Next Article in Journal
Food-Specific IgG Antibodies: Decoding Their Dual Role in Immune Tolerance and Food Intolerance
Previous Article in Journal
Integrative Transcriptomic Meta-Analysis Reveals Risk Signatures and Immune Infiltration Patterns in High-Grade Serous Ovarian Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Sjögren’s Syndrome and Ocular Inflammation: Pathophysiology, Clinical Manifestation and Mitigation Strategies

by
Konstantinos Pavlidis
1,
Theodora Adamantidi
1,
Chatzikamari Maria
2,
Karamanis Georgios
2,
Vasiliki Dania
3,
Xenophon Krokidis
2 and
Alexandros Tsoupras
1,*
1
Hephaestus Laboratory, School of Chemistry, Faculty of Sciences, Democritus University of Thrace, Kavala University Campus, St. Lucas, 65404 Kavala, Greece
2
Hematology, Biochemistry and Internal Medicine Departments, General Hospital of Kavala, St Silas, 65500 Kavala, Greece
3
First Department of Orthopedic Surgery, School of Medicine, National and Kapodistrian University of Athens, “ATTIKON” University General Hospital, 12462 Athens, Greece
*
Author to whom correspondence should be addressed.
Immuno 2025, 5(3), 24; https://doi.org/10.3390/immuno5030024
Submission received: 8 April 2025 / Revised: 16 June 2025 / Accepted: 23 June 2025 / Published: 26 June 2025

Abstract

Sjögren’s syndrome (SS) is a chronic autoimmune disease primarily affecting the lacrimal and salivary glands, characterized by ocular and oral dryness. Beyond exocrine dysfunction, SS may also involve multiple organs and systems, contributing to systemic complications that impair a patient’s quality of life. Among these, ocular inflammation represents a significant clinical challenge, manifesting as dry eye disease and other vision-affecting complexities. Despite advances in SS understanding, the inflammatory mechanisms driving ocular manifestations remain incompletely elucidated. This review aims to clarify the key inflammatory pathways underlying ocular complications in SS and the clinical implications. Additionally, it discusses both conventional and novel therapeutic strategies focusing on mitigating SS-associated ocular inflammation, including targeted immunomodulatory agents, regenerative medicine, and innovative drug delivery systems. By integrating current knowledge from recent studies, this review attempts to provide researchers and clinicians with a comprehensive resource for optimizing SS treatment approaches. The advancement of targeted therapies and emerging mitigation strategies holds promise for improving patient outcomes and enhancing SS management.

1. Introduction

Sjögren’s syndrome (SS) is a systemic autoimmune disease primarily characterized by the involvement of exocrine glands, particularly the salivary and lacrimal glands, resulting in sicca symptoms, including dryness of the mouth (xerostomia) and eyes (xerophthalmia). This disease occurs primarily in middle-aged women but also in men, children, and the elderly, less frequently [1,2]. SS prevalence and affection in the female gender follows a 9:1 ratio, and most diagnoses occur between the 4th and 5th decades of life. In contrast, its prevalence is approximately 0.5–1.5% of the general population [3,4], a difference related to the immunoregulatory properties of female sex hormones [2,5,6].
SS is a widespread rheumatoid condition; however, many aspects of this disease have not yet been fully elucidated. SS may occur either alone (primary Sjögren’s syndrome, pSS) or combined with other autoimmune and inflammatory diseases (secondary Sjögren’s syndrome, sSS). In addition to eye and mouth dryness (98% of pSS patients report at least one symptom, 89% present both symptoms) [7], highly prevalent clinical pSS features are fatigue (>70%) [8] and arthralgia (>75%) [7]. A relatively low, but important risk of lymphoma occurrence is also observed [6,9,10,11], as the risk of non-Hodgkin B-cell lymphoma, particularly mucosa-assisted lymphoid tissue (MALT), is 10- to 19-fold higher in pSS patients [12] (5–10% prevalence) [13]. While sSS syndrome presents mostly the same symptoms as pSS, it results from a combination with other autoimmune diseases, such as rheumatoid arthritis (RA) or systemic lupus erythematosus (SLE) [2,6].
Regarding the causes and symptoms of SS, debilitating inflammation is induced by autoantibody production and lymphocyte infiltration [11,14], but like all autoimmune diseases, it can also be caused due to several external (i.e., lifestyle, stress, smoking), biological (i.e., viral infections, hormones) and genetic factors [2,11,15,16]. The first report including SS manifestations was made in 1933 by Dr. Henrik Sjögren, who studied 19 women with severe symptoms of dry eyes and mouth, who presented systemic complications and lymphocytic infiltrations of the salivary and lacrimal glands [11,14]. In SS, symptoms like fatigue and arthralgia are often associated with the respiratory, nervous, and vascular systems, thus inducing complications like pulmonary embolism or vasculitis [2,6,9,15,17].
The most commonly recorded ocular effect in SS patients is systemic destruction of the lacrimal gland associated with ocular inflammation and characterized mainly by dry eye disease (DED), as well as tear production deficiency. DED is divided into two main subtypes, namely the aqueous-deficient dry eye (ADDE) caused by lacrimal gland issues and the evaporative dry eye (EDE), initiated by lacrimal gland damage [2,15,18,19,20,21]. SS pathogenesis is mainly ascribed to the activation of the type I and II interferons, cytokine production, and autoantibodies secretion by the activation, shedding, and destruction of gland-present B and T lymphocytes [9,16]. Such manifestations drive changes in the ocular surface and destroy the neural network, which in turn induces the reflexive secretion of tears [22,23]. DED has been a global public health problem for years, but no standard cure has yet been found. Symptoms vary from itching and burning to light sensitivity, blurred vision, broken eyelashes, sensation of a foreign object in the eye, etc., provoking daily discomforts for SS patients, such as difficulty in reading or driving and fatigue [18,20,24].
SS diagnosis is established based on an evaluation of specific criteria and the utilization of several diagnostic tools. Clinical techniques such as the measurement of tear volume by Schirmer’s test, sialometry (for measuring xerostomia by indicating low unstimulated salivary flow rate), and tear break-up time measurement are tests used to diagnose SS in ocular inflammation [25,26,27]. More precisely, an SS-related ophthalmologic examination should include an assessment of tear production (Schirmer’s test), ocular surface staining, as well as the integrity of meibomian glands. Evaluation of the ocular surface with vital dyes is typically performed with lissamine green, which stains devitalized conjunctival cells and has replaced the outdated Rose Bengal drops method, and fluorescein, which reveals defects in the corneal epithelium [28,29,30].
SS classification criteria rely on a combination of clinical and serological features. The 2016 American College of Rheumatology (ACR)—European League Against Rheumatism (EULAR) criteria define SS classification based on a weighted scoring system, where a total score of ≥4 is required. Key diagnostic components include ocular signs such as an ocular surface staining score (OSS) ≥ 5 or a van Bijsterveld score ≥ 4, and objective measures of salivary gland dysfunction, like a Schirmer test ≤ 5 mm in 5 min in at least one eye. The presence of anti-Ro/SSA antibodies, focal lymphocytic sialadenitis (focus score ≥ 1 focus/4 mm2 on labial salivary gland biopsy), and the unstimulated whole saliva flow rate of ≤0.1 mL/min also contribute to the diagnosis. These criteria emphasize autoimmune involvement and help differentiate SS from other causes of sicca symptoms [29,31,32].
Relative mitigation techniques are constantly updated to achieve the most effective SS treatment solution. A range of therapeutic interventions including ocular therapeutic drops, administration of anti-inflammatory drugs, mainly corticosteroids, cyclosporine A (CyA), tetracyclines and derivatives, fatty acids, vitamins, or anti-inflammatory therapies [23,25,26,33,34,35], are employed, usually involving a marked opening of the tear ducts and the insertion of collagen or silicone plugs that block and prevent tear drainage [19].
This review aims to demonstrate a comprehensive approach to the pathophysiology of SS and its association with ocular inflammation. For this purpose, hormones affecting the lacrimal glands, several genetic, epigenetic, environmental, and pathophysiological agents, as well as autoimmune mechanisms, inflammatory markers, and autoantibodies linked to SS manifestation, are thoroughly discussed. Furthermore, emphasis is also given to the clinical manifestations of SS along with treatment and mitigation interventions utilized for combating this disease, which are also extensively analyzed. The main highlights and key points of this research are illustrated in Figure 1.

2. Materials and Methods

All data included in this review were collected from scientific articles and reviews published in online databases like Scopus, MDPI, Science Direct, PubMed, and Google Scholar. Some of the main keywords and key phrases utilized in this review are the following: “Sjögren’s syndrome”, “ocular inflammation”, “dry eye disease”, “lacrimal glands”, “hormones”, “ Sjögren’s syndrome pathogenesis”, “autoimmune mechanisms”, “inflammatory mechanisms”, “type I interferon system”, “B cells”, “autoantibodies”, “genetic factors”, “epigenetic factors”, “environmental factors”, physiological factors”, “microRNA”, “clinical manifestations”, “mitigation techniques”, “topical lubricants”, “cyclosporine A”, “omega-3 fatty acids”, “omega-6 fatty acids”, “vitamin A”, “vitamin D”, “vitamin E”, “innate immunity”, “therapeutic contact lenses”, with the use of combinations of these keywords by using the AND and/or OR terms in each query, in every scientific database mentioned.
This research process was conducted from October 2024 to February 2025, considering relative data obtained from articles published mainly over the last 5–10 years. The eligible review studies met pre-established criteria, including mostly research articles, written in the English language, and being published between 2015–2025. A limited number of important articles that contained important information not mentioned in more recent publications, but that were published before 2018, were also included. Duplicates and articles published in non-eligible or invalid scientific databases were strictly excluded, while conference papers, book chapters, short surveys, newspaper or magazine articles, theses, or non-academic reports were mostly excluded as well (under the PRISMA guidelines).
All articles included were thoroughly evaluated by reviewing primarily their titles, abstracts, and keywords, involving only those that contained pertinent information. Statistical data, study outcomes, and research processes were analyzed and then presented to achieve a practical demonstration of the severity of Sjögren’s syndrome (Figure 2).

3. Pathophysiology of SS and Ocular Inflammation

In SS incidence, inflammatory processes occur primarily through the glandular epithelial cells, which are involved in the expression of antigen proteins, promote adhesion, and stimulate T lymphocytes. Certain cytokines, such as interferon γ (IFN-γ), tumor necrosis factor α (TNF-α), several interleukins (e.g., IL-1β, IL-6, IL-10), and the tumor growth factor beta (TGF-β), enhance the antigen-presenting function or induce the apoptosis of epithelial cells of the lacrimal and salivary glands [36,37].

3.1. Lacrimal Glands

Tears, which lubricate, clean, and protect the eye surface, are produced by the lacrimal gland, a compound, tubuloalveolar serous gland that opens at the inner surface of the palpebra and lubricates the ocular surface. Lobules that emerge from a connective tissue capsule and are divided by connective tissue septa make up the gland [38,39]. The predominant tear glands are located in the upper outer part of the orbit, within the lacrimal fossa of the frontal bone, and consist of two parts: the ocular, higher up in the cavity, and the buccal part, closer to the eyeball. The orbital lobe is larger among the two tear glands, while the size of the primary lacrimal gland varies significantly (comprising two lobes of about 20 × 12 × 5 mm). Differences between the symmetries of the right and left glands indicate an underlying pathology [40,41]. The lacrimal receives its arterial blood supply for both the upper and lower eyelids primarily from the lacrimal branch of the suboccipital artery, and occasionally from the recurrent meningeal artery. Finally, the lacrimal vein, which traverses the gland, drains into the superior ophthalmic vein [41,42].
The structure and function of the lacrimal gland are influenced by hormones produced in the pituitary and hypothalamus regions. Such hormones are α-melanocyte-stimulating hormone (α-MSH), prolactin, androgens, adrenocorticotropic hormone (ACTH), estrogen and progesterone, as well as retinoic acid, insulin, glucagon, and glucocorticoids (Figure 3) [21,43,44]. Hundreds of factors that protect the ocular surface, such as growth factors (e.g., epidermal growth factors (EGFs)), anti-inflammatory agents (e.g., interleukin 1 receptor antagonist (IL-1RA)), mucins, etc., are produced by lacrimal glands and in fact, decreased concentrations of these factors have been recorded in SS patients [45,46]. When the tear functional unit becomes inflamed, dry eye develops, causing dysfunction and epithelial cell death, leading to the secretion of tears into the gland and conjunctiva. Lacrimal gland dysfunction is attributed to various mechanisms, such as the cholinergic blockade by autoantibodies attacking the muscarinic acetylcholine 3 receptors (mAChRs, M3), a mediation of cytokines causing epithelial cell death, inflammatory cytokine action such as IL-1 [45,46,47,48], neurotransmission disorders [45,49], oxidative stress [50], B-lymphocyte self-reactivity [51] and angiogenesis-related disorders. An overexpression of the vascular endothelial growth factor (VEGF), which alters lacrimal gland vasculature morphology, induces inflammation and hypoxia [48,52,53]. However, these are only a few of the known mechanisms considering tear gland dysfunction that have been extensively studied.

3.2. Autoimmune and Inflammatory Mechanisms

The lacrimal gland may become an immune-disease target for SS, as a result of aging, ocular inflammation, genetic predispositions, and/or external factors. Extensive research on SS autoimmune mechanisms, mainly on the relative cytokines, chemokines, and their roles in exocrine glands, is currently conducted [45,47,54]. SS development is ascribed to a disruption of innate immune barriers, accomplished by a mechanism involving the interferon (IFN) pathway [7]. More specifically, key cytokines involved in the pathogenesis of autoimmune diseases like pSS are IFNs, which are produced by the body’s cells in response to viral infections, pathogens, or abnormal cellular processes. IFN’s overexpression enhances the growth and progression of pSS [55]. IFNs are divided into three categories: type I, II, and III. Type I IFNs mobilize innate immune responses by enhancing antigen presentation and cytotoxicity while restraining the magnitude of inflammation to prevent toxicity [56]. IFN-I consists of 13 IFN-α subtypes (-α1, -α2, -α4, -α5, -α6, -α7, -α8, -α10, -α13, -α14, -α16, -α17 and -α21), IFN-β, IFN-k, IFN-ε, IFN-d and IFN-ω. IFN-α, IFN-β, IFN-γ, IFN-e, IFN-k, and IFN-ω are traced in humans, whereas IFN-d and IFN-t have been found only in pigs and cattle, respectively [2,55,57]. SS patients have exhibited an increased expression of IFN-I genes (IFN signature), found mainly in blood mononuclear cells and tissues. The presence of the IFN signature indicates an ongoing activation of the IFN-I pathway, which accelerates autoimmune activity. In pSS, an IFN signature signifies disease activity, whereas in RA, it defines a specific disease subset related to poor clinical outcomes and resistance to B-cell depletion therapy (Figure 4) [55,58].

3.2.1. Type I Interferon System in Primary SS (pSS): An Interplay Between Innate Immunity, Inflammation, and pSS Exacerbation

To fully elucidate how the pSS-IFN-I system synergy, it is necessary to present the IFN action mechanisms in SS formation and immune system activation against the exocrine glands [2,55,57]. Both salivary and lacrimal glands express ligands, receptors, and cytokines, capable of activating several innate immune cells like natural killer (NKs), dendritic (DCs), and mast cells (MCs), as well as macrophages, type 3 innate lymphoid cells (ILC3s), antigen-presenting cells (APCs) and B or T regulatory cells. MCs induce fibrosis and salivary gland infiltration, whereas the mucosa-associated invariant T cells (MAITs) exhibit impaired activation and formation of protective cytokines, interacting with APCs. Macrophages are activated by IFN-γ, releasing IL-22 that exacerbates immune signaling, while T cells are triggered by DCs and inflammatory cytokines, for further IFN-γ production. IFN-α enhances B cell activating factor (BAFF) release, B cell survival, and autoantibody formation as concurrently, as ILC3s interact with B cells via BAFF [9,11,54].
IFN-I system activation is primarily attributed to viral infection or other environmental factors that cause inflammation and trigger plasmacytoid DCs (PDCs) activation in the lacrimal glands. PDCs produce IFN-α, which leads to epithelial cell apoptosis and also autoantigen release (anti-Sjögren’s-syndrome-related antigen A (Ro/SSA) and B (La/SSB)). Such autoantigens preserve IFN-α production, activating a self-perpetuating cycle. After being exposed during epithelial cell apoptosis, they are recognized by immune B-cells, leading to the production of autoantibodies against the autoantigens SSA/Ro and SSB/La (anti-Ro/SSA and anti-La/SSB, respectively) [11,59,60].
Immune complexes activate PDCs through binding to the FcγRIIa receptor (cluster of differentiation 32 (CD32)), promoting the formation of germinal center (GC)-like structures. This binding triggers toll-like receptors (TLRs), particularly TLR7 and TLR9, which recognize RNA and DNA within the complexes, leading to enhanced IFN-α production and further inflammation. IFN-α marker, in turn, induces major histocompatibility complex (MHC) class I and II expression in lacrimal gland epithelial cells, enabling them to present antigens. Concurrently, chemokines such as the C-X-C motif chemokines 12, 9, and 10 (CXCL12, CXCL9, and CXCL10, respectively), exacerbate inflammation by recruiting T-cells, B-cells, and additional PDCs to the site [9,11,54].
Aberrant VEGF expression drives chaotic neoangiogenesis, with the upregulation of the intracellular adhesion molecule 1 (ICAM-1) on endothelial cells, facilitating immune innate cell infiltration [9,11,54]. Sustained IFN-α maintains inflammation and increases epithelial autoantigen expression (e.g., Ro52), perpetuating the autoimmune cycle. Progressive epithelial cell and immune-mediated tissue destruction ultimately impair exocrine gland function, leading to keratoconjunctivitis sicca, the hallmark ocular manifestation of Sjögren’s syndrome [9,11,54]. The main impact of cytokines and chemokines involved in SS is presented in Table 1, while an interplay between innate immunity, inflammation, and pSS is thoroughly depicted in Figure 4.
Most inflammation-related chemokines and cytokines also contribute to oxidative stress, neuroinflammation, and ocular damage. In SS, oxidative stress is promoted via the generation of reactive oxygen species (ROS) and lipid peroxidation products, leading to tear film instability, altered tear composition, epithelial damage, and the release of additional pro-inflammatory mediators, thereby exacerbating DED symptoms. Increased oxidative stress in SS is supported by increased malondialdehyde (MDA) levels and reduced activity of antioxidant enzymes [68,69,70]. Moreover, emerging evidence suggests a critical role for neuroinflammation in SS-associated ocular complications. Disruption of the autonomic nervous system (ANS) signaling—particularly reduced parasympathetic input—has been linked to lacrimal gland dysfunction and impaired tear secretion. Corneal hypoesthesia and aggravating ocular discomfort have been associated with inflammatory cytokines (e.g., IL-6, TNF-α), which may impair corneal nerve density. Therapeutic strategies targeting both oxidative stress and neuroinflammatory mechanisms may offer promising interventions for SS-related ocular manifestations (Figure 4) [2,11,71,72,73].

3.2.2. Role of B-Cells in SS

Ongoing research into SS pathophysiology underscores the central role of B cells in disease onset and progression, particularly in pSS. These cells contribute to the formation of ectopic GC-like structures within exocrine glands and are implicated in extra-renal manifestations. Regulatory B (Breg) cells are especially important in pSS development due to their immunomodulatory functions, including the secretion of cytokines such as IL-10, IL-35, and Granzyme B (GrB) [74,75,76,77,78]. Memory B Cells (MBCs) are also implicated in SS pathogenesis, accumulating in the lacrimal and salivary glands where they promote local autoimmune responses [74]. Their activation leads to autoantibody production and the release of pro-inflammatory cytokines, thereby intensifying ocular inflammation [79]. Targeting MBCs has emerged as a promising therapeutic strategy for managing SS-related ocular complications [77,80]. The key roles of B-cell subtypes in SS pathogenesis are presented in Table 2, while the interplay between B-cell innate immunity, inflammation, and SS pathophysiology is illustrated in Figure 5.

3.3. Main Autoantibodies in SS: Their Role in Inflammation, Clinical Significance, and Diagnostic Value in SS

Autoantibodies play a variety of roles in SS and are of high diagnostic significance. Among the most clinically important antibodies are the anti-Ro/SSA and anti-La/SSB ones, which are often associated with early-onset disease (younger individuals), severe exocrine dysfunction, intense lymphocytic infiltration of minor salivary glands, and prolonged disease duration. Their detection is commonly performed through ELISA, although assay performance varies depending on the antigen source. The sensitivity and specificity for SSA/SSB antibodies range from 39–77% and 79–100%, respectively, while Line Immunoassay (LIA) has shown superior sensitivity (91.4%) and specificity (87%) [84,85].
B-cell hyperactivity in SS underlies the production of these and other autoantibodies, contributing to diagnosis and clinical manifestations. Anti-Ro/SSAs and anti-La/SSBs, while not disease-specific, remain valuable biomarkers in routine evaluation [84,85,86]. The key autoantibodies and their immunological and diagnostic relevance are described in the following subsections. Their roles in SS-linked inflammation are depicted in Figure 4.

