Skip to main content

CXCL9 may serve as a potential biomarker for primary Sjögren’s syndrome with extra-glandular manifestations

Abstract

Background

Primary Sjögren's syndrome (pSS) is an autoimmune condition that causes harm to exocrine glands and also has extra-glandular manifestations (EGM). pSS patients with EGM have a worse prognosis than those with only sicca symptoms. Previous studies have shown that the minor salivary glands (MSG) of pSS patients exhibit a unique profile of cytokines and chemokines compared to healthy controls. However, there is a lack of research comparing pSS with EGM (pSS-EGM) and pSS without EGM (pSS-non-EGM). This study aims to explore potential biomarkers associated with pSS, particularly pSS with EGM.

Methods

By utilizing RNA sequencing, we conducted an analysis on the gene expression profiles of MSG in 63 patients diagnosed with pSS, as well as 12 non-pSS individuals. Furthermore, we also investigated the MSG of pSS patients, both with and without EGM. Through bioinformatics analysis, we identified genes with differential expression (DEGs) and determined the core hub genes using PPI network. We then analyzed the top 20 DEGs and their correlation with the patients' clinical characteristics, and validated our findings using peripheral blood plasma.

Results

A total of 725 differentially expressed genes (DEGs) were identified in the comparison between pSS and non-pSS groups, and 727 DEGs were observed between pSS-EGM and pSS-non-EGM. It is noteworthy that the expression levels of CXCL9 were higher in both pSS patients and pSS-EGM when compared to the control group. Taking into consideration the significance of the top 20 DEGs in relation to clinical parameters and the central hub genes, we ultimately chose CXCL9. In comparison to the non-pSS group, pSS patients exhibited notably greater expression of the CXCL9 gene in the MSG, as well as higher levels of CXCL9 protein in their plasma (p < 0.001). Furthermore, the expression of the CXCL9 gene and levels of CXCL9 protein were notably higher in pSS patients accompanied by EGM and those with SSA antibodies. Additionally, a correlation was found between the expression of the CXCL9 gene and the EULAR Sjogren’s Syndrome Disease Activity Index (ESSDAI), as well as with immunoglobulin G (IgG) levels and erythrocyte sedimentation rate (ESR). Meanwhile, the protein levels of CXCL9 were found to be correlated with IgG levels and ESSDAI.

Conclusion

CXCL9 proves to be a valuable biomarker in pSS, specifically due to its strong ability to differentiate between pSS patients with EGM and those without EGM. There is a significant correlation between CXCL9 and various clinical parameters both at the gene and protein level. Therefore, CXCL9 could be a potential target for future treatment of pSS.

Introduction

Primary Sjögren's syndrome (pSS) is an autoimmune condition that usually starts with gradual sicca symptoms [1, 2], but approximately 40% of patients may have extra-glandular manifestations (EGM) [3]. These EGM can affect different areas of the body, such as joints, skin, lungs, kidneys, and nervous system, and may range in severity.

They not only significantly impact a person's overall health, but also increase the burden of the disease, in addition to causing dryness [4, 5]. EGM in patients with pSS affects the prognosis of patients [4, 6, 7].

The precise cause of pSS is not currently fully understood. However, lymphocytes have traditionally been believed to play a key role in its development [8]. Chemokines, which are small proteins, attract immune cells and interact with chemokine receptors to facilitate their movement and differentiation [9]. Research has indicated that the levels of CXCL13 increase before the disease becomes clinically evident. This overexpression of CXCL13 promotes the formation of abnormal lymphoid tissues primarily composed of B lymphocytes in pSS. Additionally, the use of anti-CXCL13 antibodies has shown effectiveness in managing pSS [10, 11]. Furthermore, studies have found that CXCL9, CXCL10, and CXCL11 are upregulated on the corneal and conjunctival epithelium in response to desiccating stress in mice and in patients with dry eye [12, 13]. In 2023, Zhang et al. discovered that the CXCL9, 10, 11/CXCR3 axis, which activates G protein-coupled receptor kinase 2, is involved in regulating the migration of T lymphocytes, contributing to the development of pSS [14]. Inhibiting the CXCL9, 10, 11/CXCR3 axis or G protein-coupled receptor kinase 2 can reduce the migration of T lymphocytes [14].

A key factor in diagnosing pSS is the use of a minor salivary gland (MSG) biopsy [15, 16]. Pathologists analyze the minor salivary gland, and the existence of one or more lymphocytic foci with over 50 lymphocytes per 4 mm2 is considered highly indicative of pSS [16, 17]. This is a critical diagnostic criterion for the disease. Additionally, previous research [18,19,20], along with our own [21, 22], has shown that patients with pSS have a distinct gene-expression signature in their MSG that sets them apart from healthy individuals. Most of these DEGs are upregulated, such as type I interferon, chemokine CXCL9, CXCL10, and CXCL13 [23,24,25,26,27].

While most studies focus on patients with primary Sjögren's syndrome (pSS) and non-pSS patients, as well as healthy individuals, limited research exists on patients with pSS associated with EGM and pSS without EGM. In this study, we examined the clinical traits and gene expression of minor salivary gland (MSG) and plasma samples. We compared these samples not only between individuals diagnosed with pSS and those without the diagnosis but also between pSS-EGM and pSS-non-EGM patients, aiming to identify potential biomarkers through the comparison of various indicators.

Materials and methods

Patients and sample collection

Between January 2020 and August 2021, a group of 63 pSS patients and 12 non-pSS subjects were enrolled at the First Affiliated Hospital of Wenzhou Medical University in Zhejiang, China. Based on the 2012 ACR criteria or the 2016 (ACR)/EULAR classification criteria, pSS was diagnosed in this study [28, 29]. The non-pSS subjects were mainly from the physical examination center of our hospital. This group of patients was treated in our Rheumatology and Immunology department because they were positive for antibodies or experienced discomfort with dry mouth or dry eyes. Eventually, the diagnosis of pSS was ruled out based on negative antibodies, MSG biopsy, ophthalmic KCS examination, and saliva flow rate testing. MSG biopsy was performed on all patients for diagnostic purposes.

