Back to Journals » Journal of Inflammation Research » Volume 18
Updating on the Dual Role of Salivary Gland Epithelial Cell (SGEC) in Sjögren’s Disease
Authors Zhu W, Lu Y, Zhu Y, Huang X, Wang Y
Received 9 January 2025
Accepted for publication 18 February 2025
Published 1 March 2025 Volume 2025:18 Pages 3039—3053
DOI https://doi.org/10.2147/JIR.S509220
Checked for plagiarism Yes
Review by Single anonymous peer review
Peer reviewer comments 2
Editor who approved publication: Professor Ning Quan
Wen Zhu, Yun Lu, Yifan Zhu, Xiaohan Huang, Yue Wang
Department of Rheumatology & Immunology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210032, People’s Republic of China
Correspondence: Yue Wang, Department of Rheumatology & Immunology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China, Email [email protected]
Abstract: Sjögren’s disease, an autoimmune inflammatory disease, currently lacks effective treatment options. The salivary gland, a crucial exocrine organ responsible for saliva production and local immune responses on mucous membranes, is frequently impaired in individuals with Sjögren’s disease. Restoring salivary gland function poses a significant challenge for researchers. Salivary epithelial cells, recognized as pivotal components of the salivary gland, have been increasingly implicated as key initiators of inflammation and exhibit innate immune cell-like properties. On the whole, SGEC plays a protective role in the physiological state, and can also participate in the persistence of inflammation as an initiating factor in the pathological state. In the review, we explore the interplay between Ca+, endoplasmic reticulum (ER), and mitochondrial homeostasis imbalance in salivary epithelial cells. Additionally, we provide an overview of current literature on research advancements related to Pattern Recognition Receptors (PRRs), programmed cell death, posttranslational modification (PTM), and oral microecology, etc. specifically focusing on their implications in salivary epithelial cells. Given the crucial role of salivary gland epithelial cells in the onset of Sjögren’s disease, a treatment based on salivary gland epithelial cells may have the potential to alleviate the condition by addressing the inflammatory response in the salivary glands.
Keywords: Sjögren’s disease, pattern recognition receptor, salivary gland epithelial cell, immune homeostasis, innate immune
Introduction
Sjögren’s disease (SjD) is a systemic autoimmune disease that mainly involves secretory glands, especially lacrimal and salivary glands, and can involve multiple organs.1 It is often accompanied by fatigue, musculoskeletal pain and other symptoms, which seriously affects the quality of life of patients. The existing epidemiological survey data in various regions show that the incidence and prevalence of SjD vary greatly worldwide.2,3 It is currently believed that among all chronic autoimmune rheumatism, SjD is still one of the diseases with high incidence.4 Current diagnosis and treatment strategies for Sjögren’s disease are not enough to address the current status of lymphocyte infiltration and salivary gland dysfunction.5 According to the complex role of salivary gland epithelial cells in the development of SjD, further research is needed to successfully rescue the salivary gland function of SjD.6 Most of therapy methods can effectively improve the subjective symptoms of patients, but there is less afford to the imbalance of immune system.
![]() |
Figure 1 The function of Salivary Gland Epithelial Cells under physiological conditions. |
![]() |
Figure 2 Diagram illustrating the pathogenic mechanism of SGEC involving dysregulation of mitochondrial and endoplasmic reticulum homeostasis. |
![]() |
Figure 3 PRR signaling pathway are involved in the activation of inflammatory signaling pathways in SGEC. |
Both innate and adaptive immune cells are key pathogenic factors of Sjögren’s disease.7,8 However, Sjögren’s disease has a significant Interferon (IFN)-I characteristic, and the activation of innate immune signals is a key factor in inducing the IFN-I characteristic.9 At present, the role of various innate immune signaling pathways, including TLR, cGAS-STING and NLR, in the pathogenesis of Sjögren’s disease has been clarified.10–12 Given the current status that the etiology of Sjögren’s disease has not been elucidated, in-depth research is crucial to understand its potential pathogenesis and find new therapeutic targets.13 The important role of SGEC in this disease was proposed about forty years ago, but it was ignored for more than 30 years. However, the role of SGEC in this disease are less well understood. As the most important cellular component of the salivary glands, SGEC plays a dual role in this disease.14 Firstly, salivary gland epithelial cells (SGEC) are the victim during the autoimmune injury in Sjögren’s disease.15 Then, as the main cell component of SG, SGEC also plays an important role in secreting saliva, aquaporins and so on.16 In the past decade, the biological function of SGEC has been extensively studied.17 SGEC injury or inappropriate activation can show similar functions to innate immune cells, which can activate innate immune signals, recruit inflammatory cells and aggravate local inflammatory responses.18 Therefore, it is imperative to explore strategies for mitigating pathological injury in SGEC and preserving cellular homeostasis, which warrants significant attention.
The immune system needs to be kept in balance, and the goal of treatment for autoimmune diseases such as SjD is to suppress excessive immune responses and restore stability.19 In general, in SjD, many pathways and processes in the salivary glands may be disrupted, including epithelial cell activation,20 innate immune signaling,21 and adaptive immune activation.22 These studies have been summarized in several excellent researches.23–25 However, most of the emphasis has been on how to protect SGEC from injury. As our understanding of the cellular biology of salivary gland epithelial cells (SGEC) continues to advance, researchers have increasingly focused on the role of SGEC in triggering inflammatory signals and exploring therapeutic strategies aimed at regulating SGEC cell homeostasis.26 These efforts aim to enhance salivary gland (SG) injury recovery and restore immune balance in Sjögren’s disease, thus garnering attention within the scientific community. Recently conducted studies on locally specific gene over-expression in SGEC have provided further evidence supporting the role of SGEC as an inflammatory inducer.27 However, the mechanisms of regulating SGEC cell homeostasis to protect SG function are still unclear.
In this review, we first review the physiological function and the innate immune defense role of SGEC. Second, we explore the immunogenicity of SGEC in pathological conditions. Finally, we summarize the evidence and regulatory mechanisms of proinflammatory signal activation induced by SGEC dyshomeostasis.
The Physiological Functions and Defensive Roles of SGEC
The function and composition of salivary glands provide valuable insights into the overall health status of the body, and numerous studies have been conducted to identify saliva biomarkers for disease diagnosis.28,29 Salivary glands primarily consist of epithelial cells and connective tissue,30 with epithelial cells being the main cell type responsible for saliva secretion within these glands.31 Specifically, salivary gland epithelial cells can be categorized as acini cells, duct cells, myoepithelial cells, among others.32 The function of salivary glands is predominantly manifested through saliva as a carrier. Acini cells produce and secrete mucus-rich saliva while the contraction of surrounding muscle epithelial cells facilitates its release into the oral cavity via ducts.33 Extracellular hormones or neurotransmitters (such as acetylcholine or cholecystokinin) can bind to G protein-coupled receptors on the basal membrane,34,35 serving as initiating signals for various biological processes that trigger Ca2+ release from the endoplasmic reticulum36 and subsequently stimulate extensive primary saliva production. Saliva, serving as a cleansing fluid in the oral cavity, plays a pivotal role in maintaining oral hygiene and safeguarding teeth and oral mucosa against physical and chemical harm.37 Moreover, it harbors numerous proteins that are indispensable for the host’s defense mechanism against pathogens38 Being constantly exposed to foreign pathogens, saliva acts as the primary line of defense for both human and animal oral mucosa. It shields tooth surfaces from acid-induced damage caused by dietary intake and oral bacteria. Patients with insufficient saliva secretion face an elevated risk of tooth demineralization, dental caries, oral mucositis, and fungal infections.32 Additionally, saliva encompasses a plethora of signaling molecules including nerve growth factor (NGF),39 epidermal growth factor (EGF),40 fibroblast growth factor (FGF),41 and vascular endothelial growth factor (VEGF)42 which play crucial roles in promoting healing processes of both oral mucosal and esophageal wounds as well as facilitating skin wound repair. Furthermore, secretory immunoglobulin IgA assumes a vital function in orchestrating immune responses within the oral mucosa.43 The concised function of Salivary Gland Epithelial Cells under physiological conditions can be seen in Figure 1.
Immunogenicity in SGEC Pathological State
The function of SGEC in physiological state is as described above. In pathological state, SGEC can also act as a disease inducing factor and participate in the progression of the disease.17 Currently, the factors that have been found to cause damage to SGEC include calcium homeostasis imbalance, mitochondrial dysfunction, endoplasmic reticulum stress, activation of the PRR signaling pathway, programmed cell death and so on.
