Skip to main content
Erschienen in: Clinical Rheumatology 4/2024

Open Access 20.02.2024 | ORIGINAL ARTICLE

Identification and verification of inflammatory biomarkers for primary Sjögren’s syndrome

verfasst von: Xiaodan Liu, Haojie Wang, Xiao Wang, Xiaodan Jiang, Yinji Jin, Ying Han, Zhihui Zhang

Erschienen in: Clinical Rheumatology | Ausgabe 4/2024

Abstract

Introduction

Primary Sjögren’s syndrome (pSS) is an autoimmune disease characterized by inflammatory infiltration, and dysfunction of the salivary and lacrimal glands. This research aimed to explore the disease pathogenesis and improve the diagnosis and treatment of pSS by mining inflammation-associated biomarkers.

Methods

Five pSS-related datasets were retrieved from the Gene Expression Omnibus (GEO) database. Inflammation-associated biomarkers were determined by the least absolute shrinkage and selection operator (LASSO) and support vector machines recursive feature elimination (SVM-RFE). Single sample gene set enrichment analysis (ssGSEA) was implemented to profile the infiltration levels of immune cells. Real-time quantitative PCR (RT-qPCR) verified the expression of biomarkers in clinical samples.

Results

Four genes (LY6E, EIF2AK2, IL15, and CXCL10) were screened as inflammation-associated biomarkers in pSS, the predictive performance of which were determined among three pSS-related datasets (AUC > 0.7). Functional enrichment results suggested that the biomarkers were involved in immune and inflammation-related pathways. Immune infiltration analysis revealed that biomarkers were notably connected with type 2 T helper cells, regulatory T cells which were significantly expressed between pSS and control. TESTOSTERONE and CYCLOSPORINE were predicted to take effect by targeting CXCL10 and IL15 in pSS, respectively.

Conclusion

Four inflammation-associated biomarkers (LY6E, EIF2AK2, IL15, and CXCL10) were explored, and the underlying regulatory mechanisms and targeted drugs associated with these biomarkers were preliminarily investigated according to a series of bioinformatics methods based on the online datasets of pSS, which provided a reference for understanding the pathogenesis of pSS.
Key Points
Inflammation-associated biomarkers (LY6E, EIF2AK2, IL15, and CXCL10) were firstly identified in Sjögren’s syndrome based on LASSO and SVM-RFE analyses.
CXCL10, EIF2AK2 and LY6E were prominently positively correlated with immature B cells, while IL15 were significantly negatively correlated with memory B cells in Sjögren’s syndrome.
LY6E, EIF2AK2, IL15, and CXCL10 were significantly more highly expressed in clinical Sjögren’s syndrome samples compared to healthy control samples, which was consistent with the analysis results of the GEO database.
LY6E, EIF2AK2, IL15, and CXCL10 might be used as the biomarkers for the treatment and diagnosis of Sjögren’s syndrome.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s10067-024-06901-y.
Xiaodan Liu and Haojie Wang contribute equally to this work.

Publisher's Note

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

Introduction

Primary Sjögren’s syndrome (pSS) is a complex and heterogeneous autoimmune disease that leads to secretory gland dysfunction. It causes dryness of the main mucosal surfaces such as the mouth, eyes, nose, pharynx, larynx, and vagina, mainly characterized by sicca symptoms (xerostomia and xerophthalmia) [1], which can have a major impact on quality of life, including dry eye, reduced salivary flow rates, an increased risk of dental caries, and oral candidiasis [2, 3]. Approximately 20–40% of patients with pSS may experience extraglandular involvement [4], and among them lymphoma is the leading cause of death [5]. In the 2016 American College of Rheumatology (ACR)/European League Against Rheumatism (EULAR) classification criteria, serological (Anti-Ro/SSA) and histological examinations (labial salivary gland biopsy) were assigned the highest specificity and highest values [6]. However, anti-SSA antibodies can be indicative of a more advanced stage of the disease, and relying on them alone for diagnosis may result in inadequate recognition of very early pSS [7]. Labial salivary gland biopsy is an invasive examination, which may possibly cause discomfort and complications [8], and also may be affected by the subjective judgment of specimen observers [9]. Therefore, it is necessary to develop sensitive and specific biomarkers to assist the early diagnosis of pSS.
Host inflammatory responses are essential for the development and progression of pSS and are regulated by various signaling pathways, such as pro-inflammatory cytokines and interferon [10, 11], and the use of anti-inflammatory treatments has been reported to provide relief from symptoms associated with pSS [12]. At present, local tear and saliva substitutes, systemic secretagogues and immunosuppressants (glucocorticoids, chloroquine/hydroxychloroquine chloroquine and methotrexate) are commonly used treatments for pSS; however, their effectiveness is rarely seen in practice [1315]. Targeted treatment for pSS is still unavailable despite continued research into the disease’s pathogenesis, which may be due to the lack of systematic research on targeted biomarkers. Previous studies have uncovered a substantial amount of differentially expressed genes in the SS peripheral blood sample dataset [16]. Our study sought to investigate targeted inflammation-associated biomarkers through multiple bioinformatics pathways.
Given the limitations of anti-SSA antibody detection (delay and non-specificity) and labial salivary gland biopsy (invasiveness and subjectivity) in early pSS diagnosis, regulating the inflammatory response and employing anti-inflammatory therapy have emerged as crucial management strategies for pSS [1012]. Further, considering sensitivity and specificity of biomarker detection in serum, saliva, tears, or urine can potentially provide a more prompt and accurate reflection of the disease’s presence and progression, it has the potential to enhance the diagnostic accuracy of pSS and offer improved prospects for the treatment and intervention in early pSS. Hence, in this study, the inflammation-associated biomarkers with diagnostic value for pSS were filtered through two classical machine learning algorithms. The diagnostic value for the biomarkers was confirmed, and the biomarkers-related underlying mechanisms in pSS were initially investigate. Relevance analysis of inflammation-associated biomarkers and immune cell infiltration were performed. Moreover, the regulatory networks targeting the biomarkers were investigated, and the biomarkers-targeted drugs were predicted. Based on the five pSS-related online datasets containing the transcriptional expression profiles of whole peripheral blood samples, we make the case that the research could provide a basis for understanding disease pathogenesis and improving clinical diagnosis and treatment.