The inclusion criteria for this study were as follows: Willing and voluntary participation of all participants. Signing of an informed consent form by all participants. Age range between 18 and 70 years for participants. The exclusion criteria were: Patients who had received treatment (including glucocorticoids, immunosuppressants, or biological agents). Patients who were currently pregnant or lactating. Patients with another known connective tissue disease or overlap syndrome. Patients with acquired immunodeficiency. Patients with a history of diagnosed malignant tumors. Patients with recent infections.

At the same time as the MSG biopsies, we collected salivary glands and plasma samples from all participants to analyze the results. In addition, plasma samples were collected from 10 patients with systemic lupus erythematosus (SLE) (Table S1) and 20 healthy individuals. To avoid confounding effects, collect all data, including clinical information and EULAR Sjögren's Syndrome Disease Activity Index (ESSDAI) scores [30, 31], prior to administering any systemic immunosuppressant or glucocorticoid medications. Systemic involvement was defined according to the ESSDAI [31]. The data gathered encompassed patient age, dry eyes or mouth symptoms, test results for ocular examinations, and symptoms of extra-glandular involvement. Other documented variables included active joint involvement, interstitial lung disease (ILD) [32], skin vasculitis, peripheral neurological involvement, and renal involvement characterized by persistent proteinuria, tubular acidosis, interstitial nephritis, or glomerulonephritis. Additionally, laboratory investigations, including anti-nuclear antibodies (ANA), anti-SSA antibody (SSA), anti-SSB antibody (SSB), rheumatoid factor (RF), levels of immunoglobulins, and complement component levels (C3 and C4), etc., were extracted from the patient’s clinical record.

The study (#16024) was approved by the ethics committee of the First Affiliated Hospital of Wenzhou Medical University.

Extraction of RNA, preparation of cDNA libraries, and sequencing

Gene expression was investigated in this study using RNA sequencing. We extracted total RNA from frozen salivary gland samples by utilizing TRIzol Reagent. The RNA purity was determined by utilizing a Nano Photometer spectrophotometer. To evaluate RNA quality, we employed a Bioanalyzer 2100 system with an RNA 6000 Nano kit. To generate sequencing libraries for Illumina, the NEBNext Ultra RNA Library Prep Kit was utilized using a 3 mg RNA sample. We utilized the Illumina HiSeq platform for sequencing. To improve data quality, we used TrimGalore software and Cutadapt adapters to trim raw reads and filter low-quality reads. With FastQC software, we also generated quality-control reports for sequence reads. DESeq2 was used to normalize read count files after aligning them with the human reference genome “hg38”.

Bioinformatics analysis

The DEseq2 packages were utilized to identify the differentially expressed genes (DEGs). The cluster profiler package in R was used to examine the annotation of Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway [33]. A threshold for significance was established with a criterion of adjusted p-value < 0.05. We evaluated the interactive connections and protein–protein interaction (PPI) networks of the shared DEGs by utilizing the STRING database [34]. The biological network of important DEGs [25] was created and visualized using Cytoscape software [35].

Enzyme-linked immunosorbent assay

The plasma levels of C-X-C motif chemokine ligand 9 (CXCL9) and soluble C-X-C motif chemokine receptor 3 (CXCR3) were measured in pSS patients, SLE patients, non-pSS subjects, and healthy controls using the Human CXCL9/MIG ELISA Kit from MULTI SCIENCES (Hangzhou, Zhejiang, China) and the Human Chemokine C-X-C-Motif Receptor 3 (CXCR3) ELISA Kit from Jianglai Biological (Shanghai, China), following the product guides. To measure the color produced by 3,3′,5,5′-tetramethylbenzidine (BD Biosciences, United States), the ELISA plate reader was used to record the absorbance at 450 nm. Three tests were conducted on each sample.

Statistical analysis

We analyzed the data using R version 4.2.0. The mean and standard error (SE) were calculated using a two-sample t-test. DEGs were identified based on a log2 fold change that was either less than -1 or greater than 1, along with an adjusted p-value below 0.001. Functional enrichment analysis was performed using the clusterprofiler package. For data that followed a normal distribution, the Student’s t-test was employed, whereas the Mann–Whitney U test was utilized for variables that did not exhibit normality. Categorical variables were compared using chi-square tests. Spearman's correlation analysis was utilized to quantify the connections between gene expression and clinical parameters. A p-value less than 0.05 was used to define statistical significance.

Results

Patient characteristics

In this study, we compared the characteristics of patients with pSS and non-pSS. Table 1 summarizes the characteristics of 63 pSS patients and 12 non-pSS subjects. Our results show that a greater proportion of pSS patients test positive for ANA, anti-SSB, and anti-SSA antibodies (all p-values < 0.05) compared to those without pSS. Nevertheless, neither group showed any significant differences in age or gender.

Table 1 Clinical characteristics of patients with pSS and non-pSS

We further analyzed the characteristics of pSS patients and categorized them into two groups: those with EGM and those without EGM. Table 2 presents the characteristics of these two groups. Compared to pSS-non-EGM patients, pSS patients with EGM have higher levels of ANA, RF, ESR, IgG and ESSDAI (all p-values < 0.05). Nevertheless, there was no significant difference in anti-SSA, anti-SSB, age, gender, C3, C4, dry eyes, or dry mouth between the two groups.

Table 2 Clinical characteristics of patients with pSS-EGM and pSS-non-EGM

Detecting DEGs in pSS patients

Using RNA sequencing, we analyzed gene expression patterns of MSGs in 63 pSS patients and 12 non-pSS patients. We discovered 725 genes that exhibit varying levels of expression between individuals with pSS and non-pSS subjects, with 697 of these genes (96.1%) being up-regulated. Among these DEGs, CXCL9 showed the most significant difference in expression (Fig. 1A). Figure 1B displays the top 20 DEGs. In addition, we used Cytoscape v3.9.0 software to construct a PPI network graph (Fig. 1C), which revealed hub genes at the central position, including STAT1, IFNG, CD4, CXCL9, PTPRC.