Calcium Homeostasis Imbalance
Saliva secretion is a two-stage process in which glandular cells produce an isotonic saliva initially, which is then modified by the duct system into a hypotonic fluid.44 Ca2+ is a key factor in controlling salivary gland function. Studies have found that in physiological conditions, salivary mucous cells enhance PKC activity by inducing the release of intracellular Ca2+ pools through VIP, thereby mediating the co-secretion of mucin proteins with muscarinic.45 In pathological conditions, defects in Ca2+ signaling have been observed in radiation-induced loss of salivary gland function46 and salivary gland dysfunction associated with Sjögren’s disease.47 Defects in epithelial cell calcium signaling in Sjögren’s disease are associated with a decline in salivary gland secretion.48 Recent studies have found that IL-17 causes salivary gland dysfunction in Sjögren’s disease by inhibiting TRPC1-mediated calcium transport.49 Inducing Ca2+ signaling can promote saliva secretion and prevent immune cells from infiltrating the salivary gland in a Sjögren’s disease mouse model.50 However, the cause of calcium homeostasis imbalance in the salivary gland of Sjögren’s disease patients still needs further research.
Mitochondrial Dysfunction
Oxidative-Reduction Homeostasis Imbalance
In almost all subcellular organelles, including the cytoplasm, endoplasmic reticulum (ER), mitochondria, and peroxisomes, reactive oxygen species (ROS) can be generated as byproducts of their fundamental metabolic functions.51 Physiological ROS plays a pivotal role in regulating cellular function disorders under normal conditions. However, it can also contribute to the pathogenesis of various diseases such as infectious diseases,52 autoimmune diseases,53 tumors,54 and metabolic disorders.55 The functions of immune cells are intricately linked to diverse metabolic pathways, with immune cell metabolism being inseparable from redox reactions.56 Studies have revealed that patients with Sjögren’s disease exhibit downregulation of glutathione peroxidase 4 (GPX4) expression in salivary glands. This downregulation induces an increase in lipid ROS levels within SjD SGECs, subsequently promoting STAT4 phosphorylation and nuclear translocation. Consequently, STAT4 binds to the AQP5 promoter region leading to inhibition of AQP5 expression and saliva secretion.57 Experimental studies have further demonstrated significantly elevated ROS fluorescence levels accompanied by reduced mitochondrial membrane potential in NOD mice SGEC compared to the ICR group.58 Collectively, these findings suggest that ROS accumulation is a critical factor contributing to mitochondrial damage and hypofunction in SGEC during Sjögren’s disease.
Mitochondrial Metabolic Dysfunction
Inflammatory epithelial diseases are caused by a co-disorder of immune cells and epithelial cells.59 Sjögren’s disease was previously known as autoimmune epithelitis, characterized by abnormal activation of epithelial cells and extensive infiltration of lymphocytes in the salivary gland. From an immune-metabolic perspective, it is evident that any metabolic alterations in target epithelial cells in Sjögren’s disease (SjD), regardless of the underlying cause, can induce an immunogenic phenotype.60 Thus, salivary gland epithelial cells (SGEC) have the potential to directly modulate the susceptibility and/or severity of autoimmune responses. In comparison to normal SGEC, SjD-SGEC exhibit reduced mitochondrial content, swollen and elongated mitochondria, as well as fewer and aberrant cristae.61 These metabolic changes primarily involve mitochondria and are accompanied by pronounced morphological alterations in situ. Transcriptome sequencing offers insights into the transformation process of SGEC from SjD patients into innate immune cells while uncovering translational modifications associated with metabolic remodeling.14 RNA sequencing-based investigations have revealed distinct distribution patterns of innate and adaptive immune cells within salivary gland tissue from primary Sjögren’s disease (SjD) patients, which are linked to diverse mitochondrial metabolic pathways thereby influencing disease progression.62,63 Further research is warranted to elucidate the role of mitochondrial metabolism in SGEC during Sjögren’s disease.
Maladaptive Mitochondrial Autophagy
Autophagy is a crucial regulatory pathway for cellular self-protection in various immune cells, including neutrophils, eosinophils, mast cells, and NK cells, enabling the maintenance of cellular homeostasis.64 Recently, researchers have also started to focus on autophagy in salivary gland epithelial cells. Current studies have demonstrated that modulating autophagy can exert a protective effect on the parotid gland pathology in Sjögren’s disease. Conversely, inhibiting autophagy can worsen the parotid gland pathology.58,65 Interestingly, some studies indicate a significant correlation between the level of autophagy in small salivary gland lymphocytes of Sjögren’s disease patients and the extent of lymphocyte infiltration within these glands.66 The m6A methylation process may participate in immune infiltration and autophagy regulation in primary Sjögren’s disease (SjD), thereby contributing to its pathogenesis.67 Furthermore, certain researchers propose that inflammation-induced autophagy and survival mechanisms promote activation of SGECs in primary SjD SGECs and reflect histopathological severity. They consider autophagy as a central factor underlying primary SjD pathogenesis and suggest it as a potential therapeutic target.68 However, further research is needed to confirm the role of autophagy in salivary gland epithelial cells of Sjögren’s disease patients.
Endoplasmic Reticulum Stress
The endoplasmic reticulum (ER) is the largest organelle in the cell and plays a crucial role in protein synthesis, lipid metabolism, and other essential cellular processes. When triggered by internal and external stimuli such as ischemia,69 oxidative stress,70 infection,71 drug toxicity,72 and calcium homeostasis imbalance,73 disturbances in ER homeostasis lead to the unfolded protein response (UPR), resulting in the accumulation of misfolded proteins within the ER and initiating the endoplasmic reticulum stress (ERS) response to restore ER homeostasis. In pathological conditions, ERS can also modulate immune cell functions, contributing to immune system dysregulation.74 Chronic inflammation can disrupt MUC1 secretion and induce ER stress, impacting saliva quality in Sjögren’s disease patients.75 Furthermore, it has been demonstrated that the GRP78-ATF6-CHOP signaling pathway associated with ER stress is overactivated in primary Sjögren’s disease.76 Sjögren’s disease is characterized by the excessive expression of type I interferon,77 which can induce ERS in SGECs, leading to the massive production of Ro52/SSA antigen and downregulation of autophagy, thereby increasing cell apoptosis.78 Interestingly, studies have also found that merc in SGEC mediates the transfer of calcium from the endoplasmic reticulum to the mitochondria, promoting ATP production and playing a crucial role in calcium homeostasis.79 In contrast, the inflammatory cascade signal triggered by endoplasmic reticulum stress in pathological conditions is an important factor causing dysfunction of salivary gland epithelial cells in Sjögren’s disease. Calcium homeostasis imbalance and mitochondrial damage in salivary gland epithelial cells can be seen in Figure 2.
Activation of the PRR Signaling Pathway
Innate immunity distinguishes various pathogen-associated molecular patterns (PAMPs) as the first line of defense against pathogen infection.80 Common PRRs include Toll-like receptors (TLRs), C-type lectin receptors (CLRs), nod-like receptors (NLRs), and retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs).81 Recently, salivary gland epithelial cells have been considered a special innate immune cell,14,60 and they also express a variety of PRRs on their surfaces. The specific summary is as follows.
Activation of Inflammasome
Several types of inflammasomes, such as NLRP1 inflammasome, NLRP3 inflammasome, NLRC4 inflammasome, IPAF inflammasome, and AIM2 inflammasome,82 have been identified. Among these, the NLRP3 inflammasomes have been extensively studied in the context of Sjögren’s disease. The NLRP3 inflammasome is known to play a pivotal role in the pathogenesis and progression of various autoimmune diseases.83 Recent research has demonstrated activation of the NLRP3 inflammasome in the salivary glands of Sjögren’s disease patients,84 with a potential mechanism involving SGEC pyroptosis induced by type I IFN in SjD patients.85 Although there have been studies investigating the involvement of the AIM2 inflammasome in Sjogren’s syndrome pathogenesis, most of them have primarily focused on peripheral blood mononuclear cells and lacrimal epithelial cells. Therefore, further investigations are warranted to elucidate the role of AIM2 and other inflammasomes in salivary epithelial cell dysfunction.
Activation of TLR Signaling Pathway
Toll-like receptors (TLRs) are a distinct subset of PRRs, playing a pivotal role in pathogen recognition and inflammation induction.86 Currently, 11 TLR family members have been identified, with TLR1, TLR2, TLR4, TLR5, TLR6, and TLR11 located on the cell surface and TLR3, TLR7, TLR8, and TLR9 situated in the endosome/lysosome compartment.87 In the context of Sjögren syndrome (SjD), particular attention has been given to the study of TLR2, TLR3, TLR4, and TLR7. Interleukin-15 (IL-15), an inflammatory cytokine implicated in SjD pathogenesis, is induced by SGEC through activation of TLR2, resulting in NF-kB-mediated inflammatory responses.88 Additionally, TLR3 is involved in apoptosis mediation and phosphorylated Akt activation within salivary gland epithelial cells from SjD patients.89 TLR-7 is predominantly expressed in monocytes and ducts of minor salivary glands in patients with Sjögren’s disease, and is associated with downstream signaling pathways related to type I interferon, suggesting that TLR-7-mediated innate immunity plays a role in the development of glandular inflammation in SjD.90 Additionally, TLR7 activation of SGEC cells in SjD patients promotes the presentation of the TRIM21/Ro52-SS-A antigen via the MHC class I pathway.91 Conversely, lysosomal-associated membrane protein 3 induces ectopic TLR7 expression on salivary gland epithelial cells, amplifying the type I interferon response in Sjögren’s disease.92 A notable characteristic of SjD patients is the presence of mucin proteins outside their normal location within the extracellular matrix of salivary glands. These ectopic salivary mucin proteins can be recognized by TLR4 expressed on epithelial cells, initiating an inflammatory response and attracting inflammatory cells to proliferate and prolonging inflammation, thereby promoting the chronic features of SjD.93
Activation of the cGAS-STING Signaling Pathway
cGAS plays a crucial role as a sensing protein in the innate immune response, in recognizing microbial DNA and endogenous DNA (including mitochondrial DNA (mtDNA) and genomic DNA).94 Recent findings suggest that activation of the cGAS-STING pathway can promote the expression of type I interferon (IFN) and pro-inflammatory cytokines, which play an important role in the pathogenesis of Sjögren’s disease.12,79,95 Studies have found that patients with SjD have a high lactate environment in their labial glands, and lactate can damage mitochondrial DNA (mtDNA) and cause its leakage, thereby activating the cGAS-STING pathway.96 Our previous studies have found that the cGAS-STING signaling pathway is significantly activated in salivary glands of Sjögren’s disease patients. The cGAS-STING signaling pathway is significantly related to the pathogenesis of Sjögren’s disease, and regulating autophagy is a key brake that limits the activation of the cGAS-STING signaling pathway.58 However, the specific regulatory mechanisms remain to be further elucidated.
Other Pattern Recognition Receptor-Related Signaling Pathways
AIM2 (Absent in melanoma 2) is a cytoplasmic DNA-recognition protein capable of identifying double-stranded DNA, and forms the AIM2 inflammasome, a protein platform that triggers the innate immune response. Studies have demonstrated that genomic DNA activation of the AIM2 inflammasome and STING leads to an inflammatory reaction in meibomian gland MECs, potentially contributing to the development of SjD.97 However, there is currently limited research on how C-type lectin receptors and RIG-I-like receptor-related signaling pathways impact salivary gland epithelial cells in Sjögren’s disease patients. Figure 3 shows activation of pattern recognition receptor-associated signaling pathways in salivary gland epithelial cells.
Programmed Cell Death
Programmed cell death (PCD) refers to the elimination of unwanted cells through multiple distinct pathways in order to maintain cellular homeostasis. The main types of programmed cell death include apoptosis, autophagy, ferroptosis, and pyroptosis, among others.98 Increased apoptosis of epithelial cells is the main cause of exocrine gland dysfunction and epithelial gland structural damage in patients with SjD.99 Therefore, lymphocyte-induced epithelial cell apoptosis is considered a key factor in decreased exocrine function. Subsequently, the release of antigens into the apoptotic bodies from the apoptotic cells is the main route for presenting self-antigens to the immune system.10 In vitro studies have shown that epithelial cell apoptosis can trigger the release of apoptotic bodies containing nuclear material, including Ro/SSA and La/SSB antigens.23 Among these, autophagy and pyroptosis have already been reviewed earlier, so they are not discussed again.
Ferroptosis is a novel form of regulated cell death (RCD) caused by fatal accumulation of iron-dependent and lipid peroxidation products, ultimately leading to membrane damage and cell death.100 It is currently believed that epidermal cell-derived proteins involved in ferroptosis are present in plasma exosomes from SjD patients. Complement C5 and C9 may be new molecules involved in ferroptosis that play an important role in the pathology of SjD epithelial cells. Plasma exosomes from SjD patients, rather than those from non-autoimmune sicca syndrome (nSS) patients, contain ferroptosis-related proteins. The content of apoptosis-related proteins in exosomes reflects the state of epithelial cell lesions more than that in plasma.101 Interferon-γ induces ferroptosis in salivary gland epithelial cells of Sjögren’s disease (SjD) through JAK/stat1 mediated inhibition of system Xc. JAK or STAT1 inhibition in SGEC reverses the downregulation of SLC3A2 and GPX4 induced by IFN-γ, as well as IFN-γ-induced cell death.15 GPX4 is a key molecule in the ferroptosis process. Downregulation of GPX4 expression in salivary gland epithelial cells can cause salivary secretion dysfunction in Sjögren’s disease through lipid ROS/pSTAT4/AQP5 axis.57
Crosstalk Between SGECs and Lymphocytes
SGECs play a pivotal role in the pathological changes of lymphocyte infiltration within the salivary gland of patients with Sjögren’s disease (SjD). Current research primarily focuses on elucidating the interactions between epithelial cells and B cells, as well as T cells. SGECs possess the ability to regulate recruitment, activation, and differentiation of immune cells in SjD. Phenotypic flow cytometry analysis and cytokine studies have confirmed that SGECs secrete B cell activating factor (BAFF) while also modulating the activation and differentiation processes of B cells.102 Gene expression profiling in SGECs has revealed upregulation of genes associated with interferon signaling pathway and immune response (HLA-DRA, IL-7, and B cell activation factor receptor) in primary Sjögren’s disease (SjD). Additionally, activated genes CD40 and CD48 exhibited upregulation specifically in salivary gland-derived B lymphocytes from SjD patients. Furthermore, SGECs can enhance survival of B lymphocytes; notably, those derived from SjD patients exhibit higher pro-B lymphocyte survival activity compared to controls.103 Moreover, LAMP3 expression can induce apoptosis in SGECs leading to reduced expression of proteins involved in saliva secretion along with impaired lysosomal function. Consequently, damage-associated molecular patterns (DAMPs) are released via Toll-like receptors to activate immune cells.18 However, further investigations are warranted to ascertain whether these interactions hold true universally.
Abnormal Epigenetic Modifications
The key mechanisms of epigenetics encompass various processes, including methylation modification, histone modification, and non-coding RNA regulation.104 During the progression of Sjögren’s disease, significant alterations in epigenetic modifications are frequently observed in immune cells and salivary gland epithelial cells, indicating a robust association with autoimmune responses.105 Currently, more and more research is focusing on the role of epigenetic changes in the onset of Sjögren’s disease. Methylation modification encompasses DNA methylation and RNA methylation, which play pivotal roles in regulating gene expression and cellular functions by modulating the accessibility of DNA to transcription factors and other regulatory proteins.106 The present study investigated the association between DNA methylation and the European League Against Rheumatism (EULAR) Sjögren Syndrome Disease Activity Index (ESSDAI) score, revealing a significantly higher number of differential methylation regions in patients with high ESSDAI scores compared to those with low ESSDAI scores.107 The study revealed an increased proportion of B cells in the LSG tissue of SjD patients compared to non-SjD controls, accompanied by upregulated gene expression associated with B cell function and reduced methylation of genes involved in immune response and immune tolerance.108 An epigenetic analysis of LSGs in SjD patients revealed distinct methylation patterns at CpG sites with differential methylation between SjD subgroups, providing evidence for the involvement of epigenetic factors in the heterogeneity of SjD.109 The dysfunction of the IRE1alpha/XBP-1 pathway, which is associated with DNA methylation, may result in impaired salivary gland function in patients diagnosed with Sjögren’s disease.110 The aforementioned findings further underscore the significance of methylation modifications in the pathogenesis of SjD and have the potential to inform the development of novel diagnostic and therapeutic strategies.111
The RNA modification methyladenosine N6 (m6A) is widely recognized as the most prevalent form of RNA methylation. Extensive research has demonstrated its significant involvement in immune infiltration and autophagy processes associated with SjD.67 The presence of M6A methylation and the upregulation of METTL3 protein exhibit associations with blood serological markers and dry eye symptoms in patients diagnosed with primary Sjögren’s disease.112 When exposed to inflammatory stimuli, the impairment of m6A modification in SGEC facilitates the generation of double-stranded RNA (dsRNA), thereby potentially amplifying the interferon cycle and contributing to the pathogenesis of Sjögren’s disease.113 However, METTL3-mediated m6A modifications impeded the formation of dsRNA and activation of IFN signaling. Additionally, the m6A methylation detector can aid in subtyping patients with Sjögren’s disease into diagnostic subgroups through consensus clustering, thereby categorizing SjD patients into distinct m6A patterns. The m6A score of Group B patients surpasses that of Group A patients.114 However, further investigation is required to elucidate the immunoregulatory role of methylation modification in this disease.
Post-translational modifications (PTMs) are implicated in a diverse array of biological processes and play a crucial role in modulating protein structure, activity, and function.115 Numerous studies have demonstrated the widespread involvement of protein post-translational modifications in the regulation of inflammatory processes.116 However, there remains a paucity of research on PTMs in the pathogenesis of Sjögren’s disease. A recent comparative proteomic analysis of serum samples identified and validated ITIH3 as a potential biomarker, while also pinpointing hexosamine and arginine modification sites on ITIH3. Furthermore, analysis of autoantibody subtypes against arginine-modified ITIH3 peptides allowed for further differentiation between patients with rheumatoid arthritis (RA), primary Sjögren’s disease, RA-secondary SjD, and healthy controls.117 Additionally, some studies have characterized the newly discovered Kmal (lysine acetylation form) in SjD and provided proteomic data for SjD patients. Notably, several key differentially modified proteins were found to be associated with the cell adhesion pathway, which is implicated in the development of SjD.118 In terms of PTMS, it is also a research direction that needs to be paid attention to in the future to recognize and regulate the immune homeostasis of salivary epithelial cells in response to imbalance immune responses.
Oral Microecology Disorder
There is increasing evidence that the diversity of the microbiome is associated with high disease activity in Sjögren’s disease.119 Changes in the microbiome have a negative impact on the pathogenesis of Sjögren’s disease. However, the causal relationship, especially the impact of oral microecology on salivary gland epithelial cells, is not yet clear.120 Researchers have detected exogenous (microbial) or endogenous (endogenous retrovirus) genomes in salivary gland epithelial cells of Sjögren’s disease patients, which may help clarify the mechanism by which microorganisms participate in the activation of salivary gland epithelial cells in Sjögren’s disease.121 In addition, bioinformatics analysis based on the transcriptome of salivary glands can also reveal the existence of multiple innate immune signaling pathways and signal activation related to pathogen infection in salivary glands of Sjögren’s disease patients, indicating that disruption of oral microecology participates in the pathogenesis of the disease.58 However, diagnostic indicators related to this need to be further validated by large-sample clinical studies, and the effects of oral microecology on SGEC epithelial cells need to be further elucidated through experimental studies.
Future Outlook
The presence of unknown trigger factors in salivary gland epithelial cells may activate the innate immune response and subsequently lead to an adaptive immune response against self-antigens, thereby positioning the epithelial cells as both the medium and target of the response.122 In recent years, advancements in transcriptomics, proteomics, and metabolomics technologies have provided a deeper understanding of the physiological and pathological mechanisms underlying Sjögren’s disease. The altered oral microbiome in Sjögren’s disease patients has been a focal point for researchers for numerous years.123 Investigations into SjD-like microbial dysbiosis in individuals with pre-SjD or non-SjD related diseases suggest that changes in the microbial community may precede the onset of primary Sjögren’s disease. Furthermore, it has been observed that SjD patients treated with HCQ experience partial alleviation of microbial dysbiosis. However, there remains disordered composition within the microbial community.124 A reduction in saliva flow seems to be unrelated to simple microbial dysbiosis but rather affects host-related risk factors.125 A bioinformatics analysis of the composition of salivary microbiota revealed significant differences in oral microbial composition between the anti-SSA antibody positive and negative groups. The microbial diversity of patients with SjD was lower than that of non-SjD patients. And several potential genetic markers of SjD at the level of microbiota were identified, such as a decrease in the abundance of Lactobacillus or an increase in the abundance of Streptococcus.126 There are significant differences in microbial dysbiosis between SjD patients and non-SjD patients, and it is unrelated to oral candidiasis and DMFT.127 The mechanism by which oral microecology participates in the physiological and pathological process of this disease still needs further in-depth research.
In addition, while research on single-cell sequencing in Sjögren’s disease is gradually increasing, the focus has primarily been on mononuclear cells128 and CD4 T cells.129 However, there remains a significant gap in genomic sequencing that specifically targets the single-cell transcriptome and spatial transcriptome of salivary gland epithelial cells. Although single-cell RNA sequencing (scRNA-seq) can identify cell subpopulations within tissues, it does not capture their spatial distribution or reveal the local network of cell-to-cell communication for in situ action. On the other hand, spatial transcriptomics can elucidate RNA localization within tissues but lacks complete transcriptomic information.130 Therefore, there is an urgent need to integrate the results of single-cell and spatial transcriptome sequencing to map out the trajectory of cell differentiation and gene regulatory networks, as well as to characterize cell-to-cell communication and regulatory networks within tissues.131 This integration will further illuminate the role played by salivary gland epithelial cells in shaping the immune microenvironment of salivary glands in patients with Sjögren’s disease.
Restoring the function of the salivary gland is the primary objective in treating Sjögren’s disease,132 as it relies on the functional integrity of epithelial tissue. However, there remains a limited understanding of SGEC in this disease. Consequently, researchers primarily focus on comprehending secretory physiology and exploring potential regenerative strategies to repair SGs and restore saliva production.133 It is imperative to adopt a comprehensive perspective that encompasses the interplay between innate immunity and adaptive immunity while elucidating the role of salivary gland epithelial cells in this process.134 Nevertheless, current research predominantly concentrates on individual cell interactions,135 necessitating an urgent need for holistic comprehension of local immune physiology and pathology within submental gland tissue. As immunologists, we can consider utilizing organoid research or clinically-based investigations as pivotal approaches to unraveling Sjögren’s disease pathogenesis in future studies.
Conclusion
SGEC plays a protective role in the physiological state, and can also participate in the persistence of inflammation as an initiating factor in the pathological state. Given the crucial role of salivary gland epithelial cells in the onset of Sjögren’s disease, a treatment based on salivary gland epithelial cells may have the potential to alleviate the condition by addressing the inflammatory response in the salivary glands. How to restore the immune function of hyperactive salivary gland epithelial cells to a state of calmness is a worthy subject for future research.
Data Sharing Statement
This is review paper and no data has been produced.
Acknowledgments
We thank the Project of National Natural Science Foundation of China and the affiliated hospital of Nanjing University of Chinese medicine for their funding support. The authors declare that they have not use AI-generated work in this manuscript.
Author Contributions
All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.
Funding
This work was supported by the National Natural Science Foundation of China (grant 82274454) and the Postgraduate Research & Practice Innovation Program of Jiangsu Province (KYCX21_1682).
Disclosure
The authors declare no competing interests.
References
1. Cai B, Guo Z, Yan Q, et al. Clinical features and risk factors of primary Sjögren’s syndrome complicated with severe pneumonia: a case-control study. Clin Rheumatol. 2024;43(5):1665–1674. doi:10.1007/s10067-024-06942-3
2. Seror R, Chiche L, Beydon M, et al. Estimated prevalence, incidence and healthcare costs of Sjögren’s syndrome in France: a national claims-based study. RMD Open. 2024;10(1):e003591. doi:10.1136/rmdopen-2023-003591
3. Conrad N, Misra S, Verbakel JY, et al. Incidence, prevalence, and co-occurrence of autoimmune disorders over time and by age, sex, and socioeconomic status: a population-based cohort study of 22 million individuals in the UK. Lancet. 2023;401(10391):1878–1890. doi:10.1016/S0140-6736(23)00457-9
4. Beydon M, McCoy S, Nguyen Y, et al. Epidemiology of Sjögren syndrome. Nat Rev Rheumatol. 2024;20(3):158–169. doi:10.1038/s41584-023-01057-6
5. Chowdhury F, Tappuni A, Bombardieri M. Biological therapy in primary Sjögren’s syndrome: effect on salivary gland function and inflammation. Front Med. 2021;8:707104. doi:10.3389/fmed.2021.707104
6. Bodewes ILA, Björk A, Versnel MA, Wahren-Herlenius M. Innate immunity and interferons in the pathogenesis of Sjögren’s syndrome. Rheumatology. 2021;60(6):2561–2573. doi:10.1093/rheumatology/key360
7. Kawka L, Felten R, Schleiss C, et al. Alteration of innate lymphoid cell homeostasis mainly concerns salivary glands in primary Sjögren’s syndrome. RMD Open. 2023;9(2):e003051. doi:10.1136/rmdopen-2023-003051
8. Chivasso C, Sarrand J, Perret J, et al. The involvement of innate and adaptive immunity in the initiation and perpetuation of Sjögren’s syndrome. Int J mol Sci. 2021;22(2):658. doi:10.3390/ijms22020658
9. Brkic Z, Versnel MA. Type I IFN signature in primary Sjögren’s syndrome patients. Expert Rev Clin Immunol. 2014;10(4):457–467. doi:10.1586/1744666X.2014.876364
10. Witas R, Shen Y, Nguyen CQ. Bone marrow-derived macrophages from a murine model of Sjögren’s syndrome demonstrate an aberrant, inflammatory response to apoptotic cells. Sci Rep. 2022;12(1):8593. doi:10.1038/s41598-022-12608-4
11. Xia X, Long Q, Zha J, et al. IL-27 regulates NLRP3 inflammasome activation of MDSCs in experimental Sjögren’s syndrome. Int Immunol. 2023;35(11):531–542. doi:10.1093/intimm/dxad037
12. Huijser E, Bodewes ILA, Lourens MS, et al. Hyperresponsive cytosolic DNA-sensing pathway in monocytes from primary Sjögren’s syndrome. Rheumatology. 2022;61(8):3491–3496. doi:10.1093/rheumatology/keac016
13. Thorlacius GE, Björk A, Wahren-Herlenius M. Genetics and epigenetics of primary Sjögren syndrome: implications for future therapies. Nat Rev Rheumatol. 2023;19(5):288–306. doi:10.1038/s41584-023-00932-6
14. Katsiougiannis S, Stergiopoulos A, Moustaka K, et al. Salivary gland epithelial cell in Sjögren’s syndrome: metabolic shift and altered mitochondrial morphology toward an innate immune cell function. J Autoimmun. 2023;136:103014. doi:10.1016/j.jaut.2023.103014
15. Cao T, Zhou J, Liu Q, et al. Interferon-γ induces salivary gland epithelial cell ferroptosis in Sjogren’s syndrome via JAK/STAT1-mediated inhibition of system Xc(). Free Radic Biol Med. 2023;205:116–128. doi:10.1016/j.freeradbiomed.2023.05.027
16. Tzartos JS, Stergiou C, Daoussis D, et al. Antibodies to aquaporins are frequent in patients with primary Sjögren’s syndrome. Rheumatology. 2017;56(12):2114–2122. doi:10.1093/rheumatology/kex328
17. Tang Y, Zhou Y, Wang X, et al. The role of epithelial cells in the immunopathogenesis of Sjögren’s syndrome. J Leukoc Biol. 2024;115(1):57–67. doi:10.1093/jleuko/qiad049
18. Nakamura H, Tanaka T, Pranzatelli T, et al. Lysosome-associated membrane protein 3 misexpression in salivary glands induces a Sjögren’s syndrome-like phenotype in mice. Ann Rheum Dis. 2021;80(8):1031–1039. doi:10.1136/annrheumdis-2020-219649
19. Emanuel E, Arifuzzaman M, Artis D. Epithelial-neuronal-immune cell interactions: implications for immunity, inflammation, and tissue homeostasis at mucosal sites. J Allergy Clin Immunol. 2024;153(5):1169–1180. doi:10.1016/j.jaci.2024.02.004
20. Dong Y, Ming B, Gao R, et al. The IL-33/ST2 axis promotes primary Sjögren’s syndrome by enhancing salivary epithelial cell activation and type 1 immune response. J Immunol. 2022;208(12):2652–2662. doi:10.4049/jimmunol.2101070
21. Stern ME, Theofilopoulos AN, Steven P, et al. Immunologic basis for development of keratoconjunctivitis sicca in systemic autoimmune diseases: role of innate immune sensors. Ocul Surf. 2024;32:130–138. doi:10.1016/j.jtos.2024.02.003
22. Joachims ML, Khatri B, Li C, et al. Dysregulated long non-coding RNA in Sjögren’s disease impacts both interferon and adaptive immune responses. RMD Open. 2022;8(2):e002672. doi:10.1136/rmdopen-2022-002672
23. Zhou J, Pathak JL, Liu Q, et al. Modes and mechanisms of salivary gland epithelial cell death in Sjogren’s syndrome. Adv Biol. 2023;7(12):e2300173. doi:10.1002/adbi.202300173
24. Noll B, Beckman M, Bahrani Mougeot F, et al. Exploring salivary epithelial dysfunction in Sjögren’s disease. Int J mol Sci. 2024;25(9):4973. doi:10.3390/ijms25094973
25. Qi W, Tian J, Wang G, et al. Advances in cellular and molecular pathways of salivary gland damage in Sjögren’s syndrome. Front Immunol. 2024;15:1405126. doi:10.3389/fimmu.2024.1405126
26. Manfrè V, Chatzis LG, Cafaro G, et al. Sjögren’s syndrome: one year in review 2022. Clin Exp Rheumatol. 2022;40(12):2211–2224. doi:10.55563/clinexprheumatol/43z8gu
27. Nakamura H, Tanaka T, Zheng C, et al. Lysosome-associated membrane protein 3 induces lysosome-dependent cell death by impairing autophagic caspase 8 degradation in the salivary glands of individuals with Sjögren’s disease. Arthritis Rheumatol. 2023;75(9):1586–1598. doi:10.1002/art.42540
28. Ren Y, Ma Q, Zeng X, et al. Saliva‑microbiome‑derived signatures: expected to become a potential biomarker for pulmonary nodules (MCEPN-1). BMC Microbiol. 2024;24(1):132. doi:10.1186/s12866-024-03280-x
29. Marín-Echeverri C, Pérez-Zapata L, Álvarez-Acevedo L, et al. Diagnostic performance, stability, and acceptability of self-collected saliva without additives for SARS-CoV-2 molecular diagnosis. Eur J Clin Microbiol Infect Dis. 2024;43(6):1127–1138. doi:10.1007/s10096-024-04819-6
30. Shang YF, Shen YY, Zhang MC, et al. Progress in salivary glands: endocrine glands with immune functions. Front Endocrinol. 2023;14:1061235. doi:10.3389/fendo.2023.1061235
31. Woznicki JA, Saini N, Flood P, et al. TNF-α synergises with IFN-γ to induce caspase-8-JAK1/2-STAT1-dependent death of intestinal epithelial cells. Cell Death Dis. 2021;12(10):864. doi:10.1038/s41419-021-04151-3
32. Chibly AM, Aure MH, Patel VN, et al. Salivary gland function, development, and regeneration. Physiol Rev. 2022;102(3):1495–1552. doi:10.1152/physrev.00015.2021
33. Verstappen GM, Pringle S, Bootsma H, et al. Epithelial-immune cell interplay in primary Sjögren syndrome salivary gland pathogenesis. Nat Rev Rheumatol. 2021;17(6):333–348. doi:10.1038/s41584-021-00605-2
34. Fasciani I, Petragnano F, Wang Z, et al. The C-terminus of the prototypical M2 muscarinic receptor localizes to the mitochondria and regulates cell respiration under stress conditions. PLoS Biol. 2024;22(4):e3002582. doi:10.1371/journal.pbio.3002582
35. Wang H, Qian T, Zhao Y, et al. A tool kit of highly selective and sensitive genetically encoded neuropeptide sensors. Science. 2023;382(6672):eabq8173. doi:10.1126/science.abq8173
36. Sneyd J, Vera-Sigüenza E, Rugis J, et al. Calcium dynamics and water transport in salivary acinar cells. Bull Math Biol. 2021;83(4):31. doi:10.1007/s11538-020-00841-9
37. Pedersen A, Sørensen CE, Proctor GB, et al. Salivary functions in mastication, taste and textural perception, swallowing and initial digestion. Oral Dis. 2018;24(8):1399–1416. doi:10.1111/odi.12867
38. Kaibori Y, Tamoto S, Okuda S, et al. CCL28: a promising biomarker for assessing salivary gland functionality and maintaining healthy oral environments. Biology. 2024;13(3):147. doi:10.3390/biology13030147
39. Schenck K, Schreurs O, Hayashi K, et al. The role of Nerve Growth Factor (NGF) and its precursor forms in oral wound healing. Int J mol Sci. 2017;18(2):386. doi:10.3390/ijms18020386
40. Pang X, Li H, Wang L, Zhang Y. Improvement of salivary EGF levels and serum inflammatory factors in patients with recurrent oral ulcers treated with shuanghuanglian oral solution. Altern Ther Health Med. 2024;30(12):188–193.
41. Nam K, Dos Santos HT, Maslow F, et al. Fibrin hydrogels fortified with FGF-7/10 and laminin-1 peptides promote regeneration of irradiated salivary glands. Acta Biomater. 2023;172:147–158. doi:10.1016/j.actbio.2023.10.013
42. Radović K, Brković B, Roganović J, et al. Salivary VEGF and post-extraction wound healing in type 2 diabetic immediate denture wearers. Acta Odontol Scand. 2022;80(1):9–14. doi:10.1080/00016357.2021.1930149
43. Nagakubo D, Kaibori Y. Oral microbiota: the influences and interactions of saliva, iga, and dietary factors in health and disease. Microorganisms. 2023;11(9):2307. doi:10.3390/microorganisms11092307
44. Ambudkar IS. Calcium signalling in salivary gland physiology and dysfunction. J Physiol. 2016;594(11):2813–2824. doi:10.1113/JP271143
45. Culp DJ, Zhang Z, Evans RL. VIP and muscarinic synergistic mucin secretion by salivary mucous cells is mediated by enhanced PKC activity via VIP-induced release of an intracellular Ca(2+) pool. Pflugers Arch. 2020;472(3):385–403. doi:10.1007/s00424-020-02348-7
46. Staruch M, Speth MM, Neyer P, et al. Radiation-associated changes in saliva composition of head and neck cancer patients: a systematic review. Radiother Oncol. 2024;196:110279. doi:10.1016/j.radonc.2024.110279
47. Ambudkar I. Calcium signaling defects underlying salivary gland dysfunction. Biochim Biophys Acta mol Cell Res. 2018;1865(11 Pt B):1771–1777. doi:10.1016/j.bbamcr.2018.07.002
48. Jang SI, Tandon M, Teos L, et al. Dual function of miR-1248 links interferon induction and calcium signaling defects in Sjögren’s syndrome. EBioMedicine. 2019;48:526–538. doi:10.1016/j.ebiom.2019.09.010
49. Xiao F, Du W, Zhu X, et al. IL-17 drives salivary gland dysfunction via inhibiting TRPC1-mediated calcium movement in Sjögren’s syndrome. Clin Transl Immunol. 2021;10(4):e1277. doi:10.1002/cti2.1277
50. Da conceicao V N, Sun Y, Chai X, et al. Metformin-induced activation of Ca(2+) signaling prevents immune infiltration/pathology in Sjogren’s syndrome-prone mouse models. J Transl Autoimmun. 2023;7:100210. doi:10.1016/j.jtauto.2023.100210
51. Manoharan RR, Prasad A, Pospíšil P, et al. ROS signaling in innate immunity via oxidative protein modifications. Front Immunol. 2024;15:1359600. doi:10.3389/fimmu.2024.1359600
52. Yang X, Liu X, Nie Y, et al. Oxidative stress and ROS-mediated cellular events in RSV infection: potential protective roles of antioxidants. Virol J. 2023;20(1):224. doi:10.1186/s12985-023-02194-w
53. Nguyen TL, Phan NM, Kim J. Administration of ROS-scavenging cerium oxide nanoparticles simply mixed with autoantigenic peptides induce antigen-specific immune tolerance against autoimmune encephalomyelitis. ACS Appl Mater Interfaces. 2024;16(26):33106–33120. doi:10.1021/acsami.4c05428
54. Mathan SV, Singh R, Kim SH, et al. Diallyl trisulfide induces ROS-mediated mitotic arrest and apoptosis and inhibits HNSCC tumor growth and cancer stemness. Cancers. 2024;16(2):378. doi:10.3390/cancers16020378
55. Zampieri M, Karpach K, Salerno G, et al. PAR level mediates the link between ROS and inflammatory response in patients with type 2 diabetes mellitus. Redox Biol. 2024;75:103243. doi:10.1016/j.redox.2024.103243
56. Morris G, Gevezova M, Sarafian V, et al. Redox regulation of the immune response. Cell mol Immunol. 2022;19(10):1079–1101. doi:10.1038/s41423-022-00902-0
57. Zhou J, Pathak JL, Wu L, et al. Downregulated GPX4 in salivary gland epithelial cells contributes to salivary secretion dysfunction in Sjogren’s syndrome via lipid ROS/pSTAT4/AQP5 axis. Free Radic Biol Med. 2024;218:1–15. doi:10.1016/j.freeradbiomed.2024.04.003
58. Zhu W, Wang Y, Guan Y, et al. Rapamycin can alleviate the submandibular gland pathology of Sjögren’s syndrome by limiting the activation of cGAS-STING signaling pathway. Inflammopharmacology. 2024;32(2):1113–1131. doi:10.1007/s10787-023-01393-9
59. Subudhi I, Konieczny P, Prystupa A, et al. Metabolic coordination between skin epithelium and type 17 immunity sustains chronic skin inflammation. Immunity. 2024;57(7):1665–1680.e1667. doi:10.1016/j.immuni.2024.04.022
60. Katsiougiannis S, Tenta R, Skopouli FN. Autoimmune epithelitis (Sjögren’s syndrome); the impact of metabolic status of glandular epithelial cells on auto-immunogenicity. J Autoimmun. 2019;104:102335. doi:10.1016/j.jaut.2019.102335
61. Li N, Li Y, Hu J, et al. A link between mitochondrial dysfunction and the immune microenvironment of salivary glands in primary Sjogren’s syndrome. Front Immunol. 2022;13:845209. doi:10.3389/fimmu.2022.845209
62. Luo D, Li L, Wu Y, et al. Mitochondria-related genes and metabolic profiles of innate and adaptive immune cells in primary Sjögren’s syndrome. Front Immunol. 2023;14:1156774. doi:10.3389/fimmu.2023.1156774
63. Hirose M, Sekar P, Eladham MWA, et al. Interaction between mitochondria and microbiota modulating cellular metabolism in inflammatory bowel disease. J Mol Med. 2023;101(12):1513–1526. doi:10.1007/s00109-023-02381-w
64. Germic N, Frangez Z, Yousefi S, et al. Regulation of the innate immune system by autophagy: neutrophils, eosinophils, mast cells, NK cells. Cell Death Differ. 2019;26(4):703–714. doi:10.1038/s41418-019-0295-8
65. Voynova E, Lefebvre F, Qadri A, et al. Correction of autophagy impairment inhibits pathology in the NOD.H-2h4 mouse model of primary Sjögren’s syndrome. J Autoimmun. 2020;108:102418. doi:10.1016/j.jaut.2020.102418
66. Colafrancesco S, Vomero M, Iannizzotto V, et al. Autophagy occurs in lymphocytes infiltrating Sjögren’s syndrome minor salivary glands and correlates with histological severity of salivary gland lesions. Arthritis Res Ther. 2020;22(1):238. doi:10.1186/s13075-020-02317-6
67. Cheng L, Li H, Zhan H, et al. Alterations of m6A RNA methylation regulators contribute to autophagy and immune infiltration in primary Sjögren’s syndrome. Front Immunol. 2022;13:949206. doi:10.3389/fimmu.2022.949206
68. Colafrancesco S, Barbati C, Priori R, et al. Maladaptive autophagy in the pathogenesis of autoimmune epithelitis in Sjögren’s syndrome. Arthritis Rheumatol. 2022;74(4):654–664. doi:10.1002/art.42018
69. Zhang H, Zheng C, Xu Y, et al. Comprehensive molecular and cellular characterization of endoplasmic reticulum stress-related key genes in renal ischemia/reperfusion injury. Front Immunol. 2024;15:1340997. doi:10.3389/fimmu.2024.1340997
70. Torrens JN, Hetzer SM, Evanson NK. Brief oxygen exposure after traumatic brain injury hastens recovery and promotes adaptive chronic endoplasmic reticulum stress responses. Int J mol Sci. 2023;24(12):9831. doi:10.3390/ijms24129831
71. Hu T, Wang J, Li W, et al. Endoplasmic reticulum stress in hepatitis B virus and hepatitis C virus infection. Viruses. 2022;14(12):2630. doi:10.3390/v14122630
72. Li M, Liu G, Yuan LX, et al. Triphenyl phosphate (TPP) promotes hepatocyte toxicity via induction of endoplasmic reticulum stress and inhibition of autophagy flux. Sci Total Environ. 2022;840:156461. doi:10.1016/j.scitotenv.2022.156461
73. Groenendyk J, Michalak M. Interplay between calcium and endoplasmic reticulum stress. Cell Calcium. 2023;113:102753. doi:10.1016/j.ceca.2023.102753
74. Li HY, Huang LF, Huang XR, et al. Endoplasmic reticulum stress in systemic lupus erythematosus and lupus nephritis: potential therapeutic target. J Immunol Res. 2023;2023:7625817. doi:10.1155/2023/7625817
75. Castro I, Albornoz N, Aguilera S, et al. Aberrant MUC1 accumulation in salivary glands of Sjögren’s syndrome patients is reversed by TUDCA in vitro. Rheumatology. 2020;59(4):742–753. doi:10.1093/rheumatology/kez316
76. Huang L, Liu Q, Zhou T, et al. Deficiency of β-arrestin2 alleviates apoptosis through GRP78-ATF6-CHOP signaling pathway in primary Sjögren’s syndrome. Int Immunopharmacol. 2021;101(Pt A):108281. doi:10.1016/j.intimp.2021.108281
77. Lopes AP, Hillen MR, Hinrichs AC, et al. Deciphering the role of cDC2s in Sjögren’s syndrome: transcriptomic profile links altered antigen processes with IFN signature and autoimmunity. Ann Rheum Dis. 2023;82(3):374–383. doi:10.1136/ard-2022-222728
78. Cheng D, Zhou T, Liu H, et al. MANF inhibits Sjögren’s syndrome salivary gland epithelial cell apoptosis and antigen expression of Ro52/SSA through endoplasmic reticulum stress/autophagy pathway. Int Immunopharmacol. 2023;122:110582. doi:10.1016/j.intimp.2023.110582
79. Barrera MJ, Aguilera S, Castro I, et al. Dysfunctional mitochondria as critical players in the inflammation of autoimmune diseases: potential role in Sjögren’s syndrome. Autoimmun Rev. 2021;20(8):102867. doi:10.1016/j.autrev.2021.102867
80. Woo Y, Ma M, Okawa M, et al. Hepatocyte intrinsic innate antiviral immunity against hepatitis delta virus infection: the voices of bona fide human hepatocytes. Viruses. 2024;16(5):740. doi:10.3390/v16050740
81. Zindel J, Kubes P. DAMPs, PAMPs, and LAMPs in immunity and sterile inflammation. Annu Rev Pathol. 2020;15:493–518. doi:10.1146/annurev-pathmechdis-012419-032847
82. Fang X, Wang Y, Zhang Y, et al. NLRP3 inflammasome and its critical role in gynecological disorders and obstetrical complications. Front Immunol. 2020;11:555826. doi:10.3389/fimmu.2020.555826
83. Ren W, Sun Y, Zhao L, et al. NLRP3 inflammasome and its role in autoimmune diseases: a promising therapeutic target. Biomed Pharmacother. 2024;175:116679. doi:10.1016/j.biopha.2024.116679
84. Baldini C, Santini E, Rossi C, et al. The P2X7 receptor-NLRP3 inflammasome complex predicts the development of non-Hodgkin’s lymphoma in Sjogren’s syndrome: a prospective, observational, single-centre study. J Intern Med. 2017;282(2):175–186. doi:10.1111/joim.12631
85. Hong SM, Lee J, Jang SG, et al. Type I interferon increases inflammasomes associated pyroptosis in the salivary glands of patients with primary Sjögren’s syndrome. Immune Netw. 2020;20(5):e39. doi:10.4110/in.2020.20.e39
86. Zou Y, Huang Y, Liu S, et al. Periodontopathic microbiota and atherosclerosis: roles of TLR-mediated inflammation response. Oxid Med Cell Longev. 2022;2022:9611362. doi:10.1155/2022/9611362
87. Fore F, Budipranama M, Destiawan RA. TLR10 and Its Role in Immunity. Handb Exp Pharmacol. 2022;276:161–174.
88. Sisto M, Lorusso L, Lisi S. TLR2 signals via NF-κB to drive IL-15 production in salivary gland epithelial cells derived from patients with primary Sjögren’s syndrome. Clin Exp Med. 2017;17(3):341–350. doi:10.1007/s10238-016-0429-y
89. Nakamura H, Horai Y, Suzuki T, et al. TLR3-mediated apoptosis and activation of phosphorylated Akt in the salivary gland epithelial cells of primary Sjögren’s syndrome patients. Rheumatol Int. 2013;33(2):441–450. doi:10.1007/s00296-012-2381-9
90. Zhang S, Qu J, Wang L, et al. Activation of toll-like receptor 7 signaling pathway in primary Sjögren’s syndrome-associated thrombocytopenia. Front Immunol. 2021;12:637659. doi:10.3389/fimmu.2021.637659
91. Nishihata SY, Shimizu T, Umeda M, et al. The toll-like receptor 7-mediated Ro52 Antigen-presenting pathway in the salivary gland epithelial cells of Sjögren’s syndrome. J Clin Med. 2023;12(13):4423. doi:10.3390/jcm12134423
92. Nakamura H, Tanaka T, Zheng C, et al. amplified type i interferon response in Sjögren’s disease via ectopic toll-like receptor 7 expression in salivary gland epithelial cells induced by lysosome-associated membrane protein 3. Arthritis Rheumatol. 2024;76(7):1109–1119. doi:10.1002/art.42844
93. Barrera MJ, Aguilera S, Veerman E, et al. Salivary mucins induce a toll-like receptor 4-mediated pro-inflammatory response in human submandibular salivary cells: are mucins involved in Sjögren’s syndrome? Rheumatology. 2015;54(8):1518–1527. doi:10.1093/rheumatology/kev026
94. Wang MM, Zhao Y, Liu J, et al. The role of the cGAS-STING signaling pathway in viral infections, inflammatory and autoimmune diseases. Acta Pharmacol Sin. 2024;45(10):1997–2010. doi:10.1038/s41401-023-01185-5
95. Papinska J, Bagavant H, Gmyrek GB, et al. Pulmonary involvement in a mouse model of Sjögren’s syndrome induced by STING activation. Int J mol Sci. 2020;21(12):4512. doi:10.3390/ijms21124512
96. Xu J, Chen C, Yin J, et al. Lactate-induced mtDNA accumulation activates cGAS-STING signaling and the inflammatory response in Sjögren’s syndrome. Int J Med Sci. 2023;20(10):1256–1271. doi:10.7150/ijms.83801
97. Yang M, Delcroix V, Lennikov A, et al. Genomic DNA activates the AIM2 inflammasome and STING pathways to induce inflammation in lacrimal gland myoepithelial cells. Ocul Surf. 2023;30:263–275. doi:10.1016/j.jtos.2023.09.012
98. Liang T, Gu L, Kang X, et al. Programmed cell death disrupts inflammatory tumor microenvironment (TME) and promotes glioblastoma evolution. Cell Commun Signal. 2024;22(1):333. doi:10.1186/s12964-024-01602-0
99. Kyriakidis NC, Kapsogeorgou EK, Tzioufas AG. A comprehensive review of autoantibodies in primary Sjögren’s syndrome: clinical phenotypes and regulatory mechanisms. J Autoimmun. 2014;51:67–74. doi:10.1016/j.jaut.2013.11.001
100. Zhu X, Sha X, Zang Y, et al. Current progress of ferroptosis study in hepatocellular carcinoma. Int J Biol Sci. 2024;20(9):3621–3637. doi:10.7150/ijbs.96014
101. Peng X, Hou L, Wu X, et al. The plasma exosomes from patients with primary Sjögren’s syndrome contain epithelial cell-derived proteins involved in ferroptosis. J Mol Med. 2023;101(10):1289–1304. doi:10.1007/s00109-023-02361-0
102. Kapsogeorgou EK, Tzioufas AG. Interaction of human salivary gland epithelial cells with B lymphocytes: implications in the pathogenesis of Sjögren’s syndrome. Mediterr J Rheumatol. 2020;31(4):424–426. doi:10.31138/mjr.31.4.424
103. Rivière E, Pascaud J, Tchitchek N, et al. Salivary gland epithelial cells from patients with Sjögren’s syndrome induce B-lymphocyte survival and activation. Ann Rheum Dis. 2020;79(11):1468–1477. doi:10.1136/annrheumdis-2019-216588
104. Danieli MG, Casciaro M, Paladini A, et al. Exposome: epigenetics and autoimmune diseases. Autoimmun Rev. 2024;23(6):103584. doi:10.1016/j.autrev.2024.103584
105. Wang Y, Riaz F, Wang W, et al. Functional significance of DNA methylation: epigenetic insights into Sjögren’s syndrome. Front Immunol. 2024;15:1289492. doi:10.3389/fimmu.2024.1289492
106. Switzer CH. Non-canonical nitric oxide signalling and DNA methylation: inflammation induced epigenetic alterations and potential drug targets. Br J Pharmacol. 2023.
107. Miceli-Richard C, Wang-Renault SF, Boudaoud S, et al. Overlap between differentially methylated DNA regions in blood B lymphocytes and genetic at-risk loci in primary Sjögren’s syndrome. Ann Rheum Dis. 2016;75(5):933–940. doi:10.1136/annrheumdis-2014-206998
108. Kabeerdoss J, Devarajalu P, Sandhya P. DNA methylation profiling of labial salivary gland tissues revealed hypomethylation of B-cell-related genes in primary Sjögren’s syndrome. Immunol Res. 2024;72(3):450–459. doi:10.1007/s12026-024-09453-0
109. Chi C, Solomon O, Shiboski C, et al. Identification of Sjögren’s syndrome patient subgroups by clustering of labial salivary gland DNA methylation profiles. PLoS One. 2023;18(3):e0281891. doi:10.1371/journal.pone.0281891
110. Sepúlveda D, Barrera MJ, Castro I, et al. Impaired IRE1α/XBP-1 pathway associated to DNA methylation might contribute to salivary gland dysfunction in Sjögren’s syndrome patients. Rheumatology. 2018;57(6):1021–1032. doi:10.1093/rheumatology/key021
111. Guimarães JR, Coêlho MC, de Oliveira NFP. Contribution of DNA methylation to the pathogenesis of Sjögren’s syndrome: a review. Autoimmunity. 2022;55(4):215–222. doi:10.1080/08916934.2022.2062593
112. Ma J, Wang X, Yang X, et al. Increased METTL3 expression and m(6)A RNA methylation may contribute to the development of dry eye in primary Sjögren’s syndrome. BMC Ophthalmol. 2023;23(1):252. doi:10.1186/s12886-023-02988-0
113. Truffinet F, Arco-Hierves A, Shalabi H, et al. m(6)A RNA methylation controls salivary gland epithelial cell function and has a protective role in Sjögren’s disease. Ann Rheum Dis. 2024;83:41–42. doi:10.1136/annrheumdis-2024-eular.2172
114. Li J, Xie K, Xu M, et al. Significance of N6-methyladenosine RNA methylation regulators in diagnosis and subtype classification of primary Sjögren’s syndrome. Heliyon. 2024;10(3):e24860. doi:10.1016/j.heliyon.2024.e24860
115. Li X, Yu T, Li X, et al. Role of novel protein acylation modifications in immunity and its related diseases. Immunology. 2024;173(1):53–75. doi:10.1111/imm.13822
116. Xia J, Jiang S, Dong S, et al. The role of post-translational modifications in regulation of NLRP3 inflammasome activation. Int J mol Sci. 2023;24(7):6126. doi:10.3390/ijms24076126
117. Liao CC, Chou PL, Cheng CW, et al. Comparative analysis of novel autoantibody isotypes against citrullinated-inter-alpha-trypsin inhibitor heavy chain 3 (ITIH3)(542-556) peptide in serum from Taiwanese females with rheumatoid arthritis, primary Sjögren’s syndrome and secondary Sjögren’s syndrome in rheumatoid arthritis. J Proteomics. 2016;141:1–11. doi:10.1016/j.jprot.2016.03.031
118. Liao Z, Li D, Liao S, et al. Proteomics profiling and lysine malonylation analysis in primary Sjogren’s syndrome. J Proteomics. 2023;287:104977. doi:10.1016/j.jprot.2023.104977
119. Shen Y, Yu X, Wang Q, et al. Association between primary Sjögren’s syndrome and gut microbiota disruption: a systematic review and meta-analysis. Clin Rheumatol. 2024;43(2):603–619. doi:10.1007/s10067-023-06754-x
120. Deng C, Xiao Q, Fei Y. A glimpse into the microbiome of Sjögren’s syndrome. Front Immunol. 2022;13:918619. doi:10.3389/fimmu.2022.918619
121. Manoussakis MN, Svolaki IP, Hazapis O, et al. Postgenomic bioinformatic analysis of nucleic acid sequences expressed in the salivary gland epithelial cells of primary Sjögren’s syndrome patients in search of microorganisms and endogenous retroviruses. Mediterr J Rheumatol. 2022;33(3):371–374. doi:10.31138/mjr.33.3.371
122. Baldini C, Fulvio G, La Rocca G, et al. Update on the pathophysiology and treatment of primary Sjögren syndrome. Nat Rev Rheumatol. 2024;20(8):473–491. doi:10.1038/s41584-024-01135-3
123. Choudhry HS, Hosseini S, Choudhry HS, et al. Updates in diagnostics, treatments, and correlations between oral and ocular manifestations of Sjogren’s syndrome. Ocul Surf. 2022;26:75–87. doi:10.1016/j.jtos.2022.08.001
124. Wang X, Pang K, Wang J, et al. Microbiota dysbiosis in primary Sjögren’s syndrome and the ameliorative effect of hydroxychloroquine. Cell Rep. 2022;40(11):111352. doi:10.1016/j.celrep.2022.111352
125. Bustos-Lobato L, Rus MJ, Saúco C, et al. Oral microbial biomap in the drought environment: sjogren’s syndrome. mol Oral Microbiol. 2023;38(5):400–407. doi:10.1111/omi.12435
126. Kim D, Jeong YJ, Lee Y, et al. Correlation between salivary microbiome of parotid glands and clinical features in primary Sjögren’s syndrome and non-Sjögren’s sicca subjects. Front Immunol. 2022;13:874285. doi:10.3389/fimmu.2022.874285
127. Xing H, Liu H, Pan J. High-throughput sequencing of oral microbiota in candida carriage Sjögren’s syndrome patients: a pilot cross-sectional study. J Clin Med. 2023;12(4):1559. doi:10.3390/jcm12041559
128. Bahabayi A, Alimu X, Wang G, et al. VNN2-expressing circulating monocytes exhibit unique functional characteristics and are decreased in patients with primary Sjögren’s syndrome. J Autoimmun. 2024;147:103275. doi:10.1016/j.jaut.2024.103275
129. Zhou J, Pathak JL, Cao T, et al. CD4 T cell-secreted IFN-γ in Sjögren’s syndrome induces salivary gland epithelial cell ferroptosis. Biochim Biophys Acta Mol Basis Dis. 2024;1870(4):167121. doi:10.1016/j.bbadis.2024.167121
130. Longo SK, Guo MG, Ji AL, et al. Integrating single-cell and spatial transcriptomics to elucidate intercellular tissue dynamics. Nat Rev Genet. 2021;22(10):627–644. doi:10.1038/s41576-021-00370-8
131. Qi J, Sun H, Zhang Y, et al. Single-cell and spatial analysis reveal interaction of FAP(+) fibroblasts and SPP1(+) macrophages in colorectal cancer. Nat Commun. 2022;13(1):1742. doi:10.1038/s41467-022-29366-6
132. Felix FA, Zhou J, Li D, et al. Endogenous IL-22 contributes to the pathogenesis of salivary gland dysfunction in the non-obese diabetic model of Sjögren’s syndrome. Mol Immunol. 2024;173:20–29. doi:10.1016/j.molimm.2024.06.010
133. Gupta S, Yamada E, Nakamura H, et al. Inhibition of JAK-STAT pathway corrects salivary gland inflammation and interferon driven immune activation in Sjögren’s disease. Ann Rheum Dis. 2024;83(8):1034–1047. doi:10.1136/ard-2023-224842
134. Shimizu T, Nakamura H, Kawakami A. Role of the innate immunity signaling pathway in the pathogenesis of Sjögren’s syndrome. Int J mol Sci. 2021;22(6):3090. doi:10.3390/ijms22063090
135. Rivière E, Pascaud J, Virone A, et al. Interleukin-7/interferon axis drives t cell and salivary gland epithelial cell interactions in Sjögren’s syndrome. Arthritis Rheumatol. 2021;73(4):631–640. doi:10.1002/art.41558
© 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The
full terms of this license are available at https://www.dovepress.com/terms.php
and incorporate the Creative Commons Attribution
- Non Commercial (unported, 3.0) License.
By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted
without any further permission from Dove Medical Press Limited, provided the work is properly
attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.