Materials and methods

Datasets and gene collection

Five pSS-related datasets were downloaded from the GEO database, namely the GSE51092, GSE66795, GSE84844, GSE145065, and GSE132842 datasets. The microarray datasets of GSE51092 contained the transcriptional expression profile of whole peripheral blood samples from 32 healthy controls and 190 pSS patients and was utilized to screen inflammatory-associated biomarkers, immune infiltration analysis, and single-gene GSEA analysis. Two microarray expression profiling datasets of pSS were employed to validate the expression and diagnostic value of inflammatory-associated biomarkers, that is, GSE66795 and GSE84844. GSE66795 dataset included a transcriptional expression profile of whole peripheral blood samples from 29 healthy controls and 131 pSS patients. The GSE84844 dataset comprised the transcriptional expression profiles of whole blood samples from 30 healthy controls and 30 pSS patients. The information of age and gender of patients and controls within the three pSS-related datasets above was exhibited in Table 1. Furthermore, RNA sequencing (RNA-seq) data from GSE145065 dataset, consisting of mRNA and lncRNA expression profiles of peripheral blood monocytes from 5 healthy controls and 5 pSS patients, was used for differential lncRNA screening. The GSE132842 dataset comprising miRNA expression profiles of CD1c-expressing cDC2s isolated from peripheral blood from 6 healthy controls and 15 pSS patients was detected through TaqMan OpenArray Human MicroRNA Panel and was used for differential miRNA screening. Two-hundred inflammation-associated genes were derived from the MSigDB database by searching the inflammation-associated gene set in the Hallmark gene set with the keyword ‘Inflammatory’ (Supplementary Table 1).
Table 1
The clinical information in the three pSS-related datasets
 
GSE51092
GSE66795
GSE84844
Overall
 
(N = 222)
(N = 160)
(N = 60)
(N = 442)
Group
  Control
32 (14.4%)
29 (18.1%)
30 (50.0%)
91 (20.6%)
  pSS
190 (85.6%)
131 (81.9%)
30 (50.0%)
351 (79.4%)
Age
  [20, 30]
0 (0%)
0 (0%)
7 (11.7%)
7 (1.6%)
  [30, 40]
0 (0%)
0 (0%)
10 (16.7%)
10 (2.3%)
  [40, 50]
0 (0%)
0 (0%)
15 (25.0%)
15 (3.4%)
  [50, 60]
0 (0%)
0 (0%)
10 (16.7%)
10 (2.3%)
  [60, 70]
0 (0%)
0 (0%)
12 (20.0%)
12 (2.7%)
  [70, 80]
0 (0%)
0 (0%)
6 (10.0%)
6 (1.4%)
Missing
222 (100%)
160 (100%)
0 (0%)
382 (86.4%)
Gender
  Female
0 (0%)
160 (100%)
59 (98.3%)
219 (49.5%)
  Male
0 (0%)
0 (0%)
1 (1.7%)
1 (0.2%)
  Missing
222 (100%)
0 (0%)
0 (0%)
222 (50.2%)

Certification of candidate genes for inflammation-associated biomarkers in pSS

The ‘limma’ package (version 3.50.0) was used to authenticate the differentially expressed genes (DEGs) between pSS samples and healthy controls in the GSE51092 dataset, defined by |log2FoldChange (FC) > 0.5| and p-value < 0.05 [17, 18]. The pSS-related genes were then filtered by Weighted Gene Co-expression Network Analysis (WGCNA) in the GSE51092 dataset. The R package ‘WGCNA’ (version 1.7–3) [19] was implemented to generate a co-expression network. The determination of the soft threshold firstly ensures that the interaction between genes conforms to the scale-free distribution to the maximum extent. Through gene adjacency calculations and assessing gene similarity, the introduction of topological overlap matrix (TOM) allows for the construction of a systematic clustering tree. Further, the dynamic tree cutting method was employed to assign genes to modules under hierarchical clustering, setting the minimum number of genes per gene module to 150. In addition, the pSS samples and healthy controls were considered as trait data for WGCNA to retrieve modules and genes associated with pSS using correlation analysis. The intersection of DEGs, pSS-related genes, and inflammation-associated genes was obtained from a Venn diagram and was incorporated into the subsequent analyses as candidate genes for inflammation-associated biomarkers in pSS.

Functional annotation analysis

The default gene set in R package ‘clusterProfiler’ (version 4.2.1) [20] was applied as background gene set for Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis of candidate genes. GO was categorized into cellular component (CC), molecular function (MF), and biological process (BP). An adjusted p-value < 0.05 was considered statistically significant.