Fig. 1
figure 1

Identification of DEGs in patients. A Volcano plot representing differentially expressed genes between pSS and non-pSS samples. Genes with adjusted p < 0.001 and absolute log2-fold change > 1 are represented in red or blue. B Heatmap showing the top 20 DEGs between pSS and non-pSS. C The network consists of 100 top DEGs between pSS and non-pSS. D Volcano plot representing differentially expressed genes between pSS-EGM and pSS-non-EGM samples. Genes with adjusted p < 0.001 and absolute log2-fold change > 1 are represented in red or blue. E Heatmap showing the top 20 DEGs between pSS-EGM and pSS-non-EGM. F The network consists of 100 top DEGs between pSS-EGM and pSS-non-EGM. In the network, each gene is represented by a node, and the connections between nodes represent the interactions between the corresponding proteins. The size and color of the nodes indicate the degree (number of connection) of the genes. The larger size and darker color indicate a higher degree and greater importance

Furthermore, we used RNA sequencing to analyze the gene expression patterns in patients with pSS, both with and without EGMs. We found 727 genes that were expressed differently between the two groups, with 517 (71.1%) of these genes being up-regulated. Notably, CXCL9 had significantly higher expression levels in pSS patients with EGMs than in those without, as shown in Fig. 1D. The top 20 DEGs, including CD84, IRF1, IL21RB, CXCL9, and others, are displayed in Fig. 1E. Additionally, the hub genes at the central position include STAT1, ZAP70, LCP2, CXCL9, PTPRC (Fig. 1F).

Pathway enrichment analysis of upregulated DEGs

In order to investigate potential biological mechanisms associated with pSS and extra-glandular involvement in pSS, we conducted analyses on commonly upregulated DEGs using GO and KEGG pathway methods. In Fig. 2A, the analysis of upregulated DEGs between pSS and non-pSS reveals the top 20 pathways that are enriched. In pSS, several immune-related pathways showed significant upregulation, including leukocyte cellular adhesion, differentiation of T cells, and proliferation of lymphocytes in GO. Additionally, there was an upregulation in pathways such as Cytokine-cytokine receptor interaction, T cell receptor signaling, and Chemokine signaling in KEGG.

Fig. 2
figure 2

Pathway enrichment analysis of upregulated DEGs. A Bar chart depicting the top 20 enriched Gene Ontology (GO) and KEGG pathways from the analysis of upregulated DEGs between pSS and non-pSS. B Bar chart illustrating the top 20 enriched Gene Ontology (GO) and KEGG pathways from the analysis of upregulated DEGs between pSS-EGM and pSS-non-EGM. The x-axis displays the GO and KEGG pathways, while the y-axis shows the enrichment score (-log10 adjusted p-value). Each bar corresponds to a specific pathway, with its height representing the degree of enrichment

Moreover, Fig. 2B demonstrates the top 20 enriched GO and KEGG pathways resulting from the examination of upregulated DEGs between pSS-EGM and pSS-non-EGM. In a similar manner as the previous outcome, a significant number of immune-related pathways exhibit upregulation in pSS-EGM in comparison to pSS-non-EGM. Additionally, the Chemokine signaling pathway is an important pathway in both groups.

Clinical correlations of CXCL9 in pSS

To determine how potential biomarkers affect the severity of pSS-EGM disease, we analyzed the correlation between the top 20 DEGs (DEGs between pSS-EGM and pSS-non-EGM) and various clinical parameters, including ESSDAI scores, ESR, CRP, IgA, IgG, IgM, and other clinical parameters. Among the 20 genes, only CXCL9 and PTPRC were identified as the central hub genes. Moreover, it was observed that CXCL9 exhibited a stronger correlation with the clinical parameters. Therefore, we have selected CXCL9 for additional analysis and included it in the Supplementary materials. Our analysis showed that CXCL9 in MSG had a positive correlation with ESSDAI score (Fig. 3A, r = 0.47, p < 0.001), IgG levels (Fig. 3B, r = 0.52, p < 0.001), and ESR (Fig. 3C, r = 0.40, p = 0.002). Nevertheless, no significant associations were found between CXCL9 RNA expression and other clinical factors.

Fig. 3
figure 3

Association between CXCL9 and clinical features of primary Sjögren's syndrome. A, B, C The Spearman correlation between expression of CXCL9 gene in MSG and clinical characteristics such as ESSDAI score (A), IgG (B), and ESR (C). D, E, F The Spearman correlation between expression of CXCL9 gene in plasma and clinical characteristics such as ESSDAI score (D) and IgG (E), but not with ESR (F). The top and right boxplot indicates the distribution. Dashed lines represent the mean value

Assessment of circulating chemokine CXCL9 and CXCR3

ELISA was used to measure CXCL9 and CXCR3 levels in plasma from patients with pSS, non-pSS, as well as 10 SLE patients and 20 healthy individuals. A higher level of CXCL9 and CXCR3 was noted in both pSS and SLE patients (Fig. 4A, B). Furthermore, a notably increase in CXCL9 expression was found in plasma of pSS patients with EGM. In contrast, CXCR3 expression did not display significant differences between pSS patients with and without EGM (Fig. 4C, D).

Fig. 4
figure 4

Differential expression of CXCL9 and CXCR3 in plasma. A, B Boxplots depicting the expression of plasma CXCL9 (A) and CXCR3 (B) in HC, non-pSS, pSS, SLE. C, D Boxplots showing the expression of plasma CXCL9 (C) and CXCR3 (D) between pSS-non-EGM and pSS-EGM. Significance determined by Wilcoxon's test

Furthermore, a strong association was observed between CXCL9 levels in plasma and both ESSDAI score and IgG levels. The correlation coefficients were 0.46 (p < 0.001) and 0.37 (p = 0.003), respectively (Fig. 3D, E, F).

