Your activity: 52 p.v.
your limit has been reached. plz Donate us to allow your ip full access, Email: sshnevis@outlook.com

Pathogenesis of Sjögren's syndrome

Pathogenesis of Sjögren's syndrome
Authors:
Alan N Baer, MD, MACR
Umesh S Deshmukh, PhD
Section Editor:
Robert Fox, MD, PhD
Deputy Editor:
Philip Seo, MD, MHS
Literature review current through: Dec 2022. | This topic last updated: Aug 31, 2021.

INTRODUCTION — Sjögren's syndrome (SS) is a chronic, multisystem inflammatory disorder characterized by diminished lacrimal and salivary gland function, which results in the unique combination of dry eyes and dry mouth. Additional disease manifestations may also be present, including dryness of the skin and other mucosal surfaces. Systemic extraglandular features include arthritis, nephritis, cytopenia, pneumonitis, and vasculitis. Neurologic manifestations include peripheral neuropathy, myelopathy, and cognitive disturbances. There is an increased risk of lymphoma in comparison with other autoimmune disorders. (See "Clinical manifestations of Sjögren's syndrome: Exocrine gland disease" and "Clinical manifestations of Sjögren's syndrome: Extraglandular disease".)

The pathogenesis of SS is thought to be a multistep process, triggered by an environmental factor, most likely viral, in a genetically predisposed individual. An immune response involving innate and adaptive immunity, leading to autoimmunity and chronic inflammation, are central components of the disease process.

The pathogenesis of SS is reviewed here. The clinical manifestations, diagnosis, treatment, and prognosis of this disorder are discussed separately. (See "Clinical manifestations of Sjögren's syndrome: Exocrine gland disease" and "Clinical manifestations of Sjögren's syndrome: Extraglandular disease" and "Diagnosis and classification of Sjögren's syndrome" and "Overview of the management and prognosis of Sjögren's syndrome" and "Treatment of dry eye in Sjögren's syndrome: General principles and initial therapy" and "Treatment of dry mouth and other non-ocular sicca symptoms in Sjögren's syndrome".)

OVERVIEW — The pathogenesis of SS is typically modeled as a multistep process, triggered by an environmental factor, most likely viral, in a genetically predisposed individual. The initial events engage the innate immune system, but propagation and perpetuation of the autoimmune process require a continual interplay between the innate and adaptive immune systems [1].

The result is autoreactive B-cell stimulation, autoantibody production, and chronic inflammation of the salivary and lacrimal glands and often other tissues. Extraglandular manifestations may arise from autoimmune exocrinopathy, akin to that in the salivary glands (eg, interstitial nephritis, biliary cholangitis), immune-complex deposition (eg, cryoglobulinemic vasculitis), and extranodal lymphoproliferation (eg, lymphocytic interstitial pneumonitis). Chronic stimulation of B cells in the target tissue may promote lymphomagenesis, again through a multistep process in a genetically susceptible individual.

RISK FACTORS AND ETIOPATHOGENESIS — Interactions between both genetic and nongenetic factors are involved in disease susceptibility and in the initiation as well as the progression of the disease process.

Genetic factors — Many different regions of the genome, both within and outside of the major histocompatibility complex (MHC), confer susceptibility to Sjögren's syndrome (SS) but differ between populations and studies [2,3]. (See 'HLA genes' below and 'Non-HLA genes' below.)

A familial tendency to develop SS has been well-documented, along with an increased risk of a variety of autoimmune disorders in relatives of patients with SS [4]. A concordance rate for SS in monozygotic twins has not been reported; however, it is estimated to be approximately 20 percent based upon studies of other autoimmune diseases that overlap with SS, including systemic lupus erythematosus (SLE) and rheumatoid arthritis [5,6]. Thus, a substantial role for epigenetic factors and the environment is likely in SS pathogenesis.

Multiple polymorphisms have been identified by genome-wide association studies (GWAS). These have involved cohorts of SS patients of European [2] and Han Chinese [3] descent, and another consisting of Sjögren's International Collaborative Clinical Alliance (SICCA) registrants, primarily of either European or Asian (including Chinese and Japanese) descent [7,8]. Many of the implicated genes are associated with innate or adaptive immune responses. Surprisingly, the majority of the identified single nucleotide polymorphism (SNPs) were located in the non-protein coding regions, which is suggestive of regulatory activity of these regions on gene expression.

HLA genes — SS shows the most robust genetic association within MHC genes, including those in the human leukocyte antigen (HLA)-DR region. While 20 to 25 percent of the general White European and American population shows the extended haplotype of HLA-DR3, B8, DQ2, and C4 null allele, this haplotype is present in approximately 50 percent of White Northern European patients with SS [4]. Considerable heterogeneity of this association is observed across different ethnic groups [7]. Examples include:

A markedly increased frequency of HLA-DQB1*0201 and HLA-DQA1*0501 in White American patients with primary SS [9]

DRB1*0803-DQA1*0103-DQB1*0601 in Han Chinese, with multiple different associations in one GWAS [3]

HLA-DR5 in Greek patients [10]

DRB1*0405-DRB4*0101-DQA1*0301-DQB1*0401 in Japanese patients [9]

DRB1*1501-*0301 genotype in White French patients [11]

DQA1*05:01-DQB1*02:01 in Colombian patients [12]

HLA-B15 and DRB1*03 in Tunisian populations [13]

Non-HLA genes — Genes other than those within the HLA loci are also associated with an increased risk of disease [1]. The strongest associations on GWAS include IRF5 and TNIP1, which are both involved in innate immunity, and BLK, STAT4, IL12A, and CXCR5, which are involved in adaptive immunity (table 1). In patients with SS of Han Chinese descent, GTF1, a gene involved in regulating immunoglobulin heavy chain transcription is also a risk factor for SS [14]. Various other genes have been identified in such studies, but their degrees of association do not reach genome-wide significance; these include some involved in innate immunity, such as MBL2, FCGR2B, LTA [15], TNF, NCR3, and NFKBIA, and others that have a role in the adaptive immune response, including TAP2, EBF1, PTPN22, TNFRSF4, and IL10.

The information on different gene disease associations is curated by DisGeNET [16] and freely accessible at www.disgenet.org/home.