Recognition of inflammation-associated biomarkers in pSS

In the GSE51092 dataset, two machine learning methods were applied to screen for disease characteristic genes, namely least absolute shrinkage and selection operator (LASSO) [21] and support vector machines recursive feature elimination (SVM-RFE) [22]. LASSO logistic regression was performed with the R software package ‘glmnet’ (version 4.0–2), setting the parameters family as binomial and type.measure as class. The error rate with different features was measured using tenfold cross-validation. By adjusting the penalty coefficient lambda, the majority of variable coefficients are eventually forced to converge to 0. The optimal lambda value is selected when the minimal error and the strong relevant features were selected. Support vector machine (SVM) analysis was carried out using the SVM in R package ‘e1071’ (version1.7–9). The best feature subset is determined by evaluating the model’s performance as the features are progressively reduced. Specifically, the recursive feature elimination (RFE) method was deployed to obtain the importance ranking of each gene, as well as the error rate and accuracy rate of each iteration of the combination. The features are gradually reduced until achieving the highest classification accuracy and lowest error rate under the feature subset size should not exceed the predefined maximum value, and the corresponding gene was extracted as the feature gene. The genes identified by both LASSO and SVM-RFE were defined as inflammation-associated biomarkers in pSS.

Relevance analysis of inflammation-associated biomarkers and immune cell infiltration

The relative infiltration levels of 28 types of immune cells in 32 healthy controls and 190 pSS samples in the GSE51092 dataset were profiled by the ssGSEA algorithm, which was run in the ‘GSVA’ package (version 1.38.0) [23]. Variations in the infiltration levels of different immune cells between the normal and pSS samples were estimated using a Wilcoxon test, and the results were visualized by a violin plot created by the ‘vioplot’ package (version 0.3.7). The correlations between immune cells and correlations between biomarkers and differential immune cells were assessed by the Pearson method.

Gene set enrichment analysis based on a single gene

The ‘h.all.v6.2.sytmbols.gmt’ in the MSigDB database was extracted to act as the reference gene set, and the pSS samples in the GSE51092 dataset were assigned to high- and low-expression groups with the median as the cut-off value of each key gene for calculating the fold change of gene expression between the high and low expression groups and ranking them. Furthermore, GSEA was performed to investigate the differences in gene set enrichment between the high and low expression groups using the R software ‘clusterProfiler’ (version 4.0.5) package. Significance thresholds of single gene GSEA were |NES|> 1, q value < 0.2, and p value < 0.05.

Establishment of lncRNA-miRNA-mRNA network and drug-gene network

The miRNAs targeting the inflammation-associated biomarkers and the lncRNAs targeting miRNAs were predicted by the StarBase database (screening condition: CLIP-DATA ≥ 1). Under the screening criteria of |log2FC > 1| and p-value < 0.05, the predicted miRNAs were crossed with the differentially expressed miRNAs (DE-miRNAs) between the pSS samples and healthy controls in the GSE132842 dataset. Similarly, the predicted lncRNAs were crossed with the differentially expressed lncRNAs (DE-lncRNAs) between the pSS samples and healthy controls in the GSE145065 dataset. Moreover, the drugs targeting the inflammation-associated biomarkers were forecasted in the DGIdb database. After inputting the biomarkers, drugs related to the treatment of inflammation were obtained from the extracted drug-gene interaction information. The final lncRNA-miRNA-mRNA regulatory network and gene-drug network was mapped by Cytoscape software, where each node is presented as lncRNA, miRNA, mRNA or drug, and the edge is presented as the interaction between them in a visual way (version 3.8.2) [24].

RNA acquisition and real-time quantitative PCR (RT-qPCR)

A total of 10 pSS patients and 10 healthy control patients were recruited from the Peking University Third Hospital with PBMC samples to perform RT-qPCR experiments. The pSS patients fulfilled the 2016 American College of Rheumatology (ACR)/European League Against Rheumatism (EULAR) classification criteria [6]. The detailed clinical information of the patients involved is shown in Supplementary Table 2. This study was approved by the Peking University Third Hospital Medical Science Research Ethics Committee (IRB00006761-M2022106), written informed consent was received from all participants for their enrollment, and all methods were carried out in accordance with relevant guidelines and regulations. The total RNA of PBMC samples from 10 healthy control and 10 pSS patients was isolated by the TRIzol Reagent following the manufacturer’s guidance (Ambion, USA). Next, total RNA was inversely transcribed into cDNA utilizing the SweScript-First-strand-cDNA-synthesis-kit (Servicebio, China), according to the manufacturer’s protocol. qPCR was subsequently performed using the 2xUniversal Blue SYBR Green qPCR Master Mix (Servicebio, China). The primer sequences for PCR are displayed in Table 2. The relative expression level was uniformized to the internal reference GAPDH and calculated using the 2−ΔΔCq method [25].
Table 2
The primer sequences for RT-qPCR
Primer
Sequence
LY6E For
CTGTACTGCCTGAAGCCGA
LY6E Rev
CCATGGAAGCCACACCAAC
EIF2AK2 For
GCCGCTAAACTTGCATATCTTCA
EIF2AK2 Rev
TCACACGTAGTAGCAAAAGAACC
IL15 For
ATGAAGTGCTTTCTCTTGGAGT
IL15 Rev
GAAGTGTTGATGAACATTTGGA
CXCL10 For
TTCTGATTTGCTGCCTTATCTTTC
CXCL10 Rev
CTTCTCACCCTTCTTTTTCATTGT
GAPDH For
GGAAGGTGAAGGTCGGAGT
GAPDH Rev
TGAGGTCAATGAAGGGGTC

Statistical analysis

Violin plots of gene expression were produced by the R package ‘ggstatsplot’ (version 0.9.1). ROC curves were generated by the ‘pROC’ package (1.17.0.1). All analyses were conducted using the R programming language, and the data from different groups were compared by the Wilcoxon test. The Student’s t-test was utilized to filter for DE-miRNAs and compare the differences in RT-qPCR. If not specified above, a p-value less than 0.05 was considered statistically significant.