CXCL9 expression in pSS with EGM

The study measured the expression of CXCL9 in MSGs and blood plasma of pSS patients with EGM. We sorted the patients into six subgroups based on their symptoms: peripheral blood involvement, joint involvement, kidney involvement, ILD, peripheral nervous system involvement, and rash involvement. There was a notable rise in CXCL9 expression in patients with joint involvement, rash involvement, blood involvement, kidney involvement, and ILD, compared to pSS-non-EGM patients, except for those with peripheral nervous system involvement. Furthermore, patients with blood involvement exhibited higher CXCL9 expression levels than patients with kidney involvement and ILD (Fig. 5A). Similarly, CXCL9 expression in plasma was significantly higher in patients with joint involvement, blood involvement, kidney involvement, and ILD, compared to pSS-non-EGM patients, except for those with peripheral nervous system involvement and rash involvement. Moreover, pSS patients with blood involvement had the highest levels of CXCL9 expression (Fig. 5B).

Fig. 5
figure 5

Differentially expressed CXCL9 in pSS subgroups with different EGM. A Box plot showing the expression of CXCL9 in pSS-EGM subgroups and pSS-non-EGM in MSGs. B Box plot showing the expression of CXCL9 in pSS-EGM subgroups and pSS-non-EGM in plasma. Significance determined by Kruskall-Wallis’s test. The “*” symbol denotes the significance between pSS-non-EGM and pSS-EGM subgroups. The “#” symbol denotes the significance between blood involvement and other subgroups within the pSS-EGM group. * p < 0.05, **p < 0.01, *** p < 0.001, **** p < 0.001; # p < 0.05, ## p < 0.01, ### p < 0.001, #### p < 0.001

CXCL9 expression is elevated in pSS with SSA antibodies

The study discovered that pSS patients who have SSA antibodies may have higher levels of CXCL9 expression in their minor salivary gland (MSG) and plasma. Antibodies against SSA were used to classify patients. A notable distinction was found between patients with antibodies and those without antibodies in the expression of the CXCL9 gene in MSG and the plasma levels of the protein, as shown in Figure S2A and S2B.

Discussion

Although there is a wealth of literature on the application of RNA-seq in investigating pSS patients [36,37,38,39,40], research on pSS patients with EGM is relatively limited. For this research, we employed RNA-seq to examine the gene expression patterns of diagnosed pSS patients in comparison to individuals without pSS, as well as pSS patients with and without EGM. In pSS, there are disruptions in several immune-related pathways, and these disruptions also occur in cases of pSS with extra-glandular involvement. Among these pathways, the Chemokine signaling pathway was found to be particularly important in both groups, which is consistent with prior studies on pSS [41, 42]. Additionally, among several DEGs, CXCL9 showed a significant difference in expression in pSS. Furthermore, CXCL9 has the potential to act as a critical regulator or signaling molecule in the network that drives the progression of pSS and pSS with EGM.

CXCL9 is a chemokine protein that interacts with its receptor, CXCR3, to attract T cells, natural killer cells, and macrophages to the location of inflammation [43,44,45]. Interferon-gamma-induced CXCL9 is also called MIG (monokine induced by interferon gamma) and is believed to be involved in the development of multiple autoimmune disease [46,47,48], including pSS [23, 49]. Several studies [13, 50] have reported elevated levels of CXCL9 in the exocrine glands tissue of patients with pSS, and our results were consistent with what has been previously described in the literature. However, the correlation between CXCL9 and extra-glandular involvement of pSS has not been previously investigated.Our study found that the expression of CXCL9 is higher in MSGs from pSS patients compared to non-pSS subjects. Furthermore, it was observed that pSS and SLE patients had elevated levels of CXCL9 and CXCR3 in the plasma when compared to individuals of non-pSS and healthy controls. Numerous studies [51,52,53] have suggested that the development of pSS and SLE shares some similarities. Our findings indicate that the CXCL9/CXCR3 axis may have a significant role in the pathogenesis of both pSS and SLE. Moreover, we discovered a connection between the CXCL9 gene's expression and CXCL9 protein levels, as well as various clinical characteristics in these individuals, including ESR, IgG levels, and ESSDAI. This suggests that biomarker CXCL9 may be potentially serve as a helpful indicator for assessing both disease activity and progression in individuals with pSS. Such information could assist clinicians in making more informed decisions regarding treatment options for their patients, indicating that CXCL9 could play a role in the progression of pSS.

Our findings indicate that compared to patients without EGM, patients with pSS who exhibit EGM have higher levels of ANA, RF, ESR, IgG, and ESSDAI (all p-values < 0.05). Due to the importance of these clinical parameters in the diagnosis and treatment of pSS [54,55,56,57], it is essential to consider them when evaluating patients with confirmed pSS. This will facilitate early detection of EGM in pSS patients. Additionally, we found a considerable rise in the expression of the CXCL9 gene and CXCL9 protein levels in pSS patients with EGM compared to those without EGM. It is known that the CXCL9/CXCR3 pathway is critical in the migration of immune cells [43, 58, 59]. Studies have shown that the migration of a large number of immune cells and invasion of glandular tissue are the first and most important pathological manifestations of pSS [13, 49, 56]. This suggests that the occurrence of EGM in pSS patients could be related to the CXCL9/CXCR3 pathway. There are various manifestations of extra-glandular involvement in patients with pSS [60, 61]. To further understand of the role of CXCL9 in various types of extra-glandular involvement, the pSS-EGM group was divided into six subgroups. In addition to peripheral nervous system and rash involvement, the expression of CXCL9 was noticeably higher in pSS patients with joint, blood, kidney, and ILD involvement in comparison to pSS-non-EGM patients. Patients with blood involvement showed the highest levels of CXCL9 expression. Further research is required to explore the relationship between EGM and the CXCL9/CXCR3 pathway.

