Next Article in Journal
The Anti-CGRP Antibody Fremanezumab Lowers CGRP Release from Rat Dura Mater and Meningeal Blood Flow
Previous Article in Journal
Vitronectin and Its Interaction with PAI-1 Suggests a Functional Link to Vascular Changes in AMD Pathobiology
Previous Article in Special Issue
Epigenetic Alterations in Immune Cells of Systemic Lupus Erythematosus and Therapeutic Implications
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Abnormal Epigenetic Regulations in the Immunocytes of Sjögren’s Syndrome Patients and Therapeutic Potentials

1
Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Institute of Systems Biomedicine, Peking University Health Science Center, 38 Xueyuan Road, Beijing 100191, China
2
NHC Key Laboratory of Medical Immunology, Peking University, Beijing 100191, China
3
Key Laboratory of Molecular Immunology, Chinese Academy of Medical Sciences, Beijing 100191, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cells 2022, 11(11), 1767; https://doi.org/10.3390/cells11111767
Submission received: 16 April 2022 / Revised: 22 May 2022 / Accepted: 26 May 2022 / Published: 27 May 2022

Abstract

:
Sjögren’s syndrome (SjS), characterized by keratoconjunctivitis sicca and dry mouth, is a common autoimmune disease, especially in middle-aged women. The immunopathogenesis of SjS is caused by the sequential infiltration of T and B cells into exocrine glands, including salivary and lacrimal glands. Effector cytokines produced by these immunocytes, such as interferons (IFNs), IL-17, IL-22, IL-21, IL-4, TNF-α, BAFF and APRIL, play critical roles in promoting autoimmune responses and inducing tissue damages. Epigenetic regulations, including DNA methylation, histone modification and non-coding RNAs, have recently been comprehensively studied during the activation of various immunocytes. The deficiency of key epigenetic enzymes usually leads to aberrant immune activation. Epigenetic modifications in T and B cells are usually found to be altered during the immunopathogenesis of SjS, and they are closely correlated with autoimmune responses. In particular, the important role of methylation in activating IFN pathways during SjS progression has been revealed. Thus, according to the involvement of epigenetic regulations in SjS, target therapies to reverse the altered epigenetic modifications in auto-responsive T and B cells are worthy of being considered as a potential therapeutic strategy for SjS.

1. Introduction

Sjögren’s syndrome (SjS) is the second most common chronic autoimmune rheumatic disease, characterized by dry eyes (sicca) and dry mouth. It is caused by abnormal infiltration and activation of immunocytes in the lacrimal glands (LGs) and salivary glands (SGs) due to multiple genetic and environmental factors. These infiltrated immunocytes lead to substantial progressive damage of the acinar structures responsible for saliva production and secretion. In addition, as indicated by some studies, the autoimmune responses may already occur before the immunocyte infiltration [1]. For example, Elke Theander et al. reported that anti-nuclear autoantibodies are present for up to 20 years before the diagnosis of SjS [2]. SjS occurs either alone, as primary Sjögren’s syndrome (pSS), or on the background of other autoimmune diseases as secondary Sjögren’s syndrome (sSS). Most SjS studies with patients or animal models are actually about pSS. Thus, in this paper, the SjS which we discuss also refers to pSS in principal and will be particularly mentioned as pSS if it was declared in the original study. The development of pSS can also cause extraglandular involvements of multiple organs, including fibromyalgia, arthralgia and interstitial nephritis, which suggests it is a systemic autoimmune disease [3]. In particular, patients with pSS exhibit an increased risk of non-Hodgkin B-cell lymphoma [4]. A recent study revealed that long before the clinical onset of the lymphoma, miRNA miR-200b-5p is found to be significantly downregulated in the minor SGs of pSS patients, indicating that this epigenetic regulation may be involved in the progression to non-Hodgkin B-cell lymphoma [5].
SjS generally develops in middle-aged women, but it can occur at any age throughout the population. Its prevalence is estimated to be around 3% in subjects aged 50 years and older, with a female to male ratio of 9:1 [6]. X-chromosome dosage effect is supposed to be associated with the high risk in females. In line with it, the disease risk increases 15 times in men with Klinefelter’s syndrome, which is genetically characterized by the presentation of one extra X chromosome (47, XXY) [7]. Similarly, the risk is also increased in women with trisomy X (47, XXX) [8]. In a recent study, X-linked genes in SG-derived mesenchymal stromal cells of pSS patients were found to exhibit miR-6891-5p-regulated skewing that is associated with alterations in H3K27me3 deposition. Thus, epigenetic regulations may be involved in the sexual bias of SjS; however, the precise mechanism still needs further study [9].
SjS is a multi-factorial disease caused by genetic, environmental and epigenetic abnormalities. The molecular etiology of autoimmune responses in SjS is still quite elusive. Genetic factors associated with SjS include particular HLA-DR allele subtypes and specific polymorphisms of genes, such as STAT4, IL-12A, TNIP1, IRF5 (type I interferon related), BAFF and BLK (related to B-cell activation) [10,11]. Environmental factors, such as infectious agents, are also considered as correlated with the pathogenesis of SjS. For instance, the Epstein–Barr virus (EBV) is implicated in the progression of SjS [12]. In addition, epigenetic regulations in the immune system, including DNA methylation, chromatin remodeling and non-coding RNAs, have been comprehensively studied in recent years, providing new insights into the pathogenesis and therapeutics of SjS. In addition, epigenetic changes are also involved in inflammatory responses in non-immune cells. It was reported that the DNA methylation process is abnormal in salivary gland epithelial cells (SGECs) from SjS patients [13,14]. Thus, given the potential role of epigenetic regulations in the auto-responsive immunocytes in SjS patients, in this review, epigenetic changes, particularly in the abnormally infiltrated and activated immunocytes in the exocrine glands, are particularly discussed.

2. Epigenetic Regulations in Immune Responses

Epigenetic modifications are mitotically and/or meiotically heritable and reversible alterations that induce upregulated or downregulated gene expression, without any changes in underlying DNA sequences. In addition, they also play a vital role in silencing or promoting the expression of non-coding sequences [15]. Epigenetic modifications are relatively stable over time, facilitating the maintenance of cell identity. However, they are also variable enough to change in response to various external and internal stimuli. Typical epigenetic modifications include methylation on DNA, modifications of histones, and the expression of non-coding RNAs (ncRNAs). They play an essential role in immune cell differentiation and activation [16]. Thus, their roles in regulating the aberrant differentiation and activation of immunocytes in autoimmune diseases have attracted much attention in recent years (Figure 1).

2.1. DNA Methylation

Methylation changes on DNA include both methylation and demethylation, which are, respectively, regulated by DNA methyltransferases, DNMT1, 3A and 3B, and Ten Eleven Translocation (TET)-1, 2 and 3 [17]. During DNA methylation, a methyl group donated by S-adenosyl-methionine (SAM) is transferred by DNMTs to the carbon-5 position of the cytosine pyrimidine ring (5mC) within a CpG dinucleotides. Methylated CpG sites then induce the structural changes of chromatin, making it difficult for transcriptional factor binding. CpG islands enriched in CpG base pairs are mainly distributed in the promoter and first exon regions of genes. Thus, DNA methylation in the CpG islands of chromatin is generally considered as a signal of gene silencing. Contrarily, the demethylation of DNA is the removal of a methyl group on cytosine through a serial process starting from the oxidation of 5mC to 5-hydroxymethylcytosine (5hmC) [17]. Additionally, demethylation, or 5hmC modification, of genes indicates enhanced gene expression.
DNA methylation and demethylation play critical roles in regulating the activation and differentiation of CD4+ T cells [18]. The most studied DNMTs in T cells are DNMT1 and DNMT3A. For example, during Th1 differentiation, both Tbx21 and Ifng loci display demethylation and increased 5hmC level [16]. Dnmt1 deficiency in T cells, or treating T cells with demethylation agents, results in enhanced Th1 effector cytokine expression [16]. DNMT3A is required to keep the Ifng locus stably silenced in Th17. The deletion of Dnmt3a results in demethylation at the Ifng locus of Th17 cells. Thus, under IL-12 stimulation, Dnmt3a-deficient Th17 become pathogenic and express higher levels of IFN-γ [19]. For Treg development and function, DNMT1, rather than DNMT3A, is essential. Dnmt1-deficient Treg lose their suppressive function both in vitro and in vivo [20]. Mice conditionally depleting Dnmt1 in Treg die after 3 to 4 weeks due to severe systemic autoimmunity [20].
On the other hand, TET enzymes are also involved in T-cell differentiation. In Th1, T-bet recruits TET2 to the Ifng locus to keep it demethylated, while Tet2 deficiency results in reduced IFN-γ expression [21]. In Th17, Tet2 deficient leads to reduce 5hmc and RORγt binding at the Il17a locus, thus resulting in reduced IL-17 expression [21]. In Treg, Smad3 and STAT5 activated by IL-2 and TGF-β signaling recruit TET enzymes to the Foxp3 locus, where they ensure the expression of Foxp3. Both TET1 and TET2 are required for the conversion of 5mC to 5hmC at the Foxp3 locus, while their joint depletion results in impaired Treg differentiation and function [22]. TET enzymes display functional redundancy in Treg. Though depleting either Tet2 or Tet3 does not significantly impact Foxp3 expression, depleting them both leads to severely impaired Treg differentiation and stability [23]. TET expression and activity are enhanced by vitamin C and hydrogen sulfide in Treg. Thus, they are also capable of promoting the demethylation of conserved non-coding DNA sequence (CNS) elements at the Foxp3 locus [22,24].
During Tfh differentiation, the involvement of TET and DNMT family members is still quite elusive. Nevertheless, in comparison with other Th cells, Tfh displays significantly reduced 5hmC at Bcl6 binding sites [18]. DNMT family members are found to be involved in B-cell activation and differentiation. The specific depletion of Dnmt3a and Dnmt3b in B cells does not affect their development and maturation; however, it results in the abnormal accumulation of plasma cells in the spleen and bone marrow. Compared to normal plasma cells, the Dnmt3-deficient plasma cells display reduced DNA methylation at over a thousand gene loci, and thus are unable to limit the activation and differentiation of B cells [25]. In addition, TET2 and TET3 also play a role in the class switch recombination of B cells. Tet2 and Tet3 deficiency in activated B cells results in substantially reduced 5hmC levels and defective IgG1 switching [26].

2.2. Histone Modifications

Histones play important regulatory roles in DNA replication and gene expression, highly relayed on their unique amino acid sequences. Histones are enriched with basic lysine and arginine residues, especially in the N-terminal tails, which are feasible for several post-translational modifications (PTMs), including methylation, acetylation, phosphorylation and ubiquitination [27]. Histone modifications stand for another type of important epigenetic regulation. These modified histones lead to altered chromatin structures or can act as binding sites for non-histone regulators, resulting in varied gene expression [17].
Multiple enzymes are involved in histone modifications, such as histone deacetylases (HDACs), histone acetyltransferases (HATs) and histone methyltransferase (HMTs). Several HDAC members have been reported to be involved in T- and B-cell responses [28]. For example, Hdac1-deficient Th1 display increased STAT1 phosphorylation and IFN-γ expression [29]. Additionally, Hdac11-deficient CD4+ T cells express higher Eomes and T-bet, and produce more IFN-γ [30]. Sirtuin 1(SIRT1), another member of HDAC family, shows higher expression in polarized Th17 than other Th effectors. It deacetylates RORγt in Th17, which then promotes its transcription regulator activity and enhances the effector function of Th17. SIRT1 inhibitor suppresses Th17 cells differentiation in vitro. Additionally, specifically depleting Sirt1 in T cells protects mice from EAE, due to the reduced pathogenic differentiation of Th17 [31]. In addition, HDACs can interact with Foxp3 in Treg, and Hdac6 or Hdac9 deficiency leads to the increased suppressive function of Treg. As major microbial metabolites in the gut, short-chain fatty acids (SCFAs) can passively diffuse across the cell membrane of T cells and inhibit the activity of HDAC [32,33,34]. Studies in mice show that butyrate enhances H3 acetylation at the Foxp3 locus of Treg by inhibiting HDAC, and thus promotes the transcription factor stability and activity [35]. In addition, butyrate activates mTOR and Blimp-1 in CD4+ T cells [36]. In addition to butyrate, the HDAC inhibitor valerate (VPA) also increases the activity of the mTOR complex and strongly induces IL-10 expression in T cells [37].
HDACs are also involved in regulating B-cell responses. The master regulator for plasma cell differentiation is Blimp-1. In undifferentiated B cells, Bcl-6 suppresses Blimp-1 expression by interacting with HDAC4, -5 or -7 and forming stable complexes [38]. When the HDACs are released from the complexes, histone acetylation at the promoter region of Prdm1 is increased, thus resulting in the upregulated expression of Blimp-1. In line with it, the HDAC inhibitor or butyrate is capable of inducing Blimp-1 expression in splenic B cells [39]. B cells were also reported to be regulated by SCFAs or HDAC inhibitors. One study showed that, in human and mouse B cells, butyrate, as well as the HDAC inhibitor VPA, upregulated miR-155, miR-181b and miR-361, which silenced AICDA/Aicda (encoding AID) mRNA, and upregulated miR-23b, miR-30a and miR-125b, which silenced PRDM1/Prdm1 (encoding Blimp-1) mRNA [40]. Through these B-cell intrinsic epigenetic mechanisms, VPA decreased class-switched and hypermutated autoantibodies in lupus MRL/Faslpr/lpr mice, ameliorating the disease and extending mice survival. Thus, they also provide a therapeutic rationale and potential for other autoimmune diseases [40].

2.3. ncRNAs

Non-coding RNAs (ncRNAs) are single-strand RNA molecules transcribed from the genome. They do not encode proteins, but are implicated in cell development, proliferation and metabolism, playing important regulatory roles. Small non-coding RNAs (miRNAs) are 19–22 nucleotides in length, affecting gene expression at post-transcriptional levels, whereas long non-coding RNAs (lncRNAs) are defined as being above 200 nucleotides in length [17]. The miRNAs are novel molecular regulators of genes and pathways involved in immune responses in inflammatory and autoimmune diseases, such as RA, MS, SLE and SjS [41,42]. Under pathological conditions, the abnormal ex-pression of miRNAs leads to disease development by affecting the expression of multiple genes. About 2200 miRNAs have been found in the human genome [43]. Among them, about 50% are transcribed from non-coding regions, while the residues are distributed in introns. Each miRNA could target multiple genes, making them promising diagnostic biomarkers and therapeutic targets [44].