Results

Candidate genes for inflammation-associated biomarkers in pSS

The workflow diagram for the current study was displayed in Supplementary Fig. 1. To determine the differentially expressed pSS-related genes, the DEGs between pSS and healthy controls in the GSE51092 dataset were first authenticated. According to |log2FC|> 0.5 and p-value < 0.05, a grand total of 282 DEGs, including 165 upregulated and 117 downregulated genes were identified in the pSS samples (Fig. 1A–B, Supplementary Table 3). Then, WGCNA was implemented using the data of the GSE51092 dataset. Firstly, no outlier samples were excluded by sample cluster analysis (Supplementary Fig. 2A). Nine were chosen as the optimal soft threshold (R2 = 0.85) to ensure that the interactions between genes maximally conform to the scale-free distribution (Fig. 2A). Next, a total of 11 modules were developed based on a gene clustering tree and dynamic tree cutting algorithm (Fig. 2B, Supplementary Fig. 2B). Correlations between modules and sample traits (healthy control or disease pSS) were computed, and the purple module with the highest correlation was selected as the key module (Fig. 2C). Hence, the 459 genes in the key module were regarded as pSS-related genes (Supplementary Table 4). Subsequently, the DEGs, pSS-related genes, and inflammation-associated genes were overlapped, resulting in nine intersecting genes (Fig. 2D), namely LY6E, EIF2AK2, IRF7, TNFAIP6, RTP4, IL15, CXCL10, LAMP3, and CCL2. These genes were considered the candidate genes for inflammation-associated biomarkers in pSS.
To further investigate the function of these nine genes, a functional enrichment analysis was executed. As displayed in Supplementary Table 5, 180 GO items (162 BP items and 18 MF items) and 19 KEGG pathways (Fig. 3A) were derived. The top 10 GO items under each classification were displayed in a bar chart (Fig. 3B). These genes were involved in many immune-related biological processes and pathways, including ‘response to interferon-alpha’, ‘cytokine-mediated signaling pathway’, ‘regulation of lymphocyte migration’, ‘cytokine-cytokine receptor interaction’, ‘TNF signaling pathway’, ‘RIG-I-like receptor signaling pathway’, ‘Toll-like receptor signaling pathway’, ‘NOD-like receptor signaling pathway’, ‘IL-17 signaling pathway’, and ‘chemokine signaling pathway’.

Inflammation-associated biomarkers in pSS

To further recognize inflammation-associated biomarkers in pSS, a machine learning analysis based on the nine candidate genes was performed. As shown in Fig. 4A–B, the lowest error rate was reached at lambda.min of 0.0038, and five genes (LY6E, EIF2AK2, TNFAIP6, IL15, and CXCL10) were identified by LASSO logistic regression. Meanwhile, six genes (IL15, CXCL10, EIF2AK2, IRF7, LY6E, and CCL2) were selected by the SVM-RFE model (Fig. 4C–D). Hence, four overlapping genes (LY6E, EIF2AK2, IL15, and CXCL10) were obtained by comparing the genes obtained by the two machine learning methods (Fig. 4E). In the GSE51092 dataset, all four genes were expressed at increased levels in pSS samples compared to healthy controls (Fig. 5A). The ROC curves of the four genes in the GSE51092 dataset were mapped to further estimate the potential diagnostic value of the genes. As the AUC values all exceeded 0.7 for each gene, we concluded that the expression of each gene could effectively distinguish pSS samples from healthy controls (Fig. 5D). Furthermore, the expression of the four genes was examined, and the corresponding ROC curves for the GSE66795 and GSE84844 datasets were created to validate the above results. In agreement with the results of the GSE51092 dataset, the four genes were upregulated in the pSS samples compared to the healthy controls (Fig. 5B–C). Also, the AUC values of the ROC curves were all greater than 0.7, indicating that these four genes were reliable potential diagnostic biomarkers (Fig. 5E–F). Therefore, these four genes were defined as inflammation-associated biomarkers in pSS. To gain a preliminary understanding of the functions of which, the Panther classification system (http://​pantherdb.​org/​) was used to annotate the four biomarkers by GO and Pathway function. The annotated GO items and pathways are shown in Supplementary Fig. 3, indicating that the four biomarkers were linked to ‘inflammation mediated by chemokine and cytokine signaling pathway’, ‘interleukin signaling pathway’, and ‘apoptosis signaling pathway’.
Subsequently, the single-gene GSEA was performed based on the hallmark gene set and transcriptomic data of pSS samples in the GSE51092 dataset to explore the molecular mechanisms of each inflammation-associated biomarker in pSS. As revealed in Fig. 6, all four genes were linked to the activation of ‘HALLMARK_INTERFERON_GAMMA_RESPONSE’, and ‘HALLMARK_INTERFERON_ALPHA_RESPONSE’, in the meantime, the inhibition of ‘HALLMARK HEME METABOLISM’ should be possible to related to EIF2AK2, IL15 and LY6E. More details could be found in the Supplementary Table 6.

The relevance of inflammation-associated biomarkers to immune cells in pSS

To further probe the relationship between inflammation-associated biomarkers and immune cells in pSS, the infiltration level of 28 types of immune cells between pSS samples and healthy controls in the GSE51092 dataset were first compared through the ssGSEA method. The scores for each immune infiltrating cell in each sample are shown in the heatmap (Fig. 7A). The violin plot suggested that the enrichment fraction of CD56 bright natural killer cells, CD56 dim natural killer cells, plasmacytoid dendritic cells, memory B cells, type 1 T helper cells, immature B cells, immature dendritic cells, natural killer cells, regulatory T cells, type 17 T helper cells, and type 2 T helper cells had significant differences between healthy controls and pSS samples (Fig. 7B), moreover, the plasmacytoid dendritic cells were closely relevant to type 1 T helper cells (cor = 0.54) (Fig. 7C). Then, the Pearson method was employed to compute the correlation between biomarkers and differential immune cells. As illustrated in Supplementary Table 7 and Fig. 7D–G, all four biomarkers were notably positively correlated with regulatory T cells and type 2 T helper cells as defined by |correlation coefficient|> 0.3 and p value < 0.05. Meanwhile, CXCL10, EIF2AK2 and LY6E were prominently positively correlated with immature B cells, while IL15 were significantly negatively correlated with memory B cells and plasmacytoid dendritic cells, which were consistent with CXCL10 as well.

