Next Article in Journal
α-Pinene Enhances the Anticancer Activity of Natural Killer Cells via ERK/AKT Pathway
Next Article in Special Issue
Analysis of PI3K Pathway Associated Molecules Reveals Dysregulated Innate and Adaptive Functions of B Cells in Early Diffuse Cutaneous Systemic Sclerosis
Previous Article in Journal
Inhibition of InsP3R with Xestospongin B Reduces Mitochondrial Respiration and Induces Selective Cell Death in T Cell Acute Lymphoblastic Leukemia Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Involvement of Innate and Adaptive Immunity in the Initiation and Perpetuation of Sjögren’s Syndrome

by
Clara Chivasso
1,†,
Julie Sarrand
2,†,
Jason Perret
1,
Christine Delporte
1 and
Muhammad Shahnawaz Soyfoo
2,*
1
Laboratory of Pathophysiological and Nutritional Biochemistry, Université Libre de Bruxelles, 1070 Brussels, Belgium
2
Department of Rheumatology, Hôpital Erasme, Université Libre de Bruxelles, 1070 Brussels, Belgium
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2021, 22(2), 658; https://doi.org/10.3390/ijms22020658
Submission received: 16 December 2020 / Revised: 1 January 2021 / Accepted: 6 January 2021 / Published: 11 January 2021
(This article belongs to the Special Issue Lymphocyte Signalling and Function in Systemic Autoimmune Diseases)

Abstract

:
Sjogren’s syndrome (SS) is a chronic autoimmune disease characterized by the infiltration of exocrine glands including salivary and lachrymal glands responsible for the classical dry eyes and mouth symptoms (sicca syndrome). The spectrum of disease manifestations stretches beyond the classical sicca syndrome with systemic manifestations including arthritis, interstitial lung involvement, and neurological involvement. The pathophysiology underlying SS is not well deciphered, but several converging lines of evidence have supported the conjuncture of different factors interplaying together to foster the initiation and perpetuation of the disease. The innate and adaptive immune system play a cardinal role in this process. In this review, we discuss the inherent parts played by both the innate and adaptive immune system in the pathogenesis of SS.

1. Sjögren’s Syndrome

Sjögren’s syndrome (SS) is one of the most common autoimmune rheumatic diseases. SS is characterized by the immune-mediated destruction of exocrine glands, including lachrymal and salivary glands (SGs). Two types of SS have been defined: Primary SS (pSS), which occurs in the absence of other autoimmune diseases, and secondary SS (sSS), which is associated with other autoimmune disorders such as systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), and scleroderma [1,2]. SS is characterized by a high sex preponderance with a ratio of nine female for one male. This sexual imbalance suggests an involvement of estrogens and androgens in the development of the pathology [3,4] that could account for an incidence increase of pSS during the post-menopausal stage, at the age of 40–60 years old [5]. In general, the diagnosis is based on the combination of several oral and ocular sicca symptoms, the presence of the autoimmune manifestations such the production of autoantibodies anti-Ro/SSA, the labial biopsy showing a focal lymphocytic infiltration (focus score ≥ 1 per 4 mm2) [6]. The pathophysiology of SS is very complex, multifactorial, and consecutive to several genetic, hormonal, environmental, and immunological risk factors. Due to its complexity, the clinical course of the pathology can be divided in several phases: An initiation phase consecutive to endogenous and exogenous factors, a dysregulation of salivary glands epithelial cells (SGECs), and an immune system activation and chronicity of inflammation induced by B cells hyperactivity [7]. The combination of all these events culminates in the destruction of the salivary gland architecture, and development of keratoconjunctivitis sicca and xerostomia. Each phase plays a significant role in the disease. The transition from the innate immune system to the adaptive system responses and the variety of cell types involved could explain the difficulties in developing an efficient therapeutic strategy for pSS. This review highlights the role of immune cells and the crosstalk between the innate and adaptive immunity in pSS pathogenesis.

2. Innate Immune Cells Involved in Sjögren’s Syndrome

A growing body of evidence indicates that innate immunity plays a crucial role in the pathogenesis of pSS, especially in the initiation and progression towards autoimmunity [8]. We will discuss the role of each cell type implicated in this process often called autoimmune epithelitis.

2.1. Dendritic Cells

Dendritic cells (DCs) are professional antigen presenting cells. They act as sentinels capturing and processing antigens, migrating in T cell areas to initiate immunity and differentiating in response to a variety of stimuli such as Toll-like receptor (TLR) ligands, cytokines, innate lymphocytes, and immune complexes [9]. DCs play a key role in pSS as they display an aberrant phenotype causing them to accumulate in SGs [10,11,12]. Saliva from pSS patients is characterized by an upregulation of C-C chemokine receptor type 5 (CCR5) and CCR5 ligands such as CC chemokine ligand type 3 (CCL3) and type 4 (CCL4) that play an important role for the effective migration of DCs to inflamed tissues. In addition, lower numbers of blood DCs in patients with pSS may be consecutive to the aberrant regulation of apoptosis [13].
Plasmacytoid DCs (pDCs) are a specific subset of DCs that can be activated by self-antigens through TLR-7 and TRL-9 [14,15] and to a lesser extent TLR-2, TRL-4, and TRL-9 [16], leading to the production of type I interferon (IFN). Type I IFN acts through autocrine and paracrine circuits sustaining a continuous reinforcing inflammatory loop. It also induces the production of the B cell activating factor (BAFF) by monocyte circulating cells and DCs contributing to the activation and differentiation of B cells into plasma cells secreting antibodies [16,17].
Follicular DCs (fDCs) originate from fibroblast precursor cells and play an essential part in the structure of ectopic germinal centers. FDCs promote B cells survival and proliferation in the long run by retaining on their surface immune-complexes (IC), formed by antigen-antibody-complement. Contrary to other DCs, fDCs do not display phagocytic activity and lack lysosomes and lysozyme [18,19].

2.2. Macrophages

Macrophages are the main tissue resident leucocytes and are characterized by pleomorphic phenotypes. According to their microenvironment, they can display pro-inflammatory or anti-inflammatory activities, immunogenic or tolerogenic activities, and tissue destructive or tissue regenerative activities [20,21].
In SGs specimens from patients with pSS, macrophages tend to appear early and their number is positively correlated with the biopsy focus score [22]. Macrophages are activated by interferon gamma (IFN-γ) and interleukin (IL)-17 secreted by type 1 T helper cells (Th1) and type 17 T helper cells (Th17), respectively [23]. Activated macrophages produce inflammatory cytokines such as IL-1, tumor necrosis factor alpha (TNFα), IL-18, and metalloproteases (MMPs) leading to epithelial cell damage [24,25]. Activated macrophages can also act as antigenic peptide presenting cells through their major histocompatibility complex class II (MHC-II) and interact with antigen-specific CD4+ T cells [23]. The latter, once activated, evolves in autoreactive clones that may perpetuate the activation of macrophages, themselves sustaining a pro-inflammatory auto-maintained loop [25].
Manoussakis et al., studied MSG biopsies from pSS patients and demonstrated increased infiltration by macrophages together with a marked expression of IL-18 by infiltrating macrophages. Moreover, IL-18 levels correlated with lymphoma risk factors such as persistent C4 hypocomplementemia and SG enlargement [26].
Beside SGs, this process may affect other epithelia such as the eye epithelium leading to the development of squamous metaplasia which represents the end stage of ocular involvement in pSS patients [27,28].

2.3. Mast Cells

Mast cells are immune cells mainly found in connective tissues. Their role in allergy and anaphylaxis is well established. However, a great deal of evidence underlines their possible involvement in tissue healing, angiogenesis, and autoimmune exacerbation [29].
In pSS patients, Leehan et al., confirmed that fibrosis of minor salivary glands (MSG) is a pathological feature of pSS that positively correlates witha focus score and is not age-related [30]. Another study identified that mast cells are strongly associated with the fibrosis and fatty infiltration of SGs. It is hypothesized that they promote fibrosis through interaction with local fibroblasts and through the production of enzymes cleaving and activating MMPs, which are essential mediators of tissue injury [31].
Mast cells express TLR-2 and TLR-4, as well as receptors for IL-1 including interleukin-1 receptor type 1 (IL1R1) and suppressor of tumorigenicity 2 (ST2). Mast cells activation through TLR-2 and TLR4 lead to the production of IL-1, TNF-α, IL-33, and chemokines such as C-X-C motif chemokine ligand 1 (CXCL1) and C-X-C motif chemokine ligand 2 (CXCL2), which recruit neutrophilic granulocytes and DCs. The activation of mast cells through IL1R1 and ST2 allows them to interact with T and B cells, interfering with antibody production [29]. The activation of ST2 on mast cells through IL-33 leads to the production of pro-inflammatory cytokines such as IL-1, IL-6, IL-13 [32], and induces a TH2 polarization of CD4+ T cells.

2.4. Salivary Gland Epithelial Cells (SGECs)

Salivary gland epithelial cells (SGECs) form the acinar secretory structure and the ductal excretory structures in SGs [33]. SGECs constitute the main target of auto-immunity in pSS, described as an autoimmune epithelitis [34]. Over recent years, it has become clear that SGECs also fulfill an important role in the initiation of autoimmunity.
In pSS patients, the loss of polarity of SGECs plays a crucial part in the onset of the local inflammatory process. Indeed, a decrease in occludin and zonula occludens 1 (ZO-1) expression and a redistribution of claudin to the basolateral plasma membrane have been observed in SGs from pSS patients. Furthermore, the exposure of isolated SGs cells from healthy controls to pro-inflammatory cytokines such as TNF-α and IFN-γ reproduced the alterations observed in pSS patients [35,36]. By altering the tight junction integrity of SGECs, the local cytokine production may therefore account for the secretory gland dysfunction observed in pSS patients, and subsequent decrease in saliva quality and quantity [35].
In genetically susceptible subjects, environmental stimuli such as viruses may trigger salivary gland epithelial cells (SGECs) through TLR activation [37,38]. More precisely, the activation of TLR-2 and TLR-4 expressed on the surface of SGECs results in the expression of mediators of immune activation (such as the intercellular adhesion molecule 1 (ICAM-1)), CD40, and major histocompatibility complex 1 (MHC-1) [39], as well as in IL-15 secretion inducing the proliferation of activated B and T cells and the generation and maintenance of natural killer (NK) cells. Beside leading to the expression of ICAM-1, CD40, and MHC-1, the activation of endosomal TLR3 leads to the secretion of BAFF that promotes the activation and maturation of B cells. TLR3 activation also contributes to SGEC anoikis, a form of apoptosis that is triggered by a loss of cell attachment to the extracellular matrix (ECM), thereby releasing exosomes and apoptotic blebs containing autoantigens such as Ro/SSA and La/SSB that drive autoimmunity in pSS by attracting both classical DC and pDC within SGs [40,41,42].
Activated SGECs produce chemokines that attract immune cells and contribute to the formation of germinal centers, including C-X-C motif chemokine ligand 9 (CXCL-9), C-X-C motif chemokine ligand 10 (CXCL-10), C-X-C motif chemokine ligand 12 (CXCL12), C-X-C motif chemokine ligand 13 (CXCL13) and C-C chemokine ligand 19 (CCL19), and C-C chemokine ligand 21 (CCL21) [43,44]. An increased epithelial production of cytokines such as IL-1, IL-6, and TNFα may also contribute to create a pro-inflammatory environment [45].
Activated SGECs develop the ability to act as non-professional antigen-presenting cells [46] by expressing co-stimulation molecules (CD80 and CD86) [47] and MHC-I (HLA-ABC) and MHC-II (HLA-DR), adhesion molecules such as ICAM1, vascular cell adhesion molecule 1 (VCAM-1) [46,48]. Thus, SGECs appear suitably equipped for the presentation of antigenic peptides and the transmission of activation signals to T cells [44,49].

2.5. Endothelial Cells

Endothelial cells, expressing CD31, form a one-cell thick walled layer called endothelium that upholster blood and lymphatic vessels. Beside bringing immunes cells to inflammation sites, endothelial cells take an active and regulatory role in inflammatory processes [50]. In response to IL-1 and TNFα, activated endothelium express adhesion molecules such as ICAM-1, VCAM-1, and E- and P-selectins that allow the interaction and migration of blood immune cells to inflamed tissues [51].
In pSS patients, the expression of ICAM-1 positively correlates with a focus score of salivary biopsies [52]. Both strong vascular endothelial growth factor C (VEGF-C) and vascular endothelial growth factor receptor 3 (VEGFR-3) expression were reported in MSGs from pSS patients [53]. As a result, increased and anatomically aberrant lymphatic neovascularization leads to a persistent extravasation of immune cells. In another study, defective lymphatic vessels were also characterized by the overproduction of CCL-21 that increased the infiltration of immune cells into inflamed tissues [54].