2.3.1. Adaptive Immunity

There have been many studies showing the crucial role of miRNA in the development and differentiation of T and B cells [45,46]. One frequently reported miRNA in T and B cells is miR-146a. Mir146a deficiency increases T-cell reactivity and IL-17 secretion, through the reduction in the expression of TRAF6 and IRAK1 associated with the NF-κB signaling pathway [47]. Additionally, miR-146a regulates the expression of protein kinase C epsilon (PKCε), responsible for STAT4 phosphorylation and activation. Thus, miR-146a is also involved in inhibiting the Th1 differentiation-related pathway [48,49], whereas the deficiency of Mir146a in Treg results in an impaired immunological tolerance [50]. Additionally, miR-146a directly represses multiple messenger RNA (mRNA) targets, most prominently Icos. In line with it, Mir146a deficiency leads spontaneous Tfh accumulation [51]. In addition, miR-146a is also required for regulating the differentiation of B cells by promoting Blimp-1 expression [52,53,54].
There have been numerous studies indicating that certain miRNAs, such as miR-21, miR-27, miR-31, miR-138, miR-155, miR-146a, miR-181a and the miR-17~92 cluster, regulate T-cell development, differentiation and effector function [55,56]. Abnormal miRNA regulation can be detected in several autoimmune disease, including SjS [57]. MiR-146a has been reported to be upregulated in the PBMCs of SjS patients [58,59]. It serves as a negative regulator to inhibit the NF-κB signaling transducer TRAF6 (TNF-receptor-associated factor 6) and IRAK1 (IL-1-receptor-associated kinase 1) [60]. In line with it, miR-146a-deficient mice exhibit an abnormal activation of NF-κB signaling, leading to an overactivation of T cells [61,62,63]. In miR-146a-deficient mice, a deficiency of Treg was also found, correlated with the failure in suppression of Th1 responses [50]. In addition, miR-146a in Th1 and Th17 is also involved in regulating their differentiation, and thus is crucial for autoimmune responses [47,48,49]. Moreover, miR-146a negatively regulate the co-stimulatory signal of T cells by targeting Icos, thus limiting the accumulation of Tfh and GCs [51]. MiR-155 is another miRNA that is most commonly found to be dysregulated in the PBMCs of SjS patients [59]. The upregulated miR-155 is associated with the overactivation of T cells [59,64]. Additionally, the elevated expression of miR-181a is also found in the PBMCs of SjS patients [65]. MiR-181a participates in T-cell selection by dampening the TCR signal threshold against self-antigens, and thus the loss of miR-181a results in the development of autoreactive T cells [57].
Particular and abundant miRNAs are present at different stages of B-cell development. At an early stage, the miR-17∼92 cluster can promote the degradation of PTEN mRNA to increase PI3K activity, which further promotes B-cell survival [66]. In addition, the miR-17∼92 cluster regulates the survival of early B-cell progenitors by repressing the expression of the pro-apoptotic protein BIM [67]. In immature and transitional B cells, an abnormally increased miR-148a can suppress the expressions of Gadd45α, PTEN and BIM, which in turn leads to impaired B-cell tolerance [68]. The miR-17∼92 cluster is also involved in the c-Myc/miR-17∼92/PTEN axis to regulate the PI3K-mediated positive and negative selections of B cells [69]. At a mature stage, miR-155 can directly regulate PU.1, which is critical for the maintenance of germinal center (GC) responses [70]. Meanwhile, activation-induced cytidine deaminase (AID) is also a direct target of miR-155 in activated B cells, which is required for class switch recombination [71].

2.3.2. Innate Immunity

MiRNAs also regulate the development and activation of innate immunocytes, such as ILCs and macrophages [72]. MiR-142 maintains ILC1 survival and function by promoting IL-15 signaling [73], whose deficiency results in reduced ILC1s in cancers [74]. MiR-142 and miR-146 are required for IL-5 and IL-13 secretion by ILC2, through upregulating ST2 [75,76]. The role of miRNAs in ILC3 is relatively unclear. A related study is limited to ILC3s in decidua, where miR-125 and miR-574 prevent the abnormal activation of ILC3 during pregnancy by inhibiting IL-6-STAT3 signaling [77]. In macrophages, their polarization towards M1 or M2 phenotypes plays important roles in diverse inflammatory responses [78]. MiRNAs are also found to affect polarization. MiR-9, miR-127, miR-155 and miR-125b promote M1 polarization, while miR-124, miR-223, miR-34a, let-7c, miR-132, miR-146a and miR-125a-5p promote M2 polarization [79]. The role of miR-21 in macrophage polarization is controversial. Some studies show that it promotes M1 polarization [80,81], but others suggest that it promotes M2 polarization [82,83]. Dendritic cells are also regulated by miRNAs. For instance, miR-21 promotes dendritic cell maturation, which thus helps to protect the kidney from ischemia-reperfusion injury [84].

3. Immunocyte Infiltration in SjS

Though the precise mechanism for the pathogenesis of SjS remains largely unclear, the infiltration of immunocytes in exocrine glands is usually considered associated with disease progression. Immunocytes infiltrated in SGs of SjS patients mainly include T and B cells. At an early stage, CD4+ T cells constitute the predominant infiltration in SGs, while at a later stage, B cells are gradually accumulated [85,86]. This sequential infiltration and activation of CD4+ T and B cells are closely correlated with damages in the glandular tissues of SjS patients. In addition, CD8+ T cells are also crucial players in the immunopathogenesis of SjS, contributing to acinar injury. Overall, the infiltration of T and B cells are usually closely associated with SjS progression [87]. In addition, several recent studies have also revealed the infiltration and aberrant activation of other types of immunocytes during SjS. Thus, in this section, various infiltrated immunocytes in SjS are discussed (Figure 2).

3.1. T Cells

Infiltrated T cells exist throughout the progression of SjS and act as key drivers of the disease. The aberrant activation of T cells is then critical for the progression of the disease especially at an early stage, through recruiting other immunocytes, secreting pro-inflammatory cytokines and promoting B-cell activation and autoantibody secretion [88,89,90,91]. Their frequency gradually declines, but they persist throughout disease progression [92,93]. Thus, the frequency of effector T cells in severe lesions is usually lower than in mild lesions [85]. In addition, regulatory T cells (Treg) are also found to infiltrate in SjS lesions. Their frequency reaches a peak earlier than effecter T cells in intermediate lesions [94]. Though the frequency of T cells is inversely proportional to disease severity, the role of T cells on disease onset should not be ignored.

3.1.1. Th1 and Th17

Type 1 and type 17 T-helper cells (Th1 and Th17), as major sources of IFN-γ and IL-17 in many autoimmune diseases [95,96,97,98], also play critical roles during the immunopathogenesis of SjS. In SjS patients, both IFN-γ and IL-17 are increased in T-cell-rich areas around SG ducts. In addition, Th1-cell-related (e.g., Ifng, Tnf and Tbx21) and Th17-cell-related (e.g., Il17, Il6 and Rorc) mRNA transcripts are also enriched in SGs and blood [88,89]. Early in the disease, Th1 cells infiltrate into SGs via the chemokine receptor CXCR3 [90]. The increment in IFN-γ+ cells in SGs is accompanied by a decrease in IFN-γ in the blood, suggesting that Th1 cells in blood circulation migrate to the SGs [99]. Th1 further trigger the infiltration of immune cells through secreting IFN-γ [90]. On the other hand, Th17 cells are the main pathogenic factor of SjS. They are increased in the peripheral blood of SjS patients with intermediate and severe lesions. IL-17A plays important roles in the progression of SjS, as indicated by the fact that IL-17 knockout mice are resistant to SjS disease models [100]. Moreover, IFN-γ+IL-17A+ Th17 cells usually exerting pathogenic roles in many autoimmune diseases also exist in the SGs of SjS patients [101]. However, whether the plasticity of Th17 cells plays an important role in disease progression still requires further investigation. Thus, in SjS patients, Th1 and Th17 may mainly exert their pathogenic roles by promoting immunocyte infiltration and inducing damages in their infiltrated glandular tissues, respectively.

3.1.2. Tfh

B cells are gradually accumulated at a late stage of SjS [102]. T-follicular-helper cells (Tfh) play critical roles in helping B-cell responses [103]. They are hallmarked by the expression of CXC chemokine receptor 5 (CXCR5), inducible T-cell co-stimulator (ICOS) and programed death 1 (PD-1), and are regulated by master transcription factor B-cell lymphoma 6 (BCL-6) [104]. They promote B-cell activation, germinal center (GC) formation and antibody class switching [103]. Over the past decade, Tfh have been associated with a wide range of B-cell-mediated autoimmune diseases, including SjS [91]. The frequencies of Tfh cells are significantly increased in the glandular tissue and peripheral blood of SjS patients with mild lesions, and are positive correlated with disease severity [105]. Consistently, IL-6, which facilitates Tfh differentiation, is also elevated in the SGs and blood of SjS patients. However, whether Tfh are recruited into lesion tissue or differentiated within the tissue is still unclear. The increased Tfh play important roles in aggravating the disease. Tfh migrate to the B-cell zone in the ectopic germinal centers of salivary glands through the chemokine receptor CXCR5, where they promote GC B-cell differentiation, development and maturation by producing the effector cytokine IL-21 [106]. In addition, IL-4, contributing to GC formation in SjS patients, is also mainly secreted by the Tfh co-expressing transcription factor GATA3 [107]. Therefore, Tfh differentiation is closely correlated with autoimmune B-cell responses in SjS patients.

3.1.3. Treg

Although Treg infiltration in the exocrine glands of SjS patients has been substantially studied, it is still unclear whether Treg play anti-inflammatory roles in the disease [108,109]. Several reasons, including states of the disease, markers for Treg identification as well as the balance between Treg and Th17, result in difficulties in clarifying the role of Treg in SjS [92,110]. For example, CD25 was generally used as a marker to identify Treg in SjS patients in many previous studies; however, it was also upregulated in activated CD4+ T-effector cells [111]. Treg exert their roles by inhibiting the activation of effector T cells. However, even when Treg frequency reaches a maximal level in SjS patients, the disease does not resolve, but rather it is usually aggravated, with the further increased frequency of effector T cells. The increment in Treg usually leads to a reduction in Th17-cell numbers; however, this negative correlation does not exist in SjS patients [110].
A particular subpopulation of Treg, follicular regulatory T cell (Tfr), is substantially increased in the SGs and blood of SjS patients [112,113]. These cells are distinguished by CD4+Foxp3+CXCR5highPD-1highBlimp-1+. They exert a suppressive role for Tfh proliferation and GC B-cell activation. The ratio of Tfr/Tfh in the blood of SjS patient is a biomarker for disease severity [112,113]. However, specific relationship between the Tfr/Tfh ratio and B-cell hyperactivation in SjS patients is still unclear. Tfr are also capable of downregulating B7-1 or B7-2 expression on B cells in GC, thereby potentially alleviating autoimmune responses and the severity of inflammation [114]. In general, comprehensive studies about the changes in Treg of SjS patients are still needed to further understand their roles in disease progression.

3.1.4. CD8+ T Cells

CD8+ T cells exert pathogenic roles by secreting cytotoxic molecules, such as granzyme B (GzmB) and perforin, and inflammatory cytokines, including interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α) [115]. Recent studies have revealed that CD8+ T cells are enriched around the apoptotic acinar epithelial cells of SjS patients. IFN-γ in SGs is involved in the recruitment of more CD8+ T cells. Ifng deficiency was reported to abrogate CD8+ T-cell infiltration and decrease gland destruction in SjS mice. CD8+ T cells infiltrated in SGs displayed a tissue-resident phenotype CD69+CD103+/− and exhibited dramatically increased IFN-γ production. In a p40−/−Il2ra−/− murine model of SjS, CD8+ T-cell depletion, via either genetic Cd8a deficiency or antibody-mediated depletion, fully protected the mice from pathologic manifestation [116]. Moreover, even after the onset of the disease, antibody-mediated CD8+ T-cell depletion successfully restored the secretory function of SGs, suggesting it as a potential therapeutic strategy. Thus, the correlation between CD8+ T cells and gland destruction in SjS patients should receive more attention in the future.

3.2. B Cells

B cells are central to the pathophysiology of SjS at a late stage [102]. In SjS patients, B-cell subpopulations in circulation are disturbed, and the frequency of GC founder cells is usually increased. This B-cell subpopulation expresses high levels of CD19 and tyrosine-protein kinase BTK, which together increase BCR signals [117,118]. The aberrant activation of B cells also increases the risk of SjS patients to develop B-cell lymphoma, such as non-Hodgkin’s B-cell lymphoma [119,120].
B cells promote the progression of autoimmune diseases mainly through their roles as cytokine producers, antigen-presenting cells or autoantibody secretors. B cells possess the capacity to produce a range of cytokines, including either the pro-inflammatory cytokines IL-6 and TNF-α or the anti-inflammatory cytokines TGF-β and IL-10 [117]. The difference is relayed on the polarization of B cells under different circumstances [117]. However, which kind of cytokine is mainly secreted by B cells in SjS patients has not been clearly reported. In the SGs of SjS patients, B cells also act as antigen-presenting cells, activating naive T cells in an MHC-II-dependent manner and, thereby, promoting disease progression [121].
A key feature of SjS is that target organs, including salivary and lachrymal glands, are involved in B-cell activation, especially the formation of GC-like structures within the epithelium and plasma cell niches [122]. Thus, in these organs, autoantibodies produced by autoreactive B cells are significantly enhanced. For example, the production of many autoantibodies, such as anti-salivary gland protein 1, anti-carbonic anhydrase 6 and anti-parotid secretory protein, are increased in pSS patients [123]. These autoantibodies participate in immune complex formation, which is a vital step in the progression of SjS.

3.3. Innate Lymphoid Cells

Innate lymphoid cells (ILCs) are tissue-resident innate lymphocytes that play crucial roles in regulating tissue homeostasis [124]. Unlike T and B cells, they do not express antigen specific receptors. According to their difference in effector functions, ILCs are divided into three subsets, ILC1s, ILC2s and ILC3s, mirroring the CD4+ Th subsets Th1, Th2 and Th17 [124]. The discovery and investigation of ILCs over the past decade have been closely associated with autoimmune disease [125]. For example, increased Fas expression on ILC2s and ILC3s decreases their frequency in the blood of SLE and pSS patients, potentially altering the homeostasis of ILCs [116]. ILC3 is the main source of IL-22, which is supposed to play a pathogenic role in epithelial damage in SjS patients [126].

3.4. Antigen-Presenting Cells

In addition to B cells, other antigen-presenting cells, such as DCs and macrophages, are also increased in the SGs of SjS patients [127,128]. Both DCs and macrophages are differentiated from monocytes in the blood, possessing a high antigen-presenting capacity. They are positively correlated with the severity of SjS [127]. In addition, SGECs also play an antigen-presenting role, by upregulating MHC molecules during the onset of SjS [128]. Thus, these antigen-presenting cells are indispensable for the aggregation and activation of inflammatory cells in SjS patients.

4. Pro-Inflammatory Cytokines in SjS

As previously mentioned, the immunopathogenesis of SjS is initiated by Th1 and Th17, and progressed by Th2 and Tfh, which then ignite B-cell responses, resulting in more severe tissue lesions [129]. In line with it, cytokines acting on or produced by these immunocytes, including interferons (IFNs), interleukins and tumor-necrosis-factor (TNF) superfamily members [130], play critical roles in disease progression.
After processing certain autoantigens, antigen-presenting cells are activated to produce IL-7, IL-12, IL-18 and type I IFNs, which effectively promote Th1 responses [131]. IFN-γ and TNF-α produced by Th1 cells then induce CXCL9/10 secretion from the epithelial cells and stromal cells of SGs [131]. Additionally, the activated antigen-presenting cells produce cytokines required for Th17 cell differentiation, including IL-6, IL-23, TGF-β and IL-1β. Differentiated Th17 release effector cytokines, such as IL-17, IL-22 and IL-21 [101]. IL-21, also released by Tfh cells, facilitates the formation of GC-like structures within epithelium and plasma cell niches [132]. IL-21 and IL-17 derived from Tfh or Th17, together with IL-4 from Tfh and Th2, promote B-cell proliferation, differentiation and autoantibody secretion [133]. In addition, the TNF superfamily cytokines BAFF (B-cell-activating factor) and APRIL (a proliferation-inducing ligand) are also involved in promoting B-cell responses [134]. In this section, the crucial cytokines and their effector roles during SjS are summarized (Figure 3).

4.1. Interferons

High levels of IFNs and the elevated transcription of IFN-stimulated genes (ISGs) are found in the SGs and serum of SjS patients, as well as SjS animal models, indicating an important role of IFNs in the pathogenesis of SjS [47,48,49,50]. Interferons are classified into three types, type I interferons (IFN-α and IFN-β), type II interferon (IFN-γ) and type III interferon (IFN-λ) [135,136].