The lncRNA-miRNA-mRNA network and gene-drug network based on inflammation-associated biomarkers in pSS

The DE-miRNAs between the pSS and healthy controls in the GSE132842 dataset were screened to investigate the upstream regulatory mechanisms of inflammation-associated biomarkers. A total of 12 DE-miRNAs were mined according to the screening conditions p value < 0.05 and |log2FC|> 1, of which 2 were highly expressed, and 10 showed low expression in pSS samples (Supplementary Fig. 4A–B). Meanwhile, 311 miRNAs targeting inflammation-associated biomarkers were predicted from the StarBase database. Six target miRNAs were obtained by intersecting with the 12 DE-miRNAs (Supplementary Fig. 4C). Next, the DE-lncRNAs between pSS and healthy controls in the GSE145065 dataset were screened. 63 DE-lncRNAs were identified based on the screening criteria p-value < 0.05 and |log2FC|> 1, of which 37 were highly expressed and 26 were lowly expressed in pSS (Supplementary Fig. 4D–E). Furthermore, 197 lncRNAs targeting the target miRNAs were predicted from the StarBase database. Then, 3 target lncRNAs were obtained by intersecting with the 63 DE-lncRNAs (Supplementary Fig. 4F). Finally, a lncRNA-miRNA-mRNA network with 13 nodes and 13 edges was generated using Cytoscape (Fig. 8A). In this network, hsa-miR-26-5p and hsa-miR-9-5p regulated EIF2AK2. CXCL10 was regulated by hsa-miR-21-5p, which was regulated by AL136040.1 and LINC02381. Moreover, LY6E was regulated by hsa-miR-708-5p, which was regulated by AL157392.3. IL15 was regulated by hsa-miR-30d-5p, hsa-miR-708-5p, and hsa-let-7f-5p, which were regulated by AL157392.3. In addition, hsa-let-7f-5p was regulated by LINC02381.
To explore potential drugs targeting the four inflammation-associated markers, 17 drugs targeting three biomarkers were predicted through the DGIdb database. Ultimately, a disease-gene-drug network containing 21 nodes and 21 edges was constructed (Fig. 8B). In this network, CYCLOSPORINE, SIROLIMUS and AMG714 might be associated with IL15. Furthermore, 12 drugs (ATORVASTATIN, METHYLPREDNISOLONE, TESTOSTERONE, etc.) were predicted to target CXCL10.

Verification of the expression of inflammation-associated biomarkers in clinical samples

As illustrated in Fig. 5A–C, all four inflammation-associated biomarkers were upregulated in pSS samples compared to healthy controls. The expression in clinical PBMC samples from 10 healthy controls and 10 pSS patients was further confirmed by RT-qPCR. In agreement with the results of the analysis of public RNA-sequencing data, four biomarkers were significantly more highly expressed in clinical pSS samples compared to healthy control samples (Fig. 9, Table 3).
Table 3
The statistic results of four inflammation-associated biomarkers in clinical samples
 
N
pSS
FC
t, df value
p value
LY6E
1.0785 ± 0.1054
3.5786 ± 1.6249
3.318127028
t = 4.855 df = 18
0.0001
EIF2AK2
1.0471 ± 0.0298
3.7453 ± 1.2776
3.576831248
t = 2.266 df = 9
 < 0.0001
IL15
1.0352 ± 0.0329
2.8646 ± 0.7050
2.767194745
t = 8.196 df = 9
 < 0.0001
CXCL10
1.0263 ± 0.0110
2.5598 ± 0.5605
2.494202475
t = 8.622 df = 9
 < 0.0001
FC, fold change

Discussion

pSS is characterized by chronic inflammation and is manifested by impaired function of the exocrine glands, and mononuclear cells infiltrate surrounding the ducts and replacing the secretory units of the involved glands [26]. Due to the heterogeneities of clinical phenotypes and various causes, the identification of key biomarkers in pSS is critical to understanding the pathogenesis of this complex disease. Considering the biological significance of inflammatory response in pSS progress, the study was utilized to identify the potential inflammation-associated biomarkers through the bioinformatics methods based on the online datasets of pSS.
Using the differentially expressed analysis between pSS samples and healthy controls, as well as WCGNA in the GSE51092 datasets, nine pSS-related and inflammation-associated DEGs were identified in the present study, namely LY6E, EIF2AK2, IRF7, TNFAIP6, RTP4, IL15, CXCL10, LAMP3, and CCL2, which were mainly involved in the activation of the innate antiviral immunity process and inflammatory-related signaling pathways. Evidence has indicated that viral infections alter the clinical manifestations of various autoimmune diseases. On the other hand, protective effects can be achieved by suppressing autoimmune phenomena through regulatory immune responses [27]. Influenza viruses and EBV infection were considered as central roles in the pathogenesis of pSS through the autoimmunity induced by different mechanisms in previous literature [2833].
Several functions relevant to the pSS-related inflammation-associated DEGs were related to immune and inflammation signaling pathways. Toll-like receptors (TLRs) could sense nucleic acids derived from viruses and trigger antiviral innate immune responses as pattern-recognition receptors (PRRs) [34, 35], where NF-kappaB, MAPK kinases, and IRFs that control the transcription of genes encoding type I interferon and other inflammatory cytokines were activated to eliminate viruses [36]. Previous studies have shown that TLRs play an essential role in the pathogenesis of pSS [37, 38]. They are elevated in salivary tissue [39] and in the peripheral blood of pSS patients [40]. Emerging data indicate that damage-associated molecular patterns (DAMPs) may be significant drivers of chronic and unremitting inflammation in pSS, although the ligands activating TLRs in pSS remain unknown [41, 42]. Activating TLR signaling cascades likely reduce local and systemic inflammation, as shown in an animal study [43]. There is no doubt that the interaction of the Toll-like signaling pathways and the viral defense response process may be important in pSS.
Nod-like receptor protein 3 (NLRP3) is a crucial player in regulating host immune responses to infection and cells stress [44], and it was also found highly expressed in pSS patients than control [45]. The NLRP3 inflammasome can be triggered by the P2X7 receptor (P2X7R), leading to acute inflammatory responses. Baldini et al. proposed the P2X7R-inflammasome axis as a novel potential pathway in both pSS exocrinopathy and lymphomagenesis [46]. These results suggested that NLRP3 inflammasome-mediated inflammation might be implicated in the pathogenesis of pSS. Interleukin-17 (IL-17) is a multifaceted cytokine with a well-recognized role in immune surveillance at mucosal and barrier surfaces [47]. Previous research suggests that the IL-17 axis plays a pivotal role in the pathogenesis of several autoimmune disorders, including pSS [48, 49]. Studies have demonstrated that IL-17 was overexpressed in the salivary glands (SGs) [50], serum [51], plasma [52] and tears [53] of pSS patients, and IL-17 mRNA levels in MSG biopsies seemed to be related to the degree of inflammation [52, 54]. Different IL-17 family members may play several pathogenetic roles in the development of pSS. According to a recent study, IL-17F production in pSS patients is associated with a higher level of autoantibodies and EULAR SS disease activity index (ESSDAI) than IL-17A production in pSS patients [55].
Next, four genes (LY6E, EIF2AK2, IL15, and CXCL10) were authenticated as inflammation-associated pSS biomarkers, and the reliability of them in discriminating pSS samples from healthy control samples, suggesting a potential clinical diagnostic value. Functional enrichment results and immune infiltration analysis pointed to the involvement of the four genes in the immune process and inflammation-related pathways in pSS. The Lymphocyte antigen 6E (LY6E) protein belongs to the Ly6/uPAR family of plasminogen activator receptors and is known as one of the IFN type I response genes. Recent studies have reported its essential role in immunological regulation, T cells physiology, oncogenesis, and viral infection [56]. Our study found higher LY6E levels in the peripheral blood of pSS patients, which has been proven by previous clinical studies [5759]. These findings may reveal the importance of the peripheral blood LY6E levels and the monocyte IFN type I signature in pSS patients. The eukaryotic translation initiation factor 2-α kinase 2 (EIF2AK2) gene is located on chromosome 2 and encodes modifying protein kinase R (PKR, interferon-induced, double-stranded RNA-activated protein kinase) [60]. Recent studies have revealed that the coding gene PKR is associated with the treatment of pSS, which further confirms the role of EIF2AK2 in the progression of pSS [39, 61, 62]. Although LY6E and EIF2AK2 have been found as pSS diagnostic genes in previous studies [57], we further investigated the potential ceRNA regulatory network and related drugs of LY6E and EIF2AK2 in the context of inflammation, providing insight into the direction for future research. Interleukin-15 (IL-15) is a crucial regulatory inflammatory cytokine that is upregulated in autoimmunity disorders [63, 64]. Previous studies revealed a higher IL-15 expression level in the peripheral blood of pSS patients [65], which is consistent with our results. Besides, based on gene and protein analysis and immunohistochemical results in minor salivary gland (MSG) biopsy specimens and human salivary gland epithelial cells (SGEC) obtained from patients with pSS, IL15 was documented a strong expression in acinar and duct cells of salivary glands with pSS, which may be related to TLR2/IL-15 signaling pathway [6669]. It’s consistent with our functional enrichment results (Toll-like receptor signaling pathway), which provides a theoretical basis for the detection of pSS by blood, but the protein levels in blood need further analyses. C-X-C motif chemokine ligand 10 (CXCL10) protein is categorized functionally as a Th1-chemokine, and its secretion is regulated by interferon (IFN)-γ [70]. The serum and/or tissue expressions of CXCL10 in various autoimmune diseases [7073]. A study that assessed CXCL10 plasma levels in pSS patients showed that the ratio of full-length (active) CXCL10 to truncated DPP4-truncated (inactive) CXCL10 was significantly increased in pSS patients and provided the highest correlation with disease activity [74]. Elevated CXCL10 levels were also found in the salivary gland of pSS patients, which were associated with decreased circulating CXCR3 + helper cells, suggesting facilitating their concerted migration [75]. These results guarantee the accuracy of our transcriptome analysis results.
The pathogenesis of pSS is multifactorial and complex. The process primarily encompasses antigen presentation, costimulation, B cell activation, and other related mechanisms [11], in which the cytokine profiles of Th1, Th2, Th17, follicular helper T (Tfh) cells, and regulatory cells (Tregs/Bregs) play important roles [76]. Studies have shown that the frequency of Foxp3 + regulatory T cells (Treg) in salivary glands may be correlated with glandular infiltration and the grade of local inflammation [77], while B cell activation is generally associated with an increased risk of lymphoma [78]. Lymphocytic infiltration in salivary and lacrimal glands and the deposition of autoantibodies, like anti-SS-A (anti-Ro) and anti-SS-B (anti-La), cause an autoimmune outbreak and chronic inflammation, leading to the destruction of the salivary gland architecture [79].
In this study, we found that four key genes were significantly associated with regulatory T (Treg) cells and type 2 T helper (Th2) cells via immune infiltration and Pearson correlation analysis. Treg cell deficiency has been documented in pSS patients [80], with peripheral blood levels significantly lower than those of healthy controls, suggesting that Treg cell deficiency may be involved in salivary gland destruction [81]. Type 2 immune response which Th2 cells involved in has a regulatory relationship with autoinflammation [82]. Th2 cells have been found to promote renal inflammation in patients with systemic lupus erythematosus [83], and to play a part in the process of pSS by participating in costimulation and assisting B cell activation, with the cytokines they produce dominating the early stages of pSS [84]. These findings demonstrate that significant changes occur in Treg cells and Th2 cells in pSS and other related autoimmune diseases.
The identified miRNAs in the present study exhibited consistency with other research on autoimmune or immune-mediated related diseases. The miR-26 expression level was downregulated in multiple sclerosis (MS) patients compared to controls [85]. The neuroregulatory miRNA miR-9-5p was significantly upregulated in the peripheral blood samples of HLA-B27( +) radiographic axial spondyloarthropathy (rad-AxSpA) patients [86]. Immuno-miRNAs miR-21-5p and let-7f-5p were significantly elevated in the serum of patients with acetylcholine receptor myasthenia gravis (AChR+-MG) [8790], and miR-21-5p was also upregulated in type 1 autoimmune pancreatitis (AIP) [91] and psoriatic arthritis (PsA) [92]. Kim et al. found significantly downregulated expression of miR-30d-5p in the tear samples of pSS patients [93]. These miRNAs may be involved in disease pathogenesis via immune-related processes.
In our study, three lncRNAs were identified as being associated with pSS, namely AL 136040.1, LINC02381, and AL157392.3. Previous reports have suggested that these three lncRNAs may be implicated in immunological disorders. The competitive binding of LINC02381 with miR-21 has been experimentally confirmed in previous studies. Zhao et al. demonstrated this interaction through luciferase reporter gene and RNA immunoprecipitation assays, indicating that LINC02381 sponged miR-21 to enhance KLF12 expression [94]. However, the interaction between miR-21 and LINC02381/CXCL10 still requires further validation through additional functional experiments. Additionally, Jafarzadeh et al. also confirmed LINC02381 sponged miR-21 through dual luciferase assay [95]. LINC02381/hsa-let-7f-5p/IL-6 competitive network in another immune-mediated connective tissue disease systemic sclerosis (SSc) was shown to be potentially involved in inflammatory and immune processes immune microenvironmental variation [96]. Glycolysis-associated lncRNA AL157392.3 may influence immune-related signaling in pan-cancer analysis [97].
Additionally, we predicted potential drugs based on drug-gene interaction pairs, which included glucocorticoids and immunosuppressive drugs. These drugs have been successfully used to treat autoimmune diseases. AMG-714 was used to treat celiac disease [98], which is known as an associated autoimmune disease with pSS sharing a common genetic background [99]. LEVODOPA is an effective and well-tolerated drug for the treatment of Parkinson’s disease [100], which may have a potential association with pSS [101], this suggests that IL-15 may be a potential target [100]. ZIDOVUDINE for pSS has been cited in the manuscript, but studies have shown that antiretroviral therapy has a number of severe and life-threatening adverse drug reactions. For instance, taking ZIDOVUDINE was observed as a risk factor for anemia. STAVUDINE was utilized for the treatment of peripheral neuropathy, but among that, the use of nevirapine was identified as a risk factor for cutaneous reactions [102].
Significantly, CYCLOSPORINE A has been found to be a potent inhibitor of IL-15 release in the context of acute rejection following heart transplantation in mice [103]. However, varying doses of CYCLOSPORINE, which is a key immunosuppressive therapy for kidney transplant recipients, do not appear to have an impact on serum levels of IL-15 and IP-10 cytokines [104]. While certain studies have proposed a potential role of TESTOSTERONE in modulating disease progression through the promotion of anti-inflammatory responses, the observed reduction in CXCL10 levels in male patients receiving TESTOSTERONE supplementation was not notably significant [105]. IL-15 and IP-10, in conjunction with CYCLOSPORINE, have been identified as significant inflammatory biomarkers in rheumatoid arthritis [106]. Given the notable upregulation of IL-15 and CXCL10 in pSS patients, it is postulated that pSS may contribute to the regulation of these cytokine levels via alternative mechanisms. Combined with the current research on the application of TESTOSTERONE and CYCLOSPORINE in autoimmune diseases [107109], we speculate that TESTOSTERONE and CYCLOSPORINE may regulate the abnormal activity of immune cells and reduce inflammation by targeting the inhibition of CXCL10, a proinflammatory cytokine, and IL15, an activator of immune cells. In turn, this will help improve the immune function of pSS patients and alleviate their symptoms and immune-mediated inflammation-related damage. However, it remains to be clinically verified in pSS patients.
However, there are still several limitations in our study: Verifying the reliability of transcriptional changes in gene expression establishes a theoretical foundation for the rapid evaluation of biomarkers expression in peripheral blood detection, while detecting gene expression at the protein level requires the further detection of specific proteins or cell surface markers, using techniques such as ELISA and flow cytometry. At the same time, the diagnostic efficacy of biomarkers, drug targeting results, and the regulatory networks are currently only preliminary findings from bioinformatics research and prediction, and it is necessary to conduct larger studies with a broader cohort of patients, as well as additional follow-up RNA-seq and animal studies, to validate their effectiveness, safety, and robustness. Furthermore, conducting clinical trials is necessary to verify the interaction mechanism between key genes and key immune cells using real data obtained from an increased number of clinical samples. Despite the challenges presented, the advances in genomics offer us a unique opportunity to gain a better understanding of the pathomechanism of pSS and develop novel therapeutic strategies. Further research into pSS could result in innovative treatments.
In conclusion, four genes (LY6E, EIF2AK2, IL15, CXCL10) that might be potential diagnostic inflammation-associated biomarkers of pSS in peripheral blood were identified by bioinformatics analysis, and their expression were validated by RT-qPCR. Given that the samples used in this study were all derived from peripheral blood for the pSS-datasets, we argue that leveraging peripheral blood tests for rapid evaluation of biomarker expression has the potential to improve the diagnostic accuracy of early pSS. Furthermore, the molecular mechanisms of these genes were preliminarily explored by generating a lncRNA-miRNA-mRNA regulatory network. And meanwhile, the predicted drugs, such as TESTOSTERONE targeting CXCL10 and CYCLOSPORINE targeting IL15, may potentially enhance immune function and alleviate symptoms and immune-mediated inflammation-related damage in patients with pSS. The results provided a basis for understanding the pathogenesis and improving clinical diagnosis and treatment for pSS.