The anti-SSA antibody, also known as anti-Ro antibody, is an autoantibody that targets the SSA antigen [62, 63]. It is one of the diagnostic criteria for the disease [28, 55]. Previous studies [6, 64, 65] have found that the existence of SSA antibodies is a reliable indication of the existence of EGM in pSS over an extended period of observation. However, our study discovered that the rates of positivity in SSA showed no significant variation among patients with and without EGM. The limited number of pSS patients in our sample may have influenced the research findings, and race differences may have also played a role. To verify these findings in the future, it may be necessary to conduct an ongoing follow-up study. Additionally, our research revealed that pSS patients with SSA antibodies have elevated levels of CXCL9 expression in their MSG and higher levels of CXCL9 protein in their plasma. Earlier studies [66, 67] have shown that anti-SSA antibodies can trigger activated interferon (IFN) production by activating plasmacytoid dendritic cells, which are specialized immune cells that produce significant quantities of IFN. This can in turn result in the production of CXCL9 via MIG signaling. Therefore, there may be a correlation between anti-SSA in pSS and CXCL9 mediated by IFN signaling. Nevertheless, further investigation is necessary to clarify the precise mechanisms and implications of this connection.

Conclusions

Overall, these findings confirm that CXCL9 is elevated in pSS patients, particularly those with EGM and SSA antibodies. The way it is expressed may be a useful biomarker for measuring disease severity, suggesting that it could have a significant impact on the development of the disease. It could also be utilized as a biomarker or treatment target, but additional research is necessary to validate and fully understand these discoveries.

Availability of data and materials

Data used during the study can be obtained from a reasonable request to the corresponding author.

Abbreviations

ANA:

Antinuclear antibody

CXCL9:

C-X-C motif chemokine ligand 9

CXCR3:

C-X-C motif chemokine receptor 3

DEGs:

Differentially expressed genes

EGM:

Extra-glandular manifestations

ESR:

Erythrocyte sedimentation rate

ESSDAI:

EULAR Sjogren’s Syndrome Disease Activity Index

IgG:

Immunoglobulin G

ILD:

Interstitial lung disease

MSG:

Minor salivary gland

pSS:

Primary Sjögren's syndrome

SLE:

Systemic lupus erythematosus

References

  1. Mavragani CP, Moutsopoulos HM. Sjogren’s syndrome. Annu Rev Pathol. 2014;9:273–85.

    Article  CAS  PubMed  Google Scholar 

  2. Manfre V, Chatzis LG, Cafaro G, Fonzetti S, Calvacchi S, Fulvio G, et al. Sjogren’s syndrome: one year in review 2022. Clin Exp Rheumatol. 2022;40(12):2211–24.

    PubMed  Google Scholar 

  3. Seror R, Theander E, Bootsma H, Bowman SJ, Tzioufas A, Gottenberg JE, et al. Outcome measures for primary Sjogren’s syndrome: a comprehensive review. J Autoimmun. 2014;51:51–6.

    Article  PubMed  Google Scholar 

  4. Ter Borg EJ, Kelder JC. Development of new extra-glandular manifestations or associated auto-immune diseases after establishing the diagnosis of primary Sjogren’s syndrome : a long-term study of the Antonius Nieuwegein Sjogren (ANS) cohort. Rheumatol Int. 2017;37(7):1153–8.

    Article  PubMed  Google Scholar 

  5. Trevisani VFM, Pugliesi A, Pasoto SG, Lopes MLL, Guedes LKN, Miyamoto ST, et al. Recommendations for evaluation and diagnosis of extra-glandular manifestations of primary sjogren syndrome: results of an epidemiologic systematic review/meta-analysis and a consensus guideline from the Brazilian Society of Rheumatology (articular, pulmonary and renal). Adv Rheumatol. 2022;62(1):18.

    Article  PubMed  Google Scholar 

  6. Ramos-Casals M, Brito-Zeron P, Solans R, Camps MT, Casanovas A, Sopena B, et al. Systemic involvement in primary Sjogren’s syndrome evaluated by the EULAR-SS disease activity index: analysis of 921 Spanish patients (GEAS-SS Registry). Rheumatology (Oxford). 2014;53(2):321–31.

    Article  PubMed  Google Scholar 

  7. Manfre V, Cafaro G, Riccucci I, Zabotti A, Perricone C, Bootsma H, et al. One year in review 2020: comorbidities, diagnosis and treatment of primary Sjogren’s syndrome. Clin Exp Rheumatol. 2020;38 Suppl 126(4):10–22.

    PubMed  Google Scholar 

  8. Youinou P, Saraux A, Pers JO. B-lymphocytes govern the pathogenesis of Sjogren’s syndrome. Curr Pharm Biotechnol. 2012;13(10):2071–7.

    Article  CAS  PubMed  Google Scholar 

  9. Figgett WA, Deliyanti D, Fairfax KA, Quah PS, Wilkinson-Berka JL, Mackay F. Deleting the BAFF receptor TACI protects against systemic lupus erythematosus without extensive reduction of B cell numbers. J Autoimmun. 2015;61:9–16.

    Article  CAS  PubMed  Google Scholar 

  10. Klimatcheva E, Pandina T, Reilly C, Torno S, Bussler H, Scrivens M, et al. CXCL13 antibody for the treatment of autoimmune disorders. BMC Immunol. 2015;16(1):6.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Kramer JM, Klimatcheva E, Rothstein TL. CXCL13 is elevated in Sjogren’s syndrome in mice and humans and is implicated in disease pathogenesis. J Leukoc Biol. 2013;94(5):1079–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Yoon KC, De Paiva CS, Qi H, Chen Z, Farley WJ, Li DQ, et al. Expression of Th-1 chemokines and chemokine receptors on the ocular surface of C57BL/6 mice: effects of desiccating stress. Invest Ophthalmol Vis Sci. 2007;48(6):2561–9.