4.1.1. Type I interferons

In the inflamed SGs, IFN-α are mainly produced by plasmacytoid dendritic cells (pDCs), after they recognize autoantigens through the Toll-like receptors (TLRs) TLR-7 and TLR-9 [137,138]. On the other hand, IFN-β is produced by SGECs, also through TLR signaling [139,140]. Once engaged with their receptor (IFNAR) on the target cells, type I IFNs initiate the transcription of ISGs [141,142]. ISGs are generally considered to execute antiviral and antitumor activities [143]. However, they also contribute to autoimmune responses in SjS, through triggering the secretion of pro-inflammatory cytokines and chemokines, such as CXCL10 and BAFF, class switch of immunoglobulins and cellular cytotoxicity of NK and T cells [90,144,145,146]. In line with this, mice lacking IFN-α receptor 1 (Ifnar1−/−) display significantly reduced susceptibility to SjS-like disorders [147,148].

4.1.2. Type II interferon

In addition to type I IFNs, type II IFN IFN-γ also play a crucial role in inducing tissue lesions in SjS patients. Thus, the lesion tissue is usually characterized by both type I and type II IFN signatures [149,150]. In SGs, IFN-γ is mainly generated by Th1 and Tfh. It also induces a subsequent upregulation of ISGs in either immune or non-immune cells [110,151]. This process promotes a further differentiation of naïve CD4+ T cells towards Th1 [152,153]. In addition, it also polarizes macrophages to an M1 phenotype, which usually correlates with tissue inflammation [152,153], whereas for non-immune cells, such as SGECs, IFN-γ upregulates their expression of MHC class II and co-stimulatory molecules (CD80, CD86 and CD40), which facilitate antigen presentation for adaptive immune responses [154,155,156]. In addition, IFN-γ induces epithelial damages through Fas-mediated apoptosis in SGECs and reduces epithelial integrity by disrupting tight junction (TJ) structures [157,158]. Moreover, as mentioned above, IFN-γ also facilitates the recruitment of more pro-inflammatory immunocytes.

4.1.3. Type III interferon

Type III IFN, IFN-λ, shows a certain functional similarity to type I IFNs [136]. However, unlike the extensive study of type I and type II IFNs, type III IFN is the latest member added to IFN family and its role during inflammatory responses still requires further studies. As recently reported, IFN-λ is highly expressed by the ductal epithelium of SjS patients, which acts synergistically with type I IFNs to further upregulate the expressions of CXCL10 and BAFF [159].

4.2. Interleukins

The inflammatory milieu in SjS patients is characterized by a dysregulated cytokine network, especially pro-inflammatory interleukin overexpression. The major pro-inflammatory interleukins include IL-17, IL-22, IL-21 and IL-4, and exhibit aberrant upregulation in the SGs or serum of SjS patients [122,160].

4.2.1. IL-17 and IL-22

Pro-inflammatory cytokine IL-17A is mainly expressed by Th17 cells in the inflamed SGs of SjS patients, and considerable evidence suggests IL-17A as a pathogenic factor in SjS [101]. The level of IL-17A in SG biopsies or serum is positively correlated with histopathological score of SjS patients [161,162]. It binds to its receptors on epithelial and stromal cells, and affects downstream gene expression [163,164]. Elevated IL-17A results in the significant downregulation of the TJ proteins laudin-4 and zonula occludens-I (ZO-1), impairing epithelial integrity and leading to SG dysfunction [165]. In addition, IL-17A is found to induce matrix metalloproteinase 9 (MMP-9) secretion by epithelial cells, which is also associated with the tissue damage [166,167]. In line with the crucial role of IL-17A in SjS pathogenesis, Il17a-deficient mice fail to develop SjS [100], while mice with overexpressed IL-17A in SGs exhibit a SjS-like phenotype [168].
IL-22 is another pro-inflammatory cytokine released by Th17 [101,169]. IL-22 is also found to be highly expressed in the SGs of SjS patients, correlating with their clinical manifestations [170]. IL-22 acts on epithelial and myeloid cells, and induces them to secret cytokines and chemokines, such as CXCL13 [170]. The formation of ectopic GC in the SGs of SjS patients is facilitated by IL-22 in a CXCL13-dependent manner [171]. In addition, IL-22, along with IL-17A, promote B-cell infiltration and Th17 polarization [126,170,171]. Deficiency of Il22 reduces the infiltration of B cells in the SGs of SjS mice [171].

4.2.2. IL-21

Accumulating evidence suggests that IL-21 plays a critical pathogenetic role in SjS. High levels of IL-21 and IL-21R are detected in the SGs and serum of SjS patients [172]. Additionally, several SjS-associated genes identified by genome-wide association studies (GWAS) are closely related to the IL-21 pathway [173]. IL-21-producing cells consist of Th17, Tfh and CCR9+ Th cells [174,175]. IL-21 displays a pleiotropic impact on the proliferation, apoptosis and differentiation of T and B cells, correlating with disease progression [173]. In addition, IL-21 promotes the proliferation and cytotoxicity of CD8+ T cells, which induces damages of SGECs [176]. Consistently, the local suppression of IL-21 in submandibular glands retards SjS-like symptoms in mice [177].

4.2.3. IL-4

IL-4 level is found to increase in SGs of SjS patients, especially those exhibiting significant B-cell infiltration [129]. IL-4/STAT6 signaling pathway is essential in the progression of SjS-like diseases [178]. The major source of IL-4 in SjS patients is Tfh, rather than Th2 cells [107]. IL-4 promotes CXCL12 production from stromal precursor cells [179]. Additionally, IL-4, together with CXCL12, participate in regulating B-cell activation, migration and maturation [179,180,181]. The genetic deficiency of Il4 or Stat6 impedes antibody switches to IgG1 isotype against muscarinic acetylcholine type-3 receptor (anti-M3R) [182]. In addition, IL-4 also exacerbates inflammation in SjS patients by inducing the apoptosis of SGECs [183].

4.3. Tumor-Necrosis-Factor Superfamily Members

The TNF superfamily is composed of more than 20 structurally related protein ligands [184]. They exert important immune regulatory roles and are implicated in various autoimmune diseases [185,186]. In SjS patients, TNF superfamily members, such as TNF-α, BAFF and APRIL, are also upregulated, and play critical roles in the immunopathogenesis of the disease [187,188,189,190].

4.3.1. TNF-α

TNF-α is involved in numerous autoimmune diseases, while anti-TNF-α treatment has also showed great promises as a therapeutic strategy [191]. In SjS patients, elevated expression of TNF-α is detected in both SGs and serum [189,190]. TNF-α is produced by many cells, especially Th1 and SGECs [190,192]. Alone, or in combination with IFN-γ, it induces the apoptosis of SG cells, which leads to the dysfunction of the tissue [193].

4.3.2. BAFF and APRIL

BAFF and APRIL share similar biological roles in promoting B-cell differentiation, survival and autoantibody secretion [194,195]. The levels of BAFF and APRIL in serum positively correlate with the disease severity of SjS [188]. BAFF is derived by several types of cells, including antigen-presenting cells, T cells, neutrophils and SGECs, after they are activated by type I or type II IFNs [146,187]. Given its role in promoting B-cell responses, excessive BAFF results in the accumulation of autoreactive B cells and thus the disease progresses [146,187,196,197]. In addition, BAFF-transgenic mice also exhibit enhanced B-cell infiltration and aggravated hyperplasia in SGs. Furthermore, once they age, they mostly develop SjS-like diseases [198].

5. Epigenetic Modifications in SjS

According to the role of epigenetic regulations in immunocyte activation, epigenetic changes can also be involved in the immunopathogenesis of SjS. Indeed, DNA methylation, histone modifications, as well as ncRNAs play important roles in the progression of SjS. They are involved in regulating the aberrant infiltration and activation of T and B cells and promote the production of pro-inflammatory cytokines during SjS immunopathogenesis (Figure 4).

5.1. Epigenetic Modification of IFN-Related Genes

Numerous studies have confirmed the important role of the IFN pathway in the immunopathogenesis of SjS. In pSS patients, disease progression can be divided into three key stages: activation of the innate immune system, especially via the IFN pathway; the activation of T and NK cells due to HLA-related predisposition and through IL-12–IFN-γ pathways; and the activation of B cells, presumably through CXCR5-mediated recruitment to lymphoid follicles and B-cell receptor activation. Epigenetic modifications are critical in activating the IFN pathway [199].
The correlation between DNA methylation and disease progression in pSS patients has been substantially studied. DNA methylation was compared between the naïve CD4+ T cells of 11 pSS patients and 11 healthy cohorts in an epigenome-wide study [13]. The hypomethylated CpG sites in pSS patients are related to genes of the IFN pathway, such as STAT1, IFI44L, USP18 and IFITM1. In another comprehensive study of DNA methylation of whole blood, CD19+ B cells as well as minor SG biopsies from pSS patients, prominent hypomethylation on IFN-regulated genes was also confirmed, which correlated with their increased expression. Furthermore, the top 12 differentially methylated CpG sites in pSS patients, such as MX1, IFI44L, PARP9, PLSCR1, IFIT1, IFITM1 and HLA-A, were found to be mainly influenced by the presence of anti-Sjögren’s Syndrome antigen A (SSA) and/or anti-Sjögren’s Syndrome antigen B (SSB) antibodies [13,200]. The genome-wide comparison of DNA methylation profiles in the CD4+ T cells and CD19+ B cells of pSS patients and age-matched health cohorts using 450K BeadChip suggested that the major methylation alterations were present in B cells and genetic at-risk loci [199]. The DNA methylation changes in B cells were further quantified by Pyrosequencing, and alterations in IFN-related genes, such as IFITM1, IFITM3, IFI44L and IRF5, were validated. Intriguingly, the methylation status of B cells was closely correlated with disease progression. Differentially methylated genes in patients positive for at least one pSS-specific autoantibody (anti-SSA, anti-SSB or anti-SSA/anti-SSB) showed a significantly enriched IFN signature [199].
Histone modifications in SjS patients are still quite elusive, while expression changes on miRNAs are reported in some studies. The differential expression of miRNAs, including hsa-miR-378a-3p, hsa-miR-222-3p, hsa-miR-26a-5p, hsa-miR-30b-5p and hsa-miR-19b-3p, were discovered in a comparative study of CD4+ T cells and CD19+ B cells from pSS patients and healthy cohorts [11,201]. Of them, hsa-miR-30b-5p is one of the most differentially expressed miRNAs in the SGs of pSS patients [202]. Additionally, hsa-miR-146a-5p is known to aggravate inflammatory responses. It targets IRF5, STAT1 and IRAK1, which exert regulatory roles in the IFN and NF-kB pathways [61].

5.2. Epigenetic Modifications in B Cells

As mentioned above, in SjS patients, methylation changes in B cells are more obvious than in T cells, suggesting an important role of B cells in SjS progression. The hypomethylation of IFN-induced genes, including STAT1, IFI44L and IFITM1, was found in peripheral blood B cells, especially from SSA and/or SSB antibody-positive patients [200]. The hypermethylated sites in the B and T cells of SjS patients overlap with histone modifications, corroborating that the related transcriptions are inactivated, whereas hypomethylated sites usually overlap with enhancer sites [203].
SjS patients often exhibit increased SSA and SSB autoantigens, correlated with the aberrant expression of the miRNAs targeting them. In minor SGs and peripheral blood of SjS patients, a large number of miRNAs display differential expression patterns. The miRNA difference also exists in T and B cells from the peripheral blood of SjS patients, especially those positive for anti-SSA autoantibodies. The expression of 372 miRNAs in purified blood cells from SjS patients and health cohorts were analyzed in a recent study. A total of 21 miRNAs showed significant expression differences in T cells, while 24 differentially expressed miRNAs were identified in B cells [201]. Among the differentially expressed miRNAs, hsa-miR-30b-5p was previously reported to negatively regulate BAFF expression. Thus, it may play a role in regulating B-cell function [202].

5.3. Epigenetic Modification of Foxp3

Treg exerting immune suppressive functions are quantitatively and qualitatively defective in SjS patients. Foxp3 (forkhead box p3) is the master transcription factor of Treg, which dictates their development and function [204]. Its expression is substantially regulated by epigenetic modifications. For example, the methylation status of the FOXP3 promoter region determines its expression. In a study of 15 pSS patients, the FOXP3 promoter in CD4+ T cells was found to be hypermethylated, correlated with the significantly reduced expression of FOXP3 mRNA and protein [205].

5.4. Correlation between SjS-Related SNPs and Epigenetic Changes

GWAS has been used to assess the association of genetic variation and unique traits in SjS patients, and an increasing number of susceptible SNPs have been identified [11,206,207,208]. Though genes that directly induce SjS have already been identified through the large studies [10,11], several SNPs in molecules of signaling pathways, including IFN signature (IRF5 STAT1, STAT4 and IL12A), B- and T-cell signaling (BAFF, GTF2I, TNFSF4, CXCR5, CCL11 and TNFAIP3) and the NF-κB pathway (TNIP1, CARD8, IKBKE, IRAK1 and TANK), do appear to be involved in the pathogenesis of SjS. Given the fact that most SNPs are located in noncoding sequences, they are supposed to potentially affect the regulation of genes, resulting in altered transcription levels, splicing manners and epigenetic modifications [209]. In a GWAS performed to discover the difference between pSS patients and healthy controls, a SNP in the methyl-CpG-binding protein 2 (MECP2) gene (rs17435) on the Χ chromosome was identified, which, for the first time, verified the genetic association between SNPs on the Χ chromosome and pSS [210]. MECP2 is associated with DNA methylation-induced transcriptional silencing. In SLE, it has been reported to be associated with disease susceptibility [211]. Accordingly, this SNP may be also involved in the pathogenesis of pSS. Nevertheless, further studies are still needed to unravel the correlation between SjS-related SNPs and the aberrant epigenetic modification changes during the pathogenesis of SjS.

6. Epigenetic Therapeutics of SjS

SjS manifests in various ways, typically characterized by keratoconjunctivitis sicca and dry mouth. Fatigue infections also usually threaten the patients. Thus, the major therapeutic aim for most SjS patients is to improve the quality of their lives by ameliorating the sicca and fatigue symptoms. Anti-inflammatory treatments, such as topical corticosteroids, topical cyclosporine and oral secretagogues, are commonly implied to prevent the pathogenesis of the disease [157]. In addition, targeting therapy, such as Rituximab, a monoclonal anti-CD20 antibody for depleting auto-responsive B cells, has also shown promising therapeutic results. The efficacy of rituximab in reducing fatigue in pSS patients was studied in a double-blind, placebo-controlled randomized trial, which showed significant improvement in the rituximab group [158].
The cellular and molecular etiology of SjS is quite complicated, considering the immunocytes and effector cytokines involved in the autoimmune responses. In addition, they are also dynamically changing along with the immunopathogenesis of the disease. Thus, new therapies targeting pro-inflammatory cytokines or autoreactive immunocytes in SjS patients are still worth exploring. Based on our description, epigenetics is intensively involved in the pathogenesis of SjS, and it shows broad impacts on the abnormal activation of T and B cells, as well as their aberrant production of pro-inflammatory cytokines. Accordingly, we particularly considered the possibility of taking epigenetic targets as a strategy for SjS therapy. Actually, epigenetic drugs have already been used in other autoimmune diseases, especially RA and SLE. Increased DNA methylation in CD4+ T cells is also found in RA, associated with their autoimmune responses. Methotrexate (MTX), a first-line drug for RA treatment through antagonizing folate metabolism, affects one-carbon metabolism to disrupt the methyl transfer process of CpG methylation [212]. In addition, the inhibition of histone methyltransferase Ezh2 is currently under evaluation for the therapy of SLE. Ezh2 inhibition by DZNep significantly reduces renal inflammation and improves the survival of MRL/lpr spontaneous lupus mice before and after the disease onset [135]. Additionally, a preliminary study demonstrated that the HDAC inhibitors VPA and butyrate diminished plasma cell differentiation in MRL/lpr spontaneous lupus mice, with unaffected B-cell viability and proliferation [40].
Given the involvement of epigenetic regulators, such as DNMT1, DNMT3A, DNMT3B, TET2, TET3, HDAC1, HDAC11, SIRT1 and miR-146a, in SjS immunopathogenesis, they can all be possibly considered as therapeutic targets. In particular, as previously mentioned, the IFN pathway plays critical roles in promoting SjS progression, while it is well studied that methylation changes in T and B cells are closely associated with the activation of IFN-related genes. Thus, perhaps targeting therapies towards the abnormal methylation changes in the auto-responsive immunocytes of SjS patients should be particularly considered in future.