Epigenetic factors — Epigenetic factors, such as DNA methylation, histone acetylation, noncoding RNA transcripts, and gene recombination, may all play a role in the modulation of gene expression. DNA methylation has been analyzed in the peripheral blood and target tissue of patients with SS [17-20]. These studies have supported a role for methylation in the regulation of genes in SS. Interestingly, many of the epigenetically modified regions have been previously identified as genetic risk factors for SS [21-23]. Additional gene regulation in SS occurs through the modulation of gene expression by microRNAs (miRNAs). Differential expression of miRNAs has been reported in the lymphocytes and salivary glands of SS patients and controls, and these often target genes relevant to disease pathogenesis [23-25]. In summary, these studies demonstrate that complex interactions between genetic and epigenetic factors influence the development of SS.

Sex — The striking predominance of SS in women (15 to 20:1, female to male ratio) points to a role for sex hormones in the development of the disease, as it does for other systemic autoimmune diseases, such as SLE, rheumatoid arthritis, and systemic sclerosis. The clinical onset of SS in women is most often in the sixth decade of life, just after the onset of menopause. This is in sharp contrast to SLE, where the disease occurs most often in women during their reproductive-age years.

The characteristic autoantibodies of SS are detectable for up to 18 years before clinical disease onset [26], so the autoimmune process may be initiated in a woman during her reproductive-age years but not expressed clinically with sicca or other symptoms until estrogen levels drop sharply with the onset of menopause. In animal models, estrogen protects against lacrimal and salivary glandular inflammation while its withdrawal promotes apoptosis of salivary gland epithelial cells [27]. Women with SS have lower cumulative estrogen exposure, relative to non-SS women with sicca, when measured by an integration of age at menarche, age at menopause, parity, hysterectomy, and female hormone use [28]. Low serum concentrations of dehydroepiandrosterone and dihydrotestosterone have also been demonstrated [29], arguing for a protective role of androgens, similar to that of estrogen, in the pathogenesis of SS.

Alternatively, the sex bias of SS may be mediated independently of a sex hormone effect. Women with trisomy X (47, XXX) have normal sexual development and sex hormone levels, but have a risk of SLE and SS that is 2.5- and 2.9-fold higher, respectively, than in women with 46, XX, and 25- and 41-fold higher respectively than in men with 46, XY [30]. Thus, there is a sex chromosome dose effect in the predisposition of females to autoimmune rheumatic disease, which is independent of differences in sex hormone levels.

The X chromosome has the highest density of immunity-related genes, and transcriptional silencing of an entire X chromosome (X-chromosome inactivation) in each female cell, initiated during embryogenesis, serves to equalize the expression of X-linked genes between sexes. This inactivation process is mediated by allele-specific expression of the long noncoding RNA, termed XIST. Biallelic expression of X-linked immunity genes has been observed in the lymphocytes of female mammals, suggesting incomplete X-chromosome inactivation and providing an alternative mechanism for the sex bias in autoimmune diseases, including SS [31].

Although the frequency of SS development in males is significantly lower, some studies have suggested that male SS patients have increased risk of lymphoma development [32] and extraglandular manifestation [33].

The potential role of viral infection — The first signs of SS typically occur long before diagnosis, thereby impeding study of its etiology. Many observations suggest a role for viruses in the pathogenesis of SS, but no single virus has been implicated. Evidence of ongoing or past viral infection can be detected in many patients, but no virus has been found at high levels in target tissues [34]. Observations supporting a viral etiology include:

Certain viruses, particularly Epstein-Barr virus (EBV), the ubiquitous herpes-type virus that causes infectious mononucleosis, frequently infect the salivary glands. EBV is spread to noninfected individuals via the saliva; primary EBV infections progress to lifelong latent infection with periodic reactivation, and the site of latency for EBV is in the salivary gland. EBV can induce strong immune responses by T cells and activate B-cell production of autoantibodies. (See "Virology of Epstein-Barr virus" and "Clinical manifestations and treatment of Epstein-Barr virus infection" and "Infectious mononucleosis".)

EBV can be identified in the ectopic lymphoid structures present in the salivary glands of some SS patients (see 'The lymphocytic infiltrate and glandular pathology' below), but not in SS glandular tissue lacking such structures. EBV-infected plasma cells within these structures produced antibodies to anti-Ro52 and anti-La/SSB [35]. In addition, SS patients show a higher prevalence and titer of antibodies against EBV antigens [36]. These findings support a role for active EBV in supporting the local proliferation and differentiation of autoreactive B cells.

At least three viruses (human T-lymphotropic virus [HTLV] type I, human immunodeficiency virus [HIV], and hepatitis C virus [HCV]) are associated with clinical syndromes that share many features of SS [37,38].

Hepatitis delta virus (HDV) was detected in salivary glands of 50 percent of primary SS patients and induces a primary SS-like disease in mice [39].

Retroelements are noncoding DNAs that constitute approximately half of the human genome and regulate gene expression. SS patients show increased levels of retroviral long interspersed nuclear element 1 [40]. These retroviral elements can activate innate immunity and induce excessive production of type I interferons (IFNs). Animal models have demonstrated a role for type I IFNs in SS pathogenesis [41].

MECHANISMS OF IMMUNE-MEDIATED INJURY — Glandular dysfunction in Sjögren's syndrome (SS) is generally presumed to result from autoimmune-induced inflammation and resultant damage and destruction of the tissue responsible for tear and saliva production. However, this assumption is not fully supported by two observations. First, there is a weak correlation between glandular dysfunction and the degree of glandular inflammation [42]. Second, glandular dysfunction can be induced in animal models of SS before the advent of inflammatory infiltrates [43]. In light of these observations, other mechanisms may contribute to the glandular dysfunction, including antibodies to the muscarinic receptor that may impair neural innervation of the gland (see 'Anti-muscarinic acetylcholine receptor antibodies' below) and direct effects of cytokines on neurotransmitter release or other secretory cell functions [44].

Glandular inflammation — Overexpression of interferon (IFN)-inducible genes in salivary glands and peripheral blood monocytes of SS patients (termed the IFN signature) highlights the importance of the innate immune system in pathogenesis [45,46]. Damage to the salivary gland epithelial cells (eg, by an exogenous or endogenous viral trigger) is thought to induce apoptosis and migration of the SSA (also termed SS-A or Ro60) antigen from the nucleus in a complex with human small noncoding Y RNA (hYRNA) to a bleb on the cell surface [47]. The SSA molecule thus escapes normal apoptotic degradation. The presence of antibody to the SSA-hYRNA complex promotes the uptake of this immune complex by local dendritic cells and B cells, access to intracytoplasmic toll-like receptors (TLRs), and stimulation of the IFN signatures characteristic of SS [48-50].