Acknowledgements

Grateful acknowledgement is made to Dr. Hung-Hsiang Hao for his assistance in data collection.

Declarations

This study was approved by the Peking University Third Hospital Medical Science Research Ethics Committee (IRB00006761-M2022106), written informed consent was received from all participants for their enrollment, and all methods were carried out in accordance with relevant guidelines and regulations.

Competing interests

The authors declare that they have no competing interests.
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/​.

Publisher's Note

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

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Ramos-Casals M, Brito-Zeron P, Siso-Almirall A, Bosch X (2012) Primary Sjogren syndrome. BMJ 344:e3821PubMedCrossRef Ramos-Casals M, Brito-Zeron P, Siso-Almirall A, Bosch X (2012) Primary Sjogren syndrome. BMJ 344:e3821PubMedCrossRef
2.
Zurück zum Zitat Manfre V, Cafaro G, Riccucci I, Zabotti A, Perricone C, Bootsma H, De Vita S, Bartoloni E (2020) One year in review 2020: comorbidities, diagnosis and treatment of primary Sjogren’s syndrome. Clin Exp Rheumatol 38 Suppl 126(4):10–22PubMed Manfre V, Cafaro G, Riccucci I, Zabotti A, Perricone C, Bootsma H, De Vita S, Bartoloni E (2020) One year in review 2020: comorbidities, diagnosis and treatment of primary Sjogren’s syndrome. Clin Exp Rheumatol 38 Suppl 126(4):10–22PubMed
3.
Zurück zum Zitat Medeiros CCG, Dos Anjos Borges LG, Cherubini K, Salum FG, Medina da Silva R, de Figueiredo MAZ (2018) Oral yeast colonization in patients with primary and secondary Sjogren’s syndrome. Oral Dis 24(7):1367–1378PubMedCrossRef Medeiros CCG, Dos Anjos Borges LG, Cherubini K, Salum FG, Medina da Silva R, de Figueiredo MAZ (2018) Oral yeast colonization in patients with primary and secondary Sjogren’s syndrome. Oral Dis 24(7):1367–1378PubMedCrossRef
4.
Zurück zum Zitat Seror R, Ravaud P, Bowman SJ, Baron G, Tzioufas A, Theander E, Gottenberg JE, Bootsma H, Mariette X, Vitali C et al (2010) EULAR Sjogren’s syndrome disease activity index: development of a consensus systemic disease activity index for primary Sjogren’s syndrome. Ann Rheum Dis 69(6):1103–1109PubMedCrossRef Seror R, Ravaud P, Bowman SJ, Baron G, Tzioufas A, Theander E, Gottenberg JE, Bootsma H, Mariette X, Vitali C et al (2010) EULAR Sjogren’s syndrome disease activity index: development of a consensus systemic disease activity index for primary Sjogren’s syndrome. Ann Rheum Dis 69(6):1103–1109PubMedCrossRef
5.
Zurück zum Zitat Voulgarelis M, Ziakas PD, Papageorgiou A, Baimpa E, Tzioufas AG, Moutsopoulos HM (2012) Prognosis and outcome of non-Hodgkin lymphoma in primary Sjogren syndrome. Medicine (Baltimore) 91(1):1–9PubMedCrossRef Voulgarelis M, Ziakas PD, Papageorgiou A, Baimpa E, Tzioufas AG, Moutsopoulos HM (2012) Prognosis and outcome of non-Hodgkin lymphoma in primary Sjogren syndrome. Medicine (Baltimore) 91(1):1–9PubMedCrossRef
6.
Zurück zum Zitat Shiboski CH, Shiboski SC, Seror R, Criswell LA, Labetoulle M, Lietman TM, Rasmussen A, Scofield H, Vitali C, Bowman SJ et al (2017) 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 69(1):35–45PubMedCrossRef Shiboski CH, Shiboski SC, Seror R, Criswell LA, Labetoulle M, Lietman TM, Rasmussen A, Scofield H, Vitali C, Bowman SJ et al (2017) 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 69(1):35–45PubMedCrossRef
7.
Zurück zum Zitat Jin Y, Li J, Chen J, Shao M, Zhang R, Liang Y, Zhang X, Zhang X, Zhang Q, Li F et al (2019) Tissue-specific autoantibodies improve diagnosis of primary Sjogren’s syndrome in the early stage and indicate localized salivary injury. J Immunol Res 2019:3642937PubMedPubMedCentralCrossRef Jin Y, Li J, Chen J, Shao M, Zhang R, Liang Y, Zhang X, Zhang X, Zhang Q, Li F et al (2019) Tissue-specific autoantibodies improve diagnosis of primary Sjogren’s syndrome in the early stage and indicate localized salivary injury. J Immunol Res 2019:3642937PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Pijpe J, Kalk WW, van