    Article  PubMed  Google Scholar 

  13. Yoon KC, Park CS, You IC, Choi HJ, Lee KH, Im SK, et al. Expression of CXCL9, -10, -11, and CXCR3 in the tear film and ocular surface of patients with dry eye syndrome. Invest Ophthalmol Vis Sci. 2010;51(2):643–50.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Zhang J, Zhang X, Shi X, Liu Y, Cheng D, Tian Q, et al. CXCL9, 10, 11/CXCR3 Axis Contributes to the Progress of Primary Sjogren’s Syndrome by Activating GRK2 to Promote T Lymphocyte Migration. Inflammation. 2023;46(3):1047–60.

    Article  CAS  PubMed  Google Scholar 

  15. Chiu YH, Szu-Hsien Lee T, Chao E, Chen IF, Liu FC, Chiu CH, et al. Application of classification criteria of Sjogren syndrome in patients with sicca symptoms: Real-world experience at a medical center. J Formos Med Assoc. 2020;119(1 Pt 3):480–7.

    Article  PubMed  Google Scholar 

  16. Daniels TE, Whitcher JP. Association of patterns of labial salivary gland inflammation with keratoconjunctivitis sicca. Analysis of 618 patients with suspected Sjogren’s syndrome. Arthritis Rheum. 1994;37(6):869–77.

    Article  CAS  PubMed  Google Scholar 

  17. Caporali R, Bonacci E, Epis O, Bobbio-Pallavicini F, Morbini P, Montecucco C. Safety and usefulness of minor salivary gland biopsy: retrospective analysis of 502 procedures performed at a single center. Arthritis Rheum. 2008;59(5):714–20.

    Article  PubMed  Google Scholar 

  18. Hjelmervik TO, Petersen K, Jonassen I, Jonsson R, Bolstad AI. Gene expression profiling of minor salivary glands clearly distinguishes primary Sjogren’s syndrome patients from healthy control subjects. Arthritis Rheum. 2005;52(5):1534–44.

    Article  CAS  PubMed  Google Scholar 

  19. Li N, Li Y, Hu J, Wu Y, Yang J, Fan H, et al. A link between mitochondrial dysfunction and the immune microenvironment of salivary glands in primary Sjogren’s syndrome. Front Immunol. 2022;13:845209.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Michael D, Soi S, Cabera-Perez J, Weller M, Alexander S, Alevizos I, et al. Microarray analysis of sexually dimorphic gene expression in human minor salivary glands. Oral Dis. 2011;17(7):653–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Zhu X, Lu S, Zhu L, Yu M, Wei T, Zhu X, et al. CXCR2 may serve as a useful index of disease activity in interstitial lung disease associated with primary Sjogren’s syndrome. Front Mol Biosci. 2021;8:640779.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Liu Z, Li F, Pan A, Xue H, Jiang S, Zhu C, et al. Elevated CCL19/CCR7 expression during the disease process of primary Sjogren’s syndrome. Front Immunol. 2019;10:795.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Li N, Li L, Wu M, Li Y, Yang J, Wu Y, et al. Integrated bioinformatics and validation reveal potential biomarkers associated with progression of primary Sjogren’s syndrome. Front Immunol. 2021;12:697157.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Shah NR, Noll BD, Stevens CB, Brennan MT, Mougeot FB, Mougeot JC. Biosemantics guided gene expression profiling of Sjogren’s syndrome: a comparative analysis with systemic lupus erythematosus and rheumatoid arthritis. Arthritis Res Ther. 2017;19(1):192.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Zhu T, Pan Z, Zhang N. Elevated CXCL13 in primary Sjogren’s syndrome and its correlation with disease activity: a systematic review and meta-analysis. Clin Rheumatol. 2022;41(9):2791–802.

    Article  PubMed  Google Scholar 

  26. Ogawa N, Ping L, Zhenjun L, Takada Y, Sugai S. Involvement of the interferon-gamma-induced T cell-attracting chemokines, interferon-gamma-inducible 10-kd protein (CXCL10) and monokine induced by interferon-gamma (CXCL9), in the salivary gland lesions of patients with Sjogren’s syndrome. Arthritis Rheum. 2002;46(10):2730–41.

    Article  CAS  PubMed  Google Scholar 

  27. Verstappen GM, Gao L, Pringle S, Haacke EA, van der Vegt B, Liefers SC, et al. The transcriptome of paired major and minor salivary gland tissue in patients with primary Sjogren’s syndrome. Front Immunol. 2021;12:681941.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Shiboski SC, Shiboski CH, Criswell L, Baer A, Challacombe S, Lanfranchi H, et al. American College of Rheumatology classification criteria for Sjogren’s syndrome: a data-driven, expert consensus approach in the Sjogren’s International Collaborative Clinical Alliance cohort. Arthritis Care Res (Hoboken). 2012;64(4):475–87.

    Article  CAS  PubMed  Google Scholar 

  29. Shiboski CH, Shiboski SC, Seror R, Criswell LA, Labetoulle M, Lietman TM, et al. 2016 American College of Rheumatology/European League against rheumatism classification criteria for primary Sjogren’s syndrome: a consensus and data-driven methodology involving three international patient cohorts. Arthritis Rheumatol. 2017;69(1):35–45.

    Article  PubMed  Google Scholar 

  30. Seror R, Bowman SJ, Brito-Zeron P, Theander E, Bootsma H, Tzioufas A, et al. EULAR Sjogren’s syndrome disease activity index (ESSDAI): a user guide. RMD Open. 2015;1(1):e000022.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Seror R, Ravaud P, Bowman SJ, Baron G, Tzioufas A, Theander E, et al. EULAR Sjogren’s syndrome disease activity index: development of a consensus systemic disease activity index for primary Sjogren’s syndrome. Ann Rheum Dis. 2010;69(6):1103–9.

    Article  PubMed  Google Scholar 

  32. Dong X, Zhou J, Guo X, Li Y, Xu Y, Fu Q, et al. A retrospective analysis of distinguishing features of chest HRCT and clinical manifestation in primary Sjogren’s syndrome-related interstitial lung disease in a Chinese population. Clin Rheumatol. 2018;37(11):2981–8.