Plasmacytoid dendritic cells may also be activated directly by viral or other environmental factors. Activation of the type I IFN system by the innate immune system promotes adaptive immune responses through T- and B-cell activation and induction of cytokine production.

A cycle of mutual stimulation of the innate and acquired immune systems leads to the perpetuation of glandular injury and dysfunction. Tissue injury occurs through the activation of these immune pathways by lymphocytes within the glandular tissues or extraglandular sites, leading to the release of cytokines, including IFN-gamma, interleukin (IL) 17, B-cell activating factor (BAFF; also known as B lymphocyte stimulator [BLyS]), and others, and the production of characteristic autoantibodies. Apoptosis of glandular cells and dysfunction of residual epithelial cells and tissues occur due to cytokines and metalloproteinases that interfere with salivary gland organization and function. Furthermore, a premature senescence in salivary gland progenitor cells [51] and salivary gland stem cells [52] also contributes towards the chronicity of glandular dysfunction. (See 'Autoantibodies' below and 'The lymphocytic infiltrate and glandular pathology' below.)

Extraglandular manifestations — Extraglandular manifestations of SS can be classified based on their presumed pathogenesis, including autoimmune exocrinopathy, akin to that in the salivary glands (eg, interstitial nephritis, biliary cholangitis), immune-complex deposition (eg, cryoglobulinemic vasculitis), cell- or tissue-specific autoimmunity (eg, thrombocytopenia, ataxic sensory ganglionopathy, neuromyelitis optica), and lymphoproliferation (eg, lymphocytic interstitial pneumonitis) [53,54]. Fatigue and cognitive impairment most likely have multifactorial pathogeneses, but there is evidence for immune mechanisms, including antibodies directed against N-methyl-D-aspartate receptor subtype NR2 (anti-NR2) for the cognitive impairment and elevated heat shock protein 90a for the fatigue [55,56].

Autoantibodies — SS is characterized by the presence of autoantibodies, most notably anti-Ro/SSA and anti-La/SSB in 60 to 80 percent of those affected (see "The anti-Ro/SSA and anti-La/SSB antigen-antibody systems"). Antinuclear antibodies (ANA) are present in 90 percent of patients, and high-titer rheumatoid factor is also frequent. (See 'Ro/SSA and La/SSB' below and "Diagnosis and classification of Sjögren's syndrome" and "Clinical manifestations of Sjögren's syndrome: Extraglandular disease", section on 'Autoantibodies'.)

Autoantibodies can precede the clinical onset of SS by many years, as evidenced by the analysis of stored premorbid serum samples in SS patients [26]. Additionally, mothers of children with neonatal lupus have high-titer anti-Ro/SSA and anti-La/SSB antibodies and are often asymptomatic. However, these asymptomatic women are at an increased risk for SS. The fact that SS develops in only a minority of these individuals indicates that autoantibodies alone are insufficient for induction of disease [57].

Ro/SSA and La/SSB — Anti-Ro/SSA antibodies are predominantly of the immunoglobulin G1 (IgG1) subclass and recognize two distinct proteins, the 52kD, Ro52 protein and the 60kD, Ro60 protein, encoded by different genes [58]. Both antigens are located primarily in the nucleus but are also expressed in the cytoplasm and on the cell surface.

The SSA Ro52 autoantigen belongs to the large family of the tripartite motif (TRIM)-containing family of proteins [59]. Functionally, Ro52 is an E3 ubiquitin ligase, and it plays a critical role in the regulation of innate immunity, particularly the type I IFN response [60]. Ro52 also acts as an intracellular Fc receptor and has been shown to bind the Fc portion of IgG antibodies complexed with viruses [61]. Patients positive for anti-Ro52 often present with higher disease severity. However, in these patients, anti-Ro52 is also strongly associated with the presence of anti-Ro60 [62]. A pathogenic role of anti-Ro52 antibodies in the induction of salivary gland dysfunction has been demonstrated in experimental mouse model systems [63,64].

The 60kD Ro60 protein, also known as TROVE2, binds the small cytoplasmic RNA moieties termed hYRNA and is involved in the clearance of defective RNA transcripts [65]. The Ro60 protein, like Ro52, is also involved in the regulation of inflammatory gene expression by binding to endogenous Alu retroelements [66].

Anti-La/SSB antibodies are found in 50 percent of patients with SS. These antibodies recognize a 47 kD phosphoprotein associated with newly synthesized RNA polymerase III transcripts. The gene encoding SSB is unusual in that it has two promoter sites, encoding for two different size mRNAs, and raising the possibility of gene switching under disease conditions [67]. The pathogenic role of anti-La/SSB autoantibodies in SS is not clear.

Anti-muscarinic acetylcholine receptor antibodies — Antibodies to acetylcholine receptors of salivary glands might account for decreased secretion from histologically normal glands. It is uncertain whether such antibodies in SS are primary or secondary phenomena [68-70]. One report has shown a higher frequency of antibodies to the muscarinic (M) receptor for acetylcholine (M3R) in patients with SS compared with patients with systemic lupus erythematosus (SLE) (67 versus 2 percent) [71]. In another study of 15 patients with SS, antibodies were found in five of nine subjects with primary and six of six with secondary disease [72]. In animal models, these anti-M3 acetylcholine receptor antibodies decrease glandular secretion [69].

Some evidence suggests there may be binding of these autoantibodies to the extracellular domain of the human M3R, raising the possibility that SS may be among the autoimmune disorders such as Graves' disease or myasthenia gravis where antireceptor antibodies are pathogenic [73,74].

The lymphocytic infiltrate and glandular pathology — The primary pathologic lesion of SS is lymphocytic infiltration of the salivary and lacrimal glands. The infiltrates consist of focal aggregates of lymphocytes, beginning around the ducts and spreading to involve the entire lobule (image 1). The cellular composition of these infiltrates depends on their severity. T cells, primarily CD4+, predominate in milder infiltrates, which are smaller and respect the architecture of the gland. B cells become more predominant in larger and denser infiltrates associated with acinar destruction and loss of tissue architecture [75]. Migration of the lymphocytes to sites in the glands occurs due to a series of events, including a response to chemokines, adhesion to specific vascular adhesion molecules, and entry into the gland where they interact with dendritic cells and epithelial cells [76].