3.3.1. Anti-Ro/SSA Autoantibodies

The Ro/SSA antigen is a ribonucleoprotein complex that comprises two major immunogenic proteins: Ro52 (52 kDa) and Ro60 (60 kDa). Ro60 binds non-coding Y-RNA and forms cytoplasmic ribonucleoprotein particles, whereas Ro52 can localize independently in the nucleus. Both proteins are targeted by autoantibodies in SS and are upregulated by innate immune stimuli such as TLR3. Dysregulated expression may also result from microRNA post-transcriptional mechanisms [87,88]. These autoantibodies are closely linked to SS-related inflammation and are frequently used as primary diagnostic markers.

3.3.2. Anti-La/SSB Autoantibodies

The La/SSB antigen is a 48 kDa phosphorylated protein that associates with nascent RNA transcripts, playing roles in RNA metabolism, including 3′ end protection of RNA polymerase III transcripts and pre-tRNA processing. It functions as an RNA chaperone and is localized in both the nucleus and cytoplasm. Anti-La/SSB autoantibodies are typically detected alongside immunoglobulin G (IgG) and anti-Ro/SSAs, and their isolated presence is not strongly affiliated with SS phenotypic features. Similar to Ro/SSA antigens, La/SSBs expression is modulated by TLR signaling and specific miRNAs, especially under apoptotic stress or via exosome release from epithelial cells [2,15,85,87,88].

3.3.3. Rheumatoid Factor (Rf), Complement System, and Cryoglobulins

Serological positivity for anti-Ro/SSAs and anti-La/SSBs is linked to Rf, which is frequently observed in SS patients. Research has indicated that Rf is one of the few indicators associated with severe illness and exocrine gland symptoms, necessitating further monitoring [86,89]. Rf, particularly the IgM class, targets the Fc region of IgG antibodies and is present in a significant proportion in SS patients. While classically linked to RA, Rf may also be elevated in other autoimmune disorders, including SLE and mixed connective tissue disease, largely due to chronic immune activation. Its presence warrants monitoring due to its correlation with disease severity [60,78,90].
Complement proteins and cryoglobulins are also important contributors to SS pathogenesis, particularly in systemic and vasculitic manifestations. Hypocomplementemia, especially low C3 and C4 levels, has been correlated with increased SS activity and risk of lymphoma. Cryoglobulinemia, the presence of circulating immunoglobulins that precipitate at low temperatures, occurs in a subset of SS patients and is often linked to systemic vasculitis, purpura, peripheral neuropathy, and glomerulonephritis. Both biomarkers hold diagnostic and prognostic value and share clinical relevance with Rf [91].

3.3.4. Cyclic Citrullinated Peptide Antibodies (Anti-CCP)

Patients who tested positive for anti-CCP and RF showed progressively worse ocular inflammation, providing a diagnostic tool and useful information for patients with severe inflammatory eye disease [92]. Anti-CCP antibodies, although primarily affiliated with RA, have also been detected in approximately 22.1% of patients diagnosed with pSS, even in the absence of clinically established RA. Their presence in SS may serve as a predictive marker for future arthritis development, thus providing useful prognostic information and supporting early monitoring for potential RA transition [11,92,93].

3.3.5. Anti-Centromere Antibody (ACA)

ACAs, targeting centromere proteins (CENPs), are observed in a minority of SS patients (17% of ACA-positive autoimmune diseases were SS). Since this discovery, ACA’s presence in SS has reached 3.7–27.4%, and they are associated with severe sicca symptoms and extensive lymphocytic infiltration of the salivary glands. Although more commonly linked to limited systemic sclerosis, their detection in SS suggests a distinct clinical subset with possible overlap features [83,94,95,96].

3.4. Genetic, Epigenetic, Environmental, and Physiological Factors Triggering SS Development

SS onset and progression are driven by a wide combination of genetic and epigenetic mechanisms, physiological alterations, environmental triggers, as well as gut microbiota dysbiosis, with the gut-lacrimal axis also playing a key role. These elements interact with innate immunity and chronic inflammation, as illustrated in Figure 4.

3.4.1. Genetic-Epigenetic Factors

Recent transcriptomic studies in salivary glands and peripheral blood suggest that genetic polymorphisms and epigenetic alterations, especially those affecting immune-related pathways (e.g., microRNA pathways), play a critical role in SS susceptibility. Key mechanisms include the activation of the IFN-I axis, Th1-related cytokines (IL-12, IFN-γ), and dysregulation of chemokine receptors like CXCR5 [19]. Epigenetically, various modifications, like DNA methylation, histone modifications, non-coding RNA, and IFN gene hypomethylation, lead to increased B-cell expression [19,64,75]. Hypomethylation of IFN-regulated genes in epithelial cells and immune cells like peripheral blood mononuclear cells (PBMCs) contributes to a persistent IFN-I signature [55,58,66], while altered histone acetylation enhances pro-inflammatory cytokine expression (e.g., IL-6, TNF-α) [2,9].
microRNAs Implication
MicroRNAs (miRNAs), single-stranded, non-coding RNAs regulating over 60% of protein-coding genes, play a major role in immune regulation (such as T and B-cell function) by targeting mRNAs for degradation or translational inhibition [97]. Specifically, aberrant expression of miRNAs, like upregulated miR-146a/miR-155 and downregulated miR-181a levels, is linked to dysfunctional B and T-cell responses in SS. These patterns, consistently observed in exocrine glands and PBMCs, highlight the emerging role of miRNAs as both biomarkers and potential therapeutic targets in SS (Table 3) [19,97,98,99,100].

3.4.2. Psychological Factors

Psychological distress, such as anxiety, depression, and chronic stress, is highly prevalent among pSS patients and contributes to both symptom burden and disease progression [101]. Dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis due to adrenal autoimmunity and altered levels of IL-1, IL-36a, and humoral immunity markers are associated with fatigue and neuroinflammation [102]. Depression has been correlated with neurotransmitter imbalances, neurotrophic factor disruption, and the presence of autoantibodies targeting neuropeptides. Additionally, α-MSH autoantibodies and BAFF may synergistically contribute to anxiety, brain inflammation, neuronal damage, and hippocampal dysfunction [103]. Omega-3 polyunsaturated fatty acids (ω-3 PUFAs) may mitigate depressive symptoms by reducing neuroinflammation. Emerging therapeutic interventions like dazodalibep (CD40 ligand antagonist), RSLV132 (RNaseFc fusion protein), and belimumab against fatigue symptoms show promise in addressing SS-related neuropsychiatric complications [37,55,102,103,104,105].

3.4.3. Environmental Factors

Environmental triggers such as viral infections (e.g., Epstein-Barr virus (EBV), human T-lymphotropic virus 1 (HTLV-1), cytomegalovirus (CMV), and hepatitis C virus (HCV)), play a pivotal role in SS pathogenesis, particularly through chronic persistence in salivary glands and molecular mimicry mechanisms that lead to loss of self-tolerance and autoimmunity [2,26,106,107]. Moreover, although there is emerging evidence suggesting a possible link between SS and COVID-19, post-COVID-19 SS is not yet definitely established. Studies however, indicate that SARS-CoV-2 may trigger or exacerbate SS-like symptoms or autoantibodies in certain individuals, but further research is necessitated to confirm an exact causal relationship [108,109,110].
Exposure to pollutants such as heavy metals (e.g., mercury, lead, chromium, cadmium) [111,112] and organic solvents like perfluoroalkyl acids) [113], has been associated with SS onset via oxidative stress and epigenetic modifications. In addition, lifestyle factors, including chronic ultraviolet (UV) radiation exposure, smoking, and dietary patterns, may also modulate risk. Notably, the role of smoking remains controversial, with both protective and detrimental effects reported on pSS risk. Some studies indicate nicotine’s ability to suppress B cell hyperactivity (lower pSS risk), and others suggest that it triggers exocrine gland dysfunction and oxidative stress (higher pSS risk). However, further studies are required to support this observation [114].

3.4.4. Role of Gut Dysbiosis and Gut-Lacrimal Axis

Gut microbiota dysbiosis has been increasingly implicated in SS through its impact on T-cell differentiation, mucosal immunity, and ocular inflammation. SS patients commonly display reduced microbial diversity, altered Firmicutes/Bacteroidetes (F/B) ratios, and decreased levels of short-chain fatty acid (SCFA)-producing bacteria, like Faecalibacterium prausnitzii and Bifidobacterium spp., contributing to epithelial barrier dysfunction [115,116,117]. Disruption of the gut-lacrimal axis can further aggravate DED via systemic endotoxin leakage and cytokine-driven lacrimal gland inflammation. Therapeutic strategies such as probiotic supplementation, dietary interventions, and fecal microbiota transplantation (FMT) are under investigation as adjunctive treatments (Figure 4) [115,116,117].

4. Clinical Manifestations of SS

4.1. Clinical Manifestations of SS: Sicca Symptoms and More

SS presents with a wide spectrum of clinical features, broadly categorized into glandular and extraglandular manifestations. Glandular dysfunction arises from lymphocytic infiltration and destruction of exocrine glands (primary salivary and lacrimal ones) and include xerostomia (dry mouth), keratoconjunctivitis sicca (dry eyes), parotid gland enlargement, and dryness of other mucosal surfaces (e.g., vaginal or nasal dryness, dry skin (xerosis)). Over 90% of SS patients report dry mouth, making sicca syndrome the hallmark of SS, along with DED (80% experience sicca symptoms) [118]. Extraglandular manifestations stem from systemic immune dysregulation and may involve musculoskeletal (e.g., arthralgia, myalgia), pulmonary (e.g., interstitial lung diseases (ILDs)), renal (e.g., tubulointerstitial nephritis), neurological (e.g., peripheral neuropathy), hematological (e.g., MALT lymphoma), vascular (e.g., systemic vasculitis), gastrointestinal and hepatic systems, often accompanied by constitutional symptoms (e.g., fever, fatigue) and cutaneous lesions [119,120]. Furthermore, SS may have serious systemic consequences, including lymphoma, cardiovascular disease, and increased mortality in certain subgroups. Notably, although SS affects women predominantly, men exhibit a nearly 3-fold higher mortality rate, potentially due to more severe systemic involvement [121,122].
Pulmonary complications occur in 9–20% of SS patients, most frequently as respiratory impairment like chronic ILDs and tracheobronchial disease [9,15,20,123]. ILDs include non-specific interstitial pneumonia (NSIP), usual interstitial pneumonia (UIP), lymphocytic interstitial pneumonia (LIP), and cryptogenic organizing pneumonia (COP). NSIP is the most common ILD pattern in SS (~45%), characterized by ground-glass opacities and reticular changes. It can be either cellular (inflammatory, better prognosis) or fibrotic (progressive, worse outcome) [124,125]. UIP is less common (~17%) but more severe, presenting with progressive fibrosis, honeycombing, as well as subpleural fibrosis on high-resolution CT (HRCT) [126,127]. Plus, LIP is strongly associated with SS (25%), though rare, marked by diffuse lymphocytic infiltration, cystic lung disease, and risk of lymphoma [128,129]. Finally, COP involves alveolar and small airway inflammation, leading to patchy consolidations and scarring. The pSS-COP link is extremely rare [130].
Other clinical features include hypergammaglobulinemia, often associated with anti-Ro/SSA, anti-La/SSB, and RF antibodies [2,11]. Cutaneous involvement includes dry skin, annular erythema, and vasculitis lesions, often linked to keratin alterations and skin protein dysregulation [131]. Moreover, neurological and systemic complications are increasingly recognized in SS, significantly affecting patients’ quality of life. Common neurological manifestations include peripheral and small fiber neuropathy, as well as cognitive impairment, and are often linked to B-cell hyperactivity, altered lipid metabolism, and chronic IFN-driven, neuroinflammation-related demyelination. Additionally, metabolic syndrome, pulmonary involvement, renal dysfunction, and elevated cardiovascular risk further deteriorate the SS burden. These manifestations may progress silently, and thus early detection prevents long-term morbidity [9,15,20,64,123,132,133]. Figure 5 provides a schematic overview of the most common clinical symptoms experienced by SS patients.

4.2. Dry Eye Diseases of SS

DED is one of the most prevalent and early ocular manifestations in SS patients, driven by autoimmune-mediated destruction of the lacrimal glands and chronic inflammation of the ocular surface. It is also commonly observed in other systemic or ocular conditions, such as diabetes mellitus, graft versus host disease, and Graves’ orbitopathy. In SS, DED typically presents with reduced tear production, epithelial damage, and destabilization of the tear film due to goblet cell loss, leading to symptoms such as burning, grittiness, blurred vision, and photosensitivity [134].
Diagnostic evaluation of DED includes a combination of subjective and objective assessments, such as the ocular surface disease index (OSDI), Schirmer’s test (ST), tear film break-up time (TFBUT), and corneal fluorescein staining (CFS). These tests help characterize the severity and impact of dry eye but cannot be used to distinguish pSS from sSS, as both forms exhibit similar ocular involvement, and pSS-sSS distinction is based on the presence of another well-defined autoimmune disease (e.g., RA, SLE) [134].
DED in SS significantly impacts the quality of life. Recent studies report that patients with severe DED symptoms experience a four-fold reduction in daily functionality compared to individuals without symptoms. Moreover, such patients are more than twice as likely to suffer from emotional distress, increased financial burden, and social limitations. Painful dry eye is more frequently observed in women, older individuals, and patients with corneal epithelial injury. Notably, photosensitivity affects ~50% of SS patients, while 25% report painful photosensitivity [14,23,42,135].

4.3. Ocular Inflammatory Complications

Beyond the classic symptoms of DED, SS may involve a range of inflammatory ocular complications, including sterile corneal ulcer, scleritis, optic neuritis, and uveitis as illustrated in Figure 6. These conditions may pose significant threats to vision and require prompt diagnosis and management [121,135,136,137].

4.3.1. Sterile Corneal Ulcers

Sterile corneal ulcers affect ~2.5–3.6% of SS patients and are considered vision-threatening complications. They are not caused by infections but rather by immune-mediated disorders such as SS or RA. Such ulcers may worsen after procedures like cataract surgery, especially if the underlying ocular surface is poorly controlled. In these cases, pre-operative immunosuppression is often recommended to minimize risks [138]. Additional contributing factors include the use of contaminated contact lenses (e.g., Gram-negative organisms), poor lens hygiene, or chemical exposure. Gram-negative keratitis presents with red, painful eyes and is mostly linked to contact lens damage [18,139,140]. Furthermore, topical corticosteroids, although used to suppress inflammation, can paradoxically exacerbate sterile corneal ulcers, especially with prolonged use. While they aid in modulating immune response, their use should be carefully monitored (Figure 6) [141].

4.3.2. Scleritis

Scleritis is a severe ocular inflammation affecting the sclera—the outer protective layer of the eye—whose vascular structure is particularly prone to inflammatory responses. Due to its dense and fibrous composition, the sclera does not easily eliminate harmful agents, making it susceptible to prolonged inflammatory reactions. Scleritis is typically observed in middle-aged patients (diagnosed between 47–60 years of age), and is frequently correlated with systemic autoimmune diseases like SS, rather than infectious causes. Although rare, it can also be triggered by bacterial, viral, or fungal infections [19,142,143]. It is considered a heterogeneous condition, in both terms of pathogenesis and etiology, and is increasingly viewed as an autoimmune disorder in which T- and B-cell cytokines play a central role. Treatment often involves topical or systemic corticosteroids, anti-TNF, and anti-B-cell-targeted therapies. However, these treatments can be linked to notable ocular side effects and carry potential risks to vision, further compromising the patient’s quality of life (Figure 6) [24,94,144].

4.3.3. Optic Neuritis

In some cases, optic neuritis may be the first SS manifestation, even in the absence of classic complicating the diagnostic process. The condition is linked to autoimmune activity, with antibodies such as anti-Ro/SSA, anti-La/SSB, and anti-aquaporin-4 antibodies being often detected [145]. Although usually idiopathic, optic neuritis may also be triggered by infections or systemic inflammation. It presents with retro-orbital pain, unilateral vision loss, and photosensitivity requiring prompt management (Figure 6) [145,146,147].

4.3.4. Uveitis

Uveitis is an inflammatory condition that affects the eye’s uvea, which consists of the iris, ciliary body, and choroid. It may result from infections, trauma, or immune-mediated response complications [11,148]. Although corticosteroids are widely used as the primary treatment and are often effective, they can lead to significant side effects, and their long-term use should be approached with caution. Uvea is a highly heterogeneous condition. When associated with immunobiological responses, it is linked to dysregulation between immune suppression and pro-inflammatory cascades. Regulatory T lymphocytes and their produced cytokines (e.g., IL-10, IL-27 (retinal cells), TGF-β), play a pivotal role in disease modulation (Figure 6) [11,148].

5. Common Conventional and Newly-Emerged Treatment and Mitigation Techniques for SS and DED Management

Several conventional techniques have been developed and are currently employed, paving the way for more effective and resilient innovative treatment approaches to ensure better patient compliance in SS management. The most common strategy adopted by contact lens users for symptom relief is the use of tear substitutes, which play a crucial role in restoring ocular moisture. While many contact lenses contain preservatives that can cause irritation and complications, tear substitutes are typically preservative-free and better tolerated [36]. Current DED treatments in the context of SS aim to lubricate the ocular surface, prolong tear film stability, and target ocular inflammation through specific therapeutic agents. Treatment selection depends largely on the severity of the disease, as DED can present with varying levels of discomfort and damage. A major challenge in both selecting treatments and interpreting outcomes lies in the common misclassification of DED as a uniform condition, despite its multifaceted clinical manifestations, pathogenetic mechanisms, and outcomes [11,36,60,62,103,149]. All conventional topical, systemic, interventional, adjunctive, and nutritional SS therapies used are depicted in Figure 7 and Figure 8.

5.1. Conventional Topical and Systemic Treatments

5.1.1. Topical Lubricants: Artificial and Biologic Tear Substitutes

A wide range of substances is utilized in the manufacture and administration of commercial artificial lubricants designed to alleviate dry eye symptoms. These formulations typically include demulcents, which lubricate the ocular surface, and emollients that slow tear evaporation, often involving sodium hyaluronate (SH), known for its capacity to hydrate and protect the corneal endothelium. Administration of hyaluronic acid (HA) and its sodium salt, SH, in SS patients has led to significant improvement in tear film break-up time (TBUT), cytology, and corneal staining after 2 weeks of treatment [150,151,152]. Artificial tear substitutes are mainly intended for temporary relief and symptom control in patients with mild SS symptoms. While generally safe and effective, their utility is limited in chronic or moderate-to-severe SS, due to the need for frequent dosing [23,150,151,152]. In addition, lipid-based formulations used in SS treatment vary in viscosity and hydration levels, which may present clinical challenges. For instance, more viscous formulations like ointments offer extended relief and increased ocular surface retention time, but may impair visual acuity by inducing blurred vision. The use of corticosteroids in ophthalmic solutions should also be avoided, as they can increase the risk of corneal infection and degradation. Likewise, eye drop preservatives (e.g., benzalkonium chloride, sodium chlorate), though effective, may cause ocular surface damage upon repeated use [22,23,25].
In contrast, biologic tear substitutes, such as autologous serum and allogeneic platelet-rich plasma (PRP) eye drops, deliver essential nutrients and regenerative factors that artificial lubricants lack. Such formulations contain potent anti-inflammatory and tissue repair agents, such as EGF, fibronectin, TGF-β1, and platelet-derived growth factor-AB (PDGF-ΑΒ), making them suitable for severe SS-related DED management. Notably, PRP drops have demonstrated superior efficacy compared to artificial lubricants, particularly in SS cases. However, challenges including high cost, storage conditions, and frequent dosing needs, limit their broader clinical use (Figure 7) [153,154,155,156,157].

5.1.2. Topical Cyclosporine A (CyA) Immunosuppressive Treatment

CyA is a potent immunosuppressive agent that inhibits the activation and proliferation of T cells. In the context of abnormal activation of Th17 observed in SS, CyA acts as a key mitigation agent [158]. By binding to cyclophilin D and preventing T-lymphocyte activation, CyA reduces inflammation (DED contributing factor). Furthermore, CyA helps preserve conjunctival epithelial cell integrity by inhibiting the opening of the mitochondrial permeability transition (MPT) pore, a process that otherwise leads to cell death. This inhibition results in decreased tear production as inflammation subsides. CyA improves tear secretion and lacrimal gland function by lowering the expression of pro-inflammatory markers like IFN-γ and IL-1, both elevated in SS [159]. One of the most commonly used commercial formulations of CyA is marketed under the brand name “Restasis®”, which contains 0.05% CyA along with preservatives. Other formulations used in artificial tear preparations may contain lower CyA concentrations (0.5 and 0.1%) depending on disease severity [34]. Clinical studies have reported an increase in Schirmer scores and notable symptomatic relief following CyA treatment. Improvements have also been observed in tear film stability, evidenced by increased tear meniscus height, volume, and round cell density [158]. Importantly, CyA therapy has been associated with a significant increase in goblet cell density, enhancing mucin production and providing better protection for the ocular surface. However, CyA may also lead to many adverse effects during therapy, like burning and stinging sensations, which may affect patient compliance and limit widespread use (Figure 7) [22,34,158,159].

5.1.3. Conventional Systemic Corticosteroids, Disease-Modifying Anti-Rheumatic Drugs (DMARDs) and Immunosuppressants

While topical therapies provide symptomatic relief for ocular SS manifestations, systemic treatments play a vital role in controlling the underlying autoimmune pathology and preventing SS progression. Selecting a systemic therapy is guided by SS severity, extent of systemic involvement, and patient treatment response. Systemic corticosteroids, such as prednisone and methylprednisolone, are widely used for short-term management of SS-induced acute inflammation. They function by suppressing pro-inflammatory cytokines (e.g., TNF-α, IL-6, IFN-γ), thus limiting immune cell infiltration and associated tissue damage. However, due to significant long-term side effects, like osteoporosis, hyperglycemia, hypertension, and adrenal suppression, they are generally prescribed at the lowest effective dose and for a limited duration [23,33,160].
Conventional synthetic DMARDs (csDMARDs) are standard therapy for systemic autoimmune diseases such as SS, mostly in cases presenting with arthritis, systemic inflammation, or extraglandular complications. Among these, hydroxychloroquine, an antimalarial agent, is widely favored due to its B-cell modulating capacity, autoantibody suppression, and inhibition of TLR signaling. Clinical trials suggest that it may alleviate systemic inflammation, fatigue, and sicca symptoms, while also offering cardiovascular protection [23,33,161]. Methotrexate, a folate antagonist, is often considered in SS patients with arthritis and systemic inflammatory symptoms because of its immunosuppressive and anti-inflammatory properties. Interestingly, studies suggest that anti-Ro/SSA-positive patients with RA exhibit notably lower disease activity scores (score-28 C-reactive protein (DAS28-CPR)) when treated with methotrexate, compared to anti-Ro/SSA-negative individuals. However, this finding paradoxically correlates with a poorer overall therapeutic response, suggesting that anti-Ro/SSAs may serve as predictive markers of refractoriness to methotrexate in RA and potentially in SS patients as well. When methotrexate is contraindicated, leflunomide is preferred; however, scientists should consider its adverse effects beforehand [69,162]. Moreover, the macrophage migration inhibitory factor (MIF), which binds to nuclear factor-kappa beta (NF-κΒ), reduces pro-inflammatory cytokines and exerts a significant rebalancing effect on Th1, Th17, and T regulatory cells [163]. While not yet widely applied as a therapy, its involvement in TLR modulation and innate immunity regulation suggests its potential as a future treatment SS target [149,163,164].
Regarding patients with severe systemic SS or unresponsive to csDMARDs, stronger immunosuppressants (like cyclophosphamide) may be required [165]. Mycophenolate mofetil (MMF) and azathioprine suppress lymphocyte proliferation and are often used in cases of SS-related vasculitis or ILD. MMF has also been used intravenously, often in conjunction with immunoglobulin therapy after rituximab, to manage refractory sensory neuropathy in pSS patients [166]. Systemic CyA and tacrolimus, which block T-cell activation via calcineurin inhibition, can suppress ocular inflammation and modulate Th1 cytokine activity. Despite their potential in tear gland preservation, systemic administration is limited due to risks of nephrotoxicity and hypertension (Figure 7) [166,167].

5.2. Conventional Interventional Procedures

5.2.1. Punctal Occlusion

Punctal occlusion, using temporary collagen or permanent silicone plugs, is a well-adopted method to reduce tear drainage, thus enhancing tear film stability and ocular surface hydration in patients with SS-related DED. While effective, side effects such as plug extrusion, infection, and local inflammation are common (Figure 7) [19,23,94,168].

5.2.2. Therapeutic Contact Lenses (CLs)

One of the most common therapeutic options is the Therapeutic Hyper-CLTM, a soft contact lens designed to prolong the residence time of eye drops on the ocular surface. This is achieved by creating a reservoir between the lens and cornea, where active agents are trapped and slowly released [169]. Scleral lenses, which vault over the cornea, create a moisture chamber that protects the epithelium and enhances tear film retention, displaying notable benefits in refractory DED in SS patients [170,171]. A particularly promising strategy for severe dry eye in SS is the use of silicone-hydrogel bandage contact lenses (BCL), which improve visual acuity, ocular surface healing, and overall quality of life. In a recent comparative study, SS patients treated with BCLs for six weeks exhibited greater visual improvements than those using lubricating eye drops eight times daily. Notably, the BCL group showed sustained improvements even after therapy discontinuation, without reports of pain or keratitis (Figure 7) [172].

5.2.3. Surgical Interventions

For severe refractory SS cases with corneal damage or glandular dysfunction, surgical options become necessary. Tarsorrhaphy, the partial or complete surgical closure of the eyelids, is a traditional yet effective technique to minimize tear evaporation and protect the ocular surface [18,30,151]. Innovative procedures include minor salivary gland auto-transplantation to the conjunctival fornix, which serves as a natural tear substitute, and amniotic membrane transplantation (AMT), used for epithelial regeneration and healing of severe ocular surface defects (Figure 7) [151].

5.3. Adjunctive and Nutritional Therapies

5.3.1. Oral Administration: Omega-3 and/or Omega-6 Fatty Acid Supplementation

Essential omega-3 and omega-6 fatty acids (FAs), obtained through the diet, are important regulators of inflammation and contribute to the integrity of cell membranes. Certain omega-3 FAs, like eicosapentaenoic acid (EPA) derived from fish oil, inhibit the production of pro-inflammatory cytokines (e.g., IL-1, TNF-α). Several studies have consistently pointed out that oral administration, twice daily, of linoleic and γ-linoleic FAs, leads to significant improvement of ocular irritation symptoms and accelerates tear secretion. Similarly, preclinical trials in mice have exhibited that resolvin E1, a metabolite of EPA, effectively mitigates inflammation [25,173,174]. Compared to healthy individuals, pSS patients demonstrate lower levels of omega-3 and omega-6 FAs. The beneficial impact of omega-3 FAs is likely attributed to their conversion into resolvins and other potent anti-inflammatory mediators, such as docosahexaenoic acid (DHA) and EPA, which collectively reduce inflammation and tissue damage. Moreover, SS patients often experience dry mouth and dysgeusia, which can alter their eating behavior and lead to nutritional deficiencies, including reduced intake of vitamins and omega-3 FAs. Supplementation with omega-3 and/or omega-6 FAs has shown beneficial effects on TBUT and Schirmer’s levels, improving the overall quality of life in SS patients (Figure 7) [25,173,174].

5.3.2. The Role of Vitamins in SS Mitigation

Vitamin A
Vitamin A is actively involved in immune regulation and epithelial homeostasis. In the form of all-trans-retinoic acid in animal models, it has been shown to reduce the levels of Th1 (e.g., IL-2, IFN-γ) and Th2 cytokines (e.g., IL-4), hence exerting anti-inflammatory effects. Vitamin A deficiency is associated with the progression of systemic manifestations and disease severity in SS, while it also contributes to the instability of epithelial and mucosal barriers, exacerbating symptoms like dry eyes (Figure 7 and Figure 8) [151,175,176].
Vitamin E
Vitamin E is a lipid-soluble antioxidant with immunomodulatory properties that can delay the progression of autoimmune diseases. In autoimmune mouse models, a vitamin E-rich diet has shown beneficial effects like reduced lymph node volume, decreased cytokine levels (e.g., IL-6/10/12, TNF-α), and increased IL-1β levels. In SS patients, elevated serum vitamin E levels have been recorded (41.41 μmol/L compared to 33.68 μmol/L in healthy controls), possibly reflecting a compensatory antioxidant immune response. Such findings support vitamin E as a potential adjunctive therapeutic agent in SS symptom management (Figure 7 and Figure 8) [174,175,177].
Vitamin D
Vitamin D, mainly in its active form 1α,25-dihydroxyvitamin D3 (1α,25-(OH)2D3), is known to modulate immune function via various mechanisms. Its deficiency is associated with a higher risk of autoimmune diseases, like RA, SLE, and ILD. Interestingly, the serum concentration of vitamin D in SS patients (79.96 nmol/L) does not markedly differ from that of healthy individuals (71.57 nmol/L). Nevertheless, it plays a key role in regulating immune responses by modulating cytokine production and acting as an acute-phase reactant. Treatment with 1α,25(OH)2D3 suppressed the proliferation of Th lymphocytes and memory B cells in SS patients, suggesting its immunosuppressive and anti-inflammatory potential in SS therapy (Figure 7 and Figure 8) [35,73,178,179,180].

5.4. Novel and Emerging Therapies

5.4.1. Biologic Interventions, Targeted Therapies, and Immunomodulators

Advances in biological therapies have opened new avenues for managing SS-associated autoimmune ocular inflammation by targeting specific inflammatory cascades. Several biologic DMARDs (bDMARDs) and cell-based therapies have emerged. Although B cell-targeting SS strategies remain central, therapies focusing on T cells, cytokines, chemokines, and small molecules have also been explored [23,181,182,183].
Rituximab, an anti-CD20 monoclonal antibody, is often used off-label for severe systemic pSS [181]. It depletes B cells and mitigates autoimmunity, though clinical trials show inconsistent efficacy, indicating a need for further research [23,181,182,183]. Belimumab, a BAFF receptor inhibitor (like atacicept), shows promise in reducing autoantibody generation, dryness, and B cell hyperactivity—alone or combined with rituximab—though more data are required [23,105,184,185,186,187]. Additionally, obinutuzumab, as an anti-CD20 antibody, is a good treatment response to rituximab-resistant SS patients without any symptomatic cross-immunity [188]. Abatacept, a soluble fusion protein composed of the extracellular domain of cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and a fragment of the Fc portion of human IgG1, binds more strongly to CD80/CD86 than CD28, preventing T-cell activation. Though it showed limited effect on pSS biomarkers [189], it may reduce DED symptoms and systemic activity, especially in SS related to RA [190,191]. Plus, epratuzumab, targeting CD22, may downregulate B-cell receptor signaling and IgM, with promising results in SLE and related SS [188,192]. Baminercept (lymphotoxin β receptor (LTβ-R)-binding) and acazicolcept (CD28-binding in T cells) require further validation [188,193], while iscalimab and dazodalibep (anti-CD40 antibodies) have shown potential in reducing disease activity and dryness [104,194,195]. Ianalumab, a BAFF-R inhibitor, was well tolerated and induced dose-dependent SS activity reduction [196], with sustained B cell depletion and minimal side effects [197]. Similarly, telitacicept, a fusion protein, suppressed B cell maturation and autoantibody secretion, while improving fatigue and exocrine gland function in pSS patients [198].
TNF-α inhibitors present mixed results. Infliximab showed poor response and high discontinuation rates among anti-Ro/SSA-positive patients. Similarly, adalimumab, although potentially beneficial for SS patients who also suffer from Crohn’s disease, like etanercept and infliximab, can lead to anti-drug antibody development and does not improve SS dryness or salivary flow, discouraging further investigation [23,188,199].
Cytokine pathway targeting (e.g., IL-6, IL-17) shows promise. Secukinumab (IL-17 inhibitor) and tocilizumab (IL-6 inhibitor) are currently under investigation for their ability to suppress systemic fatigue, glandular dysfunction, systemic and ocular inflammation in SS patients. Secukinumab downregulates IL-1RA and IL-38, modulating Treg/Th17 signaling [200]. Lung involvement has been recorded in 10–20% of SS patients, hence, tocilizumab could be an alternative therapeutic strategy, used for SS-associated refractory organizing pneumonia [201]. Nevertheless, it showed limited effect on immunoglobulins, symptom relief, complement, and systemic SS activity [23,202].
Selective phosphoinositide 3-kinase (PI3K) pathway inhibitors like seletalisib, leniolisib, and parsaclisib are also promising agents in regulating SS-related B cell activation and inflammatory cytokines, as they suppressed autoantibody production, Th17 activation, type I IFN signaling, and glandular inflammation while improving salivary flow and fatigue in early trials, concurrently pointing to the need for further studies [188,203,204,205].
Bruton’s tyrosine kinase (BTK) inhibitors, namely remibrutinib and tirabrutinib, and other kinase inhibitors like lanraplenib (spleen tyrosine kinase (SYK)-binding) and filgotinib (Janus kinase (JAK) inhibitor) are early-phase SS treatment candidates that showed partial efficacy. Remibrutinib reportedly improved gene and protein expression, while tirabrutinib, lanraplenib, and filgotinib failed to meet both primary and secondary endpoints [206,207]. However, JAK inhibitors like tofacitinib and baricitinib displayed suppressive effects on IFN-Is and pro-inflammatory signaling. Tofacitinib reduced IL-6 and autophagy protein 5 (ATG5) expression in labial SG (LSG) epithelial cells of pSS patients, mitigating JAK/signal transducer and activator of transcription (STAT)-driven inflammation [188,208,209,210]. Baricitinib, targeting JAK1/2, shows potential against arthritis, ILD, and skin rash, though further evidence is needed [188,211], whereas there are ongoing trials for other JAK inhibitors as well, including deucravacitinib, a selective tyrosine kinase 2 (TYK2) inhibitor with potential anti-SS behavior [212].
Emerging strategies include interference with IFN-I pathways. Biologics like RSLV-132 and RO5459072 (cathepsin S (CatS)) [213] are under evaluation as immunomodulatory therapies. RSLV-132, a recombinant RNase Fc fusion protein, degrades extracellular RNA, reduces IFN-I gene expression, and alleviates fatigue [188,214]. All aforementioned biological and targeted inhibitors are illustrated in Figure 9.

5.4.2. Anterior and Posterior Segment Ocular Drug-Delivery Technologies

Drug delivery to the anterior eye segment (including the cornea, conjunctiva, and aqueous humor) faces challenges due to tear clearance, corneal barrier resistance, and enzymatic degradation. Non-systemic kinase inhibitors (NSKIs) like TOP1360, the tear glycoprotein lacritin, and thymosin β4 derivative RGN-259 modulate ocular inflammation and protect against focal lymphocyte infiltration. TOP1360 decreases ocular inflammation by targeting upregulated kinases in DED, like p38-α, SYK, sulfiredoxin (Srx), and lymphocyte-specific protein tyrosine kinase (Lck). Clinical trials confirm its safety, tolerability, and efficacy in relieving ocular dryness, discomfort, foreign body sensation, and grittiness [23,94,215]. Furthermore, thymosin β4-derived RGN-259 has demonstrated efficacy in neurotropic keratitis. Patients reported reduced ocular discomfort and improved tear volume production [216]. In murine models, it increased mucin and goblet cell density and reduced inflammatory factors [217]. While primary endpoints (ocular discomfort, inferior corneal staining) were unmet, secondary outcomes supported its safety and potential benefit in managing SS-related DED [23,218].
Lacritin levels are reduced in SS, correlating with corneal neuropathy, DED, reduced nerve fiber density (NFD), and length (NFL). Lacritin supports ocular surface integrity by targeting syndecan-1 (SDC1) during heparinase cleavage, offering strong diagnostic sensitivity and specificity [23,219,220]. In knockout mice, it enhanced tear secretion, lowered K10 expression, and significantly reduced focal CD4+ T cell infiltration [221]. Its active 19 amino acid fragment, Lacripep, induced clinically remarkable improvements in corneal and conjunctival staining, marking it as a promising future therapy [219,222].
Drug delivery to the posterior eye segment (comprising the retina, choroid, and vitreous humor) is harder to target than the anterior one, as it is limited by the blood-retinal barrier. Intravitreal biodegradable implants, such as fluocinolone acetonide, allow sustained anti-inflammatory drug release and reduce injection frequency. They show great promise for managing severe autoimmune corneal disease cases (Figure 10) [23,223].

5.4.3. Gene Therapy and Epigenetic Modulation

Advances in gene-editing technologies, particularly CRISPR-Cas9, have opened new avenues for targeting SS pathogenic cytokine expression, aiming to halt disease progression and correct immune dysregulation. Current studies are exploring the genetic silencing of persistently overactive SS patients, with IFN-regulated genes. Gene therapy targeting pro-inflammatory cytokines (e.g., TNF-α, IL-6) is being investigated for its potential to “reprogram” immune responses [188,224]. Moreover, epigenetic therapies that modulate DNA methylation and microRNA expression are promising in reducing autoimmune hyperactivity and restoring lacrimal gland function (Figure 10) [75,97,98,99,100,188,224].

5.4.4. Contact Lens Drug-Delivery and Colloidal Nanocarriers

Conventional treatments for DED and other ocular complications, like eye drops, often face limitations in achieving controlled and effective drug absorption. Therefore, there is a pressing need for targeted and sustained drug delivery systems to enhance therapeutic outcomes. Therapeutic CLs loaded with active agents (e.g., HA, CyA) into their porous matrix offer prolonged contact time, controlled and effective drug release. They can deliver anti-inflammatory (e.g., dexamethasone (Dex)), osmoprotective, and secretagogue substances, alongside rewetting agents, thus improving comfort and treatment adherence. Compared to standard eye drops, therapeutic CLs provide longer drug retention, better patient compliance, and reduced ocular irritation and toxicity [23,225,226].
Colloidal nanocarriers (CNs) represent another innovative approach in ocular drug delivery for SS-related DED. Unlike conventional eye drops that suffer from rapid tear clearance and low bioavailability, CNs enhance corneal penetration, prolong ocular surface retention, and enable controlled drug release. Various CNs under investigation include liposomes that encapsulate both hydrophilic and lipophilic compounds (i.e., azithromycin, CyA), polymeric nanoparticles (NPs) (e.g., polylactic acid (PLA), Dex, chitosan, HA), nano-micelles, nanowafers, microneedles, and dendrimers of high-drug loading capacity, as illustrated in Figure 10 [23,227,228,229,230,231,232,233,234,235].

5.4.5. Stem Cell Therapy and Regenerative Approaches

Emerging regenerative strategies for SS focus on mesenchymal stem cells (MSCs) due to their immunomodulatory and tissue-regenerative properties. MSCs reduce lymphocytic infiltration, enhance anti-inflammatory cytokine production, promote epithelial regeneration, and restore tear and glandular function. Their immunomodulatory effects on T and B cells, DCs, and NKs—partly via regulation of the T cell immunoglobulin and mucin-domain containing-3 (Tim-3) inhibitor—have shown promise in alleviating SS symptoms [236,237,238]. Specifically, MSCs suppress Th1 and Th17 lymphocytes, disrupt T-B cell interplay, downregulate autoantibody formation, and promote survival of injured and inflamed lacrimal and salivary glands [26,236].
Dental pulp-derived stem cells (DPSCs) have exhibited superior effects over bone marrow MSCs (BMMSCs) in reversing hyposalivation, reducing inflammation, and minimizing glandular fibrosis [239,240,241,242,243]. Similarly, umbilical MSCs (UCMSCs) regulate immune activity by promoting CD4+/Forkhead box P3 (Fox3+) T cells, inducing Th17 apoptosis, and improving Tregs/Th17 balance [244]. UCMSCs also suppressed Vγ4+IL-17+, increasing saliva flow, regulating Treg/Th17 immunity, and lowering pulmonary inflammation [245,246]. However, CLs have also been linked to limbal stem cell deficiency (LSCD). Drug-eluting CLs may help prevent or slow LSDC progression [247,248].
A revolutionary approach involves stem cell-coated CLs (often limbal epithelial stem cells) to enhance corneal repair and epithelial regeneration in severe ocular surface disease [238,247,248,249,250]. Advanced high-oxygen, permeable hydrogel CLs coated with UCMSCs have demonstrated long-term retention on the ocular surface, reduction of corneal inflammation, inhibition of Th1/Th17 differentiation, and promotion of Treg conversion in models of ocular graft-versus-host disease (oGVHD) and potentially SS [251,252]. Nevertheless, broader clinical adoption is hindered by donor compatibility issues and lengthy preparation times, requiring further validation [23,253].
To overcome these challenges, exosome-based therapies (e.g., exosome extracellular vesicles) have emerged as a non-invasive alternative. Stem cell-derived extracellular vehicles (EVs) carry bioactive molecules that promote epithelial healing, immune regulation, and inflammation suppression [253,254]. These exosomes, with stable fatty acid membranes, can cross dense biological barriers and restore glandular function, ensuring stability and structural rigidity [23,148,253]. For example, labial gland-derived MSCs (LGMSCs) and their exosomes reduce inflammation by promoting Treg differentiation and inhibiting Th17 cells, offering a cell-free SS therapeutic approach [255]. LGMSC-derived exosomal miR-125b has been beneficial towards restoring salivary secretion, reducing CD19+CD20CD27+CD38+, and regulating pSS plasma cells by targeting the PR domain zinc finger protein 1 (PRDM1) [255,256,257]. Likewise, olfactory ecto-MSC-derived exosomes ameliorated SS in murine models by modulating myeloid-derived suppressor cells (MDSCs), upregulating arginase expression and ROS levels, thereby mitigating disease progression [258]. Despite these advancements, extensive clinical trials are essential before systemic application of MSCs and their exosomes (Figure 10) [258].

6. Future Directions, Perspectives, and Research

SS is a multifactorial autoimmune disease with substantial unmet clinical needs. A paradigm shift toward precision medicine, targeted immunotherapies, and regenerative strategies is essential. Current treatments such as immunosuppressants, glucocorticoids, and csDMARDs offer symptom relief but do not address the disease’s root cause, which remains unclear. Moreover, SS-specific therapeutics are still lacking, and existing drugs require further validation. Identifying predictive biomarkers like IFN signatures, BAFF levels, and genetic/epigenetic markers may enhance early, individualized diagnosis and predict treatment [9,54,55,56,57,58,61,64]. Novel immunomodulatory agents, including BTK (e.g., remibrutinib, tirabrutinib), PI3K (e.g., seletalisib, parsaclisib), and JAK inhibitors (e.g., tofacitinib, baricitinib), have shown great promise in reducing B-cell hyperactivity, cytokine-driven ocular inflammation, and systemic complications [203,204,206,207,208,209,210,211,227,229]. Further exploration of cytokines such as IL-1β, -6,-17, -23, 38, and TNF-α, and their role in T and B cell regulation, is critical for developing precise immune interventions [22,23,188].
MSCs and exosome-based therapies hold vast potential for restoring exocrine gland function. However, standardized protocols, long-term safety data, and well-structured clinical trials are needed to translate these therapies into routine care [26,237,238,249,253,254,255,256,258]. In the realm of gene and epigenetic therapy, tools like CRISPR-Cas9 and miRNA modulation offer potential for altering SS-related gene expression, but delivery methods and safety profiles must be further refined [97,98,99,100,224,256]. Emerging evidence also highlights the role of gut dysbiosis in SS-related inflammation, indicating potential for microbiota-based adjunct therapies [117]. In parallel, novel approaches such as immune cell engineering, in vitro autoimmune models, immunophenotyping, and artificial intelligence (AI)-driven diagnostic platforms may revolutionize SS research [22,188].
Accelerating the approval of novel therapies will require an optimal clinical trial design, including early predictors of treatment response, biomarkers for drug efficacy evaluation, standardized disease activity scoring, and minimization of background therapy confounding effects. Ultimately, establishing safe, effective, and personalized treatment protocols, based on symptom-specific combination therapies and novel drug candidates, is expected to mark a turning point in SS care and clinical outcomes [22,188].

7. Concluding Remarks

Sjögren’s Syndrome is a chronic systemic autoimmune condition marked by complex ocular and systemic manifestations. Its heterogeneous inflammatory mechanisms, especially in patients with overlapping autoimmune disorders, contribute to important diagnostic and therapeutic challenges. Timely diagnosis and tailored therapeutic strategies are crucial for mitigating complications such as scleritis and corneal damage. Currently, a diverse spectrum of mitigation options—from artificial tears or corticosteroids to advanced biologic agents and surgical interventions—allows for personalized SS management.
Looking ahead, emerging therapies targeting neuroinflammation, oxidative stress, and epigenetic regulation offer hope for more effective treatments of both systemic and ocular SS complications. Nonetheless, large-scale clinical studies remain essential to validate these approaches and integrate them into evidence-based care. Despite notable progress, the underlying immunopathology of SS remains incompletely understood, particularly regarding innate immune activation, cytokine regulation, and lacrimal gland dysfunction. This review sought to elucidate the complex autoimmune mechanisms of SS, critically assess current SS treatment strategies, and highlight innovative remedies with potential clinical impact. As research continues to evolve, the integration of targeted, regenerative, and individualized therapies could pave the way for improving management and long-term outcomes in SS patients.

Author Contributions

Conceptualization, A.T.; methodology, K.P., T.A. and A.T.; software, K.P., T.A. and A.T.; validation, A.T.; investigation, K.P., T.A. and A.T.; writing—original draft preparation, K.P., T.A. and A.T.; writing—review and editing, T.A., C.M., K.G., V.D., X.K. and A.T.; visualization, A.T.; supervision, A.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

The authors would like to thank the School of Chemistry, Faculty of Sciences of the Democritus University of Thrace, and the General Hospital of Kavala, Greece, for their continuous support.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Brito-Zerón, P.; Baldini, C.; Bootsma, H.; Bowman, S.J.; Jonsson, R.; Mariette, X.; Sivils, K.; Theander, E.; Tzioufas, A.; Ramos-Casals, M. Sjögren Syndrome. Nat. Rev. Dis. Primers 2016, 2, 16047. [Google Scholar] [CrossRef] [PubMed]
  2. Kelly, A.L.; Nelson, R.J.; Sara, R.; Alberto, S. Sjögren Syndrome: New Insights in the Pathogenesis and Role of Nuclear Medicine. J. Clin. Med. 2022, 11, 5227. [Google Scholar] [CrossRef]
  3. Rodrigues, A.R.; Soares, R. Inflammation in Sjögren’s Syndrome: Cause or Consequence? Autoimmunity 2017, 50, 141–150. [Google Scholar] [CrossRef]
  4. Chang, Y.-J.; Tseng, J.-C.; Leong, P.-Y.; Wang, Y.-H.; Wei, J.C.-C. Increased Risk of Sjögren’s Syndrome in Patients with Obsessive-Compulsive Disorder: A Nationwide Population-Based Cohort Study. Int. J. Environ. Res. Public Health 2021, 18, 5936. [Google Scholar] [CrossRef] [PubMed]
  5. Horai, Y.; Shimizu, T.; Nakamura, H.; Kawakami, A. Recent Advances in Pathogenesis, Diagnostic Imaging, and Treatment of Sjögren’s Syndrome. J. Clin. Med. 2024, 13, 6688. [Google Scholar] [CrossRef]
  6. Yamamoto, K. Pathogenesis of Sjögren’s Syndrome. Autoimmun. Rev. 2003, 2, 13–18. [Google Scholar] [CrossRef] [PubMed]
  7. Negrini, S.; Emmi, G.; Greco, M.; Borro, M.; Sardanelli, F.; Murdaca, G.; Indiveri, F.; Puppo, F. Sjögren’s Syndrome: A Systemic Autoimmune Disease. Clin. Exp. Med. 2022, 22, 9–25. [Google Scholar] [CrossRef]
  8. Miyamoto, S.T.; Lendrem, D.W.; Ng, W.-F.; Hackett, K.L.; Valim, V. Managing Fatigue in Patients with Primary Sjögren’s Syndrome: Challenges and Solutions. Open Access Rheumatol. 2019, 11, 77–88. [Google Scholar] [CrossRef]
  9. 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]
  10. Chaigne, B.; Lasfargues, G.; Marie, I.; Hüttenberger, B.; Lavigne, C.; Marchand-Adam, S.; Maillot, F.; Diot, E. Primary Sjögren’s Syndrome and Occupational Risk Factors: A Case–Control Study. J. Autoimmun. 2015, 60, 80–85. [Google Scholar] [CrossRef]
  11. Parisis, D.; Chivasso, C.; Perret, J.; Soyfoo, M.S.; Delporte, C. Current State of Knowledge on Primary Sjögren’s Syndrome, an Autoimmune Exocrinopathy. J. Clin. Med. 2020, 9, 2299. [Google Scholar] [CrossRef]
  12. Liang, Y.; Yang, Z.; Qin, B.; Zhong, R. Primary Sjogren’s Syndrome and Malignancy Risk: A Systematic Review and Meta-Analysis. Ann. Rheum. Dis. 2014, 73, 1151–1156. [Google Scholar] [CrossRef] [PubMed]
  13. Nishishinya, M.B.; Pereda, C.A.; Muñoz-Fernández, S.; Pego-Reigosa, J.M.; Rúa-Figueroa, I.; Andreu, J.-L.; Fernández-Castro, M.; Rosas, J.; Loza Santamaría, E. Identification of Lymphoma Predictors in Patients with Primary Sjögren’s Syndrome: A Systematic Literature Review and Meta-Analysis. Rheumatol. Int. 2015, 35, 17–26. [Google Scholar] [CrossRef] [PubMed]
  14. Akpek, E.K.; Bunya, V.Y.; Saldanha, I.J. Sjögren’s Syndrome: More Than Just Dry Eye. Cornea 2019, 38, 658–661. [Google Scholar] [CrossRef] [PubMed]
  15. Skarlis, C.; Raftopoulou, S.; Mavragani, C.P. Sjogren’s Syndrome: Recent Updates. J. Clin. Med. 2022, 11, 399. [Google Scholar] [CrossRef]
  16. Björk, A.; Mofors, J.; Wahren-Herlenius, M. Environmental Factors in the Pathogenesis of Primary Sjögren’s Syndrome. J. Intern. Med. 2020, 287, 475–492. [Google Scholar] [CrossRef]
  17. Aviña-Zubieta, J.A.; Jansz, M.; Sayre, E.C.; Choi, H.K. The Risk of Deep Venous Thrombosis and Pulmonary Embolism in Primary Sjögren Syndrome: A General Population-Based Study. J. Rheumatol. 2017, 44, 1184–1189. [Google Scholar] [CrossRef]
  18. Barrientos, R.T.; Godín, F.; Rocha-De-Lossada, C.; Soifer, M.; Sánchez-González, J.-M.; Moreno-Toral, E.; González, A.-L.; Zein, M.; Larco, P.; Mercado, C.; et al. Ophthalmological Approach for the Diagnosis of Dry Eye Disease in Patients with Sjögren’s Syndrome. Life 2022, 12, 1899. [Google Scholar] [CrossRef]
  19. Roszkowska, A.M.; Oliverio, G.W.; Aragona, E.; Inferrera, L.; Severo, A.A.; Alessandrello, F.; Spinella, R.; Postorino, E.I.; Aragona, P. Ophthalmologic Manifestations of Primary Sjögren’s Syndrome. Genes 2021, 12, 365. [Google Scholar] [CrossRef]
  20. Michaelov, E.; McKenna, C.; Ibrahim, P.; Nayeni, M.; Dang, A.; Mather, R. Sjögren’s Syndrome Associated Dry Eye: Impact on Daily Living and Adherence to Therapy. J. Clin. Med. 2022, 11, 2809. [Google Scholar] [CrossRef]
  21. Zoukhri, D. Effect of Inflammation on Lacrimal Gland Function. Exp. Eye Res. 2006, 82, 885–898. [Google Scholar] [CrossRef]
  22. Hyon, J.Y.; Lee, Y.J.; Yun, P.-Y. Management of Ocular Surface Inflammation in Sjögren Syndrome. Cornea 2007, 26, S13–S15. [Google Scholar] [CrossRef]
  23. Wu, K.Y.; Chen, W.T.; Chu-Bédard, Y.-K.; Patel, G.; Tran, S.D. Management of Sjogren’s Dry Eye Disease—Advances in Ocular Drug Delivery Offering a New Hope. Pharmaceutics 2023, 15, 147. [Google Scholar] [CrossRef] [PubMed]
  24. Murray, P.I.; Rauz, S. The Eye and Inflammatory Rheumatic Diseases: The Eye and Rheumatoid Arthritis, Ankylosing Spondylitis, Psoriatic Arthritis. Best Pract. Res. Clin. Rheumatol. 2016, 30, 802–825. [Google Scholar] [CrossRef] [PubMed]
  25. Coursey, T.G.; de Paiva, C.S. Managing Sjögren’s Syndrome and Non-Sjögren Syndrome Dry Eye with Anti-Inflammatory Therapy. Clin. Ophthalmol. 2014, 8, 1447–1458. [Google Scholar] [CrossRef]
  26. Harrell, C.R.; Volarevic, A.; Arsenijevic, A.; Djonov, V.; Volarevic, V. Targeted Therapy for Severe Sjogren’s Syndrome: A Focus on Mesenchymal Stem Cells. Int. J. Mol. Sci. 2024, 25, 13712. [Google Scholar] [CrossRef] [PubMed]
  27. Kalk, W.; Vissink, A.; Spijkervet, F.; Bootsma, H.; Kallenberg, C.; Amerongen, A. Sialometry and Sialochemistry: Diagnostic Tools for Sjögren’s Syndrome. Ann. Rheum. Dis. 2001, 60, 1110–1116. [Google Scholar] [CrossRef]
  28. Whitcher, J.P.; Shiboski, C.H.; Shiboski, S.C.; Heidenreich, A.M.; Kitagawa, K.; Zhang, S.; Hamann, S.; Larkin, G.; McNamara, N.A.; Greenspan, J.S.; et al. A Simplified Quantitative Method for Assessing Keratoconjunctivitis Sicca from the Sjögren’s Syndrome International Registry. Am. J. Ophthalmol. 2010, 149, 405–415. [Google Scholar] [CrossRef]
  29. Lin, H.; Yiu, S.C. Dry Eye Disease: A Review of Diagnostic Approaches and Treatments. Saudi J. Ophthalmol. 2014, 28, 173–181. [Google Scholar] [CrossRef]
  30. Bjordal, O.; Norheim, K.B.; Rødahl, E.; Jonsson, R.; Omdal, R. Primary Sjögren’s Syndrome and the Eye. Surv. Ophthalmol. 2020, 65, 119–132. [Google Scholar] [CrossRef]
  31. Shiboski, C.H.; Shiboski, S.C.; Seror, R.; Criswell, L.A.; Labetoulle, M.; Lietman, T.M.; Rasmussen, A.; Scofield, H.; Vitali, C.; Bowman, S.J.; et al. 2016 American College of Rheumatology/European League Against Rheumatism Classification Criteria for Primary Sjögren’s Syndrome: A Consensus and Data-Driven Methodology Involving Three International Patient Cohorts. Arthritis Rheumatol. 2017, 69, 35–45. [Google Scholar] [CrossRef] [PubMed]
  32. Franceschini, F.; Cavazzana, I.; Andreoli, L.; Tincani, A. The 2016 Classification Criteria for Primary Sjogren’s Syndrome: What’s New? BMC Med. 2017, 15, 69. [Google Scholar] [CrossRef] [PubMed]
  33. Zeng, W.; Zhou, X.; Yu, S.; Liu, R.; Quek, C.W.N.; Yu, H.; Tay, R.Y.K.; Lin, X.; Feng, Y. The Future of Targeted Treatment of Primary Sjögren’s Syndrome: A Focus on Extra-Glandular Pathology. Int. J. Mol. Sci. 2022, 23, 14135. [Google Scholar] [CrossRef]
  34. Priani, D.; Muhiddin, H.S.; Sirajuddin, J.; Eka, H.B.; Bahar, B.; Bukhari, A. Effectiveness of Topical Cyclosporin-A 0.1% Compared to Combined Topical Cyclosporin-A 0.1% with Topical Sodium Hyaluronate on Interleukin-6 Levels in the Tears of Patients with Dry Eye Disease. Vision 2023, 7, 31. [Google Scholar] [CrossRef] [PubMed]
  35. Radić, M.; Kolak, E.; Đogaš, H.; Gelemanović, A.; Bučan Nenadić, D.; Vučković, M.; Radić, J. Vitamin D and Sjögren’s Disease: Revealing the Connections—A Systematic Review and Meta-Analysis. Nutrients 2023, 15, 497. [Google Scholar] [CrossRef]
  36. Kassan, S.S.; Moutsopoulos, H.M. Clinical Manifestations and Early Diagnosis of Sjögren Syndrome. Arch. Intern. Med. 2004, 164, 1275–1284. [Google Scholar] [CrossRef]
  37. Yura, Y.; Hamada, M. Outline of Salivary Gland Pathogenesis of Sjögren’s Syndrome and Current Therapeutic Approaches. Int. J. Mol. Sci. 2023, 24, 11179. [Google Scholar] [CrossRef]
  38. D’Souza, S.; Vaidya, T.; Nair, A.P.; Shetty, R.; Kumar, N.R.; Bisht, A.; Panigrahi, T.; J, T.S.; Khamar, P.; Dickman, M.M.; et al. Altered Ocular Surface Health Status and Tear Film Immune Profile Due to Prolonged Daily Mask Wear in Health Care Workers. Biomedicines 2022, 10, 1160. [Google Scholar] [CrossRef]
  39. Schlegel, I.; De Goüyon Matignon de Pontourade, C.M.F.; Lincke, J.-B.; Keller, I.; Zinkernagel, M.S.; Zysset-Burri, D.C. The Human Ocular Surface Microbiome and Its Associations with the Tear Proteome in Dry Eye Disease. Int. J. Mol. Sci. 2023, 24, 14091. [Google Scholar] [CrossRef]
  40. Hughes, G.K.; Miszkiel, K.A. Imaging of the Lacrimal Gland. Semin. Ultrasound CT MRI 2006, 27, 476–491. [Google Scholar] [CrossRef]
  41. Muntean, D.D.; Bădărînză, M.; Ștefan, P.A.; Lenghel, M.L.; Rusu, G.M.; Csutak, C.; Coroian, P.A.; Lupean, R.A.; Fodor, D. The Diagnostic Value of MRI-Based Radiomic Analysis of Lacrimal Glands in Patients with Sjögren’s Syndrome. Int. J. Mol. Sci. 2022, 23, 10051. [Google Scholar] [CrossRef]
  42. Conrady, C.D.; Joos, Z.P.; Patel, B.C.K. Review: The Lacrimal Gland and Its Role in Dry Eye. J. Ophthalmol. 2016, 2016, 7542929. [Google Scholar] [CrossRef] [PubMed]
  43. Hat, K.; Planinić, A.; Ježek, D.; Kaštelan, S. Expression of Androgen and Estrogen Receptors in the Human Lacrimal Gland. Int. J. Mol. Sci. 2023, 24, 5609. [Google Scholar] [CrossRef] [PubMed]
  44. Wang, S.; Kahale, F.; Naderi, A.; Surico, P.L.; Yin, J.; Dohlman, T.; Chen, Y.; Dana, R. Therapeutic Effects of Stimulating the Melanocortin Pathway in Regulating Ocular Inflammation and Cell Death. Biomolecules 2024, 14, 169. [Google Scholar] [CrossRef]
  45. Delcroix, V.; Mauduit, O.; Yang, M.; Srivastava, A.; Umazume, T.; de Paiva, C.S.; Shestopalov, V.I.; Dartt, D.A.; Makarenkova, H.P. Lacrimal Gland Epithelial Cells Shape Immune Responses through the Modulation of Inflammasomes and Lipid Metabolism. Int. J. Mol. Sci. 2023, 24, 4309. [Google Scholar] [CrossRef] [PubMed]
  46. Pflugfelder, S.C. What Causes Dryness in Sjögren’s Syndrome Patients and How Can It Be Targeted? Expert. Rev. Clin. Immunol. 2014, 10, 425–427. [Google Scholar] [CrossRef]
  47. Fan, Q.; Yan, R.; Li, Y.; Lu, L.; Liu, J.; Li, S.; Fu, T.; Xue, Y.; Liu, J.; Li, Z. Exploring Immune Cell Diversity in the Lacrimal Glands of Healthy Mice: A Single-Cell RNA-Sequencing Atlas. Int. J. Mol. Sci. 2024, 25, 1208. [Google Scholar] [CrossRef]
  48. Murashima, A.d.A.B.; Sant’Ana, A.M.S.; Faustino-Barros, J.F.; Machado Filho, E.B.; da Silva, L.C.M.; Fantucci, M.Z.; Módulo, C.M.; Chahud, F.; Garcia, D.M.; Rocha, E.M. Exorbital Lacrimal Gland Ablation and Regrafting Induce Inflammation but Not Regeneration or Dry Eye. Int. J. Mol. Sci. 2024, 25, 8318. [Google Scholar] [CrossRef]
  49. 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]
  50. Lemos, C.N.; da Silva, L.E.C.M.; Faustino, J.F.; Fantucci, M.Z.; Murashima, A.d.A.B.; Adriano, L.; Alves, M.; Rocha, E.M. Oxidative Stress in the Protection and Injury of the Lacrimal Gland and the Ocular Surface: Are There Perspectives for Therapeutics? Front. Cell Dev. Biol. 2022, 10, 824726. [Google Scholar] [CrossRef]
  51. Nakamura, H.; Shimizu, T.; Takagi, Y.; Takahashi, Y.; Horai, Y.; Nakashima, Y.; Sato, S.; Shiraishi, H.; Nakamura, T.; Fukuoka, J.; et al. Reevaluation for Clinical Manifestations of HTLV-I-Seropositive Patients with Sjögren’s Syndrome. BMC Musculoskelet. Disord. 2015, 16, 335. [Google Scholar] [CrossRef]
  52. Yoo, S.-A.; Kwok, S.-K.; Kim, W.-U. Proinflammatory Role of Vascular Endothelial Growth Factor in the Pathogenesis of Rheumatoid Arthritis: Prospects for Therapeutic Intervention. Mediat. Inflamm. 2008, 2008, 129873. [Google Scholar] [CrossRef] [PubMed]
  53. Chan, J.; Lim, G.; Lee, R.; Tong, L. A Systematic Review of Tear Vascular Endothelial Growth Factor and External Eye Diseases. Int. J. Mol. Sci. 2024, 25, 1369. [Google Scholar] [CrossRef] [PubMed]
  54. Nordmark, G.; Alm, G.V.; Rönnblom, L. Mechanisms of Disease: Primary Sjögren’s Syndrome and the Type I Interferon System. Nat. Clin. Pract. Rheumatol. 2006, 2, 262–269. [Google Scholar] [CrossRef]
  55. Del Papa, N.; Minniti, A.; Lorini, M.; Carbonelli, V.; Maglione, W.; Pignataro, F.; Montano, N.; Caporali, R.; Vitali, C. The Role of Interferons in the Pathogenesis of Sjögren’s Syndrome and Future Therapeutic Perspectives. Biomolecules 2021, 11, 251. [Google Scholar] [CrossRef]
  56. Sun, Y.; Lin, J.; Chen, W. Type I Interferons in the Pathogenesis and Treatment of Sjögren’s Syndrome: An Update. Rheumatology 2022, 9, 59–69. [Google Scholar] [CrossRef]
  57. Platanias, L.C. Mechanisms of Type-I- and Type-II-Interferon-Mediated Signalling. Nat. Rev. Immunol. 2005, 5, 375–386. [Google Scholar] [CrossRef]
  58. Rönnblom, L.; Eloranta, M.-L. The Interferon Signature in Autoimmune Diseases. Curr. Opin. Rheumatol. 2013, 25, 248–253. [Google Scholar] [CrossRef]
  59. Bedei, I.A.; Kniess, D.; Keil, C.; Wolter, A.; Schenk, J.; Sachs, U.J.; Axt-Fliedner, R. Monitoring of Women with Anti-Ro/SSA and Anti-La/SSB Antibodies in Germany—Status Quo and Intensified Monitoring Concepts. J. Clin. Med. 2024, 13, 1142. [Google Scholar] [CrossRef]
  60. Parisis, D.; Sarrand, J.; Cabrol, X.; Delporte, C.; Soyfoo, M.S. Clinical Profile of Patients with Primary Sjögren’s Syndrome with Non-Identified Antinuclear Autoantibodies. Diagnostics 2024, 14, 935. [Google Scholar] [CrossRef]
  61. Massa, C.; Wang, Y.; Marr, N.; Seliger, B. Interferons and Resistance Mechanisms in Tumors and Pathogen-Driven Diseases—Focus on the Major Histocompatibility Complex (MHC) Antigen Processing Pathway. Int. J. Mol. Sci. 2023, 24, 6736. [Google Scholar] [CrossRef] [PubMed]
  62. Bhujel, B.; Oh, S.-H.; Kim, C.-M.; Yoon, Y.-J.; Chung, H.-S.; Ye, E.-A.; Lee, H.; Kim, J.-Y. Current Advances in Regenerative Strategies for Dry Eye Diseases: A Comprehensive Review. Bioengineering 2024, 11, 39. [Google Scholar] [CrossRef] [PubMed]
  63. Honing, D.Y.; Luiten, R.M.; Matos, T.R. Regulatory T Cell Dysfunction in Autoimmune Diseases. Int. J. Mol. Sci. 2024, 25, 7171. [Google Scholar] [CrossRef]
  64. Lapenta, C.; Gabriele, L.; Santini, S.M. IFN-Alpha-Mediated Differentiation of Dendritic Cells for Cancer Immunotherapy: Advances and Perspectives. Vaccines 2020, 8, 617. [Google Scholar] [CrossRef] [PubMed]
  65. Kurosawa, M.; Shikama, Y.; Furukawa, M.; Arakaki, R.; Ishimaru, N.; Matsushita, K. Chemokines Up-Regulated in Epithelial Cells Control Senescence-Associated T Cell Accumulation in Salivary Glands of Aged and Sjögren’s Syndrome Model Mice. Int. J. Mol. Sci. 2021, 22, 2302. [Google Scholar] [CrossRef]
  66. Witas, R.; Gupta, S.; Nguyen, C.Q. Contributions of Major Cell Populations to Sjögren’s Syndrome. J. Clin. Med. 2020, 9, 3057. [Google Scholar] [CrossRef]
  67. Lee, N.Y.; Ture, H.Y.; Lee, E.J.; Jang, J.A.; Kim, G.; Nam, E.J. Syndecan-1 Plays a Role in the Pathogenesis of Sjögren’s Disease by Inducing B-Cell Chemotaxis through CXCL13–Heparan Sulfate Interaction. Int. J. Mol. Sci. 2024, 25, 9375. [Google Scholar] [CrossRef]
  68. Bu, J.; Liu, Y.; Zhang, R.; Lin, S.; Zhuang, J.; Sun, L.; Zhang, L.; He, H.; Zong, R.; Wu, Y.; et al. Potential New Target for Dry Eye Disease—Oxidative Stress. Antioxidants 2024, 13, 422. [Google Scholar] [CrossRef]
  69. Peng, J.; Feinstein, D.; DeSimone, S.; Gentile, P. A Review of the Tear Film Biomarkers Used to Diagnose Sjogren’s Syndrome. Int. J. Mol. Sci. 2024, 25, 10380. [Google Scholar] [CrossRef]
  70. Dammak, A.; Pastrana, C.; Martin-Gil, A.; Carpena-Torres, C.; Peral Cerda, A.; Simovart, M.; Alarma, P.; Huete-Toral, F.; Carracedo, G. Oxidative Stress in the Anterior Ocular Diseases: Diagnostic and Treatment. Biomedicines 2023, 11, 292. [Google Scholar] [CrossRef]
  71. De Oliveira, F.R.; Fantucci, M.Z.; Adriano, L.; Valim, V.; Cunha, T.M.; Louzada-Junior, P.; Rocha, E.M. Neurological and Inflammatory Manifestations in Sjögren’s Syndrome: The Role of the Kynurenine Metabolic Pathway. Int. J. Mol. Sci. 2018, 19, 3953. [Google Scholar] [CrossRef] [PubMed]
  72. Módis, L.V.; Aradi, Z.; Horváth, I.F.; Bencze, J.; Papp, T.; Emri, M.; Berényi, E.; Bugán, A.; Szántó, A. Central Nervous System Involvement in Primary Sjögren’s Syndrome: Narrative Review of MRI Findings. Diagnostics 2023, 13, 14. [Google Scholar] [CrossRef]
  73. Mihai, A.; Chitimus, D.M.; Jurcut, C.; Blajut, F.C.; Opris-Belinski, D.; Caruntu, C.; Ionescu, R.; Caruntu, A. Comparative Analysis of Hematological and Immunological Parameters in Patients with Primary Sjögren’s Syndrome and Peripheral Neuropathy. J. Clin. Med. 2023, 12, 3672. [Google Scholar] [CrossRef] [PubMed]
  74. Du, W.; Han, M.; Zhu, X.; Xiao, F.; Huang, E.; Che, N.; Tang, X.; Zou, H.; Jiang, Q.; Lu, L. The Multiple Roles of B Cells in the Pathogenesis of Sjögren’s Syndrome. Front. Immunol. 2021, 12, 684999. [Google Scholar] [CrossRef]
  75. Bruno, D.; Tolusso, B.; Lugli, G.; Di Mario, C.; Petricca, L.; Perniola, S.; Bui, L.; Benvenuto, R.; Ferraccioli, G.; Alivernini, S.; et al. B-Cell Activation Biomarkers in Salivary Glands Are Related to Lymphomagenesis in Primary Sjögren’s Disease: A Pilot Monocentric Exploratory Study. Int. J. Mol. Sci. 2024, 25, 3259. [Google Scholar] [CrossRef] [PubMed]
  76. Mohammadnezhad, L.; Shekarkar Azgomi, M.; La Manna, M.P.; Guggino, G.; Botta, C.; Dieli, F.; Caccamo, N. B-Cell Receptor Signaling Is Thought to Be a Bridge between Primary Sjogren Syndrome and Diffuse Large B-Cell Lymphoma. Int. J. Mol. Sci. 2023, 24, 8385. [Google Scholar] [CrossRef]
  77. Ambrus, J.L.; Suresh, L.; Peck, A. Multiple Roles for B-Lymphocytes in Sjogren’s Syndrome. J. Clin. Med. 2016, 5, 87. [Google Scholar] [CrossRef]
  78. Stergiou, I.E.; Poulaki, A.; Voulgarelis, M. Pathogenetic Mechanisms Implicated in Sjögren’s Syndrome Lymphomagenesis: A Review of the Literature. J. Clin. Med. 2020, 9, 3794. [Google Scholar] [CrossRef]
  79. Stern, M.E.; Schaumburg, C.S.; Dana, R.; Calonge, M.; Niederkorn, J.Y.; Pflugfelder, S.C. Autoimmunity at the Ocular Surface: Pathogenesis and Regulation. Mucosal Immunol. 2010, 3, 425–442. [Google Scholar] [CrossRef]
  80. Reyes, J.L.; Vannan, D.T.; Eksteen, B.; Avelar, I.J.; Rodríguez, T.; González, M.I.; Mendoza, A.V. Innate and Adaptive Cell Populations Driving Inflammation in Dry Eye Disease. Mediat. Inflamm. 2018, 2018, 2532314. [Google Scholar] [CrossRef]
  81. Michée-Cospolite, M.; Boudigou, M.; Grasseau, A.; Simon, Q.; Mignen, O.; Pers, J.-O.; Cornec, D.; Le Pottier, L.; Hillion, S. Molecular Mechanisms Driving IL-10- Producing B Cells Functions: STAT3 and c-MAF as Underestimated Central Key Regulators? Front. Immunol. 2022, 13, 818814. [Google Scholar] [CrossRef]
  82. 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. [Google Scholar] [CrossRef]
  83. Blinova, V.G.; Vasilyev, V.I.; Rodionova, E.B.; Zhdanov, D.D. The Role of Regulatory T Cells in the Onset and Progression of Primary Sjögren’s Syndrome. Cells 2023, 12, 1359. [Google Scholar] [CrossRef]
  84. Vílchez-Oya, F.; Balastegui Martin, H.; García-Martínez, E.; Corominas, H. Not All Autoantibodies Are Clinically Relevant. Classic and Novel Autoantibodies in Sjögren’s Syndrome: A Critical Review. Front. Immunol. 2022, 13, 1003054. [Google Scholar] [CrossRef] [PubMed]
  85. Urbanski, G.; Gury, A.; Jeannin, P.; Chevailler, A.; Lozac’h, P.; Reynier, P.; Lavigne, C.; Lacout, C.; Vinatier, E. Discordant Predictions of Extraglandular Involvement in Primary Sjögren’s Syndrome According to the Anti-SSA/Ro60 Antibodies Detection Assay in a Cohort Study. J. Clin. Med. 2022, 11, 242. [Google Scholar] [CrossRef] [PubMed]
  86. Veenbergen, S.; Kozmar, A.; van Daele, P.L.A.; Schreurs, M.W.J. Autoantibodies in Sjögren’s Syndrome and Its Classification Criteria. J. Transl. Autoimmun. 2022, 5, 100138. [Google Scholar] [CrossRef]
  87. Kyriakidis, N.C.; Kapsogeorgou, E.K.; Tzioufas, A.G. A Comprehensive Review of Autoantibodies in Primary Sjögren’s Syndrome: Clinical Phenotypes and Regulatory Mechanisms. J. Autoimmun. 2014, 51, 67–74. [Google Scholar] [CrossRef]
  88. Mahla, R.S.; Jones, E.L.; Dustin, L.B. Ro60—Roles in RNA Processing, Inflammation, and Rheumatic Autoimmune Diseases. Int. J. Mol. Sci. 2024, 25, 7705. [Google Scholar] [CrossRef] [PubMed]
  89. Fayyaz, A.; Kurien, B.T.; Scofield, R.H. Autoantibodies in Sjögren’s Syndrome. Rheum. Dis. Clin. N. Am. 2016, 42, 419–434. [Google Scholar] [CrossRef]
  90. Tiwari, V.; Jandu, J.S.; Bergman, M.J. Rheumatoid Factor. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2025. [Google Scholar]
  91. Chen, M.; Daha, M.R.; Kallenberg, C.G.M. The Complement System in Systemic Autoimmune Disease. J. Autoimmun. 2010, 34, J276–J286. [Google Scholar] [CrossRef]
  92. Itty, S.; Pulido, J.S.; Bakri, S.J.; Baratz, K.H.; Matteson, E.L.; Hodge, D.O. Anti-Cyclic Citrullinated Peptide, Rheumatoid Factor, and Ocular Symptoms Typical of Rheumatoid Arthritis. Trans. Am. Ophthalmol. Soc. 2008, 106, 75–81, discussion 81–83. [Google Scholar] [PubMed]
  93. Kim, S.-M.; Park, E.; Lee, J.-H.; Lee, S.-H.; Kim, H.-R. The Clinical Significance of Anti-Cyclic Citrullinated Peptide Antibody in Primary Sjögren Syndrome. Rheumatol. Int. 2012, 32, 3963–3967. [Google Scholar] [CrossRef]
  94. Wu, K.Y.; Serhan, O.; Faucher, A.; Tran, S.D. Advances in Sjögren’s Syndrome Dry Eye Diagnostics: Biomarkers and Biomolecules beyond Clinical Symptoms. Biomolecules 2024, 14, 80. [Google Scholar] [CrossRef]
  95. Lee, Y.J. Is the Anti-Centromere Antibody a Marker for a Distinct Subset of Polyautoimmunity in Sjögren’s Syndrome? Korean J. Intern. Med. 2021, 36, 1323–1326. [Google Scholar] [CrossRef] [PubMed]
  96. Shimizu, T.; Nishihata, S.; Nakamura, H.; Takagi, Y.; Sumi, M.; Kawakami, A. Anti-Centromere Antibody Positivity Is an Independent Variable Associated with Salivary Gland Ultrasonography Score in Sjögren’s Syndrome. Sci. Rep. 2024, 14, 5303. [Google Scholar] [CrossRef]
  97. Cha, S.; Mona, M.; Lee, K.E.; Kim, D.H.; Han, K. MicroRNAs in Autoimmune Sjögren’s Syndrome. Genom. Inform. 2018, 16, e19. [Google Scholar] [CrossRef] [PubMed]
  98. Pauley, K.M.; Stewart, C.M.; Gauna, A.E.; Dupre, L.C.; Kuklani, R.; Chan, A.L.; Pauley, B.A.; Reeves, W.H.; Chan, E.K.L.; Cha, S. Altered miR-146a Expression in Sjögren’s Syndrome and Its Functional Role in Innate Immunity. Eur. J. Immunol. 2011, 41, 2029–2039. [Google Scholar] [CrossRef]
  99. Xu, W.-D.; Feng, S.-Y.; Huang, A.-F. Role of miR-155 in Inflammatory Autoimmune Diseases: A Comprehensive Review. Inflamm. Res. 2022, 71, 1501–1517. [Google Scholar] [CrossRef]
  100. Wang, Y.; Zhang, G.; Zhang, L.; Zhao, M.; Huang, H. Decreased microRNA-181a and -16 Expression Levels in the Labial Salivary Glands of Sjögren Syndrome Patients. Exp. Ther. Med. 2018, 15, 426–432. [Google Scholar] [CrossRef]
  101. Jin, L.; Dai, M.; Li, C.; Wang, J.; Wu, B. Risk Factors for Primary Sjögren’s Syndrome: A Systematic Review and Meta-Analysis. Clin. Rheumatol. 2023, 42, 327–338. [Google Scholar] [CrossRef]
  102. Li, X.X.; Maitiyaer, M.; Tan, Q.; Huang, W.H.; Liu, Y.; Liu, Z.P.; Wen, Y.Q.; Zheng, Y.; Chen, X.; Chen, R.L.; et al. Emerging Biologic Frontiers for Sjogren’s Syndrome: Unveiling Novel Approaches with Emphasis on Extra Glandular Pathology. Front. Pharmacol. 2024, 15, 1377055. [Google Scholar] [CrossRef] [PubMed]
  103. Thorlacius, G.E.; Björk, A.; Wahren-Herlenius, M. Genetics and Epigenetics of Primary Sjögren Syndrome: Implications for Future Therapies. Nat. Rev. Rheumatol. 2023, 19, 288–306. [Google Scholar] [CrossRef] [PubMed]
  104. St Clair, E.W.; Baer, A.N.; Ng, W.-F.; Noaiseh, G.; Baldini, C.; Tarrant, T.K.; Papas, A.; Devauchelle-Pensec, V.; Wang, L.; Xu, W.; et al. CD40 Ligand Antagonist Dazodalibep in Sjögren’s Disease: A Randomized, Double-Blinded, Placebo-Controlled, Phase 2 Trial. Nat. Med. 2024, 30, 1583–1592. [Google Scholar] [CrossRef]
  105. Mariette, X.; Barone, F.; Baldini, C.; Bootsma, H.; Clark, K.L.; De Vita, S.; Gardner, D.H.; Henderson, R.B.; Herdman, M.; Lerang, K.; et al. A Randomized, Phase II Study of Sequential Belimumab and Rituximab in Primary Sjögren’s Syndrome. JCI Insight 2022, 7, e163030. [Google Scholar] [CrossRef]
  106. Nakamura, H.; Shimizu, T.; Kawakami, A. Role of Viral Infections in the Pathogenesis of Sjögren’s Syndrome: Different Characteristics of Epstein-Barr Virus and HTLV-1. J. Clin. Med. 2020, 9, 1459. [Google Scholar] [CrossRef]
  107. Otsuka, K.; Sato, M.; Tsunematsu, T.; Ishimaru, N. Virus Infections Play Crucial Roles in the Pathogenesis of Sjögren’s Syndrome. Viruses 2022, 14, 1474. [Google Scholar] [CrossRef] [PubMed]
  108. Bandinelli, F.; Pagano, M.; Vallecoccia, M.S. Post-COVID-19 and Post-COVID-19 Vaccine Arthritis, Polymyalgia Rheumatica and Horton’s Arteritis: A Single-Center Assessment of Clinical, Serological, Genetic, and Ultrasonographic Biomarkers. J. Clin. Med. 2023, 12, 7563. [Google Scholar] [CrossRef]
  109. Shen, Y.; Voigt, A.; Goranova, L.; Abed, M.; Kleiner, D.E.; Maldonado, J.O.; Beach, M.; Pelayo, E.; Chiorini, J.A.; Craft, W.F.; et al. Evidence of a Sjögren’s Disease-like Phenotype Following COVID-19. medRxiv 2022. [Google Scholar] [CrossRef]
  110. Bakhsh, R.; Dairi, K.; Almadabgy, E.; Albiladi, A.; Gamal, L.; Almatrafi, D.; AlShariff, F.; Alsefri, A. New Onset of Neuro-Sjögren’s Syndrome Nine Months After the Third COVID-19 Vaccine Dose: A Case Report. Cureus 2024, 16, e69562. [Google Scholar] [CrossRef]
  111. Sharif, K.; Amital, H. Heavy Metals in Autoimmune Diseases: Too Much Noise in Autoimmunity. In Autoimmune Disorders; John Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2024; pp. 201–223. ISBN 978-1-119-85843-0. [Google Scholar]
  112. Lee, C.-P.; Hsu, P.-Y.; Su, C.-C. Increased Prevalence of Sjogren’s Syndrome in Where Soils Contain High Levels of Chromium. Sci. Total Environ. 2019, 657, 1121–1126. [Google Scholar] [CrossRef]
  113. Zhao, Y.; Hu, S.; Jin, H.; Fan, C.; Liao, K.; Zhang, S.; Xue, J. Relationship Between Perfluoroalkyl Acids in Human Serum and Sjogren’s Syndrome: A Case–Control Study of Populations in Hangzhou, China. Toxics 2024, 12, 764. [Google Scholar] [CrossRef] [PubMed]
  114. Olsson, P.; Turesson, C.; Mandl, T.; Jacobsson, L.; Theander, E. Cigarette Smoking and the Risk of Primary Sjögren’s Syndrome: A Nested Case Control Study. Arthritis Res. Ther. 2017, 19, 50. [Google Scholar] [CrossRef] [PubMed]
  115. Mieliauskaitė, D.; Kontenis, V. Insights into Microbiota in Sjögren’s Syndrome. Medicina 2023, 59, 1661. [Google Scholar] [CrossRef] [PubMed]
  116. Wang, X.; Liu, M.; Xia, W. Causal Relationship Between Sjögren’s Syndrome and Gut Microbiota: A Two-Sample Mendelian Randomization Study. Biomedicines 2024, 12, 2378. [Google Scholar] [CrossRef]
  117. Cano-Ortiz, A.; Laborda-Illanes, A.; Plaza-Andrades, I.; Membrillo del Pozo, A.; Villarrubia Cuadrado, A.; Rodríguez Calvo de Mora, M.; Leiva-Gea, I.; Sanchez-Alcoholado, L.; Queipo-Ortuño, M.I. Connection between the Gut Microbiome, Systemic Inflammation, Gut Permeability and FOXP3 Expression in Patients with Primary Sjögren’s Syndrome. Int. J. Mol. Sci. 2020, 21, 8733. [Google Scholar] [CrossRef]
  118. Psianou, K.; Panagoulias, I.; Papanastasiou, A.D.; de Lastic, A.-L.; Rodi, M.; Spantidea, P.I.; Degn, S.E.; Georgiou, P.; Mouzaki, A. Clinical and Immunological Parameters of Sjögren’s Syndrome. Autoimmun. Rev. 2018, 17, 1053–1064. [Google Scholar] [CrossRef]
  119. Sandhya, P.; Jeyaseelan, L.; Scofield, R.H.; Danda, D. Clinical Characteristics and Outcome of Primary Sjogren’s Syndrome: A Large Asian Indian Cohort. Open Rheumatol. J. 2014, 9, 36–45. [Google Scholar] [CrossRef] [PubMed]
  120. Lodba, A.; Ancuta, C.; Tatarciuc, D.; Ghiorghe, A.; Lodba, L.-O.; Iordache, C. Comparative Analysis of Glandular and Extraglandular Manifestations in Primary and Secondary Sjögren’s Syndrome: A Study in Two Academic Centers in North-East Romania. Diagnostics 2024, 14, 2367. [Google Scholar] [CrossRef]
  121. Mathews, P.M.; Hahn, S.; Hessen, M.; Kim, J.; Grader-Beck, T.; Birnbaum, J.; Baer, A.N.; Akpek, E.K. Ocular Complications of Primary Sjögren Syndrome in Men. Am. J. Ophthalmol. 2015, 160, 447–452.e1. [Google Scholar] [CrossRef]
  122. Virdee, S.; Greenan-Barrett, J.; Ciurtin, C. A Systematic Review of Primary Sjögren’s Syndrome in Male and Paediatric Populations. Clin. Rheumatol. 2017, 36, 2225–2236. [Google Scholar] [CrossRef]
  123. La Rocca, G.; Ferro, F.; Sambataro, G.; Elefante, E.; Fonzetti, S.; Fulvio, G.; Navarro, I.C.; Mosca, M.; Baldini, C. Primary-Sjögren’s-Syndrome-Related Interstitial Lung Disease: A Clinical Review Discussing Current Controversies. J. Clin. Med. 2023, 12, 3428. [Google Scholar] [CrossRef] [PubMed]
  124. Flament, T.; Bigot, A.; Chaigne, B.; Henique, H.; Diot, E.; Marchand-Adam, S. Pulmonary Manifestations of Sjögren’s Syndrome. Eur. Respir. Rev. 2016, 25, 110–123. [Google Scholar] [CrossRef] [PubMed]
  125. Wang, Y.; Hou, Z.; Qiu, M.; Ye, Q. Risk Factors for Primary Sjögren Syndrome-Associated Interstitial Lung Disease. J. Thorac. Dis. 2018, 10, 2108–2117. [Google Scholar] [CrossRef]
  126. Stojan, G.; Baer, A.N.; Danoff, S.K. Pulmonary Manifestations of Sjögren’s Syndrome. Curr. Allergy Asthma Rep. 2013, 13, 354–360. [Google Scholar] [CrossRef]
  127. Enomoto, Y.; Takemura, T.; Hagiwara, E.; Iwasawa, T.; Okudela, K.; Yanagawa, N.; Baba, T.; Sakai, F.; Fukuda, Y.; Nagaoka, S.; et al. Features of Usual Interstitial Pneumonia in Patients with Primary Sjögren’s Syndrome Compared with Idiopathic Pulmonary Fibrosis. Respir. Investig. 2014, 52, 227–235. [Google Scholar] [CrossRef]
  128. Kim, J.-Y.; Park, S.-H.; Kim, S.-K.; Hyun, D.-S.; Kum, Y.S.; Jung, K.-J.; Choe, J.-Y. Lymphocytic Interstitial Pneumonia in Primary Sjögren’s Syndrome: A Case Report. Korean J. Intern. Med. 2011, 26, 108–111. [Google Scholar] [CrossRef] [PubMed]
  129. Garcia, D.; Young, L. Lymphocytic Interstitial Pneumonia as a Manifestation of SLE and Secondary Sjogren’s Syndrome. BMJ Case Rep. 2013, 2013, bcr2013009598. [Google Scholar] [CrossRef]
  130. Ioannou, S.; Toya, S.P.; Tomos, P.; Tzelepis, G.E. Cryptogenic Organizing Pneumonia Associated with Primary Sjogren’s Syndrome. Rheumatol. Int. 2008, 28, 1053–1055. [Google Scholar] [CrossRef]
  131. Centala, S.; Park, J.H.; Girnita, D. Sjogren’s Syndrome Presenting with Solely Cutaneous Features. Diagnostics 2021, 11, 1260. [Google Scholar] [CrossRef]
  132. Ilzkovitz, M.; Kayembe, E.E.; Geers, C.; Pozdzik, A. Kidney Stones, Proteinuria and Renal Tubular Metabolic Acidosis: What Is the Link? Healthcare 2022, 10, 836. [Google Scholar] [CrossRef]
  133. Bouchalova, K.; Flögelova, H.; Horak, P.; Civrny, J.; Mlcak, P.; Pink, R.; Michalek, J.; Camborova, P.; Mikulkova, Z.; Kriegova, E. Juvenile Primary Sjögren Syndrome in a 15-Year-Old Boy with Renal Involvement: A Case Report and Review of the Literature. Diagnostics 2024, 14, 258. [Google Scholar] [CrossRef]
  134. Garcia, D.M.; Reis de Oliveira, F.; Módulo, C.M.; Faustino, J.; Barbosa, A.P.; Alves, M.; Rocha, E.M. Is Sjögren’s Syndrome Dry Eye Similar to Dry Eye Caused by Other Etiologies? Discriminating Different Diseases by Dry Eye Tests. PLoS ONE 2018, 13, e0208420. [Google Scholar] [CrossRef]
  135. Saldanha, I.J.; Bunya, V.Y.; McCoy, S.S.; Makara, M.; Baer, A.N.; Akpek, E.K. Ocular Manifestations and Burden Related to Sjögren Syndrome: Results of a Patient Survey. Am. J. Ophthalmol. 2020, 219, 40–48. [Google Scholar] [CrossRef]
  136. Friedlaender, M.H. Ocular Manifestations of Sjögren’s Syndrome: Keratoconjunctivitis Sicca. Rheum. Dis. Clin. N. Am. 1992, 18, 591–608. [Google Scholar] [CrossRef]
  137. Kim, S.Y.; Kim, S.H.; Yoon, C.H.; Kim, M.K.; Oh, J.Y. Infectious Keratitis in Patients with Ocular Sjögren’s Syndrome. Korean J. Ophthalmol. 2022, 36, 407–412. [Google Scholar] [CrossRef]
  138. Singh, S.; Chaudhary, S.; Das, A.V.; Basu, S. Presentation, Aetiology and Outcomes of Corneal Ulceration in Sjogren’s Syndrome. Eye 2023, 37, 3217–3220. [Google Scholar] [CrossRef]
  139. Dart, J.K. Predisposing Factors in Microbial Keratitis: The Significance of Contact Lens Wear. Br. J. Ophthalmol. 1988, 72, 926–930. [Google Scholar] [CrossRef]
  140. Trojacka, E.; Izdebska, J.; Szaflik, J.; Przybek-Skrzypecka, J. The Ocular Microbiome: Micro-Steps Towards Macro-Shift in Targeted Treatment? A Comprehensive Review. Microorganisms 2024, 12, 2232. [Google Scholar] [CrossRef]
  141. Ni, N.; Srinivasan, M.; McLeod, S.D.; Acharya, N.R.; Lietman, T.M.; Rose-Nussbaumer, J. Use of Adjunctive Topical Corticosteroids in Bacterial Keratitis. Curr. Opin. Ophthalmol. 2016, 27, 353–357. [Google Scholar] [CrossRef]
  142. Jan, R.-L.; Ho, C.-H.; Wang, J.-J.; Tseng, S.-H.; Chang, Y.-S. Associations between Sjögren Syndrome, Sociodemographic Factors, Comorbid Conditions, and Scleritis in a Taiwanese Population-Based Study. J. Pers. Med. 2022, 12, 105. [Google Scholar] [CrossRef]
  143. Nagy, G.; Czirják, L.; Kumánovics, G. Patients with Systemic Sclerosis with and without Overlap Syndrome Show Similar Microvascular Abnormalities. Diagnostics 2021, 11, 1606. [Google Scholar] [CrossRef]
  144. Wakefield, D.; Di Girolamo, N.; Thurau, S.; Wildner, G.; McCluskey, P. Scleritis: Immunopathogenesis and Molecular Basis for Therapy. Prog. Retin. Eye Res. 2013, 35, 44–62. [Google Scholar] [CrossRef]
  145. Bak, E.; Yang, H.K.; Hwang, J.-M. Optic Neuropathy Associated with Primary Sjögren’s Syndrome: A Case Series. Optom. Vis. Sci. 2017, 94, 519–526. [Google Scholar] [CrossRef]
  146. Tang, W.-Q.; Wei, S.-H. Primary Sjögren’s Syndrome Related Optic Neuritis. Int. J. Ophthalmol. 2013, 6, 888–891. [Google Scholar] [CrossRef]
  147. Kaufhold, F.; Zimmermann, H.; Schneider, E.; Ruprecht, K.; Paul, F.; Oberwahrenbrock, T.; Brandt, A.U. Optic Neuritis Is Associated with Inner Nuclear Layer Thickening and Microcystic Macular Edema Independently of Multiple Sclerosis. PLoS ONE 2013, 8, e71145. [Google Scholar] [CrossRef]
  148. Wu, K.Y.; Kulbay, M.; Tanasescu, C.; Jiao, B.; Nguyen, B.H.; Tran, S.D. An Overview of the Dry Eye Disease in Sjögren’s Syndrome Using Our Current Molecular Understanding. Int. J. Mol. Sci. 2023, 24, 1580. [Google Scholar] [CrossRef]
  149. Srivastava, A.; Makarenkova, H.P. Innate Immunity and Biological Therapies for the Treatment of Sjögren’s Syndrome. Int. J. Mol. Sci. 2020, 21, 9172. [Google Scholar] [CrossRef]
  150. Akpek, E.K.; Lindsley, K.B.; Adyanthaya, R.S.; Swamy, R.; Baer, A.N.; McDonnell, P.J. Treatment of Sjögren’s Syndrome–Associated Dry Eye: An Evidence-Based Review. Ophthalmology 2011, 118, 1242–1252. [Google Scholar] [CrossRef]
  151. Almulhim, A. Therapeutic Targets in the Management of Dry Eye Disease Associated with Sjögren’s Syndrome: An Updated Review of Current Insights and Future Perspectives. J. Clin. Med. 2024, 13, 1777. [Google Scholar] [CrossRef]
  152. Yang, Y.-J.; Lee, W.-Y.; Kim, Y.; Hong, Y. A Meta-Analysis of the Efficacy of Hyaluronic Acid Eye Drops for the Treatment of Dry Eye Syndrome. Int. J. Environ. Res. Public Health 2021, 18, 2383. [Google Scholar] [CrossRef]
  153. Mussano, F.; Genova, T.; Munaron, L.; Petrillo, S.; Erovigni, F.; Carossa, S. Cytokine, Chemokine, and Growth Factor Profile of Platelet-Rich Plasma. Platelets 2016, 27, 467–471. [Google Scholar] [CrossRef] [PubMed]
  154. Metheetrairut, C.; Ngowyutagon, P.; Tunganuntarat, A.; Khowawisetsut, L.; Kittisares, K.; Prabhasawat, P. Comparison of Epitheliotrophic Factors in Platelet-Rich Plasma versus Autologous Serum and Their Treatment Efficacy in Dry Eye Disease. Sci. Rep. 2022, 12, 8906. [Google Scholar] [CrossRef]
  155. Franchini, M.; Cruciani, M.; Mengoli, C.; Marano, G.; Capuzzo, E.; Pati, I.; Masiello, F.; Veropalumbo, E.; Pupella, S.; Vaglio, S.; et al. Serum Eye Drops for the Treatment of Ocular Surface Diseases: A Systematic Review and Meta-Analysis. Blood Transfus. 2019, 17, 200–209. [Google Scholar] [CrossRef] [PubMed]
  156. You, J.; Hodge, C.; Hoque, M.; Petsoglou, C.; Sutton, G. Human Platelets and Derived Products in Treating Ocular Surface Diseases—A Systematic Review. Clin. Ophthalmol. 2020, 14, 3195–3210. [Google Scholar] [CrossRef] [PubMed]
  157. Pan, Q.; Angelina, A.; Marrone, M.; Stark, W.J.; Akpek, E.K. Autologous Serum Eye Drops for Dry Eye. Cochrane Database Syst. Rev. 2017, 2, CD009327. [Google Scholar] [CrossRef]
  158. Wang, K.; Shi, L.; Yu, Z.; Deng, Z.; He, A.; Li, S.; Liu, L. Cyclosporine A Suppresses the Activation of the Th17 Cells in Patients with Primary Sjögren’s Syndrome. Iran. J. Allergy Asthma Immunol. 2015, 14, 198–207. [Google Scholar]
  159. de Paiva, C.S.; Pflugfelder, S.C.; Ng, S.M.; Akpek, E.K. Topical Cyclosporine A Therapy for Dry Eye Syndrome. Cochrane Database Syst. Rev. 2019, 9, CD010051. [Google Scholar] [CrossRef]
  160. Jung, H.H.; Ji, Y.S.; Sung, M.S.; Kim, K.K.; Yoon, K.C. Long-Term Outcome of Treatment with Topical Corticosteroids for Severe Dry Eye Associated with Sjögren’s Syndrome. Chonnam Med. J. 2015, 51, 26–32. [Google Scholar] [CrossRef]
  161. Yang, D.-H.; Wang, Y.-H.; Pan, L.-F.; Wei, J.C.-C. Cardiovascular Protection of Hydroxychloroquine in Patients with Sjögren’s Syndrome. J. Clin. Med. 2020, 9, 3469. [Google Scholar] [CrossRef]
  162. Avalos-Salgado, F.A.; Gonzalez-Lopez, L.; Gonzalez-Vazquez, S.; Ponce-Guarneros, J.M.; Santiago-Garcia, A.P.; Amaya-Cabrera, E.L.; Arellano-Cervantes, R.; Gutiérrez-Aceves, J.A.; Alcaraz-Lopez, M.F.; Nava-Valdivia, C.A.; et al. Risk Factors Associated with Adverse Events Leading to Methotrexate Withdrawal in Elderly Rheumatoid Arthritis Patients: A Retrospective Cohort Study. J. Clin. Med. 2024, 13, 1863. [Google Scholar] [CrossRef]
  163. Mavragani, C.P.; Moutsopoulos, H.M. Sjögren’s Syndrome: Old and New Therapeutic Targets. J. Autoimmun. 2020, 110, 102364. [Google Scholar] [CrossRef] [PubMed]
  164. Kiripolsky, J.; McCabe, L.G.; Kramer, J.M. Innate Immunity in Sjögren’s Syndrome. Clin. Immunol. 2017, 182, 4–13. [Google Scholar] [CrossRef]
  165. Lee, A.S.; Scofield, R.H.; Hammitt, K.M.; Gupta, N.; Thomas, D.E.; Moua, T.; Ussavarungsi, K.; Clair, E.W.S.; Meehan, R.; Dunleavy, K.; et al. Consensus Guidelines for Evaluation and Management of Pulmonary Disease in Sjögren’s. CHEST 2021, 159, 683–698. [Google Scholar] [CrossRef] [PubMed]
  166. Chen, W.; Lin, J. Mycophenolate for the Treatment of Primary Sjögren’s Syndrome. J. Transl. Int. Med. 2020, 8, 146–149. [Google Scholar] [CrossRef] [PubMed]
  167. Xu, R.; Yan, Q.; Gong, Y.; Cai, C.-S.; Wu, J.; Yuan, X.-Y.; Long, X.-M. Tacrolimus Therapy in Primary Sjögren’s Syndrome with Refractory Immune Thrombocytopenia: A Retrospective Study. Clin. Exp. Rheumatol. 2022, 40, 2268–2274. [Google Scholar] [CrossRef]
  168. Ahn, H.; Ji, Y.W.; Jun, I.; Kim, T.; Lee, H.K.; Seo, K.Y. Comparison of Treatment Modalities for Dry Eye in Primary Sjögren’s Syndrome. J. Clin. Med. 2022, 11, 463. [Google Scholar] [CrossRef]
  169. Romano, V.; Romano, D.; Semeraro, P.; Forbice, E.; Iaria, A.; Pizzolante, T.; Frassi, M.; Franceschini, F.; Semeraro, F. Therapeutic Hyper-CL Soft Contact Lens in Sjögren’s Syndrome. Am. J. Ophthalmol. Case Rep. 2022, 28, 101685. [Google Scholar] [CrossRef]
  170. Nunziata, S.; Petrini, D.; Dell’Anno, S.; Barone, V.; Coassin, M.; Di Zazzo, A. Customized Scleral Lenses: An Alternative Tool for Severe Dry Eye Disease—A Case Series. J. Clin. Med. 2024, 13, 3935. [Google Scholar] [CrossRef]
  171. Qiu, S.X.; Fadel, D.; Hui, A. Scleral Lenses for Managing Dry Eye Disease in the Absence of Corneal Irregularities: What Is the Current Evidence? J. Clin. Med. 2024, 13, 3838. [Google Scholar] [CrossRef]
  172. Li, J.; Zhang, X.; Zheng, Q.; Zhu, Y.; Wang, H.; Ma, H.; Jhanji, V.; Chen, W. Comparative Evaluation of Silicone Hydrogel Contact Lenses and Autologous Serum for Management of Sjögren Syndrome-Associated Dry Eye. Cornea 2015, 34, 1072–1078. [Google Scholar] [CrossRef]
  173. Castrejón-Morales, C.Y.; Granados-Portillo, O.; Cruz-Bautista, I.; Ruiz-Quintero, N.; Manjarrez, I.; Lima, G.; Hernández-Ramírez, D.F.; Astudillo-Angel, M.; Llorente, L.; Hernández-Molina, G. Omega-3 and Omega-6 Fatty Acids in Primary Sjögren’s Syndrome: Clinical Meaning and Association with Inflammation. Clin. Exp. Rheumatol. 2020, 38 (Suppl. S126), 34–39. [Google Scholar] [PubMed]
  174. da Nave, C.B.; Pereira, P.; Silva, M.L. The Effect of Polyunsaturated Fatty Acid (PUFA) Supplementation on Clinical Manifestations and Inflammatory Parameters in Individuals with Sjögren’s Syndrome: A Literature Review of Randomized Controlled Clinical Trials. Nutrients 2024, 16, 3786. [Google Scholar] [CrossRef]
  175. Szodoray, P.; Horvath, I.F.; Papp, G.; Barath, S.; Gyimesi, E.; Csathy, L.; Kappelmayer, J.; Sipka, S.; Duttaroy, A.K.; Nakken, B.; et al. The Immunoregulatory Role of Vitamins A, D and E in Patients with Primary Sjögren’s Syndrome. Rheumatology 2010, 49, 211–217. [Google Scholar] [CrossRef] [PubMed]
  176. Hyon, J.Y.; Han, S.B. Dry Eye Disease and Vitamins: A Narrative Literature Review. Appl. Sci. 2022, 12, 4567. [Google Scholar] [CrossRef]
  177. Nesvold, M.B.; Jensen, J.L.; Hove, L.H.; Singh, P.B.; Young, A.; Palm, Ø.; Frost Andersen, L.; Carlsen, M.H.; Iversen, P.O. Dietary Intake, Body Composition, and Oral Health Parameters among Female Patients with Primary Sjögren’s Syndrome. Nutrients 2018, 10, 866. [Google Scholar] [CrossRef]
  178. Trotta, M.C.; Herman, H.; Balta, C.; Rosu, M.; Ciceu, A.; Mladin, B.; Gesualdo, C.; Lepre, C.C.; Russo, M.; Petrillo, F.; et al. Oral Administration of Vitamin D3 Prevents Corneal Damage in a Knock-Out Mouse Model of Sjögren’s Syndrome. Biomedicines 2023, 11, 616. [Google Scholar] [CrossRef] [PubMed]
  179. Athanassiou, L.; Kostoglou-Athanassiou, I.; Koutsilieris, M.; Shoenfeld, Y. Vitamin D and Autoimmune Rheumatic Diseases. Biomolecules 2023, 13, 709. [Google Scholar] [CrossRef]
  180. Dabravolski, S.A.; Churov, A.V.; Starodubtseva, I.A.; Beloyartsev, D.F.; Kovyanova, T.I.; Sukhorukov, V.N.; Orekhov, N.A. Vitamin D in Primary Sjogren’s Syndrome (pSS) and the Identification of Novel Single-Nucleotide Polymorphisms Involved in the Development of pSS-Associated Diseases. Diagnostics 2024, 14, 2035. [Google Scholar] [CrossRef]
  181. Verstappen, G.M.; van Nimwegen, J.F.; Vissink, A.; Kroese, F.G.M.; Bootsma, H. The Value of Rituximab Treatment in Primary Sjögren’s Syndrome. Clin. Immunol. 2017, 182, 62–71. [Google Scholar] [CrossRef]
  182. Berardicurti, O.; Pavlich, V.; Cola, I.D.; Ruscitti, P.; Benedetto, P.D.; Navarini, L.; Marino, A.; Cipriani, P.; Giacomelli, R. Long-Term Safety of Rituximab in Primary Sjögren’s Syndrome: The Experience of a Single Centre. J. Rheumatol. 2021, 49, 171–175. [Google Scholar] [CrossRef]
  183. Chen, Y.H.; Wang, X.Y.; Jin, X.; Yang, Z.; Xu, J. Rituximab Therapy for Primary Sjögren’s Syndrome. Front. Pharmacol. 2021, 12, 731122. [Google Scholar] [CrossRef] [PubMed]
  184. Mariette, X.; Seror, R.; Quartuccio, L.; Baron, G.; Salvin, S.; Fabris, M.; Desmoulins, F.; Nocturne, G.; Ravaud, P.; De Vita, S. Efficacy and Safety of Belimumab in Primary Sjögren’s Syndrome: Results of the BELISS Open-Label Phase II Study. Ann. Rheum. Dis. 2015, 74, 526–531. [Google Scholar] [CrossRef] [PubMed]
  185. Álvarez-Rivas, N.; Sang-Park, H.; Díaz del Campo, P.; Fernández-Castro, M.; Corominas, H.; Andreu, J.L.; Navarro-Compán, V. Efficacy of Belimumab in Primary Sjögren’s Syndrome: A Systematic Review. Reumatol. Clin. 2021, 17, 170–174. [Google Scholar] [CrossRef]
  186. Kaegi, C.; Steiner, U.C.; Wuest, B.; Crowley, C.; Boyman, O. Systematic Review of Safety and Efficacy of Belimumab in Treating Immune-Mediated Disorders. Allergy 2021, 76, 2673–2683. [Google Scholar] [CrossRef] [PubMed]
  187. Pontarini, E.; Fabris, M.; Quartuccio, L.; Cappeletti, M.; Calcaterra, F.; Roberto, A.; Curcio, F.; Mavilio, D.; Della Bella, S.; De Vita, S. Treatment with Belimumab Restores B Cell Subsets and Their Expression of B Cell Activating Factor Receptor in Patients with Primary Sjogren’s Syndrome. Rheumatology 2015, 54, 1429–1434. [Google Scholar] [CrossRef]
  188. Zhao, T.; Zhang, R.; Li, Z.; Qin, D.; Wang, X. Novel and Potential Future Therapeutic Options in Sjögren’s Syndrome. Heliyon 2024, 10, e38803. [Google Scholar] [CrossRef]
  189. Baer, A.N.; Gottenberg, J.-E.; St Clair, E.W.; Sumida, T.; Takeuchi, T.; Seror, R.; Foulks, G.; Nys, M.; Mukherjee, S.; Wong, R.; et al. Efficacy and Safety of Abatacept in Active Primary Sjögren’s Syndrome: Results of a Phase III, Randomised, Placebo-Controlled Trial. Ann. Rheum. Dis. 2021, 80, 339–348. [Google Scholar] [CrossRef]
  190. de Wolff, L.; van Nimwegen, J.F.; Mossel, E.; van Zuiden, G.S.; Stel, A.J.; Majoor, K.I.; Olie, L.; Los, L.I.; Vissink, A.; Spijkervet, F.K.L.; et al. Long-Term Abatacept Treatment for 48 Weeks in Patients with Primary Sjögren’s Syndrome: The Open-Label Extension Phase of the ASAP-III Trial. Semin. Arthritis Rheum. 2022, 53, 151955. [Google Scholar] [CrossRef]
  191. Tsuboi, H.; Toko, H.; Honda, F.; Abe, S.; Takahashi, H.; Yagishita, M.; Hagiwara, S.; Ohyama, A.; Kondo, Y.; Nakano, K.; et al. Abatacept Ameliorates Both Glandular and Extraglandular Involvements in Patients with Sjögren’s Syndrome Associated with Rheumatoid Arthritis: Findings from an Open-Label, Multicentre, 1-Year, Prospective Study: The ROSE (Rheumatoid Arthritis with Orencia Trial Toward Sjögren’s Syndrome Endocrinopathy) and ROSE II Trials. Mod. Rheumatol. 2023, 33, 160–168. [Google Scholar] [CrossRef]
  192. Gottenberg, J.-E.; Dörner, T.; Bootsma, H.; Devauchelle-Pensec, V.; Bowman, S.J.; Mariette, X.; Bartz, H.; Oortgiesen, M.; Shock, A.; Koetse, W.; et al. Efficacy of Epratuzumab, an Anti-CD22 Monoclonal IgG Antibody, in Systemic Lupus Erythematosus Patients With Associated Sjögren’s Syndrome. Arthritis Rheumatol. 2018, 70, 763–773. [Google Scholar] [CrossRef]
  193. St Clair, E.W.; Baer, A.N.; Wei, C.; Noaiseh, G.; Parke, A.; Coca, A.; Utset, T.O.; Genovese, M.C.; Wallace, D.J.; McNamara, J.; et al. The Clinical Efficacy and Safety of Baminercept, a Lymphotoxin-β Receptor Fusion Protein, in Primary Sjögren’s Syndrome: Results from a Randomized, Double-Blind, Placebo-Controlled Phase II Trial. Arthritis Rheumatol. 2018, 70, 1470–1480. [Google Scholar] [CrossRef]
  194. Fisher, B.A.; Szanto, A.; Ng, W.-F.; Bombardieri, M.; Posch, M.G.; Papas, A.S.; Farag, A.M.; Daikeler, T.; Bannert, B.; Kyburz, D.; et al. Assessment of the Anti-CD40 Antibody Iscalimab in Patients with Primary Sjögren’s Syndrome: A Multicentre, Randomised, Double-Blind, Placebo-Controlled, Proof-of-Concept Study. Lancet Rheumatol. 2020, 2, e142–e152. [Google Scholar] [CrossRef] [PubMed]
  195. Fisher, B.A.; Mariette, X.; Papas, A.; Grader-Beck, T.; Bootsma, H.; Ng, W.-F.; van Daele, P.L.A.; Finzel, S.; Noaiseh, G.; Elgueta, S.; et al. Safety and Efficacy of Subcutaneous Iscalimab (CFZ533) in Two Distinct Populations of Patients with Sjögren’s Disease (TWINSS): Week 24 Results of a Randomised, Double-Blind, Placebo-Controlled, Phase 2b Dose-Ranging Study. Lancet 2024, 404, 540–553. [Google Scholar] [CrossRef] [PubMed]
  196. Bowman, S.J.; Fox, R.; Dörner, T.; Mariette, X.; Papas, A.; Grader-Beck, T.; Fisher, B.A.; Barcelos, F.; De Vita, S.; Schulze-Koops, H.; et al. Safety and Efficacy of Subcutaneous Ianalumab (VAY736) in Patients with Primary Sjögren’s Syndrome: A Randomised, Double-Blind, Placebo-Controlled, Phase 2b Dose-Finding Trial. Lancet 2022, 399, 161–171. [Google Scholar] [CrossRef]
  197. Dörner, T.; Posch, M.G.; Li, Y.; Petricoul, O.; Cabanski, M.; Milojevic, J.M.; Kamphausen, E.; Valentin, M.-A.; Simonett, C.; Mooney, L.; et al. Treatment of Primary Sjögren’s Syndrome with Ianalumab (VAY736) Targeting B Cells by BAFF Receptor Blockade Coupled with Enhanced, Antibody-Dependent Cellular Cytotoxicity. Ann. Rheum. Dis. 2019, 78, 641–647. [Google Scholar] [CrossRef]
  198. Xu, D.; Fang, J.; Zhang, S.; Huang, C.; Huang, C.; Qin, L.; Li, X.; Chen, M.; Liu, X.; Liu, Y.; et al. Efficacy and Safety of Telitacicept in Primary Sjögren’s Syndrome: A Randomized, Double-Blind, Placebo-Controlled, Phase 2 Trial. Rheumatology 2024, 63, 698–705. [Google Scholar] [CrossRef]
  199. Horai, Y.; Kurushima, S.; Shimizu, T.; Nakamura, H.; Kawakami, A. A Review of the Impact of Sjögren’s Syndrome and/or the Presence of Anti-Ro/SS-A Antibodies on Therapeutic Strategies for Rheumatoid Arthritis. J. Clin. Med. 2025, 14, 568. [Google Scholar] [CrossRef] [PubMed]
  200. Zhan, Q.; Zhang, J.; Lin, Y.; Chen, W.; Fan, X.; Zhang, D. Pathogenesis and Treatment of Sjogren’s Syndrome: Review and Update. Front. Immunol. 2023, 14, 1127417. [Google Scholar] [CrossRef]
  201. Justet, A.; Ottaviani, S.; Dieudé, P.; Taillé, C. Tocilizumab for Refractory Organising Pneumonia Associated with Sjögren’s Disease. BMJ Case Rep. 2015, 2015, bcr2014209076. [Google Scholar] [CrossRef]
  202. Felten, R.; Devauchelle-Pensec, V.; Seror, R.; Duffau, P.; Saadoun, D.; Hachulla, E.; Pierre Yves, H.; Salliot, C.; Perdriger, A.; Morel, J.; et al. Interleukin 6 Receptor Inhibition in Primary Sjögren Syndrome: A Multicentre Double-Blind Randomised Placebo-Controlled Trial. Ann. Rheum. Dis. 2021, 80, 329–338. [Google Scholar] [CrossRef]
  203. Juarez, M.; Diaz, N.; Johnston, G.I.; Nayar, S.; Payne, A.; Helmer, E.; Cain, D.; Williams, P.; Devauchelle-Pensec, V.; Fisher, B.A.; et al. A Phase 2 Randomized, Double-Blind, Placebo-Controlled, Proof-of-Concept Study of Oral Seletalisib in Primary Sjögren’s Syndrome. Rheumatology 2021, 60, 1364–1375. [Google Scholar] [CrossRef] [PubMed]
  204. Scuron, M.D.; Fay, B.L.; Connell, A.J.; Oliver, J.; Smith, P.A. The PI3Kδ Inhibitor Parsaclisib Ameliorates Pathology and Reduces Autoantibody Formation in Preclinical Models of Systemic Lupus Erythematosus and Sjögren’s Syndrome. Int. Immunopharmacol. 2021, 98, 107904. [Google Scholar] [CrossRef] [PubMed]
  205. Dörner, T.; Zeher, M.; Laessing, U.; Chaperon, F.; De Buck, S.; Hasselberg, A.; Valentin, M.-A.; Ma, S.; Cabanski, M.; Kalis, C.; et al. OP0250 A Randomised, Double-Blind Study to Assess the Safety, Tolerability and Preliminary Efficacy of Leniolisib (CDZ173) in Patients with Primary sjögren’s Syndrome. Ann. Rheum. Dis. 2018, 77, 174. [Google Scholar] [CrossRef]
  206. Dörner, T.; Kaul, M.; Szántó, A.; Tseng, J.-C.; Papas, A.S.; Pylvaenaeinen, I.; Hanser, M.; Abdallah, N.; Grioni, A.; Santos Da Costa, A.; et al. Efficacy and Safety of Remibrutinib, a Selective Potent Oral BTK Inhibitor, in Sjögren’s Syndrome: Results from a Randomised, Double-Blind, Placebo-Controlled Phase 2 Trial. Ann. Rheum. Dis. 2024, 83, 360–371. [Google Scholar] [CrossRef]
  207. Price, E.; Bombardieri, M.; Kivitz, A.; Matzkies, F.; Gurtovaya, O.; Pechonkina, A.; Jiang, W.; Downie, B.; Mathur, A.; Mozaffarian, A.; et al. Safety and Efficacy of Filgotinib, Lanraplenib and Tirabrutinib in Sjögren’s Syndrome: A Randomized, Phase 2, Double-Blind, Placebo-Controlled Study. Rheumatology 2022, 61, 4797–4808. [Google Scholar] [CrossRef]
  208. Zhao, Z.; Ye, C.; Dong, L. The Off-Label Uses Profile of Tofacitinib in Systemic Rheumatic Diseases. Int. Immunopharmacol. 2020, 83, 106480. [Google Scholar] [CrossRef]
  209. Barrera, M.-J.; Aguilera, S.; Castro, I.; Matus, S.; Carvajal, P.; Molina, C.; González, S.; Jara, D.; Hermoso, M.; González, M.-J. Tofacitinib Counteracts IL-6 Overexpression Induced by Deficient Autophagy: Implications in Sjögren’s Syndrome. Rheumatology 2021, 60, 1951–1962. [Google Scholar] [CrossRef] [PubMed]
  210. Gao, R.; Pu, J.; Wang, Y.; Wu, Z.; Liang, Y.; Song, J.; Pan, S.; Han, F.; Yang, L.; Xu, X.; et al. Tofacitinib in the Treatment of Primary Sjögren’s Syndrome-Associated Interstitial Lung Disease: Study Protocol for a Prospective, Randomized, Controlled and Open-Label Trial. BMC Pulm. Med. 2023, 23, 473. [Google Scholar] [CrossRef]
  211. Bai, W.; Yang, F.; Xu, H.; Wei, W.; Li, H.; Zhang, L.; Zhao, Y.; Shi, X.; Zhang, Y.; Zeng, X.; et al. A Multi-Center, Open-Label, Randomized Study to Explore Efficacy and Safety of Baricitinib in Active Primary Sjogren’s Syndrome Patients. Trials 2023, 24, 112. [Google Scholar] [CrossRef]
  212. Bang, C.-H.; Park, C.-J.; Kim, Y.-S. The Expanding Therapeutic Potential of Deucravacitinib Beyond Psoriasis: A Narrative Review. J. Clin. Med. 2025, 14, 1745. [Google Scholar] [CrossRef]
  213. Bentley, D.; Fisher, B.A.; Barone, F.; Kolb, F.A.; Attley, G. A Randomized, Double-Blind, Placebo-Controlled, Parallel Group Study on the Effects of a Cathepsin S Inhibitor in Primary Sjögren’s Syndrome. Rheumatology 2023, 62, 3644–3653. [Google Scholar] [CrossRef]
  214. Posada, J.; Valadkhan, S.; Burge, D.; Davies, K.; Tarn, J.; Casement, J.; Jobling, K.; Gallagher, P.; Wilson, D.; Barone, F.; et al. Improvement of Severe Fatigue Following Nuclease Therapy in Patients With Primary Sjögren’s Syndrome: A Randomized Clinical Trial. Arthritis Rheumatol. 2021, 73, 143–150. [Google Scholar] [CrossRef] [PubMed]
  215. Taylor, M.; Ousler, G.; Torkildsen, G.; Walshe, C.; Fyfe, M.C.T.; Rowley, A.; Webber, S.; Sheppard, J.D.; Duggal, A. A Phase 2 Randomized, Double-Masked, Placebo-Controlled Study of Novel Nonsystemic Kinase Inhibitor TOP1630 for the Treatment of Dry Eye Disease. Clin. Ophthalmol. 2019, 13, 261–275. [Google Scholar] [CrossRef]
  216. Sosne, G.; Dunn, S.P.; Kim, C. Thymosin Β4 Significantly Improves Signs and Symptoms of Severe Dry Eye in a Phase 2 Randomized Trial. Cornea 2015, 34, 491. [Google Scholar] [CrossRef] [PubMed]
  217. Kim, C.E.; Kleinman, H.K.; Sosne, G.; Ousler, G.W.; Kim, K.; Kang, S.; Yang, J. RGN-259 (Thymosin Β4) Improves Clinically Important Dry Eye Efficacies in Comparison with Prescription Drugs in a Dry Eye Model. Sci. Rep. 2018, 8, 10500. [Google Scholar] [CrossRef] [PubMed]
  218. Holland, E.J.; Darvish, M.; Nichols, K.K.; Jones, L.; Karpecki, P.M. Efficacy of Topical Ophthalmic Drugs in the Treatment of Dry Eye Disease: A Systematic Literature Review. Ocul. Surf. 2019, 17, 412–423. [Google Scholar] [CrossRef]
  219. Georgiev, G.A.; Gh., M.S.; Romano, J.; Dias Teixeira, K.L.; Struble, C.; Ryan, D.S.; Sia, R.K.; Kitt, J.P.; Harris, J.M.; Hsu, K.-L.; et al. Lacritin Proteoforms Prevent Tear Film Collapse and Maintain Epithelial Homeostasis. J. Biol. Chem. 2021, 296, 100070. [Google Scholar] [CrossRef]
  220. McNamara, N.A.; Ge, S.; Lee, S.M.; Enghauser, A.M.; Kuehl, L.; Chen, F.Y.-T.; Gallup, M.; McKown, R.L. Reduced Levels of Tear Lacritin Are Associated with Corneal Neuropathy in Patients with the Ocular Component of Sjögren’s Syndrome. Investig. Ophthalmol. Vis. Sci. 2016, 57, 5237–5243. [Google Scholar] [CrossRef]
  221. Vijmasi, T.; Chen, F.Y.T.; Balasubbu, S.; Gallup, M.; McKown, R.L.; Laurie, G.W.; McNamara, N.A. Topical Administration of Lacritin Is a Novel Therapy for Aqueous-Deficient Dry Eye Disease. Investig. Ophthalmol. Vis. Sci. 2014, 55, 5401–5409. [Google Scholar] [CrossRef]
  222. Group, L.S. Lacripep for the Treatment of Primary Sjögren-Associated Ocular Surface Disease: Results of the First-In-Human Study. Cornea 2023, 42, 847–857. [Google Scholar] [CrossRef]
  223. Wasielica-Poslednik, J.; Pfeiffer, N.; Gericke, A. Fluocinolone Acetonide Intravitreal Implant as a Therapeutic Option for Severe Sjögren’s Syndrome-Related Keratopathy: A Case Report. J. Med. Case Rep. 2019, 13, 21. [Google Scholar] [CrossRef] [PubMed]
  224. Terceiro, L.E.L.; Blanchard, A.A.A.; Edechi, C.A.; Freznosa, A.; Triggs-Raine, B.; Leygue, E.; Myal, Y. Generation of Prolactin-Inducible Protein (Pip) Knockout Mice by CRISPR/Cas9-Mediated Gene Engineering. Can. J. Physiol. Pharmacol. 2022, 100, 86–91. [Google Scholar] [CrossRef]
  225. Mondal, H.; Kim, H.-J.; Mohanto, N.; Jee, J.-P. A Review on Dry Eye Disease Treatment: Recent Progress, Diagnostics, and Future Perspectives. Pharmaceutics 2023, 15, 990. [Google Scholar] [CrossRef]
  226. Chang, W.-H.; Liu, P.-Y.; Lin, M.-H.; Lu, C.-J.; Chou, H.-Y.; Nian, C.-Y.; Jiang, Y.-T.; Hsu, Y.-H.H. Applications of Hyaluronic Acid in Ophthalmology and Contact Lenses. Molecules 2021, 26, 2485. [Google Scholar] [CrossRef] [PubMed]
  227. Garrós, N.; Mallandrich, M.; Beirampour, N.; Mohammadi, R.; Domènech, Ò.; Rodríguez-Lagunas, M.J.; Clares, B.; Colom, H. Baricitinib Liposomes as a New Approach for the Treatment of Sjögren’s Syndrome. Pharmaceutics 2022, 14, 1895. [Google Scholar] [CrossRef]
  228. Hofauer, B.; Kirschstein, L.; Graf, S.; Strassen, U.; Johnson, F.; Zhu, Z.; Knopf, A. Inhalative Treatment of Laryngitis Sicca in Patients with Sjögren’s Syndrome—A Pilot Study. J. Clin. Med. 2022, 11, 1081. [Google Scholar] [CrossRef]
  229. Beirampour, N.; Bustos-Salgado, P.; Garrós, N.; Mohammadi-Meyabadi, R.; Domènech, Ò.; Suñer-Carbó, J.; Rodríguez-Lagunas, M.J.; Kapravelou, G.; Montes, M.J.; Calpena, A.; et al. Formulation of Polymeric Nanoparticles Loading Baricitinib as a Topical Approach in Ocular Application. Pharmaceutics 2024, 16, 1092. [Google Scholar] [CrossRef] [PubMed]
  230. Bian, F.; Shin, C.S.; Wang, C.; Pflugfelder, S.C.; Acharya, G.; De Paiva, C.S. Dexamethasone Drug Eluting Nanowafers Control Inflammation in Alkali-Burned Corneas Associated with Dry Eye. Investig. Ophthalmol. Vis. Sci. 2016, 57, 3222–3230. [Google Scholar] [CrossRef] [PubMed]
  231. Song, H.B.; Lee, K.J.; Seo, I.H.; Lee, J.Y.; Lee, S.-M.; Kim, J.H.; Kim, J.H.; Ryu, W. Impact Insertion of Transfer-Molded Microneedle for Localized and Minimally Invasive Ocular Drug Delivery. J. Control. Release 2015, 209, 272–279. [Google Scholar] [CrossRef]
  232. De Campos, A.M.; Sánchez, A.; Alonso, M.J. Chitosan Nanoparticles: A New Vehicle for the Improvement of the Delivery of Drugs to the Ocular Surface. Application to Cyclosporin A. Int. J. Pharm. 2001, 224, 159–168. [Google Scholar] [CrossRef]
  233. Lancina, M.G.; Yang, H. Dendrimers for Ocular Drug Delivery. Can. J. Chem. 2017, 95, 897–902. [Google Scholar] [CrossRef]
  234. Pérez-Carrión, M.D.; Posadas, I. Dendrimers in Neurodegenerative Diseases. Processes 2023, 11, 319. [Google Scholar] [CrossRef]
  235. Yavuz, B.; Bozdağ Pehlivan, S.; Sümer Bolu, B.; Nomak Sanyal, R.; Vural, İ.; Ünlü, N. Dexamethasone—PAMAM Dendrimer Conjugates for Retinal Delivery: Preparation, Characterization and in Vivo Evaluation. J. Pharm. Pharmacol. 2016, 68, 1010–1020. [Google Scholar] [CrossRef]
  236. Chihaby, N.; Orliaguet, M.; Le Pottier, L.; Pers, J.-O.; Boisramé, S. Treatment of Sjögren’s Syndrome with Mesenchymal Stem Cells: A Systematic Review. Int. J. Mol. Sci. 2021, 22, 10474. [Google Scholar] [CrossRef] [PubMed]
  237. Sun, T.; Liu, S.; Yang, G.; Zhu, R.; Li, Z.; Yao, G.; Chen, H.; Sun, L. Mesenchymal Stem Cell Transplantation Alleviates Sjögren’s Syndrome Symptoms by Modulating Tim-3 Expression. Int. Immunopharmacol. 2022, 111, 109152. [Google Scholar] [CrossRef]
  238. Surico, P.L.; Barone, V.; Singh, R.B.; Coassin, M.; Blanco, T.; Dohlman, T.H.; Basu, S.; Chauhan, S.K.; Dana, R.; Di Zazzo, A. Potential Applications of Mesenchymal Stem Cells in Ocular Surface Immune-Mediated Disorders. Surv. Ophthalmol. 2025, 70, 467–479. [Google Scholar] [CrossRef] [PubMed]
  239. Ogata, K.; Matsumura-Kawashima, M.; Moriyama, M.; Kawado, T.; Nakamura, S. Dental Pulp-Derived Stem Cell-Conditioned Media Attenuates Secondary Sjögren’s Syndrome via Suppression of Inflammatory Cytokines in the Submandibular Glands. Regen. Ther. 2021, 16, 73–80. [Google Scholar] [CrossRef] [PubMed]
  240. Matsumura-Kawashima, M.; Ogata, K.; Moriyama, M.; Murakami, Y.; Kawado, T.; Nakamura, S. Secreted Factors from Dental Pulp Stem Cells Improve Sjögren’s Syndrome via Regulatory T Cell-Mediated Immunosuppression. Stem Cell Res. Ther. 2021, 12, 182. [Google Scholar] [CrossRef]
  241. Wen, K.; Li, W.; Cheng, C.; Weige, X.; Jiaqi, C.; Shiyu, S.; Lingyan, H.; Hongwei, W.; Sijing, X. Human Dental Pulp Stem Cells Ameliorate the Imiquimod-Induced Psoriasis in Mice. Heliyon 2023, 9, e13337. [Google Scholar] [CrossRef]
  242. Genç, D.; Bulut, O.; Günaydin, B.; Göksu, M.; Düzgün, M.; Dere, Y.; Sezgin, S.; Aladağ, A.; Bülbül, A. Dental Follicle Mesenchymal Stem Cells Ameliorated Glandular Dysfunction in Sjögren’s Syndrome Murine Model. PLoS ONE 2022, 17, e0266137. [Google Scholar] [CrossRef]
  243. Yang, N.; Liu, X.; Chen, X.; Yu, S.; Yang, W.; Liu, Y. Stem Cells from Exfoliated Deciduous Teeth Transplantation Ameliorates Sjögren’s Syndrome by Secreting Soluble PD-L1. J. Leukoc. Biol. 2022, 111, 1043–1055. [Google Scholar] [CrossRef] [PubMed]
  244. Liu, Y.; Li, C.; Wang, S.; Guo, J.; Guo, J.; Fu, J.; Ren, L.; An, Y.; He, J.; Li, Z. Human Umbilical Cord Mesenchymal Stem Cells Confer Potent Immunosuppressive Effects in Sjögren’s Syndrome by Inducing Regulatory T Cells. Mod. Rheumatol. 2021, 31, 186–196. [Google Scholar] [CrossRef]
  245. Zou, Y.; Xiao, W.; Liu, D.; Li, X.; Li, L.; Peng, L.; Xiong, Y.; Gan, H.; Ren, X. Human Umbilical Cord Mesenchymal Stem Cells Improve Disease Characterization of Sjogren’s Syndrome in NOD Mice through Regulation of Gut Microbiota and Treg/Th17 Cellular Immunity. Immun. Inflamm. Dis. 2024, 12, e1139. [Google Scholar] [CrossRef]
  246. Cong, Y.; Tang, X.; Wang, D.; Zhang, Z.; Huang, S.; Zhang, X.; Yao, G.; Sun, L. Umbilical Cord Mesenchymal Stem Cells Alleviate Sjögren’s Syndrome and Related Pulmonary Inflammation through Regulating Vγ4+ IL-17+ T Cells. Ann. Transl. Med. 2022, 10, 594. [Google Scholar] [CrossRef] [PubMed]
  247. Lee, Y.F.; Yong, D.W.W.; Manotosh, R. A Review of Contact Lens-Induced Limbal Stem Cell Deficiency. Biology 2023, 12, 1490. [Google Scholar] [CrossRef] [PubMed]
  248. Rossen, J.; Amram, A.; Milani, B.; Park, D.; Harthan, J.; Joslin, C.; McMahon, T.; Djalilian, A. Contact Lens-Induced Limbal Stem Cell Deficiency. Ocul. Surf. 2016, 14, 419–434. [Google Scholar] [CrossRef] [PubMed]
  249. Niu, Y.; Ji, J.; Yao, K.; Fu, Q. Regenerative Treatment of Ophthalmic Diseases with Stem Cells: Principles, Progress, and Challenges. Adv. Ophthalmol. Pract. Res. 2024, 4, 52–64. [Google Scholar] [CrossRef]
  250. Keye, P.; Issleib, S.; Gier, Y.; Glegola, M.; Maier, P.; Böhringer, D.; Eberwein, P.; Reinhard, T. Visual and Ocular Surface Benefits of Mini-Scleral Contact Lenses in Patients with Chronic Ocular Graft-versus-Host Disease (GvHD). Sci. Rep. 2024, 14, 25254. [Google Scholar] [CrossRef]
  251. Liu, Y.; Song, S.; Liu, Y.; Fu, T.; Guo, Y.; Liu, R.; Chen, J.; Lin, Y.; Cheng, Y.; Li, Y.; et al. MSCohi-O Lenses for Long-Term Retention of Mesenchymal Stem Cells on Ocular Surface as a Therapeutic Approach for Chronic Ocular Graft-versus-Host Disease. Stem Cell Rep. 2023, 18, 2356–2369. [Google Scholar] [CrossRef]
  252. Møller-Hansen, M.; Larsen, A.-C.; Wiencke, A.K.; Terslev, L.; Siersma, V.; Andersen, T.T.; Hansen, A.E.; Bruunsgaard, H.; Haack-Sørensen, M.; Ekblond, A.; et al. Allogeneic Mesenchymal Stem Cell Therapy for Dry Eye Disease in Patients with Sjögren’s Syndrome: A Randomized Clinical Trial. Ocul. Surf. 2024, 31, 1–8. [Google Scholar] [CrossRef]
  253. Zhao, J.; An, Q.; Zhu, X.; Yang, B.; Gao, X.; Niu, Y.; Zhang, L.; Xu, K.; Ma, D. Research Status and Future Prospects of Extracellular Vesicles in Primary Sjögren’s Syndrome. Stem Cell Res. Ther. 2022, 13, 230. [Google Scholar] [CrossRef] [PubMed]
  254. Li, S.-J.; Cheng, R.-J.; Wei, S.-X.; Xia, Z.-J.; Pu, Y.-Y.; Liu, Y. Advances in Mesenchymal Stem Cell-Derived Extracellular Vesicles Therapy for Sjogren’s Syndrome-Related Dry Eye Disease. Exp. Eye Res. 2023, 237, 109716. [Google Scholar] [CrossRef] [PubMed]
  255. Li, B.; Xing, Y.; Gan, Y.; He, J.; Hua, H. Labial Gland-Derived Mesenchymal Stem Cells and Their Exosomes Ameliorate Murine Sjögren’s Syndrome by Modulating the Balance of Treg and Th17 Cells. Stem Cell Res. Ther. 2021, 12, 478. [Google Scholar] [CrossRef]
  256. Xing, Y.; Li, B.; He, J.; Hua, H. Labial Gland Mesenchymal Stem Cell Derived Exosomes-Mediated miRNA-125b Attenuates Experimental Sjogren’s Syndrome by Targeting PRDM1 and Suppressing Plasma Cells. Front. Immunol. 2022, 13, 871096. [Google Scholar] [CrossRef] [PubMed]
  257. Gong, B.; Zheng, L.; Lu, Z.; Huang, J.; Pu, J.; Pan, S.; Zhang, M.; Liu, J.; Tang, J. Mesenchymal Stem Cells Negatively Regulate CD4+ T Cell Activation in Patients with Primary SjöGren Syndrome through the miRNA-125b and miRNA-155 TCR Pathway. Mol. Med. Rep. 2021, 23, 43. [Google Scholar] [CrossRef]
  258. Rui, K.; Hong, Y.; Zhu, Q.; Shi, X.; Xiao, F.; Fu, H.; Yin, Q.; Xing, Y.; Wu, X.; Kong, X.; et al. Olfactory Ecto-Mesenchymal Stem Cell-Derived Exosomes Ameliorate Murine Sjögren’s Syndrome by Modulating the Function of Myeloid-Derived Suppressor Cells. Cell Mol. Immunol. 2021, 18, 440–451. [Google Scholar] [CrossRef]
Figure 1. Sjögren’s syndrome and Ocular Inflammation. (Parts of this figure were obtained by: https://smart.servier.com and https://www.freepik.com (accessed on 27 February 2025)).
Figure 1. Sjögren’s syndrome and Ocular Inflammation. (Parts of this figure were obtained by: https://smart.servier.com and https://www.freepik.com (accessed on 27 February 2025)).
Immuno 05 00024 g001
Figure 2. Article selection, inclusion, and exclusion methodology (Following the PRISMA guidelines: https://www.prisma-statement.org (accessed on 3 March 2025)).
Figure 2. Article selection, inclusion, and exclusion methodology (Following the PRISMA guidelines: https://www.prisma-statement.org (accessed on 3 March 2025)).
Immuno 05 00024 g002
Figure 3. Structures of Hormones that Affect the Functioning of the Lacrimal Gland. (Structures used in this figure were obtained from the online databases: https://pubchem.ncbi.nlm.nih.gov and https://molview.org (accessed on 15 January 2025)).
Figure 3. Structures of Hormones that Affect the Functioning of the Lacrimal Gland. (Structures used in this figure were obtained from the online databases: https://pubchem.ncbi.nlm.nih.gov and https://molview.org (accessed on 15 January 2025)).
Immuno 05 00024 g003
Figure 4. Innate Immunity, Inflammation, and pSS. (Parts of this figure were obtained from: https://smart.servier.com and https://www.freepik.com (accessed on 27 February 2025)).
Figure 4. Innate Immunity, Inflammation, and pSS. (Parts of this figure were obtained from: https://smart.servier.com and https://www.freepik.com (accessed on 27 February 2025)).
Immuno 05 00024 g004
Figure 5. Common Symptoms Experienced by an SS Patient. (Parts of this figure were obtained from: https://smart.servier.com and https://www.freepik.com (accessed on 15 January 2025)).
Figure 5. Common Symptoms Experienced by an SS Patient. (Parts of this figure were obtained from: https://smart.servier.com and https://www.freepik.com (accessed on 15 January 2025)).
Immuno 05 00024 g005
Figure 6. Ocular Inflammatory Complications associated with Immune Response in SS patients. (Parts of this figure were obtained from: https://smart.servier.com and https://www.freepik.com (accessed on 15 January 2025)).
Figure 6. Ocular Inflammatory Complications associated with Immune Response in SS patients. (Parts of this figure were obtained from: https://smart.servier.com and https://www.freepik.com (accessed on 15 January 2025)).
Immuno 05 00024 g006
Figure 7. Conventional Topical, Systemic, and Interventional Therapeutic Procedures and Adjunctive, Nutritional Therapies for SS Treatment. (Parts of this figure were obtained from: https://smart.servier.com and https://www.freepik.com (accessed on 26 February 2025)).
Figure 7. Conventional Topical, Systemic, and Interventional Therapeutic Procedures and Adjunctive, Nutritional Therapies for SS Treatment. (Parts of this figure were obtained from: https://smart.servier.com and https://www.freepik.com (accessed on 26 February 2025)).
Immuno 05 00024 g007
Figure 8. Illustration of how Vitamins A, E, and D affect SS Mitigation. (Parts of this figure were obtained from: https://molview.org/ and https://www.freepik.com (accessed on 26 February 2025)).
Figure 8. Illustration of how Vitamins A, E, and D affect SS Mitigation. (Parts of this figure were obtained from: https://molview.org/ and https://www.freepik.com (accessed on 26 February 2025)).
Immuno 05 00024 g008
Figure 9. bDMARDs and Immunomodulators for SS Treatment. (Parts of this figure were obtained from: https://smart.servier.com and https://www.freepik.com (accessed on 26 February 2025)).
Figure 9. bDMARDs and Immunomodulators for SS Treatment. (Parts of this figure were obtained from: https://smart.servier.com and https://www.freepik.com (accessed on 26 February 2025)).
Immuno 05 00024 g009
Figure 10. Novel and Emerging SS Therapeutic Interventions. (Parts of this figure were obtained from: https://smart.servier.com and https://www.freepik.com (accessed on 26 February 2025)).
Figure 10. Novel and Emerging SS Therapeutic Interventions. (Parts of this figure were obtained from: https://smart.servier.com and https://www.freepik.com (accessed on 26 February 2025)).
Immuno 05 00024 g010
Table 1. Effects and Roles of Cytokines and Chemokines Involved in SS Autoimmune Mechanisms.
Table 1. Effects and Roles of Cytokines and Chemokines Involved in SS Autoimmune Mechanisms.
Cytokines and ChemokinesMain
Producer(s)
Effect(s)/Role(s)Pathogenesis
Association
Clinical
Significance
References
IFN-γT cells
Monocytes
Macrophages
Pro-inflammatory regulation of MHC class I and IIIncreases the expression of autoantigens, facilitating the immune responseAssociated with increased T-cell infiltration[54,61]
IL-1, IL-2, IL-6, TNFT cells
Monocytes
Macrophages
Pro-inflammatoryThey activate inflammatory pathways and contribute to the destruction of glandular cells.Associated with increased dry eyes and dry mouth[54,62]
IL-10, TGF-βΤ cellsAnti-inflammatoryThey regulate the immune response, reducing autoimmunityTheir reduced expression may contribute to autoimmunity[54,63]
IFN-αPDCs
Monocytes
Antiviral, anti-cancer, pro-inflammatoryTriggers the production of autoantibodies and the release of chemokinesAssociated with relapse and extraneous events[54,55,64]
CXCL12Epithelial cellsBinds to CXCR4 on T cells and PDCsAttracts immune cells to the area of the glandsPromotes chronic inflammation and fibrosis[54,65]
CXCL13Activated and upgraded epitheliumAssociated with CXCR5 in B cellsPromotes the accumulation of B cells and the formation of ectopic lymphocytesAssociated with the likelihood of developing lymphoma[54,65]
CXCL9, CXCL10, CXCL11Upgraded epitheliumAssociated with CXCR3 in T cells and PDCsEnhances cell entry and maintenance in the inflammatory microenvironmentCan serve as a biomarker of disease activity[37,54,65,66,67]
Table 2. Effects and Roles of Active Molecules produced by Breg cells.
Table 2. Effects and Roles of Active Molecules produced by Breg cells.
Breg Cell DerivativeMain Producer(s)Effect(s)/Role(s)References
IL-10Generated by nearly all immune cell types
  • B-cell growth factor
  • Immunosuppression
  • Effective suppression of Th17 cell response
  • Inflammation reduction
  • Tissue destruction
  • Inhibition of Th cell development via the IL-10-dependent mechanism of STAT5 phosphorylation enhancement
  • Key regulatory factor in the suppression of pSS autoimmunity
  • Cell Dysfunction
  • Decrease in IL-10 production, associated with disease exacerbation
[9,26,74,81]
IL-35Immunosuppressive cytokines
  • Restriction of inflammatory response caused by increased Th1/Th17 activity
  • Reduction of destruction of salivary and lacrimal glands
  • Induction of Treg cells, essential for the maintenance of immune tolerance
[26,74,82]
GrBPerforin-induced apoptosis of target cells by a serine protease family
  • GrB uses an independent of perforin-mechanism to stop CD4+ T cells’ growth
  • Potent variations in the frequency of Breg cells producing GrB across various autoimmune disorders
  • pSS individuals’ peripheral blood contains more CD19+/CD5+ B cells that produce GrB
  • They exhibit greater expression of the IL-21 receptor (IL-21R), as well as more unaltered IL-21-producing NK T cells
[19,74,83]
Table 3. Some miRNAs involved in the Onset and Development of Sjögren’s Syndrome.
Table 3. Some miRNAs involved in the Onset and Development of Sjögren’s Syndrome.
Differently Expressed miRNAsCorrelation with SSReferences
miR-146a
  • Increased expression in SS
  • Phagocytosis enhancement
  • Suppression of the production of inflammatory cytokines
  • mi5-146a dysfunction may contribute to: chronic inflammation and SS autoimmunity
[98]
miR-155
  • Increased SS
  • Role in innate and adaptive immunity
  • Its abnormal expression leads to inflammatory autoimmune diseases
  • Its inhibition may mitigate SS symptoms, indicating that miR-155 is a potential therapeutic target
[97,99]
miR-181a
  • Role in the regulation of SS immune response
  • Increased expression in peripheral monocytes, is associated with immune overactivity
  • Decreased expression in salivary glands is associated with their dysfunction
[100]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Pavlidis, K.; Adamantidi, T.; Maria, C.; Georgios, K.; Dania, V.; Krokidis, X.; Tsoupras, A. Sjögren’s Syndrome and Ocular Inflammation: Pathophysiology, Clinical Manifestation and Mitigation Strategies. Immuno 2025, 5, 24. https://doi.org/10.3390/immuno5030024

AMA Style

Pavlidis K, Adamantidi T, Maria C, Georgios K, Dania V, Krokidis X, Tsoupras A. Sjögren’s Syndrome and Ocular Inflammation: Pathophysiology, Clinical Manifestation and Mitigation Strategies. Immuno. 2025; 5(3):24. https://doi.org/10.3390/immuno5030024

Chicago/Turabian Style

Pavlidis, Konstantinos, Theodora Adamantidi, Chatzikamari Maria, Karamanis Georgios, Vasiliki Dania, Xenophon Krokidis, and Alexandros Tsoupras. 2025. "Sjögren’s Syndrome and Ocular Inflammation: Pathophysiology, Clinical Manifestation and Mitigation Strategies" Immuno 5, no. 3: 24. https://doi.org/10.3390/immuno5030024

APA Style

Pavlidis, K., Adamantidi, T., Maria, C., Georgios, K., Dania, V., Krokidis, X., & Tsoupras, A. (2025). Sjögren’s Syndrome and Ocular Inflammation: Pathophysiology, Clinical Manifestation and Mitigation Strategies. Immuno, 5(3), 24. https://doi.org/10.3390/immuno5030024

Article Metrics

Back to TopTop