7. Conclusions

SjS is a common chronic autoimmune rheumatic disease, caused by complicated cellular and molecular etiologies. Disease progression is companied by the abnormal activation of T and B cells sequentially, including Th1, Th17, Tfh and autoreactive B cells. Key effector cytokines produced by these immunocytes are closely correlated with the autoimmune responses and the damages induced in glandular tissues. The involvement of epigenetic regulations during immune cell activation has attracted considerable attention and has been intensively studied in the past. In particular, epigenetic changes in auto-responsive immunocytes from SjS patients have also been discovered. Thus, unraveling the involvement of epigenetic changes in SjS progression has provided many new insights into its immunopathogenesis and therapy. Especially, these studies reveal that IFN-related genes are hypomethylation in multiple immunocytes, such as T and B cells, as well as SGECs, which play critical roles in the immunopathogenesis of SjS. These discoveries suggest that epigenetic targeting could be considered as a potential therapeutic strategy of SjS. The application of epigenetic treatment in other autoimmune diseases, such as RA and SLE, further demonstrates this possibility. Nevertheless, more studies about the epigenetic modification changes and their impacts on pro-inflammatory cytokine production and SjS progression are still required to precisely design and adjust the epigenetic therapy.

Author Contributions

Conceptualization, C.Z.; writing—original draft preparation, C.Z., P.L., M.H., X.Z., G.R. and S.M.; supervision, C.Z.; funding acquisition, C.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Natural Science Foundation of China, grant numbers 31770957, 91842102 and 32170896, and the Natural Science Foundation of Beijing, grant number 18G10645.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