Salivary gland epithelial cells have the capacity to play an active role in the initiation and maintenance of glandular inflammation. Their potential pivotal role in SS immunopathogenesis has led to the concept of "autoimmune epithelitis" [77] as a unifying feature of the disease. Salivary gland epithelial cells, activated by type I IFN or a viral infection, can affect the following: (1) surface expression of major histocompatibility complex (MHC) class II molecules, including human leukocyte antigen (HLA)-DR, and costimulatory factors, including CD80, CD86, and CD40, empowering them to interact with T cells; (2) release of cytokines such as BAFF [78,79], interleukin (IL) 1, IL-6, tumor necrosis factor (TNF)-alpha, and IL-22 [80], which are crucial to both innate and adaptive immune responses; (3) promotion of lymphocytic and dendritic cell infiltration by production of CXCL13 and other chemokines [81]; and (4) mediation of the release of intracellular antigens (eg, Ro/SSA-La/SSB) through apoptosis [82] and the release of exosomes, thereby driving the generation of autoreactive B cells.

In approximately 30 to 40 percent of SS patients, the glandular periductal lymphoid aggregates develop a structure highly similar to typical secondary lymphoid organs, with B-cell follicles surrounded by T-cell rich areas, high endothelial venules, and networks of follicular dendritic cells [83,84]. These ectopic lymphoid structures promote antigen-driven selection of B-cell clones via affinity maturation and provide a conducive microenvironment for antibody production in the target tissue (picture 1) [85]. The development of these ectopic lymphoid structures is dependent on the expression of lymphotoxin-beta and lymphoid chemokines (eg, CXCL13, CCL19, and CCL21) by T, B, dendritic, and stromal cells within the infiltrate [83,86].

Pathogenetic roles have also been attributed to T helper 1 (Th1) cells, natural killer (NK)-like cells, and Th17 cells that produce IFN-gamma. Considerable attention has been given to the role of a subset of CD4+ T cells, termed the follicular helper cells (Tfh), in the pathogenesis of SS [87]. The Tfh cells are characterized by the expression of CXCR5, PD1, ICOS, and Bcl-6 [88]. These cells are the major producer of the cytokine IL-21, provide help to B cells, and are involved in the formation of germinal centers. SS patients have elevated levels of circulating Tfh, and these cells are readily detected in salivary gland biopsies [87].

Cytokines in the glandular tissue — Multiple cytokines have been identified in the salivary gland tissue and represent potential targets for therapy.

The cytokine milieu of salivary glands from SS patients is largely characterized by a Th1/Th17 profile, with production of proinflammatory IL-2, IL-10, and IFN-gamma by infiltrating CD4+ cells and IL-17 by infiltrating Th17 cells. In addition, the proinflammatory cytokines, IL-1, TNF-alpha, and IL-6, can be secreted by activated salivary gland epithelial cells [89,90].

BAFF is considered a key cytokine in SS; it is induced by type I and type II IFN and promotes the activation, proliferation, and survival of B cells. BAFF levels are elevated in the salivary glands and serum of SS patients, and the latter levels correlate with those of anti-SSA/Ro, -SSB/La, and rheumatoid factor [91]. BAFF is produced not only by monocytes, macrophages, and dendritic cells, but also by salivary gland epithelial cells and B and T cells. Thus, BAFF could be an important link between the activation of the innate immune system and the development of autoimmunity through the adaptive immune system.

Lymphomagenesis — Chronic B-cell stimulation and other factors may result, through a series of steps, in malignant B-cell transformation in some patients with SS. Patients with SS have an increased risk of lymphoma, with estimates ranging from 5- to 44-fold, compared with age-matched controls [92]. This increased risk is higher than that observed in other systemic autoimmune diseases, including rheumatoid arthritis, SLE and Crohn disease [93]. These lymphomas are most frequently extranodal marginal zone non-Hodgkin lymphomas (NHL) of "mucosal-associated lymphoid tissue" (MALT). Higher-grade diffuse B-cell lymphomas and T-cell lymphomas are much less frequent in SS [94,95]. The MALT lymphomas often develop in mucosal locations where SS is active, such as salivary glands or the gastrointestinal tract (MALT) [95]; or in the lung, where bronchial-associated lymphoid tissue (BALT) lymphomas can be seen [96,97]. (See "Clinical manifestations, pathologic features, and diagnosis of extranodal marginal zone lymphoma of mucosa associated lymphoid tissue (MALT)".)

The presence of ectopic lymphoid structures in minor salivary gland biopsies is associated with increased risk of lymphoma [98,99]. Such structures were much more common in biopsies from SS patients who later presented with an NHL compared with patients without NHL (86 versus 22 percent) [98]. Ectopic lymphoid structures are sites of antigen-driven B-cell stimulation and clonal expansion, Ig class switching, and somatic hypermutation, potentially engendering lymphoma development.

Chronic stimulation of autoimmune B cells may be associated with malignant transformation, through a series of steps involving the development of a clonal population and eventually uncontrolled clonal proliferation. Salivary gland MALT lymphomas frequently express B-cell antigen receptors with rheumatoid factor activity and bind IgG with high affinity [100,101]. Locally produced IgG autoantibodies directed against the ribonucleoproteins SSA/Ro52, SSA/Ro60, and SSB/La form immune complexes particularly suited for dual-ligand stimulation of B cells with rheumatoid factor B-cell and TLR-7 receptors [102]. The proliferation of these autoreactive B-cells is driven in part by BAFF, serum levels of which correlate with disease activity and the degree of B-cell activation.

Malignant transformation has been associated with specific genetic polymorphisms. A20 (encoded by gene TNFAIP3) is a regulator of NF-kappaB activation and is downregulated in SS [1]. Further, a polymorphism of this gene has been found in a high percentage of SS patients. Mutations and downregulation of A20 have been associated with increased germinal center (GC) formation and MALT lymphomas [103]. Polymorphisms of CXCR5, involved in the organization of GC structures, are associated with SS and NHL [104].

INFORMATION FOR PATIENTS — UpToDate offers two types of patient education materials, "The Basics" and "Beyond the Basics." The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or e-mail these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching on "patient info" and the keyword(s) of interest.)