2.6. Mucosal-Associated Invariant T (MAIT) Cells

Mucosal-associated invariant T (MAIT) cells are innate-like T cells and can therefore be considered a bridge between innate and adaptive responses [55]. MAIT cells express an invariant T cell receptor (TCR) α-chain (Vα7.2–Jα33 in humans) and CD161 which is typically expressed by NK cells. They recognize vitamin B-related peptides through the evolutionary conserved non-polymorphic MHC-I-related molecule (MR1) [56]. In response to different stimuli, MAIT cells also have the capacity to express both CD4 and/or CD8 co-receptors. MAIT cells are characterized by a natural memory function and by their capacity to rapidly produce Th1, type 2 helper cells (Th2), and Th17 cytokines [55,57].
Very little is known about the contribution of MAIT cells in the pathogenesis of pSS. Wang et al. found that MAIT cells are decreased in peripheral blood circulation but are increased in SGs from pSS patients compared to healthy controls. From a functional point of view, MAIT cells from pSS patients were mainly CD4+ and naïve, in contrast with MAIT cells from controls that were almost exclusively CD8+. In addition, MAIT cells of pSS patients displayed lower levels of activation with a reduced expression of CD69 and CD154, and lower levels of TNFα and IFN-γ. The aberrant phenotype of MAIT cells in pSS patients may lead to the dysregulation of the local immune responses, which would trigger local damage in SGs and auto-immunity [57].

2.7. Natural Killer (NK) Cells

Natural Killer (NK) cells are a cytolytic component of the innate immune system. They have the ability to sense the pathological changes of self-cells and therefore take an important part in the immune surveillance of tumor cells and virus-infected cells [58]. NK cells express the NKp30 receptor that is recognized by DCs and lead to the production of Th1 cytokines such as IFN-γ and IL-12 [59].
NK cells are enriched in MSGs from pSS patients and their presence correlate with the focus score [60]. In addition, NK cells overexpress the NKp30 receptor and SGECs express B7-H6, the ligand for NKp30. Taken together, this may explain the hyperactivity of NK cells and the interrelation with SGECs and DCs that lead to a subsequent activation of innate and adaptive immunity. The expression of B7-H6 by SGECs may also be involved in the homing of NK cells in SGs [60]. Another study identified a subset of NK cells that expresses NKp44 and produces IL-22 in SGs from pSS patients. This subgroup has however been reclassified and is now part of Innate Lymphoid Cells (ILCs), which will be discussed in the Section 2.9 of this article [61].

2.8. Natural Killer T (NKT) Cells

NK T (NKT) cells are immune components that share features of both T cells and NK cells. They discriminate self from non-self-antigens and produce prompt immune responses against Gram-negative bacteria [62]. They are a major source of IL-4 and to a lesser extent of IFN [63]. Invariant NKT (iNKT) cells are a special subset of NKT cells that seems to play a pivotal role in the regulation of immunity. They express CD161 (typical of NK cells) and a semi-invariant T cell receptor (TCR). By linking CD1d in B cells with their invariant TCR, iNKT cells are able to suppress B cell auto-reactivity. In addition, under certain circumstances, they can express both CD8+ and CD4+, which leads to the production of Th1 and Th2 cytokines [62,64].
A decreased number of NKT cells was observed in the peripheral blood of pSS patients, which could be explained by apoptosis or homing in SGs [65]. Another study reported an increased number of iNKT in peripheral blood but a complete absence of iNKT cells together with an increased number of auto-reactive B cells in SGs from pSS patients [66]. These data were corroborated by showing that the lack of CD1d following B-cells hyperactivation lead to a greater release of autoantibodies [67]. In spite of the studies supporting the candidacy of NK cells in the SG of pSS patients, there is actually no sufficient proof bolstering their role as participating actively in the pathology of SS.

2.9. Innate Lymphoid Cells (ILCs)

Innate lymphoid cells (ILCs) are the innate counterparts of T helper lymphocytes [68,69]. They are mostly concentrated at epithelial barriers and rapidly release cytokines in response to environmental triggers. They can be classified into three subsets according to the expression of specific transcription factors and the production of cytokines that mirror the subsets of helper T. ILC1 express the T-Box Transcription Factor 21 (TBX21, also named T-bet), produce IFN-γ, and respond to intracellular pathogens such as viruses. ILC2 express the transcription factor GATA Binding Protein 3 (GATA-3), produce IL-4, IL-5 and IL-13, and respond to extracellular parasites and allergens. ILC3 express the transcription factor Retinoic acid-related orphan receptor gamma t (RORγt), produce IL-17A and IL-22, and react to extracellular pathogens such as bacteria and fungi [68,70,71].
ILCs have been identified in the SGs from pSS patients [61,72] and may contribute to the formation of germinal center-like structures [73]. Blokland et al. showed that the presence of ILC1 was associated with a higher disease activity score (ESSDAI). ILC1 could contribute to the pathogenesis of pSS through the massive production of IFN-γ, but the underlying mechanisms remain largely elusive [74]. In addition, an increased IFN signature and reduced frequencies of ILC2 and ILC3 was associated with a high expression of Fas cell surface death receptor (Fas also named CD95) by ILC2 and ILC3. It was hypothesized that the increased Fas expression on ILC2 and ILC3 may induce an apoptosis of these cells. These observations corroborate previous studies in mice that reported a link between type I IFN, pDC activation, and apoptosis of circulating ILC2 and ILC3 [75,76,77].
A subset of ILC3 that was originally classified as NK cells because of its expression of NKp44 was identified in SGs from pSS patients. It was found to be a major source of IL-22 together with Th17 cells. The frequency of ILC3 was positively correlated to the focus score [61]. Additional studies are needed to further evaluate the ILC3 function in SGs from pSS patients. Currently, their role in the pathogenesis of SS remains to be determined and detailed. There is not enough data purporting their role as being active players in SS pathology.
Figure 1 summarizes the action of the different players of innate immunity in the pathogenesis of pSS.

3. Adaptive Immunity: The Insidious Role of T Cell Subsets in SS

Although B cells are involved in the last chronic inflammation phase by producing autoantibodies, pSS is mostly dominated by T lymphocytes in the early stages of the disease. In the past, T cells have been considered the most abundant cells in the pathogenic picture of SS and the Th1 subset was identified as the major cell type infiltrating MSGs. Today, the discovery of other T cell subpopulations has unveiled new avenues revealing a further understanding of the pathogenesis of pSS. The infiltrating cells observed in pSS SGs biopsies are composed of about 45–50% of CD4+ T lymphocytes, 20% of CD8+ T lymphocytes, and 20% of B cells [78]. One of most common clinical features of pSS is the reduction of lymphocytic cells in peripheral blood. Several models have been proposed to explain this phenomenon but the most accredited model attributes this phenomenon to a selective migration of the CD4+ T-cell to the inflamed tissue. Although this model remains speculative since the mechanism of cell migration still remains unclear, CD4+ T-cells represent one of the significant protagonists of pSS inflammation.

3.1. CD4+ T Cells

3.1.1. Th1-Th2 Cells

During the differentiation cascade of T CD4+, the proliferating helper T cells can differentiate into two subtypes based on distinct cytokine patterns: Th1 and Th2 cells.
Upon T-cell activation, IFN-γ and IL-12 induce the expression of T-bet and the signal transducer and activator of transcription (STAT)-4, which is involved in the differentiation of naïve CD4+ T cells into Th1 lymphocytes. Th1 cells predominantly produce pro-inflammatory cytokines such as IFN-γ [79] and IL-2. In contrast, IL-2 and IL-4 induce the GATA-3 transcription factor and the consequent polarization of naïve T cells into Th2. Th2 cells produce anti-inflammatory cytokines such as IL-4, IL-5, IL-9, IL-10, IL-13, and IL-25 [80]. Several studies have suggested that pSS was related to abnormal Th1 activation and SGs infiltration [81]. This evidence was supported by the presence of elevated levels of IFN-γ in serum and Th1 cells in blood. Furthermore, T cells expressing high levels of IFN-γ and STAT-4 mRNA have been found in SGs from SS patients while Th2-related marker transcripts were observed only in GC-positive patient SGs biopsies with severe B cell infiltration and organization. These data suggest that these two cell types operate in different stages of the disease. Paradoxically, a Th2 cytokine profile (IL-6 and IL-10) was observed in blood from pSS patients while the affected tissues were mainly characterized by Th1 and/or Th17 cells. This Th1/Th2 imbalance, generally observed in various chronic inflammatory disorders, is not easily understood because of a limited number of studies (Figure 2) [82].

3.1.2. Th17

The inflamed SGs from pSS patients represent a perfect environment for the recruitment and polarization of Th17 cells. Although their contribution to the pathogenesis of pSS remains unclear, Th17 cells can fulfill several roles considered potentially pathogenic. In healthy conditions, Th17 cells play a fundamental role in maintaining mucosal barrier integrity by inducing the synthesis of tight junction proteins [83], proliferation of epithelial cells [84], and playing a defense role against microbes [85,86,87]. Th17 cells differentiate from naive CD4+ cells in the periphery in response to different signals and cytokines secreted by antigen-presenting cells [88]. The critical cytokine involved in Th17 differentiation is transforming the growth factor beta (TGFβ) in combination with IL-6 or IL-21 [88,89] which induce the activation of several transcription factors such as signal transducer and activator of transcription (STAT)-3 that induces the expression of RORγt, a member of the retinoic acid–related orphan nuclear hormone receptor family [88]. RORγt in synergy with RORα promotes Th17 differentiation [90]. Th17 cells produce IL-17 and other inflammatory cytokines such as TNF-α, IL-22, and IL-26. IL-17 also called IL-17A is a member of six cytokines family which also includes IL-17F, another Th17 specific cytokine. IL-17A and IL-17F are characterized by a 55% amino acid sequence identity and a common receptor [91]. These cytokines along with TNF-α are especially involved in inducing and mediating pro-inflammatory responses. Regarding IL-22 and IL-26, much less is known. Several studies carried out in different cohorts of sicca-SS and sicca-non SS have showed the presence of an elevated level of IL-17 cytokines and IL-17 mRNA in tears [92,93] and SGs from sicca-SS patients in association with lymphocytic infiltrates [94,95].

3.1.3. T Follicular Helper Cells

T follicular helper (Tfh) cells are a subset of CD4+ T that act in collaboration with B cells to promote and regulate humoral responses. Tfh cell differentiation process requires initial interaction with DCs [96] and B cells. Tfh are mainly situated in secondary lymphoid organs and express a specific combination of cell surface molecules [97]. The best marker that defines the Tfh cells is the C-X-C motif chemokine receptor type 5 (CXCR5) [98,99] whose expression is regulated by a B-cell lymphoma 6 (Bcl-6) transcription factor. In the first stage of development, the naive CD4+ T cells do not express CXCR5 but only the C-C chemokine receptor type 7 (CCR7) that allow them to enter secondary lymphoid organs and reach the T-cell zones. Following their activation, the Tfh cells downregulate CCR7 and upregulate CXCR5, which facilitates their migration from the T-cell zones into the CXCL13-rich B-cell follicles [100,101]. In the follicles, Tfh cells provide a large panel of survival signals to B cells such as CD40L, IL-4, IL-21, programmed death-ligand 1 (PD-1), and BAFF [102] and contribute to the generation of most memory B cells and plasma cells. In SS pathology, a significantly increased number of Tfh cells were observed in the anti-SSA/SSB positive patients, as well as in patients with increased serum IL-12, IL-21 levels, and high focus score values suggesting that increased Tfh cells may play an important role in disease development and progression [103,104]. In conclusion, acting as a regulator of T cell-dependent B cell hyperactivity, the Tfh cells can prove to be a new therapeutic target in pSS disease [105].
Another particular type of T cells sharing similar characteristics as Tfh cells are CCR9+ Th cells [106]. The latter, similarly, to Tfh cells, express Bcl-6, IL-21, and ICOS but have the specific hallmark of displaying CCR9 and a lessened expression of CXCR5 [107]. CCR9+ T cells have a propensity of producing significantly higher levels of inflammatory cytokines such as IL-7, IL-21, IFN-γ, and IL-17. Both CCR9+ and CXCR5+ T cells correlate with increased B cell activity [108].

3.1.4. T Regulatory Cells

The self-tolerance maintenance is regulated by several processes such as the deletion of self-reactive T cells. T-regulatory cells (Tregs) play a central role in this mechanism by regulating the immune homeostasis and suppressing autoreactive lymphocytes through cell-cell contact or the release of cytokines including IL-10 and TGF-β [109,110]. The role of Tregs is explained by emerging evidences in which the suppression of immune responses might be necessary, such as in autoimmune diseases [111,112,113]. Treg cells were firstly identified as cells expressing the alpha chain of the IL-2 receptor (IL-2Rα, CD25) on their cell surface and the ability to prevent autoimmunity in an experimental mice model [114]. The differentiation of naïve T lymphocytes into Tregs depends on the specific cytokine microenvironment and the expression of the forkhead box protein transcription factor P3 (FoxP3). In general, the presence of TGF-β and the absence of IL-6 promotes the Treg phenotype instead of Th17. Thus, TGF-β is required in both cases, but the presence or absence of IL-6 leads to the generation of Th17 or Treg cells, respectively [115]. It seems clear that the perturbation of this process could induce the generation of pathogenic Th17 cells and the development of autoimmunity. To date, the role of Treg cells in SS still remains controversial [116,117,118,119,120,121,122,123]. The observed discrepancies may, in part, have been due to the strategies used to analyze Treg cells. In fact, in the past, the proportion of circulating Treg cells was based on the expression of CD25 without taking into account the co-expression of FoxP3. Some studies have shown a reduction in CD25highTreg cells [117,118,120,123], but the association with clinical or serological manifestations was observed only in a few studies [117,118]. Some other studies have observed a reduction of Treg cells in peripheral blood in patients with a milder clinical picture without extra-glandular manifestations [120]. In contrast, other studies have reported an increase in circulating Treg cells in SS [121] while still others have described that the Treg proportions were comparable between pSS and controls [116,122]. Despite these conflicting evidences, a specific subset of CD4+ T cells expressing Foxp3, TGF-β, and IL-10 but low levels of CD25 have been observed in SS patients. This new subsets of CD4+ T cell is increased in patients with inactive disease compared to the control and present a strong inhibitory activity against autoreactive cells [123,124].

3.1.5. Follicular Regulatory Cells (Tfr)

Follicular regulatory T cells (Tfr) are a subtype of Treg specialized in the regulation and suppression of Tfh and B cells activity [125]. Tfr express high levels of CXCR5, inducible the T cell co-stimulator (ICOS) and PD-1 on the cell surface, which directs them to follicles and GC regions [126]. In addition, Tfr cells express specific Treg markers such Bcl6 [127], Foxp3, Nuclear Factor of activated T cells 2 (NFAT-2), Glucocorticoid-induced TNFR-related protein (GITR), B lymphocyte-induced maturation protein-1 (Blimp-1), and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4). The Tfr differentiation program is triggered by dendritic cells [128] and B cells [129] thanks to costimulatory signals such as CD28 and ICOS [125,130]. NFAT-2 facilitates CXCR5 up-regulation in Foxp3+ T cells [131]. Following their differentiation, Tfr cells enter the circulation to become memory Tfr or to migrate to the B cell zone [132]. In an experimental Bcl6fl/flFoxp3Cre (KO) mice model, the deletion of Tfr cells makes them susceptible to the development of autoimmune diseases and experimental Sjögren’s syndrome [133]. Periphery blood and SGs analysis from SS patients shows an enrichment of Tfr cells with a Tfr/Tfh ratio increased if compared to the control [134,135]. However, the involvement of Tfr cells in exacerbation of SS still remains controversial.

3.2. CD8+ T Cells

The histological analysis of SGs from pSS patients show that CD4+ T cells are the most common cell type. However, recent studies have also observed the presence of activated CD8+T cells that express a high level of human leukocyte antigen (HLA)-DR and positively correlate with several disease features [136]. In addition, the expression of C-X-C motif chemokine receptor type 3 (CXCR3) by CD8+ T cells in SS patients may be involved in the migration of them to the inflamed SGs [137]. In non-obese diabetic (NOD) mouse model of Sjögren’s syndrome, CD8+ T cells are activated and produce inflammatory cytokines. In addition, the transfer of CD8+ T cells from NOD mice lymph nodes (LNs) into NOD-severe combined immunodeficiency hosts induces the inflammation of the lacrimal glands. These results demonstrate the pathogenic role of CD8+ T to induce exocrine gland autoimmunity in NOD mice [138].
More recently, it has been suggested that CD4−CD8− (Double-Negative) T cells also play a role in the pathogenesis of pSS. These cells are expanded in several inflammatory situations and invade the inflamed tissues contributing to the tissue damage in autoimmune disease such as psoriasis, SLE, and SS [139].

Innate T Cells

Different innate T cells are present in the peripheral blood and/or the SGs from pSS patients. They include mainly gd, invariant NK T cells, and mucosal-associated invariant T cells. These cells are activated rapidly upon stimulation and are not dependent on MHC-II activation [140].

3.3. B Cells

3.3.1. B Cell Hyperactivity

B-cell hyperactivity represents one of the hallmarks of the SS and also plays a key role in the autoimmunity and lympho-proliferative processes [141,142,143]. B cells originate from the bone marrow from hematopoietic stem cells. During development, B cells go through different stages of selection which exclude a substantial fraction of self-reactive and polyreactive B cell [144]. In the first checkpoint, which takes place in the bone marrow, polyreactive B cells are removed during a process known as central tolerance. In the second checkpoint, in the periphery, only a small amount of self-reactive and polyreactive mature naïve B cells could survive. The third tolerance checkpoint, known as the pre-germinal center checkpoint, allows the exclusion of self-reactive naïve B cells from entering B cell follicles [145]. A recent study has shown a deficiency in both early and late B cell tolerance checkpoints in patients with SS. In addition, B cell depletion using antibodies against CD20 in the inhibitor of DNA binding 3 (Id3) KO mice model leads to a significant histological recovery associated with an improvement of saliva secretory functions. This observation corroborates the hypothesis that B cells could play a critical role in SS exacerbation [146].
Some studies have posited an early activation of B cells in SS by the innate immune system. The interferon signaling pathway, which is a key feature in SS, interacts with B cells to trigger the production of autoantibodies [147]. A new subtype of neutrophils found in the splenic marginal zone (MZ), favored the activation and proliferation of B cells by inducing the secretion of BAFF, (a proliferation-inducing ligand) APRIL, and IL-21 [22,148].

3.3.2. B Cell Subpopulations

Different B cell populations have been found in SGs as well as in peripheral blood from pSS patients [149]. There is a significant increase of IgD+CD38+ B cells expressing CD19 in pSS patients. Moreover, there is also a substantial increase in Bruton tyrosine kinase (BTK) in the B cells of pSS patients [150]. The resulting effect of these important numbers of B cells is enhanced B cell signaling through B cell receptors. Besides the role of memory B cells, plasmablasts and plasma cells have been implicated in the pathogenesis of pSS. In pSS patients with lymphoma, increased numbers of CD27 highCD19 lowCD20 plasmablasts have been identified [151]. The immunophenotyping study of pSS defined six different subcellular types including increased plasmablasts, activated CD4+ and CD8+ T cells, as well as decreased numbers of CD27+ B cells, CD4+ T cells, and pDC [152]. In the SG of pSS, immunophenotyping revealed B cells, CD8+ T cells, and activated epithelial cells. Moreover, the increased number of plasmablasts and plasma cells strongly correlated with disease activity as well as serum IgG and the presence of auto antibodies [152]. In SG from pSS patients, there was an important proportion of infiltrating B cells that were fully differentiated plasma cells [152].

3.3.3. Marginal Zone B Cells

Marginal zone B cells are a subset of splenic B cells. Their involvement in the pathophysiology of pSS has not yet been demonstrated. An increased number of marginal zone-like B cells in the MSG of pSS patients has been observed [153]. In addition, studies using mice models of pSS showed that the depletion of MZ B cells prevented the development of pSS manifestations in mice [154,155,156,157]. Even if there is no current evidence of the importance of MZ B cells in pSS patients, an increased percentage of CD27+ memory B cells from the SGs of the latter were identified as IgM+ cells [153]. Furthermore, the fact that pSS associated lymphoma stem from mucosa-associated lymphoid tissue (MALT) tissue, fuels the role of this subtype of B cells in pSS [158].

3.3.4. Regulatory B Cells

Regulatory B cells are another type of B cell involved in pSS. These types of B cells exert their regulatory function through the production of IL-10. As such, B regulatory cells by inducing the expansion of Treg cells and by the actions of IL-10, are able to control the proliferation of Th1 cells and restrain the actions of TNf-α, Th17 cells IL-12 producing dendritic cells as well as CD8+ T cells. Beyond the production of IL-10, it has been recently shown that some B cells can also secrete IL-35. IL-35 is a cytokine which is part of the IL-12 family and has anti-inflammatory and immunosuppressive properties. In pSS, there is a disequilibrium between IL-12 and IL-35 in favor of IL-12. There was a significant decrease in serum IL-35 in pSS patients that was inversely correlated with disease activity. It has been hypothesized that the IL-12/IL-35 axis could play a noteworthy role in disease perpetuation in pSS [159].

3.3.5. BAFF

BAFF is a pivotal cytokine that promotes the maturation, proliferation, and survival of B cells. In pSS, BAFF plays an important role in favoring the activation and proliferation of B cells, thereby leading to the production of autoantibodies. In essence, BAFF is secreted by dendritic cells, monocytes, and macrophages but in pSS both T and B cells as well as epithelial cells have been shown to release BAFF.
In pSS patients, activated SGECs are able to produce BAFF, undergirding their role in the pathogenesis of the disease as well as demonstrating the link between innate and adaptive immunity. This is illustrated and strengthened by studies showing elevated levels of BAFF in the sera of pSS patients as well as its correlation with anti-Ro/SSA and anti/La-SSB and rheumatoid factor (RF) [160]. Furthermore, a strong correlation between BAFF secretion by monocytes and type I IFN was found. A role for BAFF in the formation of ectopic germinal centers (GC) has also been advocated [161] but nullified by further studies implying that other pathogenic pathways might be involved [162].

3.3.6. Germinal Center-Like Structures

The presence of B cells in SGs is an important characteristic of pSS extending from a discrete infiltrate to the formation of the ectopic germinal center (GC) completely invading the glands. Ectopic GC-like structures play a major role in the pathogenesis of pSS by favoring chronic B cell activation. They are present in 10–30% of pSS patients, are associated with the presence of autoantibodies anti-SSA, as well as an increased risk of developing lymphoma [163]. CXCR5 and CXCL13 are mainly involved in the formation of GC-like structures by recruiting Tfh and B cells [164].

4. Cross Talk between the Innate and Adaptive Immunity

The pathophysiology underlying pSS is complex with different intricating players from the innate immunity cells and the adaptive immunity T and B cells. Figure 3 illustrates the orchestrating of these different tenants interacting together to trigger the initiation and perpetuation of disease.
In genetically susceptible individuals, together with the presence of a hormonal disequilibrium as well as environmental factors such as viruses, there is an activation of the epithelial cells. Following the activation of epithelial cells, there is an upregulation of innate immune cells such as TLRs and ensuing pro-inflammatory cytokine production. Furthermore, activated and injured SGECs release exosomes and apoptotic blebs containing ribonucleoprotein autoantigens such as Ro/SSA and La/SSB that can attract both classical DC and pDC within SGs.
The activation of DC and pDC can trigger the production of type 1 and type 2 Interferons. The production of IFN-α by pDC and the secretion of inflammatory cytokines by conventional DC such as IL-12 and IFN-γ can induce tissue damage. The production of IFN-α as well as IFN-γ can enhance the secretion of BAFF thereby fostering B cell activation and proliferation as well as the secretion of auto antibodies. The production of autoantibodies can form immune complexes with the autoantigens (Ro/SSA and La/SSB) and can further thrust the secretion of IFN-α, thereby constituting a vicious inflammatory loop perpetuating disease progression.
It has to be stressed that the time of the differentiation factor expressions, different cell activations, as well as chemokine signaling and homing of lymphocytes is, perhaps, the most critical for the development, perpetuation, and severity of the clinical phenotype.

5. Conclusions

The innate and adaptive immunity are central in the pathogenesis of pSS, each of them representing a multi-step process leading to the triggering and perpetuation of disease. The salivary epithelial cells are at the heart of disease process, featuring as the main initial triggers of the disease and maintaining the crosstalk with B and T cells, thereby further relating to the production inflammatory cytokines and the production of autoantibodies. Further knowledge on deciphering and dissecting the different intricate players of the pathogenesis of pSS could pave the way for new avenues of therapies.

Author Contributions

C.C., J.S. wrote and revised the manuscript, C.D. and J.P. revised manuscript, M.S.S. edited, wrote and revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by EU H2020 contract HarmonicSS (H2020-SC1-2016-RTD/731944), a research credit (CDR) J.0018.18 from the Fund for Scientific Research (F.R.S.–FNRS) and a grant from the Fondation Jaumotte-Demoulin.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

APRILa proliferation-inducing ligand
BAFFB-cell activating factor
Bcl-6B-cell lymphoma 6
Blimp-1B lymphocyte-induced maturation protein-1
BTKBruton tyrosine kinase t
CCL3C-C chemokine ligand type 3
CCL4C-C chemokine ligand type 4
CCR5C-C chemokine receptor type 5
CCR7C-C chemokine receptor type 7
CTLA-4cytotoxic T-lymphocyte-associated protein 4
CXCL1C-X-C motif chemokine type 1
CXCL2C-X-C motif chemokine type 2
CXCL9C-X-C motif chemokine type 9
CXCL10C-X-C motif chemokine type 10
CXCL12C-X-C motif chemokine type 12
CXCL13C-X-C motif chemokine type 13
CXCR3C-X-C motif chemokine receptor type 3
CXCR5C-X-C motif chemokine receptor type 5
CCL3chemokine (C-C motif) ligand 3
CCL4chemokine (C-C motif) ligand 4
CCL19chemokine (C-C motif) ligand 19
CCL21chemokine (C-C motif) ligand 21
DCsdendritic cells
ECMextracellular matrix
ESSDAIEULAR Sjögren’s syndrome disease activity index
fDCsfollicular dendritic cells
FoxP3forkhead box protein transcription factor P3
GATA-3GATA Binding Protein 3
GITRglucocorticoid-induced TNFR-related protein
HLAhuman leukocyte antigen
ICAM-1intercellular adhesion molecule 1
ICimmune-complexes
ICOSInducible T-cell COStimulator
Id3inhibitor of DNA binding 3
BaffInterferon
IFN-γinterferon gamma
ILInterleukin
IL1R1interleukin-1 receptor type 1
IL-2RαIL-2 receptor α
ILCinnate Lymphoid Cells
iNKTinvariant natural killer T cells
LNslymph nodes
MAITmucosal-associated invariant T cells
MALTmucosa-associated lymphoid tissue
MSGsminor salivary glands
MHC-Imajor histocompatibility complex class I
MHC-IImajor histocompatibility complex class II
MMPsMetalloproteases
MR1MHC-I-related molecule 1
MZmarginal zone
NFAT-2nuclear Factor of activated T cells 2
NKnatural killer cells
NKTnatural killer T cells
NODnon-obese diabetic
PD-1programmed death-ligand 1
pDCsplasmacytoid dendritic cells
pSSprimary Sjögren’s syndrome
RArheumatoid arthritis
RORγtretinoic acid-related orphan receptor gamma t
SGssalivary glands
SGECssalivary glands epithelial cells
SLEsystemic lupus erythematosus
SSSjögren’s syndrome
sSSsecondary Sjögren’s syndrome
ST2suppression of tumorigenicity 2
STATsignal transducer and activator of transcription
TBX21 or T-betT-Box Transcription Factor 21
TCRT cell receptor
TfhT follicular helper
Tfrfollicular regulatory T cells
TGFβtransforming growth factor beta
Th1type 1 helper cells
Th17type 17 helper cells
TLRtoll-like receptor
TNFαtumor necrosis factor alpha
TregsT-regulatory cells
VCAM-1vascular cell adhesion molecule
VEGF-Cvascular endothelial growth factor C
VEGFR-3vascular endothelial growth factor receptor 3
ZO-1zonula occludens 1

References

  1. Fox, R.I.; Kang, H.I. Pathogenesis of Sjogren’s syndrome. Rheum. Dis. Clin. N. Am. 1992, 18, 517–538. [Google Scholar]
  2. Fox, R.I.; Michelson, P. Approaches to the treatment of Sjogren’s syndrome. J. Rheumatol. Suppl. 2000, 61, 15–21. [Google Scholar] [PubMed]
  3. Basbaum, A.I. Opioid regulation of nociceptive and neuropathic pain. Clin. Neuropharmacol. 1992, 15 Pt A (Suppl. S1), 372A. [Google Scholar] [CrossRef]
  4. Sullivan, D.A. Sex hormones and Sjogren’s syndrome. J. Rheumatol. Suppl. 1997, 50, 17–32. [Google Scholar] [PubMed]
  5. Nguyen, C.Q.; Peck, A.B. Unraveling the pathophysiology of Sjogren syndrome-associated dry eye disease. Ocul. Surf. 2009, 7, 11–27. [Google Scholar] [CrossRef] [Green Version]
  6. Vitali, C.; Bombardieri, S.; Jonsson, R.; Moutsopoulos, H.M.; Alexander, E.L.; Carsons, S.E.; Daniels, T.E.; Fox, P.C.; Fox, R.I.; Kassan, S.S.; et al. Classification criteria for Sjogren’s syndrome: A revised version of the European criteria proposed by the American-European Consensus Group. Ann. Rheum Dis. 2002, 61, 554–558. [Google Scholar] [CrossRef] [Green Version]
  7. Parisis, D.; Chivasso, C.; Perret, J.; Soyfoo, M.S.; Delporte, C. Current State of Knowledge on Primary Sjogren’s Syndrome, an Autoimmune Exocrinopathy. J. Clin. Med. 2020, 9, 2299. [Google Scholar] [CrossRef]
  8. Kiripolsky, J.; McCabe, L.G.; Kramer, J.M. Innate immunity in Sjogren’s syndrome. Clin. Immunol. 2017, 182, 4–13. [Google Scholar] [CrossRef]
  9. Steinman, R.M.; Hemmi, H. Dendritic cells: Translating innate to adaptive immunity. Curr. Top. Microbiol. Immunol. 2006, 311, 17–58. [Google Scholar]
  10. Gottenberg, J.E.; Cagnard, N.; Lucchesi, C.; Letourneur, F.; Mistou, S.; Lazure, T.; Jacques, S.; Ba, N.; Ittah, M.; Lepajolec, C.; et al. Activation of IFN pathways and plasmacytoid dendritic cell recruitment in target organs of primary Sjogren’s syndrome. Proc. Natl. Acad. Sci. USA 2006, 103, 2770–2775. [Google Scholar] [CrossRef] [Green Version]
  11. Ozaki, Y.; Ito, T.; Son, Y.; Amuro, H.; Shimamoto, K.; Sugimoto, H.; Katashiba, Y.; Ogata, M.; Miyamoto, R.; Murakami, N.; et al. Decrease of blood dendritic cells and increase of tissue-infiltrating dendritic cells are involved in the induction of Sjogren’s syndrome but not in the maintenance. Clin. Exp. Immunol. 2010, 159, 315–326. [Google Scholar] [CrossRef] [PubMed]
  12. 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 Sjogren syndrome. Ann. Rheum. Dis. 2009, 68, 420–426. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Hillen, M.R.; Pandit, A.; Blokland, S.L.M.; Hartgring, S.A.Y.; Bekker, C.P.J.; van der Heijden, E.H.M.; Servaas, N.H.; Rossato, M.; Kruize, A.A.; van Roon, J.A.G.; et al. Plasmacytoid DCs From Patients With Sjogren’s Syndrome Are Transcriptionally Primed for Enhanced Pro-inflammatory Cytokine Production. Front. Immunol. 2019, 10, 2096. [Google Scholar] [CrossRef] [Green Version]
  14. Ainola, M.; Porola, P.; Takakubo, Y.; Przybyla, B.; Kouri, V.P.; Tolvanen, T.A.; Hanninen, A.; Nordstrom, D.C. Activation of plasmacytoid dendritic cells by apoptotic particles-mechanism for the loss of immunological tolerance in Sjogren’s syndrome. Clin. Exp. Immunol. 2018, 191, 301–310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Swiecki, M.; Colonna, M. Unraveling the functions of plasmacytoid dendritic cells during viral infections, autoimmunity, and tolerance. Immunol. Rev. 2010, 234, 142–162. [Google Scholar] [CrossRef]
  16. Swiecki, M.; Colonna, M. The multifaceted biology of plasmacytoid dendritic cells. Nat. Rev. Immunol. 2015, 15, 471–485. [Google Scholar] [CrossRef]
  17. Vakaloglou, K.M.; Mavragani, C.P. Activation of the type I interferon pathway in primary Sjogren’s syndrome: An update. Curr. Opin. Rheumatol. 2011, 23, 459–464. [Google Scholar] [CrossRef]
  18. Aloisi, F.; Pujol-Borrell, R. Lymphoid neogenesis in chronic inflammatory diseases. Nat. Rev. Immunol. 2006, 6, 205–217. [Google Scholar] [CrossRef]
  19. Frank, G.B.; Kokate, T.G. Blockade of high K+ contractures and Ca(+)+-dependent slow action potentials in frog skeletal muscle by opioid drugs. Prog. Clin. Biol. Res. 1990, 328, 283–286. [Google Scholar]
  20. Stout, R.D.; Jiang, C.; Matta, B.; Tietzel, I.; Watkins, S.K.; Suttles, J. Macrophages sequentially change their functional phenotype in response to changes in microenvironmental influences. J. Immunol. 2005, 175, 342–349. [Google Scholar] [CrossRef]
  21. Morell, M.; Varela, N.; Maranon, C. Myeloid Populations in Systemic Autoimmune Diseases. Clin. Rev. Allergy Immunol. 2017, 53, 198–218. [Google Scholar] [CrossRef] [PubMed]
  22. 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]
  23. Mills, K.H. Induction, function and regulation of IL-17-producing T cells. Eur. J. Immunol. 2008, 38, 2636–2649. [Google Scholar] [CrossRef] [PubMed]
  24. Gliozzi, M.; Greenwell-Wild, T.; Jin, W.; Moutsopoulos, N.M.; Kapsogeorgou, E.; Moutsopoulos, H.M.; Wahl, S.M. A link between interferon and augmented plasmin generation in exocrine gland damage in Sjogren’s syndrome. J. Autoimmun. 2013, 40, 122–133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Rizzo, C.; Grasso, G.; Destro Castaniti, G.M.; Ciccia, F.; Guggino, G. Primary Sjogren Syndrome: Focus on Innate Immune Cells and Inflammation. Vaccines 2020, 8, 272. [Google Scholar] [CrossRef] [PubMed]
  26. Manoussakis, M.N.; Boiu, S.; Korkolopoulou, P.; Kapsogeorgou, E.K.; Kavantzas, N.; Ziakas, P.; Patsouris, E.; Moutsopoulos, H.M. Rates of infiltration by macrophages and dendritic cells and expression of interleukin-18 and interleukin-12 in the chronic inflammatory lesions of Sjogren’s syndrome: Correlation with certain features of immune hyperactivity and factors associated with high risk of lymphoma development. Arthritis Rheum. 2007, 56, 3977–3988. [Google Scholar] [PubMed]
  27. Kinoshita, S.; Nakamura, T.; Nishida, K. Pathological keratinization of ocular surface epithelium. Adv. Exp. Med. Biol. 2002, 506 Pt. A, 641–646. [Google Scholar]
  28. McNamara, N.A. Molecular mechanisms of keratinizing ocular surface disease. Optom Vis. Sci. 2010, 87, 233–238. [Google Scholar] [CrossRef]
  29. Conti, P.; Stellin, L.; Caraffa, A.; Gallenga, C.E.; Ross, R.; Kritas, S.K.; Frydas, I.; Younes, A.; Di Emidio, P.; Ronconi, G. Advances in Mast Cell Activation by IL-1 and IL-33 in Sjogren’s Syndrome: Promising Inhibitory Effect of IL-37. Int. J. Mol. Sci. 2020, 21, 4297. [Google Scholar] [CrossRef]
  30. Leehan, K.M.; Pezant, N.P.; Rasmussen, A.; Grundahl, K.; Moore, J.S.; Radfar, L.; Lewis, D.M.; Stone, D.U.; Lessard, C.J.; Rhodus, N.L.; et al. Minor salivary gland fibrosis in Sjogren’s syndrome is elevated, associated with focus score and not solely a consequence of aging. Clin. Exp. Rheumatol. 2018, 36 (Suppl. S112), 80–88. [Google Scholar]
  31. Skopouli, F.N.; Li, L.; Boumba, D.; Stefanaki, S.; Hanel, K.; Moutsopoulos, H.M.; Krilis, S.A. Association of mast cells with fibrosis and fatty infiltration in the minor salivary glands of patients with Sjogren’s syndrome. Clin. Exp. Rheumatol. 1998, 16, 63–65. [Google Scholar] [PubMed]
  32. Xu, D.; Jiang, H.R.; Kewin, P.; Li, Y.; Mu, R.; Fraser, A.R.; Pitman, N.; Kurowska-Stolarska, M.; McKenzie, A.N.; McInnes, I.B.; et al. IL-33 exacerbates antigen-induced arthritis by activating mast cells. Proc. Natl. Acad. Sci. USA 2008, 105, 10913–10918. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. de Paula, F.; Teshima, T.H.N.; Hsieh, R.; Souza, M.M.; Nico, M.M.S.; Lourenco, S.V. Overview of Human Salivary Glands: Highlights of Morphology and Developing Processes. Anat. Rec. 2017, 300, 1180–1188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Manoussakis, M.N.; Moutsopoulos, H.M. Sjogren’s syndrome: Autoimmune epithelitis. Baillieres Best Pract Res. Clin. Rheumatol. 2000, 14, 73–95. [Google Scholar] [CrossRef]
  35. Ewert, P.; Aguilera, S.; Alliende, C.; Kwon, Y.J.; Albornoz, A.; Molina, C.; Urzua, U.; Quest, A.F.; Olea, N.; Perez, P.; et al. Disruption of tight junction structure in salivary glands from Sjogren’s syndrome patients is linked to proinflammatory cytokine exposure. Arthritis Rheum. 2010, 62, 1280–1289. [Google Scholar] [CrossRef]
  36. Barrera, M.J.; Bahamondes, V.; Sepulveda, D.; Quest, A.F.; Castro, I.; Cortes, J.; Aguilera, S.; Urzua, U.; Molina, C.; Perez, P.; et al. Sjogren’s syndrome and the epithelial target: A comprehensive review. J. Autoimmun. 2013, 42, 7–18. [Google Scholar] [CrossRef]
  37. Manoussakis, M.N.; Kapsogeorgou, E.K. The role of intrinsic epithelial activation in the pathogenesis of Sjogren’s syndrome. J. Autoimmun. 2010, 35, 219–224. [Google Scholar] [CrossRef]
  38. Goules, A.V.; Kapsogeorgou, E.K.; Tzioufas, A.G. Insight into pathogenesis of Sjogren’s syndrome: Dissection on autoimmune infiltrates and epithelial cells. Clin. Immunol. 2017, 182, 30–40. [Google Scholar] [CrossRef]
  39. Hashimoto, A.; Nishikawa, T.; Hayashi, T.; Fujii, N.; Harada, K.; Oka, T.; Takahashi, K. The presence of free D-serine in rat brain. FEBS Lett. 1992, 296, 33–36. [Google Scholar] [CrossRef] [Green Version]
  40. Manoussakis, M.N.; Spachidou, M.P.; Maratheftis, C.I. Salivary epithelial cells from Sjogren’s syndrome patients are highly sensitive to anoikis induced by TLR-3 ligation. J. Autoimmun. 2010, 35, 212–218. [Google Scholar] [CrossRef]
  41. Kiripolsky, J.; Kramer, J.M. Current and Emerging Evidence for Toll-Like Receptor Activation in Sjogren’s Syndrome. J. Immunol. Res. 2018, 2018, 1246818. [Google Scholar] [CrossRef] [PubMed]
  42. 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] [PubMed] [Green Version]
  43. 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 Sjogren’s syndrome. Arthritis Rheum. 2002, 46, 2730–2741. [Google Scholar] [CrossRef] [PubMed]
  44. Manoussakis, M.N.; Kapsogeorgou, E.K. The role of epithelial cells in the pathogenesis of Sjogren’s syndrome. Clin. Rev. Allergy Immunol. 2007, 32, 225–230. [Google Scholar] [CrossRef] [PubMed]
  45. Boumba, D.; Skopouli, F.N.; Moutsopoulos, H.M. Cytokine mRNA expression in the labial salivary gland tissues from patients with primary Sjogren’s syndrome. Br. J. Rheumatol. 1995, 34, 326–333. [Google Scholar] [CrossRef]
  46. 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]
  47. Matsumura, R.; Umemiya, K.; Goto, T.; Nakazawa, T.; Kagami, M.; Tomioka, H.; Tanabe, E.; Sugiyama, T.; Sueishi, M. Glandular and extraglandular expression of costimulatory molecules in patients with Sjogren’s syndrome. Ann. Rheum Dis. 2001, 60, 473–482. [Google Scholar] [CrossRef]
  48. Kapsogeorgou, E.K.; Dimitriou, I.D.; Abu-Helu, R.F.; Moutsopoulos, H.M.; Manoussakis, M.N. Activation of epithelial and myoepithelial cells in the salivary glands of patients with Sjogren’s syndrome: High expression of intercellular adhesion molecule-1 (ICAM.1) in biopsy specimens and cultured cells. Clin. Exp. Immunol. 2001, 124, 126–133. [Google Scholar] [CrossRef]
  49. Mitsias, D.I.; Kapsogeorgou, E.K.; Moutsopoulos, H.M. The role of epithelial cells in the initiation and perpetuation of autoimmune lesions: Lessons from Sjogren’s syndrome (autoimmune epithelitis). Lupus 2006, 15, 255–261. [Google Scholar] [CrossRef]
  50. Pober, J.S.; Sessa, W.C. Evolving functions of endothelial cells in inflammation. Nat. Rev. Immunol. 2007, 7, 803–815. [Google Scholar] [CrossRef]
  51. Mikulowska-Mennis, A.; Xu, B.; Berberian, J.M.; Michie, S.A. Lymphocyte migration to inflamed lacrimal glands is mediated by vascular cell adhesion molecule-1/alpha(4)beta(1) integrin, peripheral node addressin/l-selectin, and lymphocyte function-associated antigen-1 adhesion pathways. Am. J. Pathol. 2001, 159, 671–681. [Google Scholar] [CrossRef]
  52. Turkcapar, N.; Sak, S.D.; Saatci, M.; Duman, M.; Olmez, U. Vasculitis and expression of vascular cell adhesion molecule-1, intercellular adhesion molecule-1, and E-selectin in salivary glands of patients with Sjogren’s syndrome. J. Rheumatol. 2005, 32, 1063–1070. [Google Scholar]
  53. Alunno, A.; Ibba-Manneschi, L.; Bistoni, O.; Rosa, I.; Caterbi, S.; Gerli, R.; Manetti, M. Mobilization of lymphatic endothelial precursor cells and lymphatic neovascularization in primary Sjogren’s syndrome. J. Cell Mol. Med. 2016, 20, 613–622. [Google Scholar] [CrossRef] [PubMed]
  54. Gunn, M.D.; Tangemann, K.; Tam, C.; Cyster, J.G.; Rosen, S.D.; Williams, L.T. A chemokine expressed in lymphoid high endothelial venules promotes the adhesion and chemotaxis of naive T lymphocytes. Proc. Natl. Acad. Sci. USA 1998, 95, 258–263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Toubal, A.; Nel, I.; Lotersztajn, S.; Lehuen, A. Mucosal-associated invariant T cells and disease. Nat. Rev. Immunol. 2019, 19, 643–657. [Google Scholar] [CrossRef] [PubMed]
  56. Gold, M.C.; McLaren, J.E.; Reistetter, J.A.; Smyk-Pearson, S.; Ladell, K.; Swarbrick, G.M.; Yu, Y.Y.; Hansen, T.H.; Lund, O.; Nielsen, M.; et al. MR1-restricted MAIT cells display ligand discrimination and pathogen selectivity through distinct T cell receptor usage. J. Exp. Med. 2014, 211, 1601–1610. [Google Scholar] [CrossRef] [PubMed]
  57. Wang, J.J.; Macardle, C.; Weedon, H.; Beroukas, D.; Banovic, T. Mucosal-associated invariant T cells are reduced and functionally immature in the peripheral blood of primary Sjogren’s syndrome patients. Eur. J. Immunol. 2016, 46, 2444–2453. [Google Scholar] [CrossRef]
  58. Izumi, Y.; Ida, H.; Huang, M.; Iwanaga, N.; Tanaka, F.; Aratake, K.; Arima, K.; Tamai, M.; Kamachi, M.; Nakamura, H.; et al. Characterization of peripheral natural killer cells in primary Sjogren’s syndrome: Impaired NK cell activity and low NK cell number. J. Lab. Clin. Med. 2006, 147, 242–249. [Google Scholar] [CrossRef]
  59. Ferlazzo, G.; Tsang, M.L.; Moretta, L.; Melioli, G.; Steinman, R.M.; Munz, C. Human dendritic cells activate resting natural killer (NK) cells and are recognized via the NKp30 receptor by activated NK cells. J. Exp. Med. 2002, 195, 343–351. [Google Scholar] [CrossRef]
  60. Rusakiewicz, S.; Nocturne, G.; Lazure, T.; Semeraro, M.; Flament, C.; Caillat-Zucman, S.; Sene, D.; Delahaye, N.; Vivier, E.; Chaba, K.; et al. NCR3/NKp30 contributes to pathogenesis in primary Sjogren’s syndrome. Sci. Transl. Med. 2013, 5, 195ra96. [Google Scholar] [CrossRef] [Green Version]
  61. 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] [PubMed]
  62. Bendelac, A.; Savage, P.B.; Teyton, L. The biology of NKT cells. Annu. Rev. Immunol. 2007, 25, 297–336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Yoshimoto, T. The Hunt for the Source of Primary Interleukin-4: How We Discovered That Natural Killer T Cells and Basophils Determine T Helper Type 2 Cell Differentiation In Vivo. Front. Immunol. 2018, 9, 716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Rizzo, C.; La Barbera, L.; Lo Pizzo, M.; Ciccia, F.; Sireci, G.; Guggino, G. Invariant NKT Cells and Rheumatic Disease: Focus on Primary Sjogren Syndrome. Int. J. Mol. Sci. 2019, 20, 5435. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Sudzius, G.; Mieliauskaite, D.; Siaurys, A.; Viliene, R.; Butrimiene, I.; Characiejus, D.; Dumalakiene, I. Distribution of Peripheral Lymphocyte Populations in Primary Sjogren’s Syndrome Patients. J. Immunol. Res. 2015, 2015, 854706. [Google Scholar] [CrossRef] [PubMed]
  66. Guggino, G.; Ciccia, F.; Raimondo, S.; Giardina, G.; Alessandro, R.; Dieli, F.; Sireci, G.; Triolo, G. Invariant NKT cells are expanded in peripheral blood but are undetectable in salivary glands of patients with primary Sjogren’s syndrome. Clin. Exp. Rheumatol. 2016, 34, 25–31. [Google Scholar]
  67. Yang, J.Q.; Wen, X.; Kim, P.J.; Singh, R.R. Invariant NKT cells inhibit autoreactive B cells in a contact- and CD1d-dependent manner. J. Immunol. 2011, 186, 1512–1520. [Google Scholar] [CrossRef] [Green Version]
  68. Zook, E.C.; Kee, B.L. Development of innate lymphoid cells. Nat. Immunol. 2016, 17, 775–782. [Google Scholar] [CrossRef]
  69. 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]
  70. Ebbo, M.; Crinier, A.; Vely, F.; Vivier, E. Innate lymphoid cells: Major players in inflammatory diseases. Nat. Rev. Immunol. 2017, 17, 665–678. [Google Scholar] [CrossRef]
  71. Shikhagaie, M.M.; Germar, K.; Bal, S.M.; Ros, X.R.; Spits, H. Innate lymphoid cells in autoimmunity: Emerging regulators in rheumatic diseases. Nat. Rev. Rheumatol. 2017, 13, 164–173. [Google Scholar] [CrossRef] [PubMed]
  72. Wenink, M.H.; Leijten, E.F.A.; Cupedo, T.; Radstake, T. Review: Innate Lymphoid Cells: Sparking Inflammatory Rheumatic Disease? Arthritis Rheumatol. 2017, 69, 885–897. [Google Scholar] [CrossRef] [PubMed]
  73. Pitzalis, C.; Jones, G.W.; Bombardieri, M.; Jones, S.A. Ectopic lymphoid-like structures in infection, cancer and autoimmunity. Nat. Rev. Immunol. 2014, 14, 447–462. [Google Scholar] [CrossRef] [PubMed]
  74. 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 Sjogren’s syndrome. Rheumatology 2019, 58, 1740–1745. [Google Scholar] [CrossRef] [PubMed]
  75. Maazi, H.; Banie, H.; Aleman Muench, G.R.; Patel, N.; Wang, B.; Sankaranarayanan, I.; Bhargava, V.; Sato, T.; Lewis, G.; Cesaroni, M.; et al. Activated plasmacytoid dendritic cells regulate type 2 innate lymphoid cell-mediated airway hyperreactivity. J. Allergy Clin. Immunol. 2018, 141, 893–905 e6. [Google Scholar] [CrossRef] [Green Version]
  76. Zhang, Z.; Cheng, L.; Zhao, J.; Li, G.; Zhang, L.; Chen, W.; Nie, W. Plasmacytoid dendritic cells promote HIV-1-induced group 3 innate lymphoid cell depletion. J. Clin. Invest. 2015, 125, 3692–3703. [Google Scholar] [CrossRef] [Green Version]
  77. Duerr, C.U.; McCarthy, C.D.; Mindt, B.C.; Rubio, M.; Meli, A.P.; Pothlichet, J.; Eva, M.M.; Gauchat, J.F.; Qureshi, S.T.; Mazer, B.D.; et al. Type I interferon restricts type 2 immunopathology through the regulation of group 2 innate lymphoid cells. Nat. Immunol. 2016, 17, 65–75. [Google Scholar] [CrossRef]
  78. Skopouli, F.N.; Fox, P.C.; Galanopoulou, V.; Atkinson, J.C.; Jaffe, E.S.; Moutsopoulos, H.M. T cell subpopulations in the labial minor salivary gland histopathologic lesion of Sjogren’s syndrome. J. Rheumatol. 1991, 18, 210–214. [Google Scholar]
  79. Hooks, J.J.; Moutsopoulos, H.M.; Geis, S.A.; Stahl, N.I.; Decker, J.L.; Notkins, A.L. Immune interferon in the circulation of patients with autoimmune disease. N. Engl. J. Med. 1979, 301, 5–8. [Google Scholar] [CrossRef]
  80. Youinou, P.; Pers, J.O. Disturbance of cytokine networks in Sjogren’s syndrome. Arthritis Res. Ther. 2011, 13, 227. [Google Scholar] [CrossRef] [Green Version]
  81. 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 Sjogren’s syndrome compared with non-Sjogren’s sicca syndrome. Ann. Rheum Dis. 2005, 64, 1474–1479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Mosmann, T.R.; Coffman, R.L. TH1 and TH2 cells: Different patterns of lymphokine secretion lead to different functional properties. Annu. Rev. Immunol. 1989, 7, 145–173. [Google Scholar] [CrossRef] [PubMed]
  83. Lee, J.S.; Tato, C.M.; Joyce-Shaikh, B.; Gulen, M.F.; Cayatte, C.; Chen, Y.; Blumenschein, W.M.; Judo, M.; Ayanoglu, G.; McClanahan, T.K.; et al. Interleukin-23-Independent IL-17 Production Regulates Intestinal Epithelial Permeability. Immunity 2015, 43, 727–738. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Dudakov, J.A.; Hanash, A.M.; van den Brink, M.R. Interleukin-22: Immunobiology and pathology. Annu. Rev. Immunol. 2015, 33, 747–785. [Google Scholar] [CrossRef] [Green Version]
  85. Abusleme, L.; Moutsopoulos, N.M. IL-17: Overview and role in oral immunity and microbiome. Oral Dis. 2017, 23, 854–865. [Google Scholar] [CrossRef]
  86. Curtis, M.M.; Way, S.S. Interleukin-17 in host defence against bacterial, mycobacterial and fungal pathogens. Immunology 2009, 126, 177–185. [Google Scholar] [CrossRef]
  87. Khader, S.A.; Gaffen, S.L.; Kolls, J.K. Th17 cells at the crossroads of innate and adaptive immunity against infectious diseases at the mucosa. Mucosal Immunol. 2009, 2, 403–411. [Google Scholar] [CrossRef] [Green Version]
  88. Ivanov, I.I.; McKenzie, B.S.; Zhou, L.; Tadokoro, C.E.; Lepelley, A.; Lafaille, J.J.; Cua, D.J.; Littman, D.R. The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell 2006, 126, 1121–1133. [Google Scholar] [CrossRef] [Green Version]
  89. Dong, C. Genetic controls of Th17 cell differentiation and plasticity. Exp. Mol. Med. 2011, 43, 1–6. [Google Scholar] [CrossRef] [Green Version]
  90. Yang, X.O.; Pappu, B.P.; Nurieva, R.; Akimzhanov, A.; Kang, H.S.; Chung, Y.; Ma, L.; Shah, B.; Panopoulos, A.D.; Schluns, K.S.; et al. T helper 17 lineage differentiation is programmed by orphan nuclear receptors ROR alpha and ROR gamma. Immunity 2008, 28, 29–39. [Google Scholar] [CrossRef] [Green Version]
  91. Kuestner, R.E.; Taft, D.W.; Haran, A.; Brandt, C.S.; Brender, T.; Lum, K.; Harder, B.; Okada, S.; Ostrander, C.D.; Kreindler, J.L.; et al. Identification of the IL-17 receptor related molecule IL-17RC as the receptor for IL-17F. J. Immunol. 2007, 179, 5462–5473. [Google Scholar] [CrossRef] [PubMed]
  92. 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] [PubMed] [Green Version]
  93. Liu, R.; Gao, C.; Chen, H.; Li, Y.; Jin, Y.; Qi, H. Analysis of Th17-associated cytokines and clinical correlations in patients with dry eye disease. PLoS ONE 2017, 12, e0173301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Nguyen, C.Q.; Hu, M.H.; Li, Y.; Stewart, C.; Peck, A.B. Salivary gland tissue expression of interleukin-23 and interleukin-17 in Sjogren’s syndrome: Findings in humans and mice. Arthritis Rheum. 2008, 58, 734–743. [Google Scholar] [CrossRef] [Green Version]
  95. 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]
  96. Goenka, R.; Barnett, L.G.; Silver, J.S.; O’Neill, P.J.; Hunter, C.A.; Cancro, M.P.; Laufer, T.M. Cutting edge: Dendritic cell-restricted antigen presentation initiates the follicular helper T cell program but cannot complete ultimate effector differentiation. J. Immunol. 2011, 187, 1091–1095. [Google Scholar] [CrossRef] [Green Version]
  97. Chen, M.; Guo, Z.; Ju, W.; Ryffel, B.; He, X.; Zheng, S.G. The development and function of follicular helper T cells in immune responses. Cell. Mol. Immunol. 2012, 9, 375–379. [Google Scholar] [CrossRef] [Green Version]
  98. Kim, C.H.; Rott, L.S.; Clark-Lewis, I.; Campbell, D.J.; Wu, L.; Butcher, E.C. Subspecialization of CXCR5+ T cells: B helper activity is focused in a germinal center-localized subset of CXCR5+ T cells. J. Exp. Med. 2001, 193, 1373–1381. [Google Scholar] [CrossRef] [Green Version]
  99. Arnold, C.N.; Campbell, D.J.; Lipp, M.; Butcher, E.C. The germinal center response is impaired in the absence of T cell-expressed CXCR5. Eur. J. Immunol. 2007, 37, 100–109. [Google Scholar] [CrossRef]
  100. Hardtke, S.; Ohl, L.; Forster, R. Balanced expression of CXCR5 and CCR7 on follicular T helper cells determines their transient positioning to lymph node follicles and is essential for efficient B-cell help. Blood 2005, 106, 1924–1931. [Google Scholar] [CrossRef] [Green Version]
  101. Haynes, N.M.; Allen, C.D.; Lesley, R.; Ansel, K.M.; Killeen, N.; Cyster, J.G. Role of CXCR5 and CCR7 in follicular Th cell positioning and appearance of a programmed cell death gene-1high germinal center-associated subpopulation. J. Immunol. 2007, 179, 5099–5108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Crotty, S. Follicular helper CD4 T cells (TFH). Annu. Rev. Immunol. 2011, 29, 621–663. [Google Scholar] [CrossRef] [PubMed]
  103. 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] [PubMed] [Green Version]
  104. Jin, L.; Yu, D.; Li, X.; Yu, N.; Li, X.; Wang, Y.; Wang, Y. CD4+CXCR5+ follicular helper T cells in salivary gland promote B cells maturation in patients with primary Sjogren’s syndrome. Int. J. Clin. Exp. Pathol. 2014, 7, 1988–1996. [Google Scholar] [PubMed]
  105. 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] [PubMed]
  106. Cosorich, I.; McGuire, H.M.; Warren, J.; Danta, M.; King, C. CCR9 Expressing T Helper and T Follicular Helper Cells Exhibit Site-Specific Identities During Inflammatory Disease. Front. Immunol. 2018, 9, 2899. [Google Scholar] [CrossRef] [Green Version]
  107. 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]
  108. Blokland, S.L.M.; Hillen, M.R.; Kruize, A.A.; Meller, S.; Homey, B.; Smithson, G.M.; Radstake, T.; van Roon, J.A.G. Increased CCL25 and T Helper Cells Expressing CCR9 in the Salivary Glands of Patients with Primary Sjogren’s Syndrome: Potential New Axis in Lymphoid Neogenesis. Arthritis Rheumatol. 2017, 69, 2038–2051. [Google Scholar] [CrossRef] [Green Version]
  109. Schwartz, R.H. Natural regulatory T cells and self-tolerance. Nat. Immunol. 2005, 6, 327–330. [Google Scholar] [CrossRef]
  110. von Boehmer, H. Mechanisms of suppression by suppressor T cells. Nat. Immunol. 2005, 6, 338–344. [Google Scholar] [CrossRef]
  111. Sakaguchi, S. Naturally arising Foxp3-expressing CD25+CD4+ regulatory T cells in immunological tolerance to self and non-self. Nat. Immunol. 2005, 6, 345–352. [Google Scholar] [CrossRef] [PubMed]
  112. Lan, R.Y.; Ansari, A.A.; Lian, Z.X.; Gershwin, M.E. Regulatory T cells: Development, function and role in autoimmunity. Autoimmun. Rev. 2005, 4, 351–363. [Google Scholar] [CrossRef] [PubMed]
  113. Wraith, D.C.; Nicolson, K.S.; Whitley, N.T. Regulatory CD4+ T cells and the control of autoimmune disease. Curr. Opin. Immunol. 2004, 16, 695–701. [Google Scholar] [CrossRef] [PubMed]
  114. Kim, J.M.; Rasmussen, J.P.; Rudensky, A.Y. Regulatory T cells prevent catastrophic autoimmunity throughout the lifespan of mice. Nat. Immunol. 2007, 8, 191–197. [Google Scholar] [CrossRef] [PubMed]
  115. Noack, M.; Miossec, P. Th17 and regulatory T cell balance in autoimmune and inflammatory diseases. Autoimmun. Rev. 2014, 13, 668–677. [Google Scholar] [CrossRef] [PubMed]
  116. Gottenberg, J.E.; Lavie, F.; Abbed, K.; Gasnault, J.; Le Nevot, E.; Delfraissy, J.F.; Taoufik, Y.; Mariette, X. CD4 CD25high regulatory T cells are not impaired in patients with primary Sjogren’s syndrome. J. Autoimmun. 2005, 24, 235–242. [Google Scholar] [CrossRef]
  117. Li, X.; Li, X.; Qian, L.; Wang, G.; Zhang, H.; Wang, X.; Chen, K.; Zhai, Z.; Li, Q.; Wang, Y.; et al. T regulatory cells are markedly diminished in diseased salivary glands of patients with primary Sjogren’s syndrome. J. Rheumatol. 2007, 34, 2438–2445. [Google Scholar]
  118. Liu, M.F.; Lin, L.H.; Weng, C.T.; Weng, M.Y. Decreased CD4+CD25+bright T cells in peripheral blood of patients with primary Sjogren’s syndrome. Lupus 2008, 17, 34–39. [Google Scholar] [CrossRef]
  119. 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] [Green Version]
  120. Szodoray, P.; Papp, G.; Horvath, I.F.; Barath, S.; Sipka, S.; Nakken, B.; Zeher, M. Cells with regulatory function of the innate and adaptive immune system in primary Sjogren’s syndrome. Clin. Exp. Immunol. 2009, 157, 343–349. [Google Scholar] [CrossRef]
  121. Sarigul, M.; Yazisiz, V.; Bassorgun, C.I.; Ulker, M.; Avci, A.B.; Erbasan, F.; Gelen, T.; Gorczynski, R.M.; Terzioglu, E. The numbers of Foxp3 + Treg cells are positively correlated with higher grade of infiltration at the salivary glands in primary Sjogren’s syndrome. Lupus 2010, 19, 138–145. [Google Scholar] [CrossRef] [PubMed]
  122. Furuzawa-Carballeda, J.; Hernandez-Molina, G.; Lima, G.; Rivera-Vicencio, Y.; Ferez-Blando, K.; Llorente, L. Peripheral regulatory cells immunophenotyping in primary Sjogren’s syndrome: A cross-sectional study. Arthritis Res. Ther. 2013, 15, R68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Alunno, A.; Petrillo, M.G.; Nocentini, G.; Bistoni, O.; Bartoloni, E.; Caterbi, S.; Bianchini, R.; Baldini, C.; Nicoletti, I.; Riccardi, C.; et al. Characterization of a new regulatory CD4+ T cell subset in primary Sjogren’s syndrome. Rheumatology 2013, 52, 1387–1396. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Alunno, A.; Nocentini, G.; Bistoni, O.; Petrillo, M.G.; Bartoloni Bocci, E.; Ronchetti, S.; Lo Vaglio, E.; Riccardi, C.; Gerli, R. Expansion of CD4+CD25-GITR+ regulatory T-cell subset in the peripheral blood of patients with primary Sjogren’s syndrome: Correlation with disease activity. Reumatismo 2012, 64, 293–298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Linterman, M.A.; Pierson, W.; Lee, S.K.; Kallies, A.; Kawamoto, S.; Rayner, T.F.; Srivastava, M.; Divekar, D.P.; Beaton, L.; Hogan, J.J.; et al. Foxp3+ follicular regulatory T cells control the germinal center response. Nat. Med. 2011, 17, 975–982. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Wollenberg, I.; Agua-Doce, A.; Hernandez, A.; Almeida, C.; Oliveira, V.G.; Faro, J.; Graca, L. Regulation of the germinal center reaction by Foxp3+ follicular regulatory T cells. J. Immunol. 2011, 187, 4553–4560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Chung, Y.; Tanaka, S.; Chu, F.; Nurieva, R.I.; Martinez, G.J.; Rawal, S.; Wang, Y.H.; Lim, H.; Reynolds, J.M.; Zhou, X.H.; et al. Follicular regulatory T cells expressing Foxp3 and Bcl-6 suppress germinal center reactions. Nat. Med. 2011, 17, 983–988. [Google Scholar] [CrossRef]
  128. Gerner, M.Y.; Torabi-Parizi, P.; Germain, R.N. Strategically localized dendritic cells promote rapid T cell responses to lymph-borne particulate antigens. Immunity 2015, 42, 172–185. [Google Scholar] [CrossRef] [Green Version]
  129. Kerfoot, S.M.; Yaari, G.; Patel, J.R.; Johnson, K.L.; Gonzalez, D.G.; Kleinstein, S.H.; Haberman, A.M. Germinal center B cell and T follicular helper cell development initiates in the interfollicular zone. Immunity 2011, 34, 947–960. [Google Scholar] [CrossRef] [Green Version]
  130. 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] [Green Version]
  131. Vaeth, M.; Eckstein, M.; Shaw, P.J.; Kozhaya, L.; Yang, J.; Berberich-Siebelt, F.; Clancy, R.; Unutmaz, D.; Feske, S. Store-Operated Ca(2+) Entry in Follicular T Cells Controls Humoral Immune Responses and Autoimmunity. Immunity 2016, 44, 1350–1364. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Sage, P.T.; Alvarez, D.; Godec, J.; von Andrian, U.H.; Sharpe, A.H. Circulating T follicular regulatory and helper cells have memory-like properties. J. Clin. Invest. 2014, 124, 5191–5204. [Google Scholar] [CrossRef] [PubMed]
  133. Fu, W.; Liu, X.; Lin, X.; Feng, H.; Sun, L.; Li, S.; Chen, H.; Tang, H.; Lu, L.; Jin, W.; et al. Deficiency in T follicular regulatory cells promotes autoimmunity. J. Exp. Med. 2018, 215, 815–825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. 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 Sjogren’s Syndrome? Comment on the Article by Fonseca et al. Arthritis Rheumatol. 2018, 70, 1354–1355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Fonseca, V.R.; Romao, V.C.; Agua-Doce, A.; Santos, M.; Lopez-Presa, D.; Ferreira, A.C.; Fonseca, J.E.; Graca, L. The Ratio of Blood T Follicular Regulatory Cells to T Follicular Helper Cells Marks Ectopic Lymphoid Structure Formation While Activated Follicular Helper T Cells Indicate Disease Activity in Primary Sjogren’s Syndrome. Arthritis Rheumatol. 2018, 70, 774–784. [Google Scholar] [CrossRef] [PubMed]
  136. Tasaki, S.; Suzuki, K.; Nishikawa, A.; Kassai, Y.; Takiguchi, M.; Kurisu, R.; Okuzono, Y.; Miyazaki, T.; Takeshita, M.; Yoshimoto, K.; et al. Multiomic disease signatures converge to cytotoxic CD8 T cells in primary Sjogren’s syndrome. Ann. Rheum Dis. 2017, 76, 1458–1466. [Google Scholar] [CrossRef] [Green Version]
  137. Zhou, J.; Yu, Q. Disruption of CXCR3 function impedes the development of Sjogren’s syndrome-like xerostomia in non-obese diabetic mice. Lab. Invest. 2018, 98, 620–628. [Google Scholar] [CrossRef] [Green Version]
  138. Barr, J.Y.; Wang, X.; Meyerholz, D.K.; Lieberman, S.M. CD8 T cells contribute to lacrimal gland pathology in the nonobese diabetic mouse model of Sjogren syndrome. Immunol. Cell Biol. 2017, 95, 684–694. [Google Scholar] [CrossRef] [Green Version]
  139. Brandt, D.; Hedrich, C.M. TCRalphabeta(+)CD3(+)CD4(-)CD8(-) (double negative) T cells in autoimmunity. Autoimmun. Rev. 2018, 17, 422–430. [Google Scholar] [CrossRef]
  140. Gao, Y.; Williams, A.P. Role of Innate T Cells in Anti-Bacterial Immunity. Front Immunol. 2015, 6, 302. [Google Scholar] [CrossRef] [Green Version]
  141. Hansen, A.; Lipsky, P.E.; Dorner, T. B cells in Sjogren’s syndrome: Indications for disturbed selection and differentiation in ectopic lymphoid tissue. Arthritis Res. Ther. 2007, 9, 218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Hansen, A.; Reiter, K.; Ziprian, T.; Jacobi, A.; Hoffmann, A.; Gosemann, M.; Scholze, J.; Lipsky, P.E.; Dorner, T. Dysregulation of chemokine receptor expression and function by B cells of patients with primary Sjogren’s syndrome. Arthritis Rheum. 2005, 52, 2109–2119. [Google Scholar] [CrossRef] [PubMed]
  143. Kroese, F.G.; Abdulahad, W.H.; Haacke, E.; Bos, N.A.; Vissink, A.; Bootsma, H. B-cell hyperactivity in primary Sjogren’s syndrome. Expert Rev. Clin. Immunol. 2014, 10, 483–499. [Google Scholar] [CrossRef] [PubMed]
  144. Scheid, J.F.; Mouquet, H.; Kofer, J.; Yurasov, S.; Nussenzweig, M.C.; Wardemann, H. Differential regulation of self-reactivity discriminates between IgG+ human circulating memory B cells and bone marrow plasma cells. Proc. Natl. Acad. Sci. USA 2011, 108, 18044–18048. [Google Scholar] [CrossRef] [Green Version]
  145. Mouquet, H.; Nussenzweig, M.C. Polyreactive antibodies in adaptive immune responses to viruses. Cell. Mol. Life Sci. 2012, 69, 1435–1445. [Google Scholar] [CrossRef]
  146. Hayakawa, I.; Tedder, T.F.; Zhuang, Y. B-lymphocyte depletion ameliorates Sjogren’s syndrome in Id3 knockout mice. Immunology 2007, 122, 73–79. [Google Scholar] [CrossRef]
  147. Nocturne, G.; Mariette, X. Advances in understanding the pathogenesis of primary Sjogren’s syndrome. Nat. Rev. Rheumatol. 2013, 9, 544–556. [Google Scholar] [CrossRef]
  148. Puga, I.; Cols, M.; Barra, C.M.; He, B.; Cassis, L.; Gentile, M.; Comerma, L.; Chorny, A.; Shan, M.; Xu, W.; et al. B cell-helper neutrophils stimulate the diversification and production of immunoglobulin in the marginal zone of the spleen. Nat. Immunol. 2011, 13, 170–180. [Google Scholar] [CrossRef] [Green Version]
  149. Szabo, K.; Papp, G.; Szanto, A.; Tarr, T.; Zeher, M. A comprehensive investigation on the distribution of circulating follicular T helper cells and B cell subsets in primary Sjogren’s syndrome and systemic lupus erythematosus. Clin. Exp. Immunol. 2016, 183, 76–89. [Google Scholar] [CrossRef] [Green Version]
  150. Corneth, O.B.J.; Verstappen, G.M.P.; Paulissen, S.M.J.; de Bruijn, M.J.W.; Rip, J.; Lukkes, M.; van Hamburg, J.P.; Lubberts, E.; Bootsma, H.; Kroese, F.G.M.; et al. Enhanced Bruton’s Tyrosine Kinase Activity in Peripheral Blood B Lymphocytes from Patients with Autoimmune Disease. Arthritis Rheumatol. 2017, 69, 1313–1324. [Google Scholar] [CrossRef]
  151. Hansen, A.; Odendahl, M.; Reiter, K.; Jacobi, A.M.; Feist, E.; Scholze, J.; Burmester, G.R.; Lipsky, P.E.; Dorner, T. Diminished peripheral blood memory B cells and accumulation of memory B cells in the salivary glands of patients with Sjogren’s syndrome. Arthritis Rheum. 2002, 46, 2160–2171. [Google Scholar] [CrossRef] [PubMed]
  152. 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.e12. [Google Scholar] [CrossRef] [PubMed]
  153. Daridon, C.; Pers, J.O.; Devauchelle, V.; Martins-Carvalho, C.; Hutin, P.; Pennec, Y.L.; Saraux, A.; Youinou, P. Identification of transitional type II B cells in the salivary glands of patients with Sjogren’s syndrome. Arthritis Rheum. 2006, 54, 2280–2288. [Google Scholar] [CrossRef]
  154. Fletcher, C.A.; Sutherland, A.P.; Groom, J.R.; Batten, M.L.; Ng, L.G.; Gommerman, J.; Mackay, F. Development of nephritis but not sialadenitis in autoimmune-prone BAFF transgenic mice lacking marginal zone B cells. Eur. J. Immunol. 2006, 36, 2504–2514. [Google Scholar] [CrossRef] [PubMed]
  155. Nguyen, C.Q.; Kim, H.; Cornelius, J.G.; Peck, A.B. Development of Sjogren’s syndrome in nonobese diabetic-derived autoimmune-prone C57BL/6.NOD-Aec1Aec2 mice is dependent on complement component-3. J. Immunol. 2007, 179, 2318–2329. [Google Scholar] [CrossRef] [Green Version]
  156. Shen, L.; Zhang, C.; Wang, T.; Brooks, S.; Ford, R.J.; Lin-Lee, Y.C.; Kasianowicz, A.; Kumar, V.; Martin, L.; Liang, P.; et al. Development of autoimmunity in IL-14alpha-transgenic mice. J. Immunol. 2006, 177, 5676–5686. [Google Scholar] [CrossRef] [Green Version]
  157. Shen, L.; Gao, C.; Suresh, L.; Xian, Z.; Song, N.; Chaves, L.D.; Yu, M.; Ambrus, J.L., Jr. Central role for marginal zone B cells in an animal model of Sjogren’s syndrome. Clin. Immunol. 2016, 168, 30–36. [Google Scholar] [CrossRef] [Green Version]
  158. Nocturne, G.; Mariette, X. Sjogren Syndrome-associated lymphomas: An update on pathogenesis and management. Br. J. Haematol. 2015, 168, 317–327. [Google Scholar] [CrossRef]
  159. Fogel, O.; Riviere, E.; Seror, R.; Nocturne, G.; Boudaoud, S.; Ly, B.; Gottenberg, J.E.; Le Guern, V.; Dubost, J.J.; Nititham, J.; et al. Role of the IL-12/IL-35 balance in patients with Sjogren syndrome. J. Allergy Clin. Immunol. 2018, 142, 258–268.e5. [Google Scholar] [CrossRef] [Green Version]
  160. Mariette, X.; Roux, S.; Zhang, J.; Bengoufa, D.; Lavie, F.; Zhou, T.; Kimberly, R. The level of BLyS (BAFF) correlates with the titre of autoantibodies in human Sjogren’s syndrome. Ann. Rheum. Dis. 2003, 62, 168–171. [Google Scholar] [CrossRef]
  161. Goenka, R.; Scholz, J.L.; Sindhava, V.J.; Cancro, M.P. New roles for the BLyS/BAFF family in antigen-experienced B cell niches. Cytokine Growth Factor Rev. 2014, 25, 107–113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Ding, J.; Zhang, W.; Haskett, S.; Pellerin, A.; Xu, S.; Petersen, B.; Jandreski, L.; Hamann, S.; Reynolds, T.L.; Zheng, T.S.; et al. BAFF overexpression increases lymphocytic infiltration in Sjogren’s target tissue, but only inefficiently promotes ectopic B-cell differentiation. Clin. Immunol. 2016, 169, 69–79. [Google Scholar] [CrossRef] [PubMed]
  163. Risselada, A.P.; Looije, M.F.; Kruize, A.A.; Bijlsma, J.W.; van Roon, J.A. The role of ectopic germinal centers in the immunopathology of primary Sjogren’s syndrome: A systematic review. Semin. Arthritis Rheum. 2013, 42, 368–376. [Google Scholar] [CrossRef] [PubMed]
  164. Bombardieri, M.; Lewis, M.; Pitzalis, C. Ectopic lymphoid neogenesis in rheumatic autoimmune diseases. Nat. Rev. Rheumatol. 2017, 13, 141–154. [Google Scholar] [CrossRef]
Figure 1. Innate immunity in Sjögren’s syndrome. SGECs constitute the main target of auto-immunity in pSS, described as an autoimmune epithelitis. SGECs exhibit a subverted architecture mainly characterized by altered tight junctions. In genetic susceptible subjects, environmental stimuli such as viruses may trigger salivary gland epithelial cells (SGECs) through TLR activation. Activated SGECs secrete the BAFF that promotes activation and maturation of B cells. SGECs also produce chemokines such as CXCR9, 10, 11, and 12 that attract immune cells and contribute to the formation of germinal centers. Activated SGECs have the ability to act as non-professional antigen-presenting cells by expressing MHC-I, (HLA-ABC) and MHC-II (HLA-DR), adhesion molecules such as ICAM1 allowing them to activate T cells. TLR activation also contributes to SGEC apoptosis, releasing autoantigens that drive autoimmunity in pSS. Activated macrophages produce inflammatory cytokines such as IL-1, TNFα, and MMPs leading to epithelial cell damage. They can also act as antigenic peptide presenting cells through their MHC-II and interact with antigen-specific CD4+ T cells. pDCs lead to the production of type I IFN that acts through autocrine and paracrine circuits feeding a continuous reinforcing inflammatory loop. It also induces the production of BAFF, production contributing to the activation of B cells into plasma cells. fDCs play an essential part in the structure of ectopic germinal centers and retain on their surface immune-complexes, formed by antigen-antibody-complement. Mast cells contribute to the fibrosis and fatty infiltration of salivary glands (SGs). The aberrant phenotype of MAIT cells in pSS patients may lead to the dysregulation of the local immune responses, which would trigger local damage in SGs and auto-immunity. NK cells express the NKp30 receptor that is recognized by DCs and lead to the production of Th1 cytokines such as IFN-γ and IL-12. SGECs express B7-H6, the ligand for NKp30. Taken together, this may explain the hyperactivity of NK cells and the cross-talk with SGECs and DCs that lead to a subsequent activation of innate and adaptive immunity. A subset of ILC3 was found to be a major source of IL-22 in SGECs. Abbreviations: APC: Antigen presenting cells; BAFF: B-cell activating factor; CXCL9: C-X-C motif chemokine type 9; CXCL10: C-X-C motif chemokine type 10; CXCL12: C-X-C motif chemokine type 12; CXCL13: C-X-C motif chemokine type 13; DCs: dendritic cells; fDCs: follicular dendritic cells; ICAM-1: intercellular adhesion molecule 1; IFN-γ: interferon gamma; IL-: interleukin; ILC3: innate Lymphoid Cells type 3 MAIT: Mucosal-associated invariant T cells; MHC-I: major histocompatibility complex class I; MHC-II: major histocompatibility complex class II; MMPs: metalloproteases; NK: natural killer cells; NKp44L: NKp44 ligand; pDCs: plasmacytoid dendritic cells; SGECs: salivary glands epithelial cells; TCR: T cell receptor; TLR: Toll like receptor; TNFα: tumor necrosis factor alpha; VEGF-C: vascular endothelial growth factor C.
Figure 1. Innate immunity in Sjögren’s syndrome. SGECs constitute the main target of auto-immunity in pSS, described as an autoimmune epithelitis. SGECs exhibit a subverted architecture mainly characterized by altered tight junctions. In genetic susceptible subjects, environmental stimuli such as viruses may trigger salivary gland epithelial cells (SGECs) through TLR activation. Activated SGECs secrete the BAFF that promotes activation and maturation of B cells. SGECs also produce chemokines such as CXCR9, 10, 11, and 12 that attract immune cells and contribute to the formation of germinal centers. Activated SGECs have the ability to act as non-professional antigen-presenting cells by expressing MHC-I, (HLA-ABC) and MHC-II (HLA-DR), adhesion molecules such as ICAM1 allowing them to activate T cells. TLR activation also contributes to SGEC apoptosis, releasing autoantigens that drive autoimmunity in pSS. Activated macrophages produce inflammatory cytokines such as IL-1, TNFα, and MMPs leading to epithelial cell damage. They can also act as antigenic peptide presenting cells through their MHC-II and interact with antigen-specific CD4+ T cells. pDCs lead to the production of type I IFN that acts through autocrine and paracrine circuits feeding a continuous reinforcing inflammatory loop. It also induces the production of BAFF, production contributing to the activation of B cells into plasma cells. fDCs play an essential part in the structure of ectopic germinal centers and retain on their surface immune-complexes, formed by antigen-antibody-complement. Mast cells contribute to the fibrosis and fatty infiltration of salivary glands (SGs). The aberrant phenotype of MAIT cells in pSS patients may lead to the dysregulation of the local immune responses, which would trigger local damage in SGs and auto-immunity. NK cells express the NKp30 receptor that is recognized by DCs and lead to the production of Th1 cytokines such as IFN-γ and IL-12. SGECs express B7-H6, the ligand for NKp30. Taken together, this may explain the hyperactivity of NK cells and the cross-talk with SGECs and DCs that lead to a subsequent activation of innate and adaptive immunity. A subset of ILC3 was found to be a major source of IL-22 in SGECs. Abbreviations: APC: Antigen presenting cells; BAFF: B-cell activating factor; CXCL9: C-X-C motif chemokine type 9; CXCL10: C-X-C motif chemokine type 10; CXCL12: C-X-C motif chemokine type 12; CXCL13: C-X-C motif chemokine type 13; DCs: dendritic cells; fDCs: follicular dendritic cells; ICAM-1: intercellular adhesion molecule 1; IFN-γ: interferon gamma; IL-: interleukin; ILC3: innate Lymphoid Cells type 3 MAIT: Mucosal-associated invariant T cells; MHC-I: major histocompatibility complex class I; MHC-II: major histocompatibility complex class II; MMPs: metalloproteases; NK: natural killer cells; NKp44L: NKp44 ligand; pDCs: plasmacytoid dendritic cells; SGECs: salivary glands epithelial cells; TCR: T cell receptor; TLR: Toll like receptor; TNFα: tumor necrosis factor alpha; VEGF-C: vascular endothelial growth factor C.
Ijms 22 00658 g001
Figure 2. Th1-Th2 imbalance. Upon T-cell activation, IFN-γ, and IL-12 induce the expression of T-bet and STAT-4, which is involved in the differentiation of naïve CD4+ T cells into Th1 lymphocytes. Th1 cells predominantly produce pro-inflammatory cytokines such as IFN-γ and IL-2. In contrast, IL-4 induces the GATA-3 transcription factor and the consequent polarization of naïve T cells into Th2. Th2 cells produce anti-inflammatory cytokines such as IL-4, IL-5, and IL-13. Several studies have suggested that pSS is related to abnormal Th1 activation and SGs infiltration. It is supported by the presence of elevated levels of IFN-γ in serum and Th1 cells in blood. Furthermore, T cells expressing a high level of IFN-γ and STAT-4 mRNA have been found in SGs from pSS patients. This Th1/Th2 imbalance, generally observed in various chronic inflammatory disorders, is not easily understood because of a limited number of studies. Abbreviations: IFN-γ:interferon gamma; IL-: interleukin; pSS: primary Sjögren’s syndrome; STAT: signal transducer and activator of transcription; T-bet: T-Box Transcription Factor 21; Th1: type 1 helper cells; Th2: type 2 helper cells.
Figure 2. Th1-Th2 imbalance. Upon T-cell activation, IFN-γ, and IL-12 induce the expression of T-bet and STAT-4, which is involved in the differentiation of naïve CD4+ T cells into Th1 lymphocytes. Th1 cells predominantly produce pro-inflammatory cytokines such as IFN-γ and IL-2. In contrast, IL-4 induces the GATA-3 transcription factor and the consequent polarization of naïve T cells into Th2. Th2 cells produce anti-inflammatory cytokines such as IL-4, IL-5, and IL-13. Several studies have suggested that pSS is related to abnormal Th1 activation and SGs infiltration. It is supported by the presence of elevated levels of IFN-γ in serum and Th1 cells in blood. Furthermore, T cells expressing a high level of IFN-γ and STAT-4 mRNA have been found in SGs from pSS patients. This Th1/Th2 imbalance, generally observed in various chronic inflammatory disorders, is not easily understood because of a limited number of studies. Abbreviations: IFN-γ:interferon gamma; IL-: interleukin; pSS: primary Sjögren’s syndrome; STAT: signal transducer and activator of transcription; T-bet: T-Box Transcription Factor 21; Th1: type 1 helper cells; Th2: type 2 helper cells.
Ijms 22 00658 g002
Figure 3. Innate and adaptive crosstalk. Activated SGECs secrete BAFF that promotes the activation and maturation of B cells into long-lasting memory B cells and plasma B cells producing auto-antibodies. SGECs also produce chemokines IL-1, IL-6, IL18, and TNFα that attract immune cells and contribute to the formation of germinal centers. Activated SGECs have the ability to act as non-professional antigen-presenting cells by expressing MHC-I (HLA-ABC) and MHC-II (HLA-DR) adhesion molecules such as ICAM1, allowing them to activate T cells. TLR activation also contributes to SGEC apoptosis, releasing autoantigens that drive autoimmunity in pSS. Activated macrophages can act as antigenic peptide presenting cells through their MHC-II and interact with antigen-specific CD4+ T cells. pDCs lead to the production of type I IFN that acts through autocrine and paracrine circuits feeding a continuous reinforcing inflammatory loop. It also induces BAFF production, contributing to the activation of B cells into plasma cells. DCs also play an essential part in the structure of ectopic germinal centers and retain on their surface immune-complexes, formed by antigen-antibody-complement.
Figure 3. Innate and adaptive crosstalk. Activated SGECs secrete BAFF that promotes the activation and maturation of B cells into long-lasting memory B cells and plasma B cells producing auto-antibodies. SGECs also produce chemokines IL-1, IL-6, IL18, and TNFα that attract immune cells and contribute to the formation of germinal centers. Activated SGECs have the ability to act as non-professional antigen-presenting cells by expressing MHC-I (HLA-ABC) and MHC-II (HLA-DR) adhesion molecules such as ICAM1, allowing them to activate T cells. TLR activation also contributes to SGEC apoptosis, releasing autoantigens that drive autoimmunity in pSS. Activated macrophages can act as antigenic peptide presenting cells through their MHC-II and interact with antigen-specific CD4+ T cells. pDCs lead to the production of type I IFN that acts through autocrine and paracrine circuits feeding a continuous reinforcing inflammatory loop. It also induces BAFF production, contributing to the activation of B cells into plasma cells. DCs also play an essential part in the structure of ectopic germinal centers and retain on their surface immune-complexes, formed by antigen-antibody-complement.
Ijms 22 00658 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Chivasso, C.; Sarrand, J.; Perret, J.; Delporte, C.; Soyfoo, M.S. The Involvement of Innate and Adaptive Immunity in the Initiation and Perpetuation of Sjögren’s Syndrome. Int. J. Mol. Sci. 2021, 22, 658. https://doi.org/10.3390/ijms22020658

AMA Style

Chivasso C, Sarrand J, Perret J, Delporte C, Soyfoo MS. The Involvement of Innate and Adaptive Immunity in the Initiation and Perpetuation of Sjögren’s Syndrome. International Journal of Molecular Sciences. 2021; 22(2):658. https://doi.org/10.3390/ijms22020658

Chicago/Turabian Style

Chivasso, Clara, Julie Sarrand, Jason Perret, Christine Delporte, and Muhammad Shahnawaz Soyfoo. 2021. "The Involvement of Innate and Adaptive Immunity in the Initiation and Perpetuation of Sjögren’s Syndrome" International Journal of Molecular Sciences 22, no. 2: 658. https://doi.org/10.3390/ijms22020658

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