    Article  PubMed  Google Scholar 

  33. Wu T, Hu E, Xu S, Chen M, Guo P, Dai Z, et al. clusterProfiler 4.0: A universal enrichment tool for interpreting omics data. Innovation (Camb). 2021;2(3):100141.

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Szklarczyk D, Gable AL, Nastou KC, Lyon D, Kirsch R, Pyysalo S, et al. The STRING database in 2021: customizable protein-protein networks, and functional characterization of user-uploaded gene/measurement sets. Nucleic Acids Res. 2021;49(D1):D605–12.

    Article  CAS  PubMed  Google Scholar 

  35. Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13(11):2498–504.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Luo J, Liao X, Zhang L, Xu X, Ying S, Yu M, et al. Transcriptome sequencing reveals potential roles of ICOS in primary Sjogren’s syndrome. Front Cell Dev Biol. 2020;8:592490.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Chen X, Jiang S, Zhou Z, Xu X, Ying S, Du L, et al. Increased expression of interleukin-21-inducible genes in minor salivary glands are associated with primary Sjogren’s syndrome disease characteristics. Rheumatology (Oxford). 2021;60(6):2979–89.

    Article  CAS  PubMed  Google Scholar 

  38. Huang L, Liu Q, Zhou T, Zhang J, Tian Q, Zhang Q, et al. Deficiency of beta-arrestin2 alleviates apoptosis through GRP78-ATF6-CHOP signaling pathway in primary Sjogren’s syndrome. Int Immunopharmacol. 2021;101(Pt A):108281.

    Article  CAS  PubMed  Google Scholar 

  39. Soret P, Le Dantec C, Desvaux E, Foulquier N, Chassagnol B, Hubert S, et al. A new molecular classification to drive precision treatment strategies in primary Sjogren’s syndrome. Nat Commun. 2021;12(1):3523.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Horeth E, Oyelakin A, Song EC, Che M, Bard J, Min S, et al. Transcriptomic and Single-cell analysis reveals regulatory networks and cellular heterogeneity in mouse primary Sjogren’s syndrome salivary glands. Front Immunol. 2021;12:729040.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Zhang S, Qu J, Wang L, Li M, Xu D, Zhao Y, et al. Activation of toll-like receptor 7 signaling pathway in primary Sjogren’s syndrome-associated thrombocytopenia. Front Immunol. 2021;12:637659.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Chen X, Cheng Q, Du Y, Liu L, Wu H. Differential long non-coding RNA expression profile and function analysis in primary Sjogren’s syndrome. BMC Immunol. 2021;22(1):47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Niederlova J, Koubek K. Chemokines and chemokine receptors. Review article. Sb Lek. 1999;100(3):169–89.

    CAS  PubMed  Google Scholar 

  44. Tokunaga R, Zhang W, Naseem M, Puccini A, Berger MD, Soni S, et al. CXCL9, CXCL10, CXCL11/CXCR3 axis for immune activation - a target for novel cancer therapy. Cancer Treat Rev. 2018;63:40–7.

    Article  CAS  PubMed  Google Scholar 

  45. Colvin RA, Campanella GSV, Sun JT, Luster AD. Intracellular domains of CXCR3 that mediate CXCL9, CXCL10, and CXCL11 function. J Biol Chem. 2004;279(29):30219–27.

    Article  CAS  PubMed  Google Scholar 

  46. Rabquer BJ, Tsou PS, Hou Y, Thirunavukkarasu E, Haines GK 3rd, Impens AJ, et al. Dysregulated expression of MIG/CXCL9, IP-10/CXCL10 and CXCL16 and their receptors in systemic sclerosis. Arthritis Res Ther. 2011;13(1):R18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Ferreira GA, Teixeira AL, Sato EI. Atorvastatin therapy reduces interferon-regulated chemokine CXCL9 plasma levels in patients with systemic lupus erythematosus. Lupus. 2010;19(8):927–34.

    Article  CAS  PubMed  Google Scholar 

  48. Oda K, Kotani T, Takeuchi T, Ishida T, Shoda T, Isoda K, et al. Chemokine profiles of interstitial pneumonia in patients with dermatomyositis: a case control study. Sci Rep. 2017;7(1):1635.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Zhang J, Zhang X, Shi X, Liu Y, Cheng D, Tian Q, et al. CXCL9, 10, 11/CXCR3 Axis Contributes to the Progress of Primary Sjogren’s Syndrome by Activating GRK2 to Promote T Lymphocyte Migration. Inflammation. 2023;46:1047.

    Article  CAS  PubMed  Google Scholar 

  50. Castrejon-Morales CY, Granados-Portillo O, Cruz-Bautista I, Ruiz-Quintero N, Manjarrez I, Lima G, et al. Omega-3 and omega-6 fatty acids in primary Sjogren’s syndrome: clinical meaning and association with inflammation. Clin Exp Rheumatol. 2020;38 Suppl 126(4):34–9.

    PubMed  Google Scholar 

  51. Lee KE, Mun S, Kim SM, Shin W, Jung W, Paek J, et al. The inflammatory signature in monocytes of Sjogren’s syndrome and systemic lupus erythematosus, revealed by the integrated Reactome and drug target analysis. Genes Genomics. 2022;44(10):1215–29.

    Article  CAS  PubMed  Google Scholar 

  52. Lundtoft C, Sjowall C, Rantapaa-Dahlqvist S, Bengtsson AA, Jonsen A, Pucholt P, et al. Strong association of combined genetic deficiencies in the classical complement pathway with risk of systemic lupus erythematosus and primary Sjogren’s syndrome. Arthritis Rheumatol. 2022;74(11):1842–50.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Imgenberg-Kreuz J, Almlof JC, Leonard D, Sjowall C, Syvanen AC, Ronnblom L, et al. Shared and unique patterns of DNA methylation in systemic lupus erythematosus and primary Sjogren’s syndrome. Front Immunol. 2019;10:1686.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Meiners PM, Arends S, Brouwer E, Spijkervet FK, Vissink A, Bootsma H. Responsiveness of disease activity indices ESSPRI and ESSDAI in patients with primary Sjogren’s syndrome treated with rituximab. Ann Rheum Dis. 2012;71(8):1297–302.

    Article  CAS  PubMed  Google Scholar 

  55. Shiboski CH, Shiboski SC, Seror R, Criswell LA, Labetoulle M, Lietman TM, et al. 2016 American College of Rheumatology/European League against rheumatism classification criteria for primary Sjogren’s syndrome: a consensus and data-driven methodology involving three international patient cohorts. Ann Rheum Dis. 2017;76(1):9–16.

    Article  PubMed  Google Scholar 

  56. Mariette X, Criswell LA. Primary Sjogren’s syndrome. N Engl J Med. 2018;379(1):97.

    PubMed  Google Scholar 

  57. Wang X, Zhang T, Guo Z, Pu J, Riaz F, Feng R, et al. The efficiency of hydroxychloroquine for the treatment of primary Sjogren’s syndrome: a systematic review and meta-analysis. Front Pharmacol. 2021;12:693796.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Lacotte S, Brun S, Muller S, Dumortier H. CXCR3, inflammation, and autoimmune diseases. Ann N Y Acad Sci. 2009;1173:310–7.

    Article  CAS  PubMed  Google Scholar 

  59. Nibbs RJB, Graham GJ. Immune regulation by atypical chemokine receptors. Nat Rev Immunol. 2013;13(11):815–29.

    Article  PubMed  Google Scholar 

  60. Gairy K, Knight C, Anthony P, Hoskin B. Burden of illness among subgroups of patients with primary Sjogren’s syndrome and systemic involvement. Rheumatology (Oxford). 2021;60(4):1871–81.

    Article  PubMed  Google Scholar 

  61. Brito-Zeron P, Acar-Denizli N, Ng WF, Horvath IF, Rasmussen A, Seror R, et al. Epidemiological profile and north-south gradient driving baseline systemic involvement of primary Sjogren’s syndrome. Rheumatology (Oxford). 2020;59(9):2350–9.

    Article  PubMed  Google Scholar 

  62. Foulquier N, Le Dantec C, Bettacchioli E, Jamin C, Alarcon-Riquelme ME, Pers JO. Machine learning for the identification of a common signature for anti-SSA/Ro 60 antibody expression across autoimmune diseases. Arthritis Rheumatol. 2022;74(10):1706–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Scopelitis E, Perez M, Biundo JJ Jr. Anti-SSA(Ro) antibody: a connective tissue disease marker. J Rheumatol. 1985;12(6):1105–8.

    CAS  PubMed  Google Scholar 

  64. Cui G, Sugai S, Ogawa Y, Takeshita S, Masaki Y, Fukutoku M. Long-term follow-up of 43 patients with Sjogren’s syndrome. Ryumachi. 1997;37(6):770–80.

    CAS  PubMed  Google Scholar 

  65. Thorlacius GE, Hultin-Rosenberg L, Sandling JK, Bianchi M, Imgenberg-Kreuz J, Pucholt P, et al. Genetic and clinical basis for two distinct subtypes of primary Sjogren’s syndrome. Rheumatology (Oxford). 2021;60(2):837–48.

    CAS  PubMed  Google Scholar 

  66. Imgenberg-Kreuz J, Sandling JK, Norheim KB, Johnsen SJA, Omdal R, Syvanen AC, et al. DNA methylation-based interferon scores associate with sub-phenotypes in Primary Sjogren’s syndrome. Front Immunol. 2021;12:702037.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Bjork A, Richardsdotter Andersson E, Imgenberg-Kreuz J, Thorlacius GE, Mofors J, Syvanen AC, et al. Protein and DNA methylation-based scores as surrogate markers for interferon system activation in patients with primary Sjogren’s syndrome. RMD Open. 2020;6(1):e000995.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

The research was funded by grants from the Wenzhou Science and Technology Plan Project (Y20210852); the Education Department Project of Zhejiang Province (Y202045501); the First Affiliated Hospital of Wenzhou Medical University (FHY2019093).

Author information

Authors and Affiliations

Authors

Contributions

JwH, XfZ, XbW, HC, PW and YzW were responsible for conducting most of the experiments, analyzing the data, and creating the figures and tables. In addition, XfZ, XbW, JwH, HC, PW, YzW, SsL and YZ collaborated to design the experiments and write the manuscript. The final version of the article was approved by all authors who made significant contributions.

Corresponding authors

Correspondence to Xiao bing Wang or Xiaofang Zhu.

Ethics declarations

Ethics approval and consent to participate

The study was approved by the the ethics committee of Wenzhou Medical University’s First Affiliated Hospital approved the study (#16024). All subjects provided written informed consent to participate in the study.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1: Table S1.

Clinical characteristics of patients with pSS and SLE.

Additional file 2: Table S2.

The correlation between top 20 DEGs and clinical features.

Additional file 3: Table S3.

The results of the Shapiro–Wilk test.

Additional file 4: Figure S1.

Association Between CXCL9 and ClinESSDAI score of Primary Sjögren's Syndrome. (A) The Spearman correlation between expression of CXCL9 gene in MSG and ClinESSDAI score. (B) The Spearman correlation between expression of CXCL9 gene in plasma and ClinESSDAI score. The top and right boxplot indicates the distribution. Dashed lines represent the mean value.

Additional file 5: Figure S2.

Differentially expressed CXCL9 between pSS patients with or without SSA antibodies. (A) Box plot illustrates the CXCL9 expression in MSGs among pSS patients with or without SSA antibodies. (B) Box plot showing the expression of CXCL9 between pSS patients with or without SSA antibodies in plasma. Significance determined by Wilcoxon's test.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hong, J., Cheng, H., Wang, P. et al. CXCL9 may serve as a potential biomarker for primary Sjögren’s syndrome with extra-glandular manifestations. Arthritis Res Ther 26, 26 (2024). https://doi.org/10.1186/s13075-023-03229-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13075-023-03229-x

Keywords