Beyond the Basics topics (see "Patient education: Sjögren's syndrome (Beyond the Basics)")

SUMMARY

Both genetic and nongenetic factors are involved in disease susceptibility and the disease process. Molecular genetic analyses suggest important roles for human leukocyte antigen (HLA)-DR molecules, and genes encoding elements of both innate and acquired immunity. The strongest associations are with genes in the HLA-DR region, but there is considerable heterogeneity across different ethnic groups. Epigenetic factors may play a role through modulation of gene expression. (See 'Risk factors and etiopathogenesis' above and 'Genetic factors' above and 'HLA genes' above and 'Non-HLA genes' above.)

Many observations suggest a role for viruses in the pathogenesis of Sjögren's syndrome (SS), but no single virus has been implicated. Evidence of ongoing or past viral infection can be detected in many patients, but no virus has been found at high levels in target tissues. An explanation might be that the viral trigger took place years before the development of SS. (See 'The potential role of viral infection' above.)

A cycle of mutual stimulation of the innate and acquired immune systems leads to the perpetuation of glandular injury and dysfunction. Tissue injury occurs through the activation of these immune pathways by lymphocytes within the glandular tissues or extraglandular sites, leading to the release of cytokines, including interferon (IFN)-gamma, interleukin (IL) 17, IL-21, and B-cell activating factor. Salivary and lacrimal gland epithelial cells, as well as the local vascular adhesive molecules, play essential early roles. (See 'Mechanisms of immune-mediated injury' above.)

SS is characterized by the presence of specific autoantibodies, which by some criteria are required for the diagnosis; these include anti-Ro/SSA and anti-La/SSB. Antinuclear antibodies (ANA) are present in 90 percent of patients, and high-titer rheumatoid factor is also frequent. Other antibodies, including antibodies to acetylcholine receptors of salivary glands, may also be seen. (See 'Autoantibodies' above and 'Ro/SSA and La/SSB' above.)

The principal pathologic lesion of SS is a lymphocytic infiltration; the salivary and lacrimal glands are the most frequently affected tissues, but these infiltrates are common to all affected organs, including extraglandular sites, and can result in the glandular and systemic extraglandular features. The infiltrates consist of focal aggregates of lymphocytes, beginning around the ducts and spreading to involve the entire lobule. Migration of the lymphocytes occurs to the gland in response to chemokines, adhesion to specific vascular adhesion molecules, and entry into the glandular cells where they interact with dendritic cells and epithelial cells. (See 'The lymphocytic infiltrate and glandular pathology' above.)

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges Paul Creamer, MD, who contributed to an earlier version of this topic review.