The authors are particularly thankful to Xiaolin Sun for his helpful discussion.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hayashi, T. Dysfunction of lacrimal and salivary glands in Sjögren’s syndrome: Nonimmunologic injury in preinflammatory phase and mouse model. J. Biomed. Biotechnol. 2011, 2011, 407031. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Theander, E.; Jonsson, R.; Sjöström, B.; Brokstad, K.; Olsson, P.; Henriksson, G. Prediction of Sjögren’s Syndrome Years before Diagnosis and Identification of Patients with Early Onset and Severe Disease Course by Autoantibody Profiling. Arthritis Rheumatol. 2015, 67, 2427–2436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Fragkioudaki, S.; Moutsopoulos, H.M.; Mavragani, C.P. Sjgren’s Syndrome; The Heart in Rheumatic, Autoimmune and Inflammatory Diseases; Academic Press: Cambridge, MA, USA, 2017. [Google Scholar]
  4. Solans-Laqué, R.; López-Hernandez, A.; Bosch-Gil, J.A.; Palacios, A.; Campillo, M.; Vilardell-Tarres, M. Risk, predictors, and clinical characteristics of lymphoma development in primary Sjögren’s syndrome. Semin. Arthritis Rheum. 2011, 41, 415–423. [Google Scholar] [CrossRef] [PubMed]
  5. Kapsogeorgou, E.K.; Papageorgiou, A.; Protogerou, A.D.; Voulgarelis, M.; Tzioufas, A.G. Low miR200b-5p levels in minor salivary glands: A novel molecular marker predicting lymphoma development in patients with Sjögren’s syndrome. Ann. Rheum. Dis. 2018, 77, 1200–1207. [Google Scholar] [CrossRef] [PubMed]
  6. Qin, B.; Wang, J.; Yang, Z.; Yang, M.; Ma, N.; Huang, F.; Zhong, R. Epidemiology of primary Sjogren’s syndrome: A systematic review and meta-analysis. Ann. Rheum. Dis. 2015, 74, 1983–1989. [Google Scholar] [CrossRef] [PubMed]
  7. Harris, V.M.; Sharma, R.; Cavett, J.; Kurien, B.T.; Liu, K.; Koelsch, K.A.; Rasmussen, A.; Radfar, L.; Lewis, D.; Stone, D.U.; et al. Klinefelter’s syndrome (47,XXY) is in excess among men with Sjögren’s syndrome. Clin. Immunol. 2016, 168, 25–29. [Google Scholar] [CrossRef] [Green Version]
  8. Liu, K.; Kurien, B.T.; Zimmerman, S.L.; Kaufman, K.M.; Taft, D.H.; Kottyan, L.C.; Lazaro, S.; Weaver, C.A.; Ice, J.A.; Adler, A.J.; et al. X Chromosome Dose and Sex Bias in Autoimmune Diseases: Increased Prevalence of 47,XXX in Systemic Lupus Erythematosus and Sjögren’s Syndrome. Arthritis Rheumatol. 2016, 68, 1290–1300. [Google Scholar] [CrossRef]
  9. Shaw, T.M.; Zhang, W.; McCoy, S.S.; Pagenkopf, A.; Carp, D.M.; Garg, S.; Parker, M.H.; Qiu, X.; Scofield, R.H.; Galipeau, J.; et al. X-linked genes exhibit miR6891-5p-regulated skewing in Sjögren’s syndrome. J. Mol. Med. 2022, 100, 1–13. [Google Scholar] [CrossRef]
  10. Li, Y.; Zhang, K.; Chen, H.; Sun, F.; Xu, J.; Wu, Z.; Li, P.; Zhang, L.; Du, Y.; Luan, H.; et al. A genome-wide association study in Han Chinese identifies a susceptibility locus for primary Sjögren’s syndrome at 7q11.23. Nat. Genet. 2013, 45, 1361–1365. [Google Scholar] [CrossRef]
  11. Lessard, C.J.; Li, H.; Adrianto, I.; Ice, J.A.; Rasmussen, A.; Grundahl, K.M.; Kelly, J.A.; Dozmorov, M.G.; Miceli-Richard, C.; Bowman, S.; et al. Variants at multiple loci implicated in both innate and adaptive immune responses are associated with Sjögren’s syndrome. Nat. Genet. 2013, 45, 1284–1292. [Google Scholar] [CrossRef]
  12. Xuan, J.; Ji, Z.; Wang, B.; Zeng, X.; Shi, G.J.F.I.I. Serological Evidence for the Association between Epstein-Barr Virus Infection and Sjgren’s Syndrome. Front. Immunol. 2020, 11, 590444. [Google Scholar] [CrossRef] [PubMed]
  13. Altorok, N.; Coit, P.; Hughes, T.; Koelsch, K.A.; Stone, D.U.; Rasmussen, A.; Radfar, L.; Scofield, R.H.; Sivils, K.L.; Farris, A.D.; et al. Genome-Wide DNA Methylation Patterns in Naive CD4+ T Cells from Patients with Primary Sjogren’s Syndrome. Arthritis Rheumatol. 2013, 66, 731–739. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Arvaniti, P.; Zachou, K.; Lyberopoulou, A.; Gatselis, N.K.; Brooks, W.H.; Dalekos, G.N.; Renaudineau, Y. Epigenetic Modifications in Generalized Autoimmune Epithelitis: Sjogren’s Syndrome and Primary Biliary Cholangitis. Epigenomes 2019, 3, 15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Mazzone, R.; Zwergel, C.; Artico, M.; Taurone, S.; Ralli, M.; Greco, A.; Mai, A. The emerging role of epigenetics in human autoimmune disorders. Clin. Epigenetics 2019, 11, 34. [Google Scholar] [CrossRef] [Green Version]
  16. Schmolka, N.; Silva-Santos, B.; Gomes, A.Q. Epigenetic mechanisms in the regulation of lymphocyte differentiation. Epigenetics Immune Syst. 2020, 16, 77–116. [Google Scholar]
  17. Imgenberg-Kreuz, J.; Sandling, J.K.; Nordmark, G. Epigenetic alterations in primary Sjogren’s syndrome—An overview. Clin. Immunol. 2018, 196, 12–20. [Google Scholar] [CrossRef]
  18. Correa, L.O.; Jordan, M.S.; Carty, S.A. DNA Methylation in T-Cell Development and Differentiation. Crit. Rev. Immunol. 2020, 40, 135–156. [Google Scholar] [CrossRef]
  19. Thomas, R.M.; Gamper, C.J.; Ladle, B.H.; Powell, J.D.; Wells, A.D. De novo DNA methylation is required to restrict T helper lineage plasticity. J. Biol. Chem. 2012, 287, 22900–22909. [Google Scholar] [CrossRef] [Green Version]
  20. Wang, L.; Liu, Y.; Beier, U.H.; Han, R.; Bhatti, T.R.; Akimova, T.; Hancock, W.W. Foxp3+ T-regulatory cells require DNA methyltransferase 1 expression to prevent development of lethal autoimmunity. Blood 2013, 121, 3631–3639. [Google Scholar] [CrossRef] [Green Version]
  21. Ichiyama, K.; Chen, T.; Wang, X.; Yan, X.; Kim, B.S.; Tanaka, S.; Ndiaye-Lobry, D.; Deng, Y.; Zou, Y.; Zheng, P.; et al. The methylcytosine dioxygenase Tet2 promotes DNA demethylation and activation of cytokine gene expression in T cells. Immunity 2015, 42, 613–626. [Google Scholar] [CrossRef] [Green Version]
  22. Yang, R.; Qu, C.; Zhou, Y.; Konkel, J.E.; Shi, S.; Liu, Y.; Chen, C.; Liu, S.; Liu, D.; Chen, Y.; et al. Hydrogen Sulfide Promotes Tet1- and Tet2-Mediated Foxp3 Demethylation to Drive Regulatory T Cell Differentiation and Maintain Immune Homeostasis. Immunity 2015, 43, 251–263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Yue, X.; Trifari, S.; Äijö, T.; Tsagaratou, A.; Pastor, W.A.; Zepeda-Martínez, J.A.; Lio, C.W.; Li, X.; Huang, Y.; Vijayanand, P.; et al. Control of Foxp3 stability through modulation of TET activity. J. Exp. Med. 2016, 213, 377–397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Nair, V.S.; Oh, K.I. Vitamin C and DNA Demethylation in Regulatory T Cells. In Handbook of Nutrition, Diet, and Epigenetics; Patel, V., Preedy, V., Eds.; Springer International Publishing: Cham, Switzerland, 2017; pp. 1–15. [Google Scholar]
  25. Barwick, B.G.; Scharer, C.D.; Martinez, R.J.; Price, M.J.; Wein, A.N.; Haines, R.R.; Bally, A.P.R.; Kohlmeier, J.E.; Boss, J.M. B cell activation and plasma cell differentiation are inhibited by de novo DNA methylation. Nat. Commun. 2018, 9, 1900. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Lio, C.J.; Shukla, V.; Samaniego-Castruita, D.; González-Avalos, E.; Chakraborty, A.; Yue, X.; Schatz, D.G.; Ay, F.; Rao, A. TET enzymes augment activation-induced deaminase (AID) expression via 5-hydroxymethylcytosine modifications at the Aicda superenhancer. Sci. Immunol. 2019, 4, eaau7523. [Google Scholar] [CrossRef]
  27. Li, J.; Ding, Y.; Ling, Z.J.T.E. Histone-Mediated Transgenerational Epigenetics; Academic Press: Cambridge, MA, USA, 2014; pp. 87–103. [Google Scholar]
  28. Ellmeier, W.; Seiser, C. Histone deacetylase function in CD4(+) T cells. Nat. Rev. Immunol. 2018, 18, 617–634. [Google Scholar] [CrossRef]
  29. Grausenburger, R.; Bilic, I.; Boucheron, N.; Zupkovitz, G.; El-Housseiny, L.; Tschismarov, R.; Zhang, Y.; Rembold, M.; Gaisberger, M.; Hartl, A.; et al. Conditional deletion of histone deacetylase 1 in T cells leads to enhanced airway inflammation and increased Th2 cytokine production. J. Immunol. 2010, 185, 3489–3497. [Google Scholar] [CrossRef]
  30. Woods, D.M.; Woan, K.V.; Cheng, F.; Sodré, A.L.; Wang, D.; Wu, Y.; Wang, Z.; Chen, J.; Powers, J.; Pinilla-Ibarz, J.; et al. T cells lacking HDAC11 have increased effector functions and mediate enhanced alloreactivity in a murine model. Blood 2017, 130, 146–155. [Google Scholar] [CrossRef] [Green Version]
  31. Lim, H.W.; Kang, S.G.; Ryu, J.K.; Schilling, B.; Fei, M.; Lee, I.S.; Kehasse, A.; Shirakawa, K.; Yokoyama, M.; Schnölzer, M.; et al. SIRT1 deacetylates RORγt and enhances Th17 cell generation. J. Exp. Med. 2015, 212, 607–617. [Google Scholar] [CrossRef]
  32. El Kaoutari, A.; Armougom, F.; Gordon, J.I.; Raoult, D.; Henrissat, B. The abundance and variety of carbohydrate-active enzymes in the human gut microbiota. Nat. Rev. Microbiol. 2013, 11, 497–504. [Google Scholar] [CrossRef]
  33. Kim, C.H.; Park, J.; Kim, M. Gut microbiota-derived short-chain Fatty acids, T cells, and inflammation. Immune Netw. 2014, 14, 277–288. [Google Scholar] [CrossRef] [Green Version]
  34. Tanoue, T.; Atarashi, K.; Honda, K. Development and maintenance of intestinal regulatory T cells. Nat. Rev. Immunol. 2016, 16, 295–309. [Google Scholar] [CrossRef] [PubMed]
  35. Arpaia, N.; Campbell, C.; Fan, X.; Dikiy, S.; van der Veeken, J.; de Roos, P.; Liu, H.; Cross, J.R.; Pfeffer, K.; Coffer, P.J.; et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013, 504, 451–455. [Google Scholar] [CrossRef] [PubMed]
  36. Sun, M.; Wu, W.; Chen, L.; Yang, W.; Huang, X.; Ma, C.; Chen, F.; Xiao, Y.; Zhao, Y.; Ma, C.; et al. Microbiota-derived short-chain fatty acids promote Th1 cell IL-10 production to maintain intestinal homeostasis. Nat. Commun. 2018, 9, 3555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Luu, M.; Pautz, S.; Kohl, V.; Singh, R.; Romero, R.; Lucas, S.; Hofmann, J.; Raifer, H.; Vachharajani, N.; Carrascosa, L.C.; et al. The short-chain fatty acid pentanoate suppresses autoimmunity by modulating the metabolic-epigenetic crosstalk in lymphocytes. Nat. Commun. 2019, 10, 760. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Lemercier, C.; Brocard, M.P.; Puvion-Dutilleul, F.; Kao, H.Y.; Albagli, O.; Khochbin, S. Class II histone deacetylases are directly recruited by BCL6 transcriptional repressor. J. Biol. Chem. 2002, 277, 22045–22052. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Lee, S.C.; Bottaro, A.; Insel, R.A. Activation of terminal B cell differentiation by inhibition of histone deacetylation. Mol. Immunol. 2003, 39, 923–932. [Google Scholar] [CrossRef]
  40. White, C.A.; Pone, E.J.; Lam, T.; Tat, C.; Hayama, K.L.; Li, G.; Zan, H.; Casali, P. Histone deacetylase inhibitors upregulate B cell microRNAs that silence AID and Blimp-1 expression for epigenetic modulation of antibody and autoantibody responses. J. Immunol. 2014, 193, 5933–5950. [Google Scholar] [CrossRef]
  41. Wei, J.; Bhattacharyya, S.; Tourtellotte, W.G.; Varga, J. Fibrosis in systemic sclerosis: Emerging concepts and implications for targeted therapy. Autoimmun. Rev. 2011, 10, 267–275. [Google Scholar] [CrossRef] [Green Version]
  42. Scrivo, R.; Vasile, M.; Bartosiewicz, I.; Valesini, G. Inflammation as “common soil” of the multifactorial diseases. Autoimmun. Rev. 2011, 10, 369–374. [Google Scholar] [CrossRef]
  43. Shu, J.; Silva, B.; Gao, T.; Xu, Z.; Cui, J. Dynamic and Modularized MicroRNA Regulation and Its Implication in Human Cancers. Sci. Rep. 2017, 7, 13356. [Google Scholar] [CrossRef] [Green Version]
  44. Schickel, R.; Boyerinas, B.; Park, S.M.; Peter, M.E. MicroRNAs: Key players in the immune system, differentiation, tumorigenesis and cell death. Oncogene 2008, 27, 5959–5974. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Jeker, L.T.; Bluestone, J.A. MicroRNA regulation of T-cell differentiation and function. Immunol. Rev. 2013, 253, 65–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Danger, R.; Braza, F.; Giral, M.; Soulillou, J.P.; Brouard, S. MicroRNAs, Major Players in B Cells Homeostasis and Function. Front. Immunol. 2014, 5, 98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Li, B.; Wang, X.; Choi, I.Y.; Wang, Y.C.; Liu, S.; Pham, A.T.; Moon, H.; Smith, D.J.; Rao, D.S.; Boldin, M.P.; et al. miR-146a modulates autoreactive Th17 cell differentiation and regulates organ-specific autoimmunity. J. Clin. Investig. 2017, 127, 3702–3716. [Google Scholar] [CrossRef] [Green Version]
  48. Basak, J.; Majsterek, I. miRNA-Dependent CD4(+) T Cell Differentiation in the Pathogenesis of Multiple Sclerosis. Mult. Scler. Int. 2021, 2021, 8825588. [Google Scholar] [CrossRef]
  49. Mohnle, P.; Schutz, S.V.; van der Heide, V.; Hubner, M.; Luchting, B.; Sedlbauer, J.; Limbeck, E.; Hinske, L.C.; Briegel, J.; Kreth, S. MicroRNA-146a controls Th1-cell differentiation of human CD4+ T lymphocytes by targeting PRKCepsilon. Eur. J. Immunol. 2015, 45, 260–272. [Google Scholar] [CrossRef]
  50. Lu, L.F.; Boldin, M.P.; Chaudhry, A.; Lin, L.L.; Taganov, K.D.; Hanada, T.; Yoshimura, A.; Baltimore, D.; Rudensky, A.Y. Function of miR-146a in controlling Treg cell-mediated regulation of Th1 responses. Cell 2010, 142, 914–929. [Google Scholar] [CrossRef] [Green Version]
  51. Pratama, A.; Srivastava, M.; Williams, N.J.; Papa, I.; Lee, S.K.; Dinh, X.T.; Hutloff, A.; Jordan, M.A.; Zhao, J.L.; Casellas, R.; et al. MicroRNA-146a regulates ICOS-ICOSL signalling to limit accumulation of T follicular helper cells and germinal centres. Nat. Commun. 2015, 6, 6436. [Google Scholar] [CrossRef]
  52. King, J.K.; Ung, N.M.; Paing, M.H.; Contreras, J.R.; Alberti, M.O.; Fernando, T.R.; Zhang, K.; Pellegrini, M.; Rao, D.S. Regulation of Marginal Zone B-Cell Differentiation by MicroRNA-146a. Front. Immunol. 2016, 7, 670. [Google Scholar] [CrossRef] [Green Version]
  53. Gururajan, M.; Haga, C.L.; Das, S.; Leu, C.M.; Hodson, D.; Josson, S.; Turner, M.; Cooper, M.D. MicroRNA 125b inhibition of B cell differentiation in germinal centers. Int. Immunol. 2010, 22, 583–592. [Google Scholar] [CrossRef]
  54. Nie, K.; Zhang, T.; Allawi, H.; Gomez, M.; Liu, Y.; Chadburn, A.; Wang, Y.L.; Knowles, D.M.; Tam, W. Epigenetic down-regulation of the tumor suppressor gene PRDM1/Blimp-1 in diffuse large B cell lymphomas: A potential role of the microRNA let-7. Am. J. Pathol. 2010, 177, 1470–1479. [Google Scholar] [CrossRef] [PubMed]
  55. Baulina, N.M.; Kulakova, O.G.; Favorova, O.O. MicroRNAs: The Role in Autoimmune Inflammation. Acta Nat. 2016, 8, 21–33. [Google Scholar] [CrossRef]
  56. Weissman, R.; Pilar, N.; Durham, B.H.; Ki, M.; Mazor, R.D.; Abdel-Wahab, O.I.; Shomron, N.; Shpilberg, O.; Diamond, E.L.; Rokah, O. The Role of microRNAs in the Pathogenesis of Erdheim-Chester Disease and Their Potential Use As Biomarkers for Diagnosis and Prognosis of the Disease. Blood 2018, 132, 2397. [Google Scholar] [CrossRef]
  57. Garo, L.P.; Murugaiyan, G. Contribution of MicroRNAs to autoimmune diseases. Cell Mol. Life Sci. 2016, 73, 2041–2051. [Google Scholar] [CrossRef] [PubMed]
  58. Zilahi, E.; Tarr, T.; Papp, G.; Griger, Z.; Sipka, S.; Zeher, M. Increased microRNA-146a/b, TRAF6 gene and decreased IRAK1 gene expressions in the peripheral mononuclear cells of patients with Sjogren’s syndrome. Immunol. Lett. 2012, 141, 165–168. [Google Scholar] [CrossRef]
  59. Shi, H.; Zheng, L.Y.; Zhang, P.; Yu, C.Q. miR-146a and miR-155 expression in PBMCs from patients with Sjögren’s syndrome. J. Oral Pathol. Med. 2014, 43, 792–797. [Google Scholar] [CrossRef]
  60. Taganov, K.D.; Boldin, M.P.; Chang, K.J.; Baltimore, D. NF-kappaB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proc. Natl. Acad. Sci. USA 2006, 103, 12481–12486. [Google Scholar] [CrossRef] [Green Version]
  61. Boldin, M.P.; Taganov, K.D.; Rao, D.S.; Yang, L.L.; Zhao, J.L.; Kalwani, M.; Garcia-Flores, Y.; Luong, M.; Devrekanli, A.; Xu, J.; et al. miR-146a is a significant brake on autoimmunity, myeloproliferation, and cancer in mice. J. Exp. Med. 2011, 208, 1189–1201. [Google Scholar] [CrossRef]
  62. Zhao, J.L.; Rao, D.S.; Boldin, M.P.; Taganov, K.D.; O’Connell, R.M.; Baltimore, D. NF-kappa B dysregulation in microRNA-146a-deficient mice drives the development of myeloid malignancies. Proc. Natl. Acad. Sci. USA 2011, 108, 9184–9189. [Google Scholar] [CrossRef] [Green Version]
  63. Yang, L.; Boldin, M.P.; Yu, Y.; Liu, C.S.; Ea, C.K.; Ramakrishnan, P.; Taganov, K.D.; Zhao, J.L.; Baltimore, D. miR-146a controls the resolution of T cell responses in mice. J. Exp. Med. 2012, 209, 1655–1670. [Google Scholar] [CrossRef] [Green Version]
  64. Fulci, V.; Chiaretti, S.; Goldoni, M.; Azzalin, G.; Carucci, N.; Tavolaro, S.; Castellano, L.; Magrelli, A.; Citarella, F.; Messina, M.; et al. Quantitative technologies establish a novel microRNA profile of chronic lymphocytic leukemia. Blood 2007, 109, 4944–4951. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Peng, L.; Ma, W.; Yi, F.; Yang, Y.J.; Lin, W.; Chen, H.; Zhang, X.; Zhang, L.H.; Zhang, F.; Du, Q. MicroRNA profiling in Chinese patients with primary Sjögren syndrome reveals elevated miRNA-181a in peripheral blood mononuclear cells. J. Rheumatol. 2014, 41, 2208–2213. [Google Scholar] [CrossRef]
  66. Benhamou, D.; Labi, V.; Getahun, A.; Benchetrit, E.; Dowery, R.; Rajewsky, K.; Cambier, J.C.; Melamed, D. The c-Myc/miR17-92/PTEN Axis Tunes PI3K Activity to Control Expression of Recombination Activating Genes in Early B Cell Development. Front. Immunol. 2018, 9, 2715. [Google Scholar] [CrossRef] [PubMed]
  67. Ventura, A.; Young, A.G.; Winslow, M.M.; Lintault, L.; Meissner, A.; Erkeland, S.J.; Newman, J.; Bronson, R.T.; Crowley, D.; Stone, J.R.; et al. Targeted deletion reveals essential and overlapping functions of the miR-17 through 92 family of miRNA clusters. Cell 2008, 132, 875–886. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Gonzalez-Martin, A.; Adams, B.D.; Lai, M.; Shepherd, J.; Salvador-Bernaldez, M.; Salvador, J.M.; Lu, J.; Nemazee, D.; Xiao, C. The microRNA miR-148a functions as a critical regulator of B cell tolerance and autoimmunity. Nat. Immunol. 2016, 17, 433–440. [Google Scholar] [CrossRef]
  69. Benhamou, D.; Labi, V.; Novak, R.; Dai, I.; Shafir-Alon, S.; Weiss, A.; Gaujoux, R.; Arnold, R.; Shen-Orr, S.S.; Rajewsky, K.; et al. A c-Myc/miR17-92/Pten Axis Controls PI3K-Mediated Positive and Negative Selection in B Cell Development and Reconstitutes CD19 Deficiency. Cell Rep. 2016, 16, 419–431. [Google Scholar] [CrossRef] [Green Version]
  70. Lu, D.; Nakagawa, R.; Lazzaro, S.; Staudacher, P.; Abreu-Goodger, C.; Henley, T.; Boiani, S.; Leyland, R.; Galloway, A.; Andrews, S.; et al. The miR-155-PU.1 axis acts on Pax5 to enable efficient terminal B cell differentiation. J. Exp. Med. 2014, 211, 2183–2198. [Google Scholar] [CrossRef] [Green Version]
  71. Teng, G.; Hakimpour, P.; Landgraf, P.; Rice, A.; Tuschl, T.; Casellas, R.; Papavasiliou, F.N. MicroRNA-155 is a negative regulator of activation-induced cytidine deaminase. Immunity 2008, 28, 621–629. [Google Scholar] [CrossRef] [Green Version]
  72. Momen-Heravi, F.; Bala, S. miRNA regulation of innate immunity. J. Leukoc. Biol. 2018, 103, 1205–1217. [Google Scholar] [CrossRef]
  73. Berrien-Elliott, M.M.; Sun, Y.; Neal, C.; Ireland, A.; Trissal, M.C.; Sullivan, R.P.; Wagner, J.A.; Leong, J.W.; Wong, P.; Mah-Som, A.Y.; et al. MicroRNA-142 Is Critical for the Homeostasis and Function of Type 1 Innate Lymphoid Cells. Immunity 2019, 51, 479–490.e476. [Google Scholar] [CrossRef]
  74. Fionda, C.; Stabile, H.; Cerboni, C.; Soriani, A.; Gismondi, A.; Cippitelli, M.; Santoni, A. Hitting More Birds with a Stone: Impact of TGF-β on ILC Activity in Cancer. J. Clin. Med. 2020, 9, 143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Roberts, L.B.; Jowett, G.M.; Read, E.; Zabinski, T.; Berkachy, R.; Selkirk, M.E.; Jackson, I.; Niazi, U.; Anandagoda, N.; Araki, M.; et al. MicroRNA-142 Critically Regulates Group 2 Innate Lymphoid Cell Homeostasis and Function. J. Immunol. 2021, 206, 2725–2739. [Google Scholar] [CrossRef]
  76. Singh, P.B.; Pua, H.H.; Happ, H.C.; Schneider, C.; von Moltke, J.; Locksley, R.M.; Baumjohann, D.; Ansel, K.M. MicroRNA regulation of type 2 innate lymphoid cell homeostasis and function in allergic inflammation. J. Exp. Med. 2017, 214, 3627–3643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Pelosi, A.; Alicata, C.; Tumino, N.; Ingegnere, T.; Loiacono, F.; Mingari, M.C.; Moretta, L.; Vacca, P. An Anti-inflammatory microRNA Signature Distinguishes Group 3 Innate Lymphoid Cells From Natural Killer Cells in Human Decidua. Front. Immunol. 2020, 11, 133. [Google Scholar] [CrossRef] [PubMed]
  78. Li, C.; Xu, M.M.; Wang, K.; Adler, A.J.; Vella, A.T.; Zhou, B. Macrophage polarization and meta-inflammation. Transl. Res. J. Lab. Clin. Med. 2018, 191, 29–44. [Google Scholar] [CrossRef]
  79. Essandoh, K.; Li, Y.; Huo, J.; Fan, G.C. MiRNA-Mediated Macrophage Polarization and its Potential Role in the Regulation of Inflammatory Response. Shock 2016, 46, 122–131. [Google Scholar] [CrossRef]
  80. Wang, Z.; Brandt, S.; Medeiros, A.; Wang, S.; Wu, H.; Dent, A.; Serezani, C.H. MicroRNA 21 is a homeostatic regulator of macrophage polarization and prevents prostaglandin E2-mediated M2 generation. PLoS ONE 2015, 10, e0115855. [Google Scholar] [CrossRef]
  81. Lu, T.X.; Munitz, A.; Rothenberg, M.E. MicroRNA-21 is up-regulated in allergic airway inflammation and regulates IL-12p35 expression. J. Immunol. 2009, 182, 4994–5002. [Google Scholar] [CrossRef] [Green Version]
  82. Sheedy, F.J. Turning 21: Induction of miR-21 as a Key Switch in the Inflammatory Response. Front. Immunol. 2015, 6, 19. [Google Scholar] [CrossRef] [Green Version]
  83. Caescu, C.I.; Guo, X.; Tesfa, L.; Bhagat, T.D.; Verma, A.; Zheng, D.; Stanley, E.R. Colony stimulating factor-1 receptor signaling networks inhibit mouse macrophage inflammatory responses by induction of microRNA-21. Blood 2015, 125, e1–e13. [Google Scholar] [CrossRef]
  84. Song, N.; Zhang, T.; Xu, X.; Lu, Z.; Yu, X.; Fang, Y.; Hu, J.; Jia, P.; Teng, J.; Ding, X. miR-21 Protects Against Ischemia/Reperfusion-Induced Acute Kidney Injury by Preventing Epithelial Cell Apoptosis and Inhibiting Dendritic Cell Maturation. Front. Physiol. 2018, 9, 790. [Google Scholar] [CrossRef] [PubMed]
  85. Christodoulou, M.I.; Kapsogeorgou, E.K.; Moutsopoulos, H.M. Characteristics of the minor salivary gland infiltrates in Sjogren’s syndrome. J. Autoimmun. 2010, 34, 400–407. [Google Scholar] [CrossRef] [PubMed]
  86. Rodrigues, A.R.; Soares, R. Inflammation in Sjogren’s syndrome: Cause or consequence? Autoimmunity 2017, 50, 141–150. [Google Scholar] [CrossRef] [PubMed]
  87. Mingueneau, M.; Boudaoud, S.; Haskett, S.; Reynolds, T.L.; Nocturne, G.; Norton, E.; Zhang, X.; Constant, M.; Park, D.; Wang, W.; et al. Cytometry by time-of-flight immunophenotyping identifies a blood Sjogren’s signature correlating with disease activity and glandular inflammation. J. Allergy Clin. Immunol. 2016, 137, 1809–1821.e1812. [Google Scholar] [CrossRef] [PubMed]
  88. Nguyen, C.Q.; Hu, M.H.; Li, Y.; Stewart, C.; Peck, A.B. Salivary gland tissue expression of interleukin-23 and interleukin-17 in Sjögren’s syndrome: Findings in humans and mice. Arthritis Rheum. 2008, 58, 734–743. [Google Scholar] [CrossRef] [Green Version]
  89. van Woerkom, J.M.; Kruize, A.A.; Wenting-van Wijk, M.J.; Knol, E.; Bihari, I.C.; Jacobs, J.W.; Bijlsma, J.W.; Lafeber, F.P.; van Roon, J.A. Salivary gland and peripheral blood T helper 1 and 2 cell activity in Sjögren’s syndrome compared with non-Sjögren’s sicca syndrome. Ann. Rheum. Dis. 2005, 64, 1474–1479. [Google Scholar] [CrossRef] [Green Version]
  90. Ogawa, N.; Ping, L.; Zhenjun, L.; Takada, Y.; Sugai, S. Involvement of the interferon-gamma-induced T cell-attracting chemokines, interferon-gamma-inducible 10-kd protein (CXCL10) and monokine induced by interferon-gamma (CXCL9), in the salivary gland lesions of patients with Sjögren’s syndrome. Arthritis Rheum. 2002, 46, 2730–2741. [Google Scholar] [CrossRef]
  91. Crotty, S. T follicular helper cell differentiation, function, and roles in disease. Immunity 2014, 41, 529–542. [Google Scholar] [CrossRef] [Green Version]
  92. Yao, Y.; Ma, J.F.; Chang, C.; Xu, T.; Gao, C.Y.; Gershwin, M.E.; Lian, Z.X. Immunobiology of T Cells in Sjogren’s Syndrome. Clin. Rev. Allergy Immunol. 2021, 60, 111–131. [Google Scholar] [CrossRef]
  93. Singh, N.; Cohen, P.L. The T cell in Sjogren’s syndrome: Force majeure, not spectateur. J. Autoimmun. 2012, 39, 229–233. [Google Scholar] [CrossRef] [Green Version]
  94. Christodoulou, M.I.; Kapsogeorgou, E.K.; Moutsopoulos, N.M.; Moutsopoulos, H.M. Foxp3+ T-regulatory cells in Sjogren’s syndrome: Correlation with the grade of the autoimmune lesion and certain adverse prognostic factors. Am. J. Pathol. 2008, 173, 1389–1396. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Kamali, A.N.; Noorbakhsh, S.M.; Hamedifar, H.; Jadidi-Niaragh, F.; Yazdani, R.; Bautista, J.M.; Azizi, G. A role for Th1-like Th17 cells in the pathogenesis of inflammatory and autoimmune disorders. Mol. Immunol. 2019, 105, 107–115. [Google Scholar] [CrossRef] [PubMed]
  96. Fallahi, P.; Ferrari, S.M.; Ragusa, F.; Ruffilli, I.; Elia, G.; Paparo, S.R.; Antonelli, A. Th1 Chemokines in Autoimmune Endocrine Disorders. J. Clin. Endocrinol. Metab. 2020, 105, 1046–1060. [Google Scholar] [CrossRef] [PubMed]
  97. Yasuda, K.; Takeuchi, Y.; Hirota, K. The pathogenicity of Th17 cells in autoimmune diseases. Semin. Immunopathol. 2019, 41, 283–297. [Google Scholar] [CrossRef]
  98. Lee, J.Y.; Hall, J.A.; Kroehling, L.; Wu, L.; Najar, T.; Nguyen, H.H.; Lin, W.Y.; Yeung, S.T.; Silva, H.M.; Li, D.; et al. Serum Amyloid A Proteins Induce Pathogenic Th17 Cells and Promote Inflammatory Disease. Cell 2020, 183, 2036–2039. [Google Scholar] [CrossRef]
  99. Ogawa, Y.; Shimizu, E.; Tsubota, K. Interferons and Dry Eye in Sjögren’s Syndrome. Int. J. Mol. Sci. 2018, 19, 3548. [Google Scholar] [CrossRef] [Green Version]
  100. Voigt, A.; Esfandiary, L.; Wanchoo, A.; Glenton, P.; Donate, A.; Craft, W.F.; Craft, S.L.; Nguyen, C.Q. Sexual dimorphic function of IL-17 in salivary gland dysfunction of the C57BL/6.NOD-Aec1Aec2 model of Sjogren’s syndrome. Sci. Rep. 2016, 6, 38717. [Google Scholar] [CrossRef] [Green Version]
  101. Verstappen, G.M.; Corneth, O.B.J.; Bootsma, H.; Kroese, F.G.M. Th17 cells in primary Sjogren’s syndrome: Pathogenicity and plasticity. J. Autoimmun. 2018, 87, 16–25. [Google Scholar] [CrossRef]
  102. Fasano, S.; Mauro, D.; Macaluso, F.; Xiao, F.; Zhao, Y.; Lu, L.; Guggino, G.; Ciccia, F. Pathogenesis of primary Sjögren’s syndrome beyond B lymphocytes. Clin. Exp. Rheumatol. 2020, 38 (Suppl. S126), 315–323. [Google Scholar]
  103. Crotty, S. T Follicular Helper Cell Biology: A Decade of Discovery and Diseases. Immunity 2019, 50, 1132–1148. [Google Scholar] [CrossRef]
  104. Jogdand, G.M.; Mohanty, S.; Devadas, S. Regulators of Tfh Cell Differentiation. Front. Immunol. 2016, 7, 520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Chen, W.; Yang, F.; Xu, G.; Ma, J.; Lin, J. Follicular helper T cells and follicular regulatory T cells in the immunopathology of primary Sjögren’s syndrome. J. Leukoc. Biol. 2021, 109, 437–447. [Google Scholar] [CrossRef] [PubMed]
  106. Szabo, K.; Papp, G.; Barath, S.; Gyimesi, E.; Szanto, A.; Zeher, M. Follicular helper T cells may play an important role in the severity of primary Sjogren’s syndrome. Clin. Immunol. 2013, 147, 95–104. [Google Scholar] [CrossRef] [Green Version]
  107. King, I.L.; Mohrs, M. IL-4-producing CD4+ T cells in reactive lymph nodes during helminth infection are T follicular helper cells. J. Exp. Med. 2009, 206, 1001–1007. [Google Scholar] [CrossRef] [PubMed]
  108. Katsifis, G.E.; Moutsopoulos, N.M.; Wahl, S.M. T lymphocytes in Sjögren’s syndrome: Contributors to and regulators of pathophysiology. Clin. Rev. Allergy Immunol. 2007, 32, 252–264. [Google Scholar] [CrossRef] [PubMed]
  109. Ríos-Ríos, W.J.; Sosa-Luis, S.A.; Torres-Aguilar, H. T Cells Subsets in the Immunopathology and Treatment of Sjogren’s Syndrome. Biomolecules 2020, 10, 1539. [Google Scholar] [CrossRef]
  110. Verstappen, G.M.; Kroese, F.G.M.; Bootsma, H. T cells in primary Sjogren’s syndrome: Targets for early intervention. Rheumatology 2019, 60, 3088–3098. [Google Scholar] [CrossRef] [Green Version]
  111. Raimondi, G.; Shufesky, W.J.; Tokita, D.; Morelli, A.E.; Thomson, A.W. Regulated compartmentalization of programmed cell death-1 discriminates CD4+CD25+ resting regulatory T cells from activated T cells. J. Immunol. 2006, 176, 2808–2816. [Google Scholar] [CrossRef] [Green Version]
  112. Wang, Y.; Luo, J.; Gao, C.; Zhao, X.C. Sat0195 Abnormity Tfh Subsets Indicate Disease Activity While Sirolimus Therapy Restores the Pd-1+Icos+Tfh/Activated Tfr Balance in Primary Sjogren’s Syndrome Patients. BMJ 2020, 79, 1039–1040. [Google Scholar] [CrossRef]
  113. Verstappen, G.M.; Nakshbandi, U.; Mossel, E.; Haacke, E.A.; van der Vegt, B.; Vissink, A.; Bootsma, H.; Kroese, F.G.M. Is the T Follicular Regulatory:Follicular Helper T Cell Ratio in Blood a Biomarker for Ectopic Lymphoid Structure Formation in Sjögren’s Syndrome? Comment on the Article by Fonseca et al. Arthritis Rheumatol. 2018, 70, 1354–1355. [Google Scholar] [CrossRef] [Green Version]
  114. Sage, P.T.; Paterson, A.M.; Lovitch, S.B.; Sharpe, A.H. The coinhibitory receptor CTLA-4 controls B cell responses by modulating T follicular helper, T follicular regulatory, and T regulatory cells. Immunity 2014, 41, 1026–1039. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Borges da Silva, H. Navigating in Deep Waters: How Tissue Damage and Inflammation Shape Effector and Memory CD8(+) T Cell Responses. ImmunoHorizons 2021, 5, 338–348. [Google Scholar] [CrossRef] [PubMed]
  116. Blokland, S.L.M.; van den Hoogen, L.L.; Leijten, E.F.A.; Hartgring, S.A.Y.; Fritsch, R.; Kruize, A.A.; van Roon, J.A.G.; Radstake, T. Increased expression of Fas on group 2 and 3 innate lymphoid cells is associated with an interferon signature in systemic lupus erythematosus and Sjögren’s syndrome. Rheumatology 2019, 58, 1740–1745. [Google Scholar] [CrossRef] [PubMed]
  117. Nocturne, G.; Mariette, X. B cells in the pathogenesis of primary Sjögren syndrome. Nat. Rev. Rheumatol. 2018, 14, 133–145. [Google Scholar] [CrossRef] [PubMed]
  118. Bohnhorst, J.; Bjørgan, M.B.; Thoen, J.E.; Natvig, J.B.; Thompson, K.M. Bm1-Bm5 classification of peripheral blood B cells reveals circulating germinal center founder cells in healthy individuals and disturbance in the B cell subpopulations in patients with primary Sjögren’s syndrome. J. Immunol. 2001, 167, 3610–3618. [Google Scholar] [CrossRef]
  119. Carbone, J.; Perez-Fernandez, R.; Muñoz, A.; Sabin, P.; Carreño, L.; Fernandez-Cruz, E. Combined therapy with rituximab plus cyclophosphamide/vincristine/prednisone for Sjogren’s syndrome-associated B-cell non-Hodgkin’s lymphoma. Clin. Rev. Allergy Immunol. 2008, 34, 80–84. [Google Scholar] [CrossRef]
  120. Xian, Z.; Fu, D.; Liu, S.; Yao, Y.; Gao, C. Association between B Cell Growth Factors and Primary Sjögren’s Syndrome-Related Autoantibodies in Patients with Non-Hodgkin’s Lymphoma. J. Immunol. Res. 2019, 2019, 7627384. [Google Scholar] [CrossRef] [Green Version]
  121. Hurdayal, R.; Nieuwenhuizen, N.E.; Khutlang, R.; Brombacher, F. Inflammatory Dendritic Cells, Regulated by IL-4 Receptor Alpha Signaling, Control Replication, and Dissemination of Leishmania major in Mice. Front. Cell. Infect. Microbiol. 2020, 9, 479. [Google Scholar] [CrossRef]
  122. Youinou, P.; Pers, J.O. Disturbance of cytokine networks in Sjögren’s syndrome. Arthritis Res. Ther. 2011, 13, 227. [Google Scholar] [CrossRef] [Green Version]
  123. Shen, L.; Suresh, L.; Lindemann, M.; Xuan, J.; Kowal, P.; Malyavantham, K.; Ambrus, J.L., Jr. Novel autoantibodies in Sjogren’s syndrome. Clin. Immunol. 2012, 145, 251–255. [Google Scholar] [CrossRef]
  124. Vivier, E.; Artis, D.; Colonna, M.; Diefenbach, A.; Di Santo, J.P.; Eberl, G.; Koyasu, S.; Locksley, R.M.; McKenzie, A.N.J.; Mebius, R.E.; et al. Innate Lymphoid Cells: 10 Years On. Cell 2018, 174, 1054–1066. [Google Scholar] [CrossRef] [Green Version]
  125. Panda, S.K.; Colonna, M. Innate Lymphoid Cells in Mucosal Immunity. Front. Immunol. 2019, 10, 861. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Ciccia, F.; Guggino, G.; Giardina, A.; Ferrante, A.; Carrubbi, F.; Giacomelli, R.; Triolo, G. The role of innate and lymphoid IL-22-producing cells in the immunopathology of primary Sjögren’s syndrome. Expert Rev. Clin. Immunol. 2014, 10, 533–541. [Google Scholar] [CrossRef] [PubMed]
  127. Wildenberg, M.E.; Welzen-Coppens, J.M.; van Helden-Meeuwsen, C.G.; Bootsma, H.; Vissink, A.; van Rooijen, N.; van de Merwe, J.P.; Drexhage, H.A.; Versnel, M.A. Increased frequency of CD16+ monocytes and the presence of activated dendritic cells in salivary glands in primary Sjögren syndrome. Ann. Rheum. Dis. 2009, 68, 420–426. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Kapsogeorgou, E.K.; Abu-Helu, R.F.; Moutsopoulos, H.M.; Manoussakis, M.N. Salivary gland epithelial cell exosomes: A source of autoantigenic ribonucleoproteins. Arthritis Rheum. 2005, 52, 1517–1521. [Google Scholar] [CrossRef] [PubMed]
  129. Maehara, T.; Moriyama, M.; Hayashida, J.N.; Tanaka, A.; Shinozaki, S.; Kubo, Y.; Matsumura, K.; Nakamura, S. Selective localization of T helper subsets in labial salivary glands from primary Sjogren’s syndrome patients. Clin. Exp. Immunol. 2012, 169, 89–99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Psianou, K.; Panagoulias, I.; Papanastasiou, A.D.; de Lastic, A.L.; Rodi, M.; Spantidea, P.I.; Degn, S.E.; Georgiou, P.; Mouzaki, A. Clinical and immunological parameters of Sjogren’s syndrome. Autoimmun. Rev. 2018, 17, 1053–1064. [Google Scholar] [CrossRef] [PubMed]
  131. Jin, J.O.; Kawai, T.; Cha, S.; Yu, Q. Interleukin-7 enhances the Th1 response to promote the development of Sjogren’s syndrome-like autoimmune exocrinopathy in mice. Arthritis Rheum. 2013, 65, 2132–2142. [Google Scholar] [CrossRef] [Green Version]
  132. Gong, Y.Z.; Nititham, J.; Taylor, K.; Miceli-Richard, C.; Sordet, C.; Wachsmann, D.; Bahram, S.; Georgel, P.; Criswell, L.A.; Sibilia, J.; et al. Differentiation of follicular helper T cells by salivary gland epithelial cells in primary Sjogren’s syndrome. J. Autoimmun. 2014, 51, 57–66. [Google Scholar] [CrossRef]
  133. Verstappen, G.M.; Meiners, P.M.; Corneth, O.B.J.; Visser, A.; Arends, S.; Abdulahad, W.H.; Hendriks, R.W.; Vissink, A.; Kroese, F.G.M.; Bootsma, H. Attenuation of Follicular Helper T Cell-Dependent B Cell Hyperactivity by Abatacept Treatment in Primary Sjogren’s Syndrome. Arthritis Rheumatol. 2017, 69, 1850–1861. [Google Scholar] [CrossRef]
  134. Thompson, N.; Isenberg, D.A.; Jury, E.C.; Ciurtin, C. Exploring BAFF: Its expression, receptors and contribution to the immunopathogenesis of Sjogren’s syndrome. Rheumatology 2016, 55, 1548–1555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Pestka, S.; Krause, C.D.; Walter, M.R. Interferons, interferon-like cytokines, and their receptors. Immunol. Rev. 2004, 202, 8–32. [Google Scholar] [CrossRef] [PubMed]
  136. Lazear, H.M.; Schoggins, J.W.; Diamond, M.S. Shared and Distinct Functions of Type I and Type III Interferons. Immunity 2019, 50, 907–923. [Google Scholar] [CrossRef]
  137. Lovgren, T.; Eloranta, M.L.; Kastner, B.; Wahren-Herlenius, M.; Alm, G.V.; Ronnblom, L. Induction of interferon-alpha by immune complexes or liposomes containing systemic lupus erythematosus autoantigen- and Sjogren’s syndrome autoantigen-associated RNA. Arthritis Rheum. 2006, 54, 1917–1927. [Google Scholar] [CrossRef] [PubMed]
  138. Ali, S.; Mann-Nuttel, R.; Schulze, A.; Richter, L.; Alferink, J.; Scheu, S. Sources of Type I Interferons in Infectious Immunity: Plasmacytoid Dendritic Cells Not Always in the Driver’s Seat. Front. Immunol. 2019, 10, 778. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Ittah, M.; Miceli-Richard, C.; Gottenberg, J.E.; Sellam, J.; Eid, P.; Lebon, P.; Pallier, C.; Lepajolec, C.; Mariette, X. Viruses induce high expression of BAFF by salivary gland epithelial cells through TLR- and type-I IFN-dependent and -independent pathways. Eur. J. Immunol. 2008, 38, 1058–1064. [Google Scholar] [CrossRef] [PubMed]
  140. Kyriakidis, N.C.; Kapsogeorgou, E.K.; Gourzi, V.C.; Konsta, O.D.; Baltatzis, G.E.; Tzioufas, A.G. Toll-like receptor 3 stimulation promotes Ro52/TRIM21 synthesis and nuclear redistribution in salivary gland epithelial cells, partially via type I interferon pathway. Clin. Exp. Immunol. 2014, 178, 548–560. [Google Scholar] [CrossRef] [Green Version]
  141. Schreiber, G. The molecular basis for differential type I interferon signaling. J. Biol. Chem. 2017, 292, 7285–7294. [Google Scholar] [CrossRef] [Green Version]
  142. Negishi, H.; Taniguchi, T.; Yanai, H. The Interferon (IFN) Class of Cytokines and the IFN Regulatory Factor (IRF) Transcription Factor Family. Cold Spring Harb. Perspect. Biol. 2018, 10, a028423. [Google Scholar] [CrossRef]
  143. Li, S.F.; Gong, M.J.; Zhao, F.R.; Shao, J.J.; Xie, Y.L.; Zhang, Y.G.; Chang, H.Y. Type I Interferons: Distinct Biological Activities and Current Applications for Viral Infection. Cell Physiol. Biochem. 2018, 51, 2377–2396. [Google Scholar] [CrossRef]
  144. Le Bon, A.; Schiavoni, G.; D’Agostino, G.; Gresser, I.; Belardelli, F.; Tough, D.F. Type i interferons potently enhance humoral immunity and can promote isotype switching by stimulating dendritic cells in vivo. Immunity 2001, 14, 461–470. [Google Scholar] [CrossRef] [Green Version]
  145. Hervas-Stubbs, S.; Perez-Gracia, J.L.; Rouzaut, A.; Sanmamed, M.F.; Le Bon, A.; Melero, I. Direct effects of type I interferons on cells of the immune system. Clin. Cancer Res. 2011, 17, 2619–2627. [Google Scholar] [CrossRef] [Green Version]
  146. Ittah, M.; Miceli-Richard, C.; Eric Gottenberg, J.; Lavie, F.; Lazure, T.; Ba, N.; Sellam, J.; Lepajolec, C.; Mariette, X. B cell-activating factor of the tumor necrosis factor family (BAFF) is expressed under stimulation by interferon in salivary gland epithelial cells in primary Sjögren’s syndrome. Arthritis Res. Ther. 2006, 8, R51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Cha, S.; van Blockland, S.C.; Versnel, M.A.; Homo-Delarche, F.; Nagashima, H.; Brayer, J.; Peck, A.B.; Humphreys-Beher, M.G. Abnormal organogenesis in salivary gland development may initiate adult onset of autoimmune exocrinopathy. Exp. Clin. Immunogenet. 2001, 18, 143–160. [Google Scholar] [CrossRef]
  148. Szczerba, B.M.; Rybakowska, P.D.; Dey, P.; Payerhin, K.M.; Peck, A.B.; Bagavant, H.; Deshmukh, U.S. Type I interferon receptor deficiency prevents murine Sjogren’s syndrome. J. Dent. Res. 2013, 92, 444–449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Hall, J.C.; Casciola-Rosen, L.; Berger, A.E.; Kapsogeorgou, E.K.; Cheadle, C.; Tzioufas, A.G.; Baer, A.N.; Rosen, A. Precise probes of type II interferon activity define the origin of interferon signatures in target tissues in rheumatic diseases. Proc. Natl. Acad. Sci. USA 2012, 109, 17609–17614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Peck, A.B.; Nguyen, C.Q. What can Sjogren’s syndrome-like disease in mice contribute to human Sjogren’s syndrome? Clin. Immunol. 2017, 182, 14–23. [Google Scholar] [CrossRef] [Green Version]
  151. Pontarini, E.; Murray-Brown, W.J.; Croia, C.; Lucchesi, D.; Conway, J.; Rivellese, F.; Fossati-Jimack, L.; Astorri, E.; Prediletto, E.; Corsiero, E.; et al. Unique expansion of IL-21+ Tfh and Tph cells under control of ICOS identifies Sjogren’s syndrome with ectopic germinal centres and MALT lymphoma. Ann. Rheum. Dis. 2020, 79, 1588–1599. [Google Scholar] [CrossRef]
  152. Reksten, T.R.; Lessard, C.J.; Sivils, K.L. Genetics in Sjogren Syndrome. Rheum. Dis. Clin. N. Am. 2016, 42, 435–447. [Google Scholar] [CrossRef]
  153. Wang, F.; Zhang, S.; Jeon, R.; Vuckovic, I.; Jiang, X.; Lerman, A.; Folmes, C.D.; Dzeja, P.D.; Herrmann, J. Interferon Gamma Induces Reversible Metabolic Reprogramming of M1 Macrophages to Sustain Cell Viability and Pro-Inflammatory Activity. EBioMedicine 2018, 30, 303–316. [Google Scholar] [CrossRef] [Green Version]
  154. Manoussakis, M.N.; Dimitriou, I.D.; Kapsogeorgou, E.K.; Xanthou, G.; Paikos, S.; Polihronis, M.; Moutsopoulos, H.M. Expression of B7 costimulatory molecules by salivary gland epithelial cells in patients with Sjogren’s syndrome. Arthritis Rheum. 1999, 42, 229–239. [Google Scholar] [CrossRef]
  155. Dimitriou, I.D.; Kapsogeorgou, E.K.; Moutsopoulos, H.M.; Manoussakis, M.N. CD40 on salivary gland epithelial cells: High constitutive expression by cultured cells from Sjogren’s syndrome patients indicating their intrinsic activation. Clin. Exp. Immunol. 2002, 127, 386–392. [Google Scholar] [CrossRef] [PubMed]
  156. Tsunawaki, S.; Nakamura, S.; Ohyama, Y.; Sasaki, M.; Ikebe-Hiroki, A.; Hiraki, A.; Kadena, T.; Kawamura, E.; Kumamaru, W.; Shinohara, M.; et al. Possible function of salivary gland epithelial cells as nonprofessional antigen-presenting cells in the development of Sjogren’s syndrome. J. Rheumatol. 2002, 29, 1884–1896. [Google Scholar] [PubMed]
  157. Abu-Helu, R.F.; Dimitriou, I.D.; Kapsogeorgou, E.K.; Moutsopoulos, H.M.; Manoussakis, M.N. Induction of Salivary Gland Epithelial Cell Injury in Sjogren’s Syndrome: In Vitro Assessment of T Cell-derived Cytokines and Fas Protein Expression. J. Autoimmun. 2001, 17, 141–153. [Google Scholar] [CrossRef] [PubMed]
  158. Ewert, P.; Aguilera, S.; Alliende, C.; Kwon, Y.J.; Albornoz, A.; Molina, C.; Urzúa, U.; Quest, A.; Olea, N.; Rheum, P.P.J.A. Disruption of tight junction structure in salivary glands from Sjgren’s syndrome patients is linked to proinflammatory cytokine exposure. Arthritis Rheum. 2010, 62, 1280–1289. [Google Scholar] [CrossRef]
  159. Apostolou, E.; Kapsogeorgou, E.K.; Konsta, O.D.; Giotakis, I.; Saridaki, M.I.; Andreakos, E.; Tzioufas, A.G. Expression of type III interferons (IFNlambdas) and their receptor in Sjogren’s syndrome. Clin. Exp. Immunol. 2016, 186, 304–312. [Google Scholar] [CrossRef] [Green Version]
  160. Roescher, N.; Tak, P.P.; Illei, G.G. Cytokines in Sjogren’s syndrome. Oral Dis. 2009, 15, 519–526. [Google Scholar] [CrossRef] [Green Version]
  161. Katsifis, G.E.; Rekka, S.; Moutsopoulos, N.M.; Pillemer, S.; Wahl, S.M. Systemic and local interleukin-17 and linked cytokines associated with Sjogren’s syndrome immunopathogenesis. Am. J. Pathol. 2009, 175, 1167–1177. [Google Scholar] [CrossRef] [Green Version]
  162. Fei, Y.; Zhang, W.; Lin, D.; Wu, C.; Li, M.; Zhao, Y.; Zeng, X.; Zhang, F. Clinical parameter and Th17 related to lymphocytes infiltrating degree of labial salivary gland in primary Sjogren’s syndrome. Clin. Rheumatol. 2014, 33, 523–529. [Google Scholar] [CrossRef]
  163. Sakai, A.; Sugawara, Y.; Kuroishi, T.; Sasano, T.; Sugawara, S. Identification of IL-18 and Th17 cells in salivary glands of patients with Sjogren’s syndrome, and amplification of IL-17-mediated secretion of inflammatory cytokines from salivary gland cells by IL-18. J. Immunol. 2008, 181, 2898–2906. [Google Scholar] [CrossRef]
  164. Iwakura, Y.; Ishigame, H.; Saijo, S.; Nakae, S. Functional specialization of interleukin-17 family members. Immunity 2011, 34, 149–162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Zhang, L.W.; Cong, X.; Zhang, Y.; Wei, T.; Wu, L.L. Interleukin-17 Impairs Salivary Tight Junction Integrity in Sjgren’s Syndrome. J. Dent. Res. 2016, 95, 784. [Google Scholar] [CrossRef] [PubMed]
  166. Fogli, L.K.; Sundrud, M.S.; Goel, S.; Bajwa, S.; Jensen, K.; Derudder, E.; Sun, A.; Coffre, M.; Uyttenhove, C.; Van Snick, J.; et al. T Cell-Derived IL-17 Mediates Epithelial Changes in the Airway and Drives Pulmonary Neutrophilia. J. Immunol. 2013, 191, 3100–3111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Pérez, P.; Kwon, Y.J.; Alliende, C.; Leyton, L.; Aguilera, S.; Molina, C.; Labra, C.; Julio, M.; Leyton, C.; Arthritis, M.-J.G.J.; et al. Increased acinar damage of salivary glands of patients with Sjögren’s syndrome is paralleled by simultaneous imbalance of matrix metalloproteinase 3/tissue inhibitor of metalloproteinases 1 and matrix metalloproteinase 9/tissue inhibitor of metalloproteinases 1 ratios. Arthritis Rheum. 2005, 59, 2751–2760. [Google Scholar]
  168. Nguyen, C.Q.; Yin, H.; Lee, B.H.; Carcamo, W.C.; Chiorini, J.A.; Peck, A.B. Pathogenic effect of interleukin-17A in induction of Sjögren’s syndrome-like disease using adenovirus-mediated gene transfer. Arthritis Res. Ther. 2010, 12, R220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Liang, S.C.; Tan, X.Y.; Luxenberg, D.P.; Karim, R.; Dunussi-Joannopoulos, K.; Collins, M.; Fouser, L.A. Interleukin (IL)-22 and IL-17 are coexpressed by Th17 cells and cooperatively enhance expression of antimicrobial peptides. J. Exp. Med. 2006, 203, 2271–2279. [Google Scholar] [CrossRef]
  170. Ciccia, F.; Guggino, G.; Rizzo, A.; Ferrante, A.; Raimondo, S.; Giardina, A.; Dieli, F.; Campisi, G.; Alessandro, R.; Triolo, G. Potential involvement of IL-22 and IL-22-producing cells in the inflamed salivary glands of patients with Sjogren’s syndrome. Ann. Rheum. Dis. 2012, 71, 295–301. [Google Scholar] [CrossRef]
  171. Barone, F.; Nayar, S.; Campos, J.; Cloake, T.; Withers, D.R.; Toellner, K.M.; Zhang, Y.; Fouser, L.; Fisher, B.; Bowman, S.; et al. IL-22 regulates lymphoid chemokine production and assembly of tertiary lymphoid organs. Proc. Natl. Acad. Sci. USA 2015, 112, 11024–11029. [Google Scholar] [CrossRef] [Green Version]
  172. Kang, K.Y.; Kim, H.O.; Kwok, S.K.; Ju, J.H.; Park, K.S.; Sun, D.I.; Jhun, J.Y.; Oh, H.J.; Park, S.H.; Kim, H.Y. Impact of interleukin-21 in the pathogenesis of primary Sjögren’s syndrome: Increased serum levels of interleukin-21 and its expression in the labial salivary glands. Arthritis Res. Ther. 2011, 13, R179. [Google Scholar] [CrossRef] [Green Version]
  173. Kwok, S.K.; Lee, J.; Yu, D.; Kang, K.Y.; Cho, M.L.; Kim, H.R.; Ju, J.H.; Lee, S.H.; Park, S.H.; Kim, H.Y. A pathogenetic role for IL-21 in primary Sjogren syndrome. Nat. Rev. Rheumatol. 2015, 11, 368–374. [Google Scholar] [CrossRef]
  174. McGuire, H.M.; Vogelzang, A.; Ma, C.S.; Hughes, W.E.; Silveira, P.A.; Tangye, S.G.; Christ, D.; Fulcher, D.; Falcone, M.; King, C. A subset of interleukin-21+ chemokine receptor CCR9+ T helper cells target accessory organs of the digestive system in autoimmunity. Immunity 2011, 34, 602–615. [Google Scholar] [CrossRef] [Green Version]
  175. Wei, L.; Laurence, A.; Elias, K.M.; O’Shea, J.J. IL-21 is produced by Th17 cells and drives IL-17 production in a STAT3-dependent manner. J. Biol. Chem. 2007, 282, 34605–34610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Zeng, R.; Spolski, R.; Finkelstein, S.E.; Oh, S.; Kovanen, P.E.; Hinrichs, C.S.; Pise-Masison, C.A.; Radonovich, M.F.; Brady, J.N.; Restifo, N.P.; et al. Synergy of IL-21 and IL-15 in regulating CD8+ T cell expansion and function. J. Exp. Med. 2005, 201, 139–148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Liu, H.; Liu, G.; Gong, L.; Zhang, Y.; Jiang, G. Local suppression of IL-21 in submandibular glands retards the development of Sjögren’s syndrome in non-obese diabetic mice. J. Oral Pathol. Med. 2012, 41, 728–735. [Google Scholar] [CrossRef] [PubMed]
  178. Nguyen, C.Q.; Gao, J.-H.; Kim, H.; Saban, D.; Immunology, R.J. IL-4-STAT6 Signal Transduction-Dependent Induction of the Clinical Phase of Sjogren’s Syndrome-Like Disease of the Nonobese Diabetic Mouse. J. Immunol. 2007, 179, 382–390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Pandey, S.; Mourcin, F.; Marchand, T.; Nayar, S.; Guirriec, M.; Pangault, C.; Monvoisin, C.; Ame-Thomas, P.; Guilloton, F.; Dulong, J.; et al. IL-4/CXCL12 loop is a key regulator of lymphoid stroma function in follicular lymphoma. Blood 2017, 129, 2507–2518. [Google Scholar] [CrossRef] [Green Version]
  180. Barone, F.; Bombardieri, M.; Rosado, M.M.; Morgan, P.R.; Challacombe, S.J.; De Vita, S.; Carsetti, R.; Spencer, J.; Valesini, G.; Pitzalis, C. CXCL13, CCL21, and CXCL12 expression in salivary glands of patients with Sjogren’s syndrome and MALT lymphoma: Association with reactive and malignant areas of lymphoid organization. J. Immunol. 2008, 180, 5130–5140. [Google Scholar] [CrossRef]
  181. Wurster, A.L.; Rodgers, V.L.; White, M.F.; Rothstein, T.L.; Grusby, M.J. Interleukin-4-mediated protection of primary B cells from apoptosis through Stat6-dependent up-regulation of Bcl-xL. J. Biol. Chem. 2002, 277, 27169–27175. [Google Scholar] [CrossRef] [Green Version]
  182. Brayer, J.B.; Cha, S.; Nagashima, H.; Yasunari, U.; Lindberg, A.; Diggs, S.; Martinez, J.; Goa, J.; Humphreys-Beher, M.G.; Peck, A.B. IL-4-dependent effector phase in autoimmune exocrinopathy as defined by the NOD.IL-4-gene knockout mouse model of Sjögren’s syndrome. Scand. J. Immunol. 2001, 54, 133–140. [Google Scholar] [CrossRef]
  183. Xu, T. Leptin/OB-R pathway promotes IL-4 secretion from B lymphocytes and induces salivary gland epithelial cell apoptosis in Sjögren’s syndrome. In Proceedings of the 17th International Congress of Immunology, Beijing, China, 19–23 October 2019. [Google Scholar]
  184. Vanamee, E.S.; Faustman, D.L. Structural principles of tumor necrosis factor superfamily signaling. Sci. Signal. 2018, 11, aao4910. [Google Scholar] [CrossRef] [Green Version]
  185. Dostert, C.; Grusdat, M.; Letellier, E.; Brenner, D. The TNF Family of Ligands and Receptors: Communication Modules in the Immune System and Beyond. Physiol. Rev. 2019, 99, 115–160. [Google Scholar] [CrossRef]
  186. Holbrook, J.; Lara-Reyna, S.; Jarosz-Griffiths, H.; McDermott, M. Tumour necrosis factor signalling in health and disease. F1000Res 2019, 8, 111. [Google Scholar] [CrossRef] [PubMed]
  187. Morimoto, S.; Nakano, S.; Watanabe, T.; Tamayama, Y.; Mitsuo, A.; Nakiri, Y.; Suzuki, J.; Nozawa, K.; Amano, H.; Tokano, Y.; et al. Expression of B-cell activating factor of the tumour necrosis factor family (BAFF) in T cells in active systemic lupus erythematosus: The role of BAFF in T cell-dependent B cell pathogenic autoantibody production. Rheumatology 2007, 46, 1083–1086. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Szodoray, P.; Alex, P.; Jonsson, M.V.; Knowlton, N.; Dozmorov, I.; Nakken, B.; Delaleu, N.; Jonsson, R.; Centola, M. Distinct profiles of Sjogren’s syndrome patients with ectopic salivary gland germinal centers revealed by serum cytokines and BAFF. Clin. Immunol. 2005, 117, 168–176. [Google Scholar] [CrossRef]
  189. Kang, E.H.; Lee, Y.J.; Hyon, J.Y.; Yun, P.Y.; Song, Y.W. Salivary cytokine profiles in primary Sjogren’s syndrome differ from those in non-Sjogren sicca in terms of TNF-alpha levels and Th-1/Th-2 ratios. Clin. Exp. Rheumatol. 2011, 29, 970–976. [Google Scholar] [PubMed]
  190. Fox, R.I.; Kang, H.I.; Ando, D.; Abrams, J.; Pisa, E. Cytokine mRNA expression in salivary gland biopsies of Sjögren’s syndrome. J. Immunol. 1994, 152, 5532–5539. [Google Scholar] [PubMed]
  191. Chatzantoni, K.; Mouzaki, A. Anti-TNF-alpha antibody therapies in autoimmune diseases. Curr. Top. Med. Chem. 2006, 6, 1707–1714. [Google Scholar] [CrossRef]
  192. Jin, J.O.; Yu, Q. T Cell-Associated Cytokines in the Pathogenesis of Sjogren’s Syndrome. J. Clin. Cell. Immunol. 2013, 9, 1–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Kamachi, M.; Kawakami, A.; Yamasaki, S.; Hida, A.; Nakashima, T.; Nakamura, H.; Ida, H.; Furuyama, M.; Nakashima, K.; Shibatomi, K.; et al. Regulation of apoptotic cell death by cytokines in a human salivary gland cell line: Distinct and synergistic mechanisms in apoptosis induced by tumor necrosis factor alpha and interferon gamma. J. Lab. Clin. Med. 2002, 139, 13–19. [Google Scholar] [CrossRef]
  194. Bossen, C.; Schneider, P. BAFF, APRIL and their receptors: Structure, function and signaling. Semin. Immunol. 2006, 18, 263–275. [Google Scholar] [CrossRef] [Green Version]
  195. Mackay, F.; Browning, J.L. BAFF: A fundamental survival factor for B cells. Nat. Rev. Immunol. 2002, 2, 465–475. [Google Scholar] [CrossRef] [PubMed]
  196. Mackay, F.; Schneider, P. Cracking the BAFF code. Nat. Rev. Immunol. 2009, 9, 491–502. [Google Scholar] [CrossRef] [Green Version]
  197. Litinskiy, M.B.; Nardelli, B.; Hilbert, D.M.; He, B.; Schaffer, A.; Casali, P.; Cerutti, A. DCs induce CD40-independent immunoglobulin class switching through BLyS and APRIL. Nat. Immunol. 2002, 3, 822–829. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  198. Groom, J.; Kalled, S.L.; Cutler, A.H.; Olson, C.; Woodcock, S.A.; Schneider, P.; Tschopp, J.; Cachero, T.G.; Batten, M.; Wheway, J.; et al. Association of BAFF/BLyS overexpression and altered B cell differentiation with Sjogren’s syndrome. J. Clin. Investig. 2002, 109, 59–68. [Google Scholar] [CrossRef] [PubMed]
  199. Miceli-Richard, C.; Wang-Renault, S.F.; Boudaoud, S.; Busato, F.; Lallemand, C.; Bethune, K.; Belkhir, R.; Nocturne, G.; Mariette, X.; Tost, J. Overlap between differentially methylated DNA regions in blood B lymphocytes and genetic at-risk loci in primary Sjögren’s syndrome. Ann. Rheum. Dis. 2016, 75, 933–940. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  200. Imgenberg-Kreuz, J.; Sandling, J.K.; Almlöf, J.C.; Nordlund, J.; Signér, L.; Norheim, K.B.; Omdal, R.; Rönnblom, L.; Eloranta, M.L.; Syvänen, A.C.; et al. Genome-wide DNA methylation analysis in multiple tissues in primary Sjögren’s syndrome reveals regulatory effects at interferon-induced genes. Ann. Rheum. Dis. 2016, 75, 2029–2036. [Google Scholar] [CrossRef] [Green Version]
  201. Wang-Renault, S.F.; Boudaoud, S.; Nocturne, G.; Roche, E.; Sigrist, N.; Daviaud, C.; Bugge Tinggaard, A.; Renault, V.; Deleuze, J.F.; Mariette, X.; et al. Deregulation of microRNA expression in purified T and B lymphocytes from patients with primary Sjögren’s syndrome. Ann. Rheum. Dis. 2018, 77, 133–140. [Google Scholar] [CrossRef] [Green Version]
  202. Alevizos, I.; Alexander, S.; Turner, R.J.; Illei, G.G. MicroRNA expression profiles as biomarkers of minor salivary gland inflammation and dysfunction in Sjögren’s syndrome. Arthritis Rheum. 2011, 63, 535–544. [Google Scholar] [CrossRef] [Green Version]
  203. Marketos, N.; Cinoku, I.; Rapti, A.; Mavragani, C.P. Type I interferon signature in Sjögren’s syndrome: Pathophysiological and clinical implications. Clin. Exp. Rheumatol. 2019, 37 (Suppl. S118), 185–191. [Google Scholar]
  204. Toker, A.; Engelbert, D.; Garg, G.; Polansky, J.K.; Floess, S.; Miyao, T.; Baron, U.; Düber, S.; Geffers, R.; Giehr, P.; et al. Active demethylation of the Foxp3 locus leads to the generation of stable regulatory T cells within the thymus. J. Immunol. 2013, 190, 3180–3188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Yu, X.; Liang, G.; Yin, H.; Ngalamika, O.; Li, F.; Zhao, M.; Lu, Q. DNA hypermethylation leads to lower FOXP3 expression in CD4+ T cells of patients with primary Sjögren’s syndrome. Clin. Immunol. 2013, 148, 254–257. [Google Scholar] [CrossRef] [PubMed]
  206. Taylor, K.E.; Wong, Q.; Levine, D.M.; McHugh, C.; Laurie, C.; Doheny, K.; Lam, M.Y.; Baer, A.N.; Challacombe, S.; Lanfranchi, H.; et al. Genome-Wide Association Analysis Reveals Genetic Heterogeneity of Sjögren’s Syndrome According to Ancestry. Arthritis Rheumatol. 2017, 69, 1294–1305. [Google Scholar] [CrossRef] [Green Version]
  207. Nordmark, G.; Kristjansdottir, G.; Theander, E.; Eriksson, P.; Brun, J.G.; Wang, C.; Padyukov, L.; Truedsson, L.; Alm, G.; Eloranta, M.L.; et al. Additive effects of the major risk alleles of IRF5 and STAT4 in primary Sjögren’s syndrome. Genes Immun. 2009, 10, 68–76. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  208. Reksten, T.R.; Johnsen, S.J.; Jonsson, M.V.; Omdal, R.; Brun, J.G.; Theander, E.; Eriksson, P.; Wahren-Herlenius, M.; Jonsson, R.; Nordmark, G. Genetic associations to germinal centre formation in primary Sjogren’s syndrome. Ann. Rheum. Dis. 2014, 73, 1253–1258. [Google Scholar] [CrossRef]
  209. Lindén, M.; Ramírez Sepúlveda, J.I.; James, T.; Thorlacius, G.E.; Brauner, S.; Gómez-Cabrero, D.; Olsson, T.; Kockum, I.; Wahren-Herlenius, M. Sex influences eQTL effects of SLE and Sjögren’s syndrome-associated genetic polymorphisms. Biol. Sex Differ. 2017, 8, 34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  210. Cobb, B.L.; Fei, Y.; Jonsson, R.; Bolstad, A.I.; Brun, J.G.; Rischmueller, M.; Lester, S.E.; Witte, T.; Illei, G.; Brennan, M.; et al. Genetic association between methyl-CpG binding protein 2 (MECP2) and primary Sjogren’s syndrome. Ann. Rheum. Dis. 2010, 69, 1731–1732. [Google Scholar] [CrossRef] [Green Version]
  211. Webb, R.; Wren, J.D.; Jeffries, M.; Kelly, J.A.; Kaufman, K.M.; Tang, Y.; Frank, M.B.; Merrill, J.; Kimberly, R.P.; Edberg, J.C.; et al. Variants within MECP2, a key transcription regulator, are associated with increased susceptibility to lupus and differential gene expression in patients with systemic lupus erythematosus. Arthritis Rheum. 2009, 60, 1076–1084. [Google Scholar] [CrossRef] [Green Version]
  212. Zhao, J.; Wei, K.; Chang, C.; Xu, L.; Jiang, P.; Guo, S.; Schrodi, S.J.; He, D. DNA Methylation of T Lymphocytes as a Therapeutic Target: Implications for Rheumatoid Arthritis Etiology. Front. Immunol. 2022, 13, 863703. [Google Scholar] [CrossRef]
Figure 1. Epigenetic regulations in immune responses. Epigenetic modifications, such as DNA methylation (bottom left), histone modification (bottom right) and non−coding RNA expression (top), play critical roles in immune response. Abnormal epigenetic changes could lead to the dysregulation of immune responses.
Figure 1. Epigenetic regulations in immune responses. Epigenetic modifications, such as DNA methylation (bottom left), histone modification (bottom right) and non−coding RNA expression (top), play critical roles in immune response. Abnormal epigenetic changes could lead to the dysregulation of immune responses.
Cells 11 01767 g001
Figure 2. Graphics of immunocyte infiltration during the pathogenesis of SjS. At an early stage, the majority of infiltrated immunocytes in SGs of SjS patients are T cells (left), while, at a later stage, the prominent infiltrated immunocytes are B cells (right).
Figure 2. Graphics of immunocyte infiltration during the pathogenesis of SjS. At an early stage, the majority of infiltrated immunocytes in SGs of SjS patients are T cells (left), while, at a later stage, the prominent infiltrated immunocytes are B cells (right).
Cells 11 01767 g002
Figure 3. Mutual interactions between T cells, B cells and SGECs result in the immunopathogenesis of SjS. Infiltrated T and B cells in the SGs of SjS patients produce pro-inflammatory cytokines, such as IFNs and interleukins, acting on them mutually to further aggravate the autoimmune response. In addition, these pro-inflammatory cytokines also act on SGECs, inducing the destructions of glandular tissues.
Figure 3. Mutual interactions between T cells, B cells and SGECs result in the immunopathogenesis of SjS. Infiltrated T and B cells in the SGs of SjS patients produce pro-inflammatory cytokines, such as IFNs and interleukins, acting on them mutually to further aggravate the autoimmune response. In addition, these pro-inflammatory cytokines also act on SGECs, inducing the destructions of glandular tissues.
Cells 11 01767 g003
Figure 4. Epigenetic modification changes in immunocytes during SjS. IFN-related genes exhibit hypomethylation in the T and B cells of SjS patients. In addition, multiple miRNAs, correlated with IFN-related genes, are also altered in T and B cells, whereas the FOXP3 locus in Treg is hypermethylated.
Figure 4. Epigenetic modification changes in immunocytes during SjS. IFN-related genes exhibit hypomethylation in the T and B cells of SjS patients. In addition, multiple miRNAs, correlated with IFN-related genes, are also altered in T and B cells, whereas the FOXP3 locus in Treg is hypermethylated.
Cells 11 01767 g004
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Li, P.; Han, M.; Zhao, X.; Ren, G.; Mei, S.; Zhong, C. Abnormal Epigenetic Regulations in the Immunocytes of Sjögren’s Syndrome Patients and Therapeutic Potentials. Cells 2022, 11, 1767. https://doi.org/10.3390/cells11111767

AMA Style

Li P, Han M, Zhao X, Ren G, Mei S, Zhong C. Abnormal Epigenetic Regulations in the Immunocytes of Sjögren’s Syndrome Patients and Therapeutic Potentials. Cells. 2022; 11(11):1767. https://doi.org/10.3390/cells11111767

Chicago/Turabian Style

Li, Peng, Mengwei Han, Xingyu Zhao, Guanqun Ren, Si Mei, and Chao Zhong. 2022. "Abnormal Epigenetic Regulations in the Immunocytes of Sjögren’s Syndrome Patients and Therapeutic Potentials" Cells 11, no. 11: 1767. https://doi.org/10.3390/cells11111767

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop