J. Edberg, et al. Genome-wide DNA methylation analysis of systemic lupus erythematosus reveals persistent hypomethylation of interferon genes and compositional changes to CD4+ T-cell populations. PLoS Genet 2013; 9: e1003678. N. Altorok, P. Coit, T. Hughes, K.A. Koelsch, D.U. Stone, A. Rasmussen, et al. Genome-wide DNA methylation patterns in naive CD4+ T cells from patients with primary Sjogren's syndrome. Arthritis Rheumatol 2014; 66: 731-9. P. Coit, M. Jeffries, N. Altorok, M.G. Dozmorov, K.A. Koelsch, J.D. Wren, et al. Genome-wide DNA methylation study suggests epigenetic accessibility and transcriptional poising of interferon-regulated genes in naive CD4+ T cells from lupus patients. J Autoimmun 2013; 43: 78-84. S. Garaud, C. Le Dantec, S. Jousse-Joulin, C. Hanrotel-Saliou, A. Saraux, R.A. Mageed, et al. IL-6 modulates CD5 expression in B cells from patients with lupus by regulating DNA methylation. J Immunol 2009; 182: 5623-32. T. Fali, C. Le Dantec, Y. Thabet, S. Jousse, C. Hanrotel, P. Youinou, et al. DNA methylation modulates HRES1/p28 expression in B cells from patients with Lupus. Autoimmunity 2014; 47: 265-71. R. Belkhir, N. Gestermann, M. Koutero, R. Seror, J. Tost, X. Mariette, et al. Upregulation of membrane-bound CD40L on CD4+ T cells in women with primary Sjogren's syndrome. Scand J Immunol 2014; 79: 37-42. N. Gestermann, M. Koutero, R. Belkhir, J. Tost, X. Mariette and C. Miceli-Richard. Methylation profile of the promoter region of IRF5 in primary Sjogren's syndrome. Eur Cytokine Netw 2012; 23: 166-72. Y.Y. Wang, Q. Wang, X.H. Sun, R.Z. Liu, Y. Shu, T. Kanekura, 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: 39-47. Y. Li, M. Zhao, C. Hou, G. Liang, L. Yang, Y. Tan, et al. Abnormal DNA methylation in CD4+ T cells from people with latent autoimmune diabetes in adults. Diabetes Res Clin Pract 2011; 94: 242-8. P.C. Janson, L.B. Linton, E.A. Bergman, P. Marits, M. Eberhardson, F. Piehl, et al. Profiling of CD4+ T cells with epigenetic immune lineage analysis. J Immunol 2011; 186: 92-102. Y. Wang, P.S. Fan and B. Kahaleh. Association between enhanced type I collagen expression and epigenetic repression of the FLI1 gene in scleroderma fibroblasts. Arthritis Rheum 2006; 54: 2271-9. C. Dees, I. Schlottmann, R. Funke, A. Distler, K. Palumbo-Zerr, P. Zerr, et al. The Wnt antagonists DKK1 and SFRP1 are downregulated by promoter hypermethylation in systemic sclerosis. Ann Rheum Dis 2014; 73: 1232-9. M. Trenkmann, M. Brock, R.E. Gay, C. Kolling, R. Speich, B.A. Michel, et al. Expression and function 11
ACCEPTED MANUSCRIPT
[40]
[48] [49]
[50] [51] [52]
[53] [54] [55]
[56] [57]
[58] [59]
SC R
NU
MA
[47]
D
[45] [46]
TE
[44]
CE P
[43]
AC
[42]
IP
T
[41]
of EZH2 in synovial fibroblasts: epigenetic repression of the Wnt inhibitor SFRP1 in rheumatoid arthritis. Ann Rheum Dis 2011; 70: 1482-8. C.J. Nile, R.C. Read, M. Akil, G.W. Duff and A.G. Wilson. Methylation status of a single CpG site in the IL6 promoter is related to IL6 messenger RNA levels and rheumatoid arthritis. Arthritis Rheum 2008; 58: 2686-93. N. Takami, K. Osawa, Y. Miura, K. Komai, M. Taniguchi, M. Shiraishi, et al. Hypermethylated promoter region of DR3, the death receptor 3 gene, in rheumatoid arthritis synovial cells. Arthritis Rheum 2006; 54: 779-87. S.E. Baranzini, J. Mudge, J.C. van Velkinburgh, P. Khankhanian, I. Khrebtukova, N.A. Miller, et al. Genome, epigenome and RNA sequences of monozygotic twins discordant for multiple sclerosis. Nature 2010; 464: 1351-6. F.G. Mastronardi, A. Noor, D.D. Wood, T. Paton and M.A. Moscarello. Peptidyl argininedeiminase 2 CpG island in multiple sclerosis white matter is hypomethylated. J Neurosci Res 2007; 85: 2006-16. A.A. Musse, J.M. Boggs and G. Harauz. Deimination of membrane-bound myelin basic protein in multiple sclerosis exposes an immunodominant epitope. Proc Natl Acad Sci U S A 2006; 103: 4422-7. T. Kouzarides. Chromatin modifications and their function. Cell 2007; 128: 693-705. N. Hu, X. Qiu, Y. Luo, J. Yuan, Y. Li, W. Lei, et al. Abnormal histone modification patterns in lupus CD4+ T cells. J Rheumatol 2008; 35: 804-10. Y. Zhou, X. Qiu, Y. Luo, J. Yuan, Y. Li, Q. Zhong, et al. Histone modifications and methyl-CpG-binding domain protein levels at the TNFSF7 (CD70) promoter in SLE CD4+ T cells. Lupus 2011; 20: 1365-71. Y. Wang, Y. Yang, Y. Luo, Y. Yin, Q. Wang, Y. Li, et al. Aberrant histone modification in peripheral blood B cells from patients with systemic sclerosis. Clin Immunol 2013; 149: 46-54. X. Pedre, F. Mastronardi, W. Bruck, G. Lopez-Rodas, T. Kuhlmann and P. Casaccia. Changed histone acetylation patterns in normal-appearing white matter and early multiple sclerosis lesions. J Neurosci 2011; 31: 3435-45. F. Miao, Z. Chen, L. Zhang, Z. Liu, X. Wu, Y.C. Yuan, et al. Profiles of epigenetic histone post-translational modifications at type 1 diabetes susceptible genes. J Biol Chem 2012; 287: 16335-45. M. Kramer, C. Dees, J. Huang, I. Schlottmann, K. Palumbo-Zerr, P. Zerr, et al. Inhibition of H3K27 histone trimethylation activates fibroblasts and induces fibrosis. Ann Rheum Dis 2013; 72: 614-20. F. Miao, D.D. Smith, L. Zhang, A. Min, W. Feng and R. Natarajan. Lymphocytes from patients with type 1 diabetes display a distinct profile of chromatin histone H3 lysine 9 dimethylation: an epigenetic study in diabetes. Diabetes 2008; 57: 3189-98. J.S. Mattick. Non-coding RNAs: the architects of eukaryotic complexity. EMBO Rep 2001; 2: 986-91. C.Z. Chen, S. Schaffert, R. Fragoso and C. Loh. Regulation of immune responses and tolerance: the microRNA perspective. Immunol Rev 2013; 253: 112-28. W. Pan, S. Zhu, M. Yuan, H. Cui, L. Wang, X. Luo, et al. MicroRNA-21 and microRNA-148a contribute to DNA hypomethylation in lupus CD4+ T cells by directly and indirectly targeting DNA methyltransferase 1. J Immunol 2010; 184: 6773-81. X.N. Wu, Y.X. Ye, J.W. Niu, Y. Li, X. Li, X. You, et al. Defective PTEN regulation contributes to B cell hyperresponsiveness in systemic lupus erythematosus. Sci Transl Med 2014; 6: 246ra99. E. Stagakis, G. Bertsias, P. Verginis, M. Nakou, M. Hatziapostolou, H. Kritikos, et al. Identification of novel microRNA signatures linked to human lupus disease activity and pathogenesis: miR-21 regulates aberrant T cell responses through regulation of PDCD4 expression. Ann Rheum Dis 2011; 70: 1496-506. H. Zhu, H. Luo, Y. Li, Y. Zhou, Y. Jiang, J. Chai, et al. MicroRNA-21 in scleroderma fibrosis and its function in TGF-beta-regulated fibrosis-related genes expression. J Clin Immunol 2013; 33: 1100-9. G. Murugaiyan, A.P.d. Cunha, A.K. Ajay, N. Joller, L.P. Garo, S. Kumaradevan, et al. MicroRNA-21 12
ACCEPTED MANUSCRIPT
[60]
[67] [68] [69] [70] [71]
[72]
[73] [74]
[75] [76] [77] [78]
SC R
NU
MA
[66]
D
[65]
TE
[64]
CE P
[63]
AC
[62]
IP
T
[61]
promotes Th17 differentiation and mediates experimental autoimmune encephalomyelitis. The Journal of Clinical Investigation 2015. L. Dong, X. Wang, J. Tan, H. Li, W. Qian, J. Chen, et al. Decreased expression of microRNA-21 correlates with the imbalance of Th17 and Treg cells in patients with rheumatoid arthritis. J Cell Mol Med 2014; 18: 2213-24. Q. Ruan, T. Wang, V. Kameswaran, Q. Wei, D.S. Johnson, F. Matschinsky, et al. The microRNA-21-PDCD4 axis prevents type 1 diabetes by blocking pancreatic beta cell death. Proc Natl Acad Sci U S A 2011; 108: 12030-5. H. Qin, X. Zhu, J. Liang, J. Wu, Y. Yang, S. Wang, et al. MicroRNA-29b contributes to DNA hypomethylation of CD4+ T cells in systemic lupus erythematosus by indirectly targeting DNA methyltransferase 1. J Dermatol Sci 2013; 69: 61-7. K.M. Smith, M. Guerau-de-Arellano, S. Costinean, J.L. Williams, A. Bottoni, G. Mavrikis Cox, et al. miR-29ab1 deficiency identifies a negative feedback loop controlling Th1 bias that is dysregulated in multiple sclerosis. J Immunol 2012; 189: 1567-76. B. Maurer, J. Stanczyk, A. Jungel, A. Akhmetshina, M. Trenkmann, M. Brock, et al. MicroRNA-29, a key regulator of collagen expression in systemic sclerosis. Arthritis Rheum 2010; 62: 1733-43. A.L. Carlsen, A.J. Schetter, C.T. Nielsen, C. Lood, S. Knudsen, A. Voss, et al. Circulating microRNA expression profiles associated with systemic lupus erythematosus. Arthritis Rheum 2013; 65: 1324-34. M. Filkova, B. Aradi, L. Senolt, C. Ospelt, S. Vettori, H. Mann, et al. Association of circulating miR-223 and miR-16 with disease activity in patients with early rheumatoid arthritis. Ann Rheum Dis 2014; 73: 1898-904. X. Ma, J. Zhou, Y. Zhong, L. Jiang, P. Mu, Y. Li, et al. Expression, regulation and function of microRNAs in multiple sclerosis. Int J Med Sci 2014; 11: 810-8. J.A. Heward and M.A. Lindsay. Long non-coding RNAs in the regulation of the immune response. Trends Immunol 2014; 35: 408-19. P. Wang, Y. Xue, Y. Han, L. Lin, C. Wu, S. Xu, et al. The STAT3-binding long noncoding RNA lnc-DC controls human dendritic cell differentiation. Science 2014; 344: 310-3. S. Carpenter, D. Aiello, M.K. Atianand, E.P. Ricci, P. Gandhi, L.L. Hall, et al. A long noncoding RNA mediates both activation and repression of immune response genes. Science 2013; 341: 789-92. J.H. Yuan, F. Yang, F. Wang, J.Z. Ma, Y.J. Guo, Q.F. Tao, et al. A long noncoding RNA activated by TGF-beta promotes the invasion-metastasis cascade in hepatocellular carcinoma. Cancer Cell 2014; 25: 666-81. S. Sharma, G.M. Findlay, H.S. Bandukwala, S. Oberdoerffer, B. Baust, Z. Li, et al. Dephosphorylation of the nuclear factor of activated T cells (NFAT) transcription factor is regulated by an RNA-protein scaffold complex. Proc Natl Acad Sci U S A 2011; 108: 11381-6. N.A. Rapicavoli, K. Qu, J. Zhang, M. Mikhail, R.M. Laberge and H.Y. Chang. A mammalian pseudogene lncRNA at the interface of inflammation and anti-inflammatory therapeutics. Elife 2013; 2: e00762. F. Modarresi, M.A. Faghihi, M.A. Lopez-Toledano, R.P. Fatemi, M. Magistri, S.P. Brothers, et al. Inhibition of natural antisense transcripts in vivo results in gene-specific transcriptional upregulation. Nat Biotechnol 2012; 30: 453-9. C. Carrieri, L. Cimatti, M. Biagioli, A. Beugnet, S. Zucchelli, S. Fedele, et al. Long non-coding antisense RNA controls Uchl1 translation through an embedded SINEB2 repeat. Nature 2012; 491: 454-7. I. Ulitsky and D.P. Bartel. lincRNAs: genomics, evolution, and mechanisms. Cell 2013; 154: 26-46. B. Huang, C. Jiang and R. Zhang. Epigenetics: the language of the cell? Epigenomics 2014; 6: 73-88. C.M. Hedrich, T. Rauen, S.A. Apostolidis, A.P. Grammatikos, N. Rodriguez Rodriguez, C. Ioannidis, et al. Stat3 promotes IL-10 expression in lupus T cells through trans-activation and chromatin remodeling. Proc 13
ACCEPTED MANUSCRIPT
[85] [86]
[87] [88]
[89] [90]
[91]
[92] [93]
[94] [95]
T
IP
SC R
NU
[84]
MA
[83]
D
[82]
TE
[81]
CE P
[80]
Natl Acad Sci U S A 2014; 111: 13457-62. H. Hemmatazad, H.M. Rodrigues, B. Maurer, F. Brentano, M. Pileckyte, J.H. Distler, et al. Histone deacetylase 7, a potential target for the antifibrotic treatment of systemic sclerosis. Arthritis Rheum 2009; 60: 1519-29. A.M. Grabiec, O. Korchynskyi, P.P. Tak and K.A. Reedquist. Histone deacetylase inhibitors suppress rheumatoid arthritis fibroblast-like synoviocyte and macrophage IL-6 production by accelerating mRNA decay. Ann Rheum Dis 2012; 71: 424-31. N. Mishra, D.R. Brown, I.M. Olorenshaw and G.M. Kammer. Trichostatin A reverses skewed expression of CD154, interleukin-10, and interferon-gamma gene and protein expression in lupus T cells. Proc Natl Acad Sci U S A 2001; 98: 2628-33. T. Patel, V. Patel, R. Singh and S. Jayaraman. Chromatin remodeling resets the immune system to protect against autoimmune diabetes in mice. Immunol Cell Biol 2011; 89: 640-9. J. Vojinovic, N. Damjanov, C. D'Urzo, A. Furlan, G. Susic, S. Pasic, et al. Safety and efficacy of an oral histone deacetylase inhibitor in systemic-onset juvenile idiopathic arthritis. Arthritis Rheum 2011; 63: 1452-8. M.W. Chan, C.B. Chang, C.H. Tung, J. Sun, J.L. Suen and S.F. Wu. Low-dose 5-aza-2'-deoxycytidine pretreatment inhibits experimental autoimmune encephalomyelitis by induction of regulatory T cells. Mol Med 2014; 20: 248-56. M. Kroesen, P. Gielen, I.C. Brok, I. Armandari, P.M. Hoogerbrugge and G.J. Adema. HDAC inhibitors and immunotherapy; a double edged sword? Oncotarget 2014; 5: 6558-72. Y. Tang, X. Luo, H. Cui, X. Ni, M. Yuan, Y. Guo, et al. MicroRNA-146A contributes to abnormal activation of the type I interferon pathway in human lupus by targeting the key signaling proteins. Arthritis Rheum 2009; 60: 1065-75. M.S. Ebert, J.R. Neilson and P.A. Sharp. MicroRNA sponges: competitive inhibitors of small RNAs in mammalian cells. Nat Methods 2007; 4: 721-6. C. Du, C. Liu, J. Kang, G. Zhao, Z. Ye, S. Huang, et al. MicroRNA miR-326 regulates TH-17 differentiation and is associated with the pathogenesis of multiple sclerosis. Nat Immunol 2009; 10: 1252-9. K. Gumireddy, D.D. Young, X. Xiong, J.B. Hogenesch, Q. Huang and A. Deiters. Small-molecule inhibitors of microrna miR-21 function. Angew Chem Int Ed Engl 2008; 47: 7482-4. D.D. Young, C.M. Connelly, C. Grohmann and A. Deiters. Small molecule modifiers of microRNA miR-122 function for the treatment of hepatitis C virus infection and hepatocellular carcinoma. J Am Chem Soc 2010; 132: 7976-81. Y.T. Li, S.Y. Chen, C.R. Wang, M.F. Liu, C.C. Lin, I.M. Jou, et al. Brief report: amelioration of collagen-induced arthritis in mice by lentivirus-mediated silencing of microRNA-223. Arthritis Rheum 2012; 64: 3240-5. H.L. Janssen, H.W. Reesink, E.J. Lawitz, S. Zeuzem, M. Rodriguez-Torres, K. Patel, et al. Treatment of HCV infection by targeting microRNA. N Engl J Med 2013; 368: 1685-94. M.H. van der Ree, A.J. van der Meer, J. de Bruijne, R. Maan, A. van Vliet, T.M. Welzel, et al. Long-term safety and efficacy of microRNA-targeted therapy in chronic hepatitis C patients. Antiviral Res 2014; 111C: 53-59. Z. Wang. The principles of MiRNA-masking antisense oligonucleotides technology. Methods Mol Biol 2011; 676: 43-9. J.R. Glossop, N.B. Nixon, R.D. Emes, K.E. Haworth, J.C. Packham, P.T. Dawes, et al. Epigenome-wide profiling identifies significant differences in DNA methylation between matched-pairs of T- and B-lymphocytes from healthy individuals. Epigenetics 2013; 8: 1188-97.
AC
[79]
14
ACCEPTED MANUSCRIPT
[102]
[103] [104]
[105] [106]
[107]
[108] [109] [110]
[111]
[112]
[113]
T
IP
SC R
NU
[101]
MA
[100]
D
[99]
TE
[98]
CE P
[97]
M. Graves, M. Benton, R. Lea, M. Boyle, L. Tajouri, D. Macartney-Coxson, et al. Methylation differences at the HLA-DRB1 locus in CD4+ T-Cells are associated with multiple sclerosis. Mult Scler 2013; 20: 1033-1041. M. Zhao, J. Tang, F. Gao, X. Wu, Y. Liang, H. Yin, et al. Hypomethylation of IL10 and IL13 promoters in CD4+ T cells of patients with systemic lupus erythematosus. J Biomed Biotechnol 2010; 2010: 931018. Q. Lu, M. Kaplan, D. Ray, S. Zacharek, D. Gutsch and B. Richardson. Demethylation of ITGAL (CD11a) regulatory sequences in systemic lupus erythematosus. Arthritis Rheum 2002; 46: 1282-91. M.J. Kaplan, Q. Lu, A. Wu, J. Attwood and B. Richardson. Demethylation of promoter regulatory elements contributes to perforin overexpression in CD4+ lupus T cells. J Immunol 2004; 172: 3652-61. B.M. Javierre, A.F. Fernandez, J. Richter, F. Al-Shahrour, J.I. Martin-Subero, J. Rodriguez-Ubreva, et al. Changes in the pattern of DNA methylation associate with twin discordance in systemic lupus erythematosus. Genome Res 2010; 20: 170-9. C. Kumagai, B. Kalman, F.A. Middleton, T. Vyshkina and P.T. Massa. Increased promoter methylation of the immune regulatory gene SHP-1 in leukocytes of multiple sclerosis subjects. J Neuroimmunol 2012; 246: 51-7. V.K. Rakyan, H. Beyan, T.A. Down, M.I. Hawa, S. Maslau, D. Aden, et al. Identification of type 1 diabetes-associated DNA methylation variable positions that precede disease diagnosis. PLoS Genet 2011; 7: e1002300. C.G. Miao, Y.Y. Yang, X. He, C. Huang, Y. Huang, D. Qin, et al. MicroRNA-152 modulates the canonical Wnt pathway activation by targeting DNA methyltransferase 1 in arthritic rat model. Biochimie 2014. S. Gonzalez, S. Aguilera, C. Alliende, U. Urzua, A.F. Quest, L. Herrera, et al. Alterations in type I hemidesmosome components suggestive of epigenetic control in the salivary glands of patients with Sjogren's syndrome. Arthritis Rheum 2011; 63: 1106-15. Z. Zhang, L. Song, K. Maurer, M.A. Petri and K.E. Sullivan. Global H4 acetylation analysis by ChIP-chip in systemic lupus erythematosus monocytes. Genes Immun 2010; 11: 124-33. F. Niederer, C. Ospelt, F. Brentano, M.O. Hottiger, R.E. Gay, S. Gay, et al. SIRT1 overexpression in the rheumatoid arthritis synovium contributes to proinflammatory cytokine production and apoptosis resistance. Ann Rheum Dis 2011; 70: 1866-73. K. Makino, M. Jinnin, A. Hirano, K. Yamane, M. Eto, T. Kusano, 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: 3905-15. C.B. Chafin, N.L. Regna, D.L. Caudell and C.M. Reilly. MicroRNA-let-7a promotes E2F-mediated cell proliferation and NFkappaB activation in vitro. Cell Mol Immunol 2014; 11: 79-83. E. Roggli, S. Gattesco, D. Caille, C. Briet, C. Boitard, P. Meda, et al. Changes in microRNA expression contribute to pancreatic beta-cell dysfunction in prediabetic NOD mice. Diabetes 2012; 61: 1742-51. Y. Liu, J. Dong, R. Mu, Y. Gao, X. Tan, Y. Li, et al. MicroRNA-30a promotes B cell hyperactivity in patients with systemic lupus erythematosus by direct interaction with Lyn. Arthritis Rheum 2013; 65: 1603-11. G. Alsaleh, A. Francois, L. Philippe, Y.Z. Gong, S. Bahram, S. Cetin, et al. MiR-30a-3p Negatively Regulates BAFF Synthesis in Systemic Sclerosis and Rheumatoid Arthritis Fibroblasts. PLoS One 2014; 9: e111266. S. Tanaka, A. Suto, K. Ikeda, Y. Sanayama, D. Nakagomi, T. Iwamoto, et al. Alteration of circulating miRNAs in SSc: miR-30b regulates the expression of PDGF receptor beta. Rheumatology (Oxford) 2013; 52: 1963-72. Q. Zhou, S. Haupt, J.T. Kreuzer, A. Hammitzsch, F. Proft, C. Neumann, et al. Decreased expression of miR-146a and miR-155 contributes to an abnormal Treg phenotype in patients with rheumatoid arthritis.
AC
[96]
15
ACCEPTED MANUSCRIPT
[118]
[119]
T
CE P
[122]
AC
[121]
TE
D
[120]
IP
[117]
SC R
[116]
NU
[115]
Ann Rheum Dis 2014. K.M. Pauley, C.M. Stewart, A.E. Gauna, L.C. Dupre, R. Kuklani, A.L. Chan, et al. Altered miR-146a expression in Sjogren's syndrome and its functional role in innate immunity. Eur J Immunol 2011; 41: 2029-39. C.S. Moore, V.T. Rao, B.A. Durafourt, B.J. Bedell, S.K. Ludwin, A. Bar-Or, et al. miR-155 as a multiple sclerosis-relevant regulator of myeloid cell polarization. Ann Neurol 2013; 74: 709-20. A. Junker, M. Krumbholz, S. Eisele, H. Mohan, F. Augstein, R. Bittner, et al. MicroRNA profiling of multiple sclerosis lesions identifies modulators of the regulatory protein CD47. Brain 2009; 132: 3342-52. J. Stanczyk, D.M. Pedrioli, F. Brentano, O. Sanchez-Pernaute, C. Kolling, R.E. Gay, et al. Altered expression of MicroRNA in synovial fibroblasts and synovial tissue in rheumatoid arthritis. Arthritis Rheum 2008; 58: 1001-9. J. Gillespie, S. Savic, C. Wong, A. Hempshall, M. Inman, P. Emery, et al. Histone deacetylases are dysregulated in rheumatoid arthritis and a novel histone deacetylase 3-selective inhibitor reduces interleukin-6 production by peripheral blood mononuclear cells from rheumatoid arthritis patients. Arthritis Rheum 2012; 64: 418-22. Z. Ge, Y. Da, Z. Xue, K. Zhang, H. Zhuang, M. Peng, et al. Vorinostat, a histone deacetylase inhibitor, suppresses dendritic cell function and ameliorates experimental autoimmune encephalomyelitis. Exp Neurol 2013; 241: 56-66. Q.Y. Choo, P.C. Ho, Y. Tanaka and H.S. Lin. Histone deacetylase inhibitors MS-275 and SAHA induced growth arrest and suppressed lipopolysaccharide-stimulated NF-kappaB p65 nuclear accumulation in human rheumatoid arthritis synovial fibroblastic E11 cells. Rheumatology (Oxford) 2010; 49: 1447-60. S.J. Saouaf, B. Li, G. Zhang, Y. Shen, N. Furuuchi, W.W. Hancock, et al. Deacetylase inhibition increases regulatory T cell function and decreases incidence and severity of collagen-induced arthritis. Exp Mol Pathol 2009; 87: 99-104. E.C. Lewis, L. Blaabjerg, J. Storling, S.G. Ronn, P. Mascagni, C.A. Dinarello, et al. The oral histone deacetylase inhibitor ITF2357 reduces cytokines and protects islet beta cells in vivo and in vitro. Mol Med 2011; 17: 369-77.
MA
[114]
16
ACCEPTED MANUSCRIPT Table 1. Epigenetic alterations in autoimmune diseases at the level of DNA methylation Genes
Epigenetic alteration
CD40LG
Hypomethylation
CD40L, B cell costimulatory molecule encoded [20-22] on the X chromosome
CD70
Hypomethylation
CD70, B cell costimulatory molecule associated [24-26] with overproduction of IgG
SSc, T1DM, RA,
FOXP3
Hypermethylation Forkhead box protein 3, involved in quantitative [34-36] defects of regulatory T cells.
MS
HLA-DRB1
Hypomethylation
HLA class II beta chain
SLE
IL10, IL13
Hypomethylation
Involved in autoantibody production and tissue [78, 97] damage
SLE
IRF5, IFIT2
Hypomethylation
Involved in type I interferon pathway
[27]
SLE
ITGAL
Hypomethylation
Integrin α-L, associated with cell-cell adhesion
[98]
SLE
PRF1
Hypomethylation
Perforin 1, involved in autoreactive killing
[99]
SLE, pSS
STAT1, IFI44L, USP18
Hypomethylation
Involved in type I interferon pathway
[28, 29]
pSS
LTA
Hypomethylation
pSS
RUNX1
Hypermethylation Transcription factor associated to lymphoma
SLE
CD5
Hypomethylation
SLE
HRES-1
Hypomethylation
RA
IL6
SLE
IFNGR2, MMP14
B cell
PBMC
SLE
CD14+ monocyte
T
IP
SC R
NU
Lymphotoxin-α
[96]
[28] [28]
CD5, involved in activation and expansion of [30] autoreactive B cells Human endogenous retroviruses proteins, involved [31] in induction of cross-reactive autoantibodies
Hypomethylation
IL-6, involved in B cell response
Hypomethylation
IFN-γ receptor 1, Matrix metalloproteinase-14, [100] involved in inflammation
LCN2
Hypomethylation
Neutrophil gelatinase-associated lipocalin, iron [100] transporter and marker for SLE
SHP-1
Hypermethylation A negative regulator of cytokine signaling through [101] NF-κB and STATs
AC
MS
MA
Naïve CD4 T cell
+
References
D
Peripheral SLE, RA, blood CD4+ SSc T cell SLE, SSc, pSS
Product and/or function
TE
Disease
CE P
Cell type
[40]
T1DM
HLA-DQB1
Hypomethylation
HLA class II
[102]
T1DM
RFXAP
Hypomethylation
HLA class II regulating element
[102]
T1DM
NFKB1A
Hypomethylation
Regulator of apoptosis and inflammation
[102]
T1DM
GAD2
Hypomethylation
GAD65, a major autoantigen involved in T1D
[102]
T1DM
TNF
Hypermethylation Key inflammatory cytokine
T1DM
CD6
Hypermethylation Involved in lymphocyte activation and differentiation [102]
Fibroblast
SSc
FLI1
Hypermethylation Involved in type I collagen expression
[37]
Fibroblast, PBMC
SSc
DKK1, SFRP1
Hypermethylation Wnt signaling antagonists
[38]
Synovial fibroblast
RA
SFRP1, SFRP4
Hypermethylation Wnt signaling antagonists
[39, 103]
RA
DR3
Hypermethylation Death receptor 3, associated with cell apoptosis
[41]
NAWM
MS
PAD2
Hypomethylation
LSG
pSS
DST
Hypermethylation BP230, bullous pemphigoid antigen 1 protein
[102]
Peptidyl argininedeiminase type II, responsible for [43] the increased citrullinated myelin basic protein [104]
Abbreviations: SLE, systemic lupus erythematosus; RA, rheumatoid arthritis; MS, multiple sclerosis; SSc, Systemic sclerosis; T1DM, type 1 diabetes; pSS, primary Sjögren's syndrome; PBMC, peripheral blood mononuclear cell; NAWM, normal appearing white matter; LSG, labial salivary gland. 17
ACCEPTED MANUSCRIPT Table 2. Epigenetic alterations in autoimmune diseases at the level of histone modification Disease Epigenetic alteration
Gene affected and expression change
References
T cell
SLE
H3 and H4 hypoacetylation, H3K9 hypomethylation
ND
[46]
SLE
Increased histone H3 acetylation at lysine 18 (H3K18ac)
IL10, upregulation
[78]
SLE
Increased H3 acetylation, dimethylated H3 lysine4 (H3K4me2)
T1DM
Increased H3K9me2
B cell
SSc
H4 hyperacetylation, decreased HDAC2 and HDAC7; ND H3K9 hypomethylation, decreased SUV39H2 (member of HMT), increased JHDM2A (member of HDM)
Monocyte
T1DM
Increased H3K9 acetylation (H3K9Ac)
SLE
Global H4 hyperacetylation
SSc
Increased H3 and H4 deacetylation
SSc
Inhibition of H3K27me3
RA RA
Synovial fibroblast
Oligodendrocyte MS
IP
CD70, upregulation
NU
SC R
CTLA4, downregulation
[47] [52] [48]
HLA-DRB1, HLA-DQB1, [50] upregulation IRF1, RFX1 and BLIMP1, [105] upregulation FLI1, downregulation
[37]
FOSL2, upregulation
[51]
Increased zeste homologue 2 (EZH2, member of HMT)
SFRP1, downregulation
[39]
Increased sirtuin 1 (Sirt1, member of HDAC)
ND
[106]
Increased histone H3 deacetylation
ND
[49]
MA
Fibroblast
T
Cell type
TE
D
Abbreviations: SLE, systemic lupus erythematosus; T1DM, type 1 diabetes; SSc, Systemic sclerosis; RA, rheumatoid arthritis; MS, multiple sclerosis; ND, not determined; HMT, histone methyltransferases; HDAC, histone deacetylase; HDM, histone demethylase.
Table 3. Common miRNAs dysregulated in autoimmune diseases Cell type
let-7a
Fibroblasts
Epigenetic alteration / Function
Disease
References
Downregulation
Increased type I collagen expression
SSc
[107]
Upregulation
Decreased E2F5 expression, increased IL6 expression, regulated by E2F2 or NF-κB
SLE
[108]
CD4+ T cell Upregulation
Repressing DNMT1 expression by targeting RASGRP1
SLE
[55]
CD4+ T cell Upregulation
Targeting and depleting enhancing TGF-β signaling
CD4+ T cell Downregulation
Increased STAT3 expression, decreased expression and STAT5/pSTAT5 protein
B cell
Upregulation
Downregulating PTEN expression, contributing to B SLE cell hyperresponsiveness
[56]
PBMCs
Upregulation
Targeting PDCD4 involved in aberrant T cell response
SLE
[57]
Fibroblasts
Upregulation
Directly downregulating SMAD7 and increasing fibrosis-related genes expression
thereby SSc
[58]
miR-29
Pancreatic islets cell
Upregulation
Decreased antiapoptotic protein Mcl1
T1DM
[109]
miR-29a
Fibroblasts
Downregulation
Targeting COL3A1 involved in collagen expression
SSc
[64]
miR-29b
CD4+ T cell Upregulation
Repressing DNMT1 expression
SLE
[62]
Mesangial cells
AC
miR-21
Expression change
CE P
miRNA
+
SMAD7,
and
thereby MS
[59]
Foxp3 RA
[60]
CD4 T cell Upregulation
Targeting T-bet and IFN-γ, controlling Th1 differentiation MS
[63]
miR-30a
B cell
Upregulation
Targeting LYN involved in B cell hyperactivity
SLE
[110]
miR-30a-3p
Fibroblasts
Downregulation
Targeting BAFF involved in autoimmune responses
RA, SSc
[111]
miR-30b
Fibroblasts
Downregulation
Targeting PDGFR-β involved in fibroblast proliferation
SSc
[112]
18
ACCEPTED MANUSCRIPT Targeting STAT1 involved in phenotype of regulatory T cells
pro-inflammatory
RA
[113]
PBMCs
Upregulation
Involved in phagocytic activity and pro-inflammatory pSS cytokine production
[114]
PBMCs
Downregulation
Targeting and IRF5 and STAT1 involved in activation SLE of type I IFN pathway
[86]
Monocytes, microglia
Upregulation
Targeting CD47, increased TNFα and IL-6 secretion, MS increased CD80 and CCR7 expression
[115, 116]
Synovial fibroblasts
Upregulation
Downregulation of MMP-3 involved in inflammation
T
Downregulation
RA
[117]
SC R
miR-155
Regulatory T cells
IP
miR-146a
Abbreviations: SSc, Systemic sclerosis; SLE, systemic lupus erythematosus; RA, rheumatoid arthritis; MS, multiple sclerosis; T1DM, type 1 diabetes; pSS, primary Sjögren's syndrome; PBMCs, peripheral blood mononuclear cells; DNMT, DNA methyltransferase.
Cell type
Effects
Disease
References
Trichostatin A (HDAC inhibitor)
Fibroblasts
Increased acetylation of FLI1, silencing HDAC-7
SSc
[37, 79]
Macrophages, Synovial fibroblasts
Modulate NF-κB signaling, impairing the stability of IL-6 mRNA
RA
[80]
T cell
Downregulating CD40L and IL10 expression, upregulating IFN-γ expression
SLE
[81]
Entinostat (HDAC inhibitor)
D
[82]
Inhibiting TNF and IL-6 production
RA
[118]
Dendritic cell
Decreased expression of CD80 and CD86, the MS matural marker CD83, and HLA-DR
[119]
Synovial fibroblasts
Suppressing NF-κB pathway
RA
[120]
Regulatory T cell
Increasing regulatory T cell activity and expansion RA of FOXP3+Treg cells
[121]
Synovial fibroblasts
Involved in induction of p21 and suppression of RA the NF-κB pathway
[120]
Macrophages, Synovial fibroblasts
Modulate NF-κB signaling, impairing the stability of IL-6 mRNA
RA
[80]
Macrophages, Splenocytes
Inhibiting the production of TNFα and IFNγ
T1DM
[122]
Fibroblasts
Hypomethylation of FLI1, DKK1, SFRP1
SSc
[37, 38]
Synovial fibroblasts
Hypomethylation of SFRP4
RA
[103]
Regulatory T cell
Increasing the population and immunosuppressive MS function of Foxp3+ regulatory T cells
CE P
PBMCs
AC
Vorinostat (HDAC inhibitor)
Valproic acid (HDAC inhibitor)
Increasing the expression of IFN-γ and its T1DM transcription factor Tbx21
TE
T cell MI192 (HDAC3-selective inhibitor)
Givinostat (ITF2357) (HDAC inhibitor)
5-aza-2'-deoxycytidine (DNMT inhibitor)
MA
Drug (Type)
NU
Table 4. Examples of chemicals modulating DNA methylation and histone modification in the context of autoimmune diseases
[84]
Abbreviations: SSc, Systemic sclerosis; RA, rheumatoid arthritis; SLE, systemic lupus erythematosus; MS, multiple sclerosis; T1DM, type 1 diabetes; PBMCs, peripheral blood mononuclear cells.
19
CE P
TE
D
MA
NU
SC R
IP
T
ACCEPTED MANUSCRIPT
AC
Figure 1. Mechanisms of miRNA inhibition strategies. A. miRNA sponges could selectively deplete endogenous miRNAs based on high complementation to multiple artificial miRNA-binding sites, thereby allowing translation of the target mRNAs. B. Small-molecule inhibitors could target multiple steps of miRNA assembly and function, derepressing its inhibitory effect on mRNA translation. C. anti-miRs are designed to be high complementary to the target miRNA, binding to and specifically suppressing target miRNAs function. D. miR-mask covers up the sites on mRNAs where are fully complementary to target miRNA, preventing the inhibitory effect of miRNA on target mRNA translation. Abbreviations: miRISC, miRNA-induced silencing complex; Anti-miRs, miRNA-targeting antisense oligonucleotides.
20