  1. Nocturne G, Mariette X. Advances in understanding the pathogenesis of primary Sjögren's syndrome. Nat Rev Rheumatol 2013; 9:544.
  2. Lessard CJ, Li H, Adrianto I, et al. Variants at multiple loci implicated in both innate and adaptive immune responses are associated with Sjögren's syndrome. Nat Genet 2013; 45:1284.
  3. Li Y, Zhang K, Chen H, et al. A genome-wide association study in Han Chinese identifies a susceptibility locus for primary Sjögren's syndrome at 7q11.23. Nat Genet 2013; 45:1361.
  4. Foster H, Walker D, Charles P, et al. Association of DR3 with susceptibility to and severity of primary Sjögren's syndrome in a family study. Br J Rheumatol 1992; 31:309.
  5. Lee WS, Yoo WH. Primary Sjögren's syndrome in monozygotic twins. Int J Rheum Dis 2014; 17:578.
  6. Cobb BL, Lessard CJ, Harley JB, Moser KL. Genes and Sjögren's syndrome. Rheum Dis Clin North Am 2008; 34:847.
  7. Taylor KE, Wong Q, Levine DM, et al. Genome-Wide Association Analysis Reveals Genetic Heterogeneity of Sjögren's Syndrome According to Ancestry. Arthritis Rheumatol 2017; 69:1294.
  8. Daniels TE, Criswell LA, Shiboski C, et al. An early view of the international Sjögren's syndrome registry. Arthritis Rheum 2009; 61:711.
  9. Kang HI, Fei HM, Saito I, et al. Comparison of HLA class II genes in Caucasoid, Chinese, and Japanese patients with primary Sjögren's syndrome. J Immunol 1993; 150:3615.
  10. Papasteriades CA, Skopouli FN, Drosos AA, et al. HLA-alloantigen associations in Greek patients with Sjögren's syndrome. J Autoimmun 1988; 1:85.
  11. Guggenbuhl P, Jean S, Jego P, et al. Primary Sjögren's syndrome: role of the HLA-DRB1*0301-*1501 heterozygotes. J Rheumatol 1998; 25:900.
  12. Cruz-Tapias P, Rojas-Villarraga A, Maier-Moore S, Anaya JM. HLA and Sjögren's syndrome susceptibility. A meta-analysis of worldwide studies. Autoimmun Rev 2012; 11:281.
  13. Charfi A, Mahfoudh N, Kamoun A, et al. Association of HLA Alleles with Primary Sjögren Syndrome in the South Tunisian Population. Med Princ Pract 2020; 29:32.
  14. Song IW, Chen HC, Lin YF, et al. Identification of susceptibility gene associated with female primary Sjögren's syndrome in Han Chinese by genome-wide association study. Hum Genet 2016; 135:1287.
  15. Bolstad AI, Le Hellard S, Kristjansdottir G, et al. Association between genetic variants in the tumour necrosis factor/lymphotoxin α/lymphotoxin β locus and primary Sjogren's syndrome in Scandinavian samples. Ann Rheum Dis 2012; 71:981.
  16. Piñero J, Ramírez-Anguita JM, Saüch-Pitarch J, et al. The DisGeNET knowledge platform for disease genomics: 2019 update. Nucleic Acids Res 2020; 48:D845.
  17. Altorok N, Coit P, Hughes T, et al. Genome-wide DNA methylation patterns in naive CD4+ T cells from patients with primary Sjögren's syndrome. Arthritis Rheumatol 2014; 66:731.
  18. 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; 75:933.
  19. Imgenberg-Kreuz J, Sandling JK, Almlöf JC, et al. Genome-wide DNA methylation analysis in multiple tissues in primary Sjögren's syndrome reveals regulatory effects at interferon-induced genes. Ann Rheum Dis 2016; 75:2029.
  20. Cole MB, Quach H, Quach D, et al. Epigenetic Signatures of Salivary Gland Inflammation in Sjögren's Syndrome. Arthritis Rheumatol 2016; 68:2936.
  21. Konsta OD, Le Dantec C, Charras A, et al. Defective DNA methylation in salivary gland epithelial acini from patients with Sjögren's syndrome is associated with SSB gene expression, anti-SSB/LA detection, and lymphocyte infiltration. J Autoimmun 2016; 68:30.
  22. Chi C, Taylor KE, Quach H, et al. Hypomethylation mediates genetic association with the major histocompatibility complex genes in Sjögren's syndrome. PLoS One 2021; 16:e0248429.
  23. Bordron A, Devauchelle-Pensec V, Le Dantec C, et al. Epigenetics in Primary Sjögren's Syndrome. Adv Exp Med Biol 2020; 1253:285.
  24. Alevizos I, Alexander S, Turner RJ, Illei GG. MicroRNA expression profiles as biomarkers of minor salivary gland inflammation and dysfunction in Sjögren's syndrome. Arthritis Rheum 2011; 63:535.
  25. Wang-Renault SF, Boudaoud S, Nocturne G, et al. Deregulation of microRNA expression in purified T and B lymphocytes from patients with primary Sjögren's syndrome. Ann Rheum Dis 2018; 77:133.
  26. Jonsson R, Theander E, Sjöström B, et al. Autoantibodies present before symptom onset in primary Sjögren syndrome. JAMA 2013; 310:1854.
  27. Brandt JE, Priori R, Valesini G, Fairweather D. Sex differences in Sjögren's syndrome: a comprehensive review of immune mechanisms. Biol Sex Differ 2015; 6:19.
  28. McCoy SS, Sampene E, Baer AN. Association of Sjögren's Syndrome With Reduced Lifetime Sex Hormone Exposure: A Case-Control Study. Arthritis Care Res (Hoboken) 2020; 72:1315.
  29. Porola P, Virkki L, Przybyla BD, et al. Androgen deficiency and defective intracrine processing of dehydroepiandrosterone in salivary glands in Sjögren's syndrome. J Rheumatol 2008; 35:2229.
  30. Liu K, Kurien BT, Zimmerman SL, et al. X Chromosome Dose and Sex Bias in Autoimmune Diseases: Increased Prevalence of 47,XXX in Systemic Lupus Erythematosus and Sjögren's Syndrome. Arthritis Rheumatol 2016; 68:1290.
  31. Wang J, Syrett CM, Kramer MC, et al. Unusual maintenance of X chromosome inactivation predisposes female lymphocytes for increased expression from the inactive X. Proc Natl Acad Sci U S A 2016; 113:E2029.
  32. Chatzis L, Pezoulas VC, Ferro F, et al. Sjögren's Syndrome: The Clinical Spectrum of Male Patients. J Clin Med 2020; 9.
  33. Ramírez Sepúlveda JI, Kvarnström M, Eriksson P, et al. Long-term follow-up in primary Sjögren's syndrome reveals differences in clinical presentation between female and male patients. Biol Sex Differ 2017; 8:25.
  34. Triantafyllopoulou A, Moutsopoulos H. Persistent viral infection in primary Sjogren's syndrome: review and perspectives. Clin Rev Allergy Immunol 2007; 32:210.
  35. Croia C, Astorri E, Murray-Brown W, et al. Implication of Epstein-Barr virus infection in disease-specific autoreactive B cell activation in ectopic lymphoid structures of Sjögren's syndrome. Arthritis Rheumatol 2014; 66:2545.
  36. Xuan J, Ji Z, Wang B, et al. Serological Evidence for the Association Between Epstein-Barr Virus Infection and Sjögren's Syndrome. Front Immunol 2020; 11:590444.
  37. Nakamura H, Kawakami A. What is the evidence for Sjögren's syndrome being triggered by viral infection? Subplot: infections that cause clinical features of Sjögren's syndrome. Curr Opin Rheumatol 2016; 28:390.
  38. Ghrenassia E, Martis N, Boyer J, et al. The diffuse infiltrative lymphocytosis syndrome (DILS). A comprehensive review. J Autoimmun 2015; 59:19.
  39. Weller ML, Gardener MR, Bogus ZC, et al. Hepatitis Delta Virus Detected in Salivary Glands of Sjögren's Syndrome Patients and Recapitulates a Sjögren's Syndrome-Like Phenotype in Vivo. Pathog Immun 2016; 1:12.
  40. Mavragani CP, Sagalovskiy I, Guo Q, et al. Expression of Long Interspersed Nuclear Element 1 Retroelements and Induction of Type I Interferon in Patients With Systemic Autoimmune Disease. Arthritis Rheumatol 2016; 68:2686.
  41. Szczerba BM, Rybakowska PD, Dey P, et al. Type I interferon receptor deficiency prevents murine Sjogren's syndrome. J Dent Res 2013; 92:444.
  42. Fox RI, Maruyama T. Pathogenesis and treatment of Sjögren's syndrome. Curr Opin Rheumatol 1997; 9:393.
  43. Hayashi T. Dysfunction of lacrimal and salivary glands in Sjögren's syndrome: nonimmunologic injury in preinflammatory phase and mouse model. J Biomed Biotechnol 2011; 2011:407031.
  44. Dawson LJ, Fox PC, Smith PM. Sjogrens syndrome--the non-apoptotic model of glandular hypofunction. Rheumatology (Oxford) 2006; 45:792.
  45. Hjelmervik TO, Petersen K, Jonassen I, et al. Gene expression profiling of minor salivary glands clearly distinguishes primary Sjögren's syndrome patients from healthy control subjects. Arthritis Rheum 2005; 52:1534.
  46. Emamian ES, Leon JM, Lessard CJ, et al. Peripheral blood gene expression profiling in Sjögren's syndrome. Genes Immun 2009; 10:285.
  47. Ohlsson M, Jonsson R, Brokstad KA. Subcellular redistribution and surface exposure of the Ro52, Ro60 and La48 autoantigens during apoptosis in human ductal epithelial cells: a possible mechanism in the pathogenesis of Sjögren's syndrome. Scand J Immunol 2002; 56:456.
  48. Bolstad AI, Jonsson R. The role of apoptosis in Sjögren's syndrome. Ann Med Interne (Paris) 1998; 149:25.
  49. Jonsson R, Vogelsang P, Volchenkov R, et al. The complexity of Sjögren's syndrome: novel aspects on pathogenesis. Immunol Lett 2011; 141:1.
  50. Båve U, Nordmark G, Lövgren T, et al. Activation of the type I interferon system in primary Sjögren's syndrome: a possible etiopathogenic mechanism. Arthritis Rheum 2005; 52:1185.
  51. Wang X, Bootsma H, Terpstra J, et al. Progenitor cell niche senescence reflects pathology of the parotid salivary gland in primary Sjögren's syndrome. Rheumatology (Oxford) 2020; 59:3003.
  52. Pringle S, Wang X, Verstappen GMPJ, et al. Salivary Gland Stem Cells Age Prematurely in Primary Sjögren's Syndrome. Arthritis Rheumatol 2019; 71:133.
  53. Asmussen K, Andersen V, Bendixen G, et al. A new model for classification of disease manifestations in primary Sjögren's syndrome: evaluation in a retrospective long-term study. J Intern Med 1996; 239:475.
  54. Tzioufas AG, Voulgarelis M. Update on Sjögren's syndrome autoimmune epithelitis: from classification to increased neoplasias. Best Pract Res Clin Rheumatol 2007; 21:989.
  55. Lauvsnes MB, Maroni SS, Appenzeller S, et al. Memory dysfunction in primary Sjögren's syndrome is associated with anti-NR2 antibodies. Arthritis Rheum 2013; 65:3209.
  56. Bårdsen K, Nilsen MM, Kvaløy JT, et al. Heat shock proteins and chronic fatigue in primary Sjögren's syndrome. Innate Immun 2016; 22:162.
  57. Rivera TL, Izmirly PM, Birnbaum BK, et al. Disease progression in mothers of children enrolled in the Research Registry for Neonatal Lupus. Ann Rheum Dis 2009; 68:828.
  58. Itoh K, Itoh Y, Frank MB. Protein heterogeneity in the human Ro/SSA ribonucleoproteins. The 52- and 60-kD Ro/SSA autoantigens are encoded by separate genes. J Clin Invest 1991; 87:177.
  59. Ozato K, Shin DM, Chang TH, Morse HC 3rd. TRIM family proteins and their emerging roles in innate immunity. Nat Rev Immunol 2008; 8:849.
  60. Zhang Z, Bao M, Lu N, et al. The E3 ubiquitin ligase TRIM21 negatively regulates the innate immune response to intracellular double-stranded DNA. Nat Immunol 2013; 14:172.
  61. Rhodes DA, Isenberg DA. TRIM21 and the Function of Antibodies inside Cells. Trends Immunol 2017; 38:916.
  62. Hakulinen A, Heinonen K, Jokela V, Launiala K. Prematurity-associated morbidity during the first two years of life. A population-based study. Acta Paediatr Scand 1988; 77:340.
  63. Szczerba BM, Kaplonek P, Wolska N, et al. Interaction between innate immunity and Ro52-induced antibody causes Sjögren's syndrome-like disorder in mice. Ann Rheum Dis 2016; 75:617.
  64. Sroka M, Bagavant H, Biswas I, et al. Immune response against the coiled coil domain of Sjögren's syndrome associated autoantigen Ro52 induces salivary gland dysfunction. Clin Exp Rheumatol 2018; 36 Suppl 112:41.
  65. Sim S, Wolin SL. Emerging roles for the Ro 60-kDa autoantigen in noncoding RNA metabolism. Wiley Interdiscip Rev RNA 2011; 2:686.
  66. Hung T, Pratt GA, Sundararaman B, et al. The Ro60 autoantigen binds endogenous retroelements and regulates inflammatory gene expression. Science 2015; 350:455.
  67. Tröster H, Metzger TE, Semsei I, et al. One gene, two transcripts: isolation of an alternative transcript encoding for the autoantigen La/SS-B from a cDNA library of a patient with primary Sjögrens' syndrome. J Exp Med 1994; 180:2059.
  68. Dawson L, Tobin A, Smith P, Gordon T. Antimuscarinic antibodies in Sjögren's syndrome: where are we, and where are we going? Arthritis Rheum 2005; 52:2984.
  69. Robinson CP, Brayer J, Yamachika S, et al. Transfer of human serum IgG to nonobese diabetic Igmu null mice reveals a role for autoantibodies in the loss of secretory function of exocrine tissues in Sjögren's syndrome. Proc Natl Acad Sci U S A 1998; 95:7538.
  70. Bacman S, Perez Leiros C, Sterin-Borda L, et al. Autoantibodies against lacrimal gland M3 muscarinic acetylcholine receptors in patients with primary Sjögren's syndrome. Invest Ophthalmol Vis Sci 1998; 39:151.
  71. He J, Guo JP, Ding Y, et al. Diagnostic significance of measuring antibodies to cyclic type 3 muscarinic acetylcholine receptor peptides in primary Sjogren's syndrome. Rheumatology (Oxford) 2011; 50:879.
  72. Waterman SA, Gordon TP, Rischmueller M. Inhibitory effects of muscarinic receptor autoantibodies on parasympathetic neurotransmission in Sjögren's syndrome. Arthritis Rheum 2000; 43:1647.
  73. Kovács L, Marczinovits I, György A, et al. Clinical associations of autoantibodies to human muscarinic acetylcholine receptor 3(213-228) in primary Sjogren's syndrome. Rheumatology (Oxford) 2005; 44:1021.
  74. Sumida T, Tsuboi H, Iizuka M, et al. Anti-M3 muscarinic acetylcholine receptor antibodies in patients with Sjögren's syndrome. Mod Rheumatol 2013; 23:841.
  75. Christodoulou MI, Kapsogeorgou EK, Moutsopoulos HM. Characteristics of the minor salivary gland infiltrates in Sjögren's syndrome. J Autoimmun 2010; 34:400.
  76. Jonsson R, Gordon TP, Konttinen YT. Recent advances in understanding molecular mechanisms in the pathogenesis and antibody profile of Sjögren's syndrome. Curr Rheumatol Rep 2003; 5:311.
  77. Manoussakis MN, Kapsogeorgou EK. The role of intrinsic epithelial activation in the pathogenesis of Sjögren's syndrome. J Autoimmun 2010; 35:219.
  78. Ittah M, Miceli-Richard C, Gottenberg JE, et al. Viruses induce high expression of BAFF by salivary gland epithelial cells through TLR- and type-I IFN-dependent and -independent pathways. Eur J Immunol 2008; 38:1058.
  79. Ittah M, Miceli-Richard C, Eric Gottenberg J, et al. B cell-activating factor of the tumor necrosis factor family (BAFF) is expressed under stimulation by interferon in salivary gland epithelial cells in primary Sjögren's syndrome. Arthritis Res Ther 2006; 8:R51.
  80. Sandhya P, Kurien BT, Danda D, Scofield RH. Update on Pathogenesis of Sjogren's Syndrome. Curr Rheumatol Rev 2017; 13:5.
  81. Amft N, Curnow SJ, Scheel-Toellner D, et al. Ectopic expression of the B cell-attracting chemokine BCA-1 (CXCL13) on endothelial cells and within lymphoid follicles contributes to the establishment of germinal center-like structures in Sjögren's syndrome. Arthritis Rheum 2001; 44:2633.
  82. McArthur C, Wang Y, Veno P, et al. Intracellular trafficking and surface expression of SS-A (Ro), SS-B (La), poly(ADP-ribose) polymerase and alpha-fodrin autoantigens during apoptosis in human salivary gland cells induced by tumour necrosis factor-alpha. Arch Oral Biol 2002; 47:443.
  83. Bombardieri M, Lewis M, Pitzalis C. Ectopic lymphoid neogenesis in rheumatic autoimmune diseases. Nat Rev Rheumatol 2017; 13:141.
  84. Pontarini E, Lucchesi D, Bombardieri M. Current views on the pathogenesis of Sjögren's syndrome. Curr Opin Rheumatol 2018; 30:215.
  85. Salomonsson S, Jonsson MV, Skarstein K, et al. Cellular basis of ectopic germinal center formation and autoantibody production in the target organ of patients with Sjögren's syndrome. Arthritis Rheum 2003; 48:3187.
  86. Barone F, Bombardieri M, Manzo A, et al. Association of CXCL13 and CCL21 expression with the progressive organization of lymphoid-like structures in Sjögren's syndrome. Arthritis Rheum 2005; 52:1773.
  87. Chen W, Yang F, Xu G, et al. Follicular helper T cells and follicular regulatory T cells in the immunopathology of primary Sjögren's syndrome. J Leukoc Biol 2021; 109:437.
  88. Vinuesa CG, Linterman MA, Yu D, MacLennan IC. Follicular Helper T Cells. Annu Rev Immunol 2016; 34:335.
  89. Fox RI, Kang HI, Ando D, et al. Cytokine mRNA expression in salivary gland biopsies of Sjögren's syndrome. J Immunol 1994; 152:5532.
  90. Boumba D, Skopouli FN, Moutsopoulos HM. Cytokine mRNA expression in the labial salivary gland tissues from patients with primary Sjögren's syndrome. Br J Rheumatol 1995; 34:326.
  91. Nocturne G, Mariette X. B cells in the pathogenesis of primary Sjögren syndrome. Nat Rev Rheumatol 2018; 14:133.
  92. Nocturne G, Mariette X. Sjögren Syndrome-associated lymphomas: an update on pathogenesis and management. Br J Haematol 2015; 168:317.
  93. Ekström Smedby K, Vajdic CM, Falster M, et al. Autoimmune disorders and risk of non-Hodgkin lymphoma subtypes: a pooled analysis within the InterLymph Consortium. Blood 2008; 111:4029.
  94. Voulgarelis M, Ziakas PD, Papageorgiou A, et al. Prognosis and outcome of non-Hodgkin lymphoma in primary Sjögren syndrome. Medicine (Baltimore) 2012; 91:1.
  95. Royer B, Cazals-Hatem D, Sibilia J, et al. Lymphomas in patients with Sjogren's syndrome are marginal zone B-cell neoplasms, arise in diverse extranodal and nodal sites, and are not associated with viruses. Blood 1997; 90:766.
  96. Ahmed S, Kussick SJ, Siddiqui AK, et al. Bronchial-associated lymphoid tissue lymphoma: a clinical study of a rare disease. Eur J Cancer 2004; 40:1320.
  97. Lee IJ, Kim SH, Koo SH, et al. Bronchus-associated lymphoid tissue (BALT) lymphoma of the lung showing mosaic pattern of inhomogeneous attenuation on thin-section CT: a case report. Korean J Radiol 2000; 1:159.
  98. Theander E, Vasaitis L, Baecklund E, et al. Lymphoid organisation in labial salivary gland biopsies is a possible predictor for the development of malignant lymphoma in primary Sjögren's syndrome. Ann Rheum Dis 2011; 70:1363.
  99. Bombardieri M, Barone F, Humby F, et al. Activation-induced cytidine deaminase expression in follicular dendritic cell networks and interfollicular large B cells supports functionality of ectopic lymphoid neogenesis in autoimmune sialoadenitis and MALT lymphoma in Sjögren's syndrome. J Immunol 2007; 179:4929.
  100. Bende RJ, Aarts WM, Riedl RG, et al. Among B cell non-Hodgkin's lymphomas, MALT lymphomas express a unique antibody repertoire with frequent rheumatoid factor reactivity. J Exp Med 2005; 201:1229.
  101. Bende RJ, Janssen J, Beentjes A, et al. Salivary Gland Mucosa-Associated Lymphoid Tissue-Type Lymphoma From Sjögren's Syndrome Patients in the Majority Express Rheumatoid Factors Affinity-Selected for IgG. Arthritis Rheumatol 2020; 72:1330.
  102. Lau CM, Broughton C, Tabor AS, et al. RNA-associated autoantigens activate B cells by combined B cell antigen receptor/Toll-like receptor 7 engagement. J Exp Med 2005; 202:1171.
  103. Nocturne G, Boudaoud S, Miceli-Richard C, et al. Germline and somatic genetic variations of TNFAIP3 in lymphoma complicating primary Sjogren's syndrome. Blood 2013; 122:4068.
  104. Song H, Tong D, Cha Z, Bai J. C-X-C chemokine receptor type 5 gene polymorphisms are associated with non-Hodgkin lymphoma. Mol Biol Rep 2012; 39:8629.
Topic 5620 Version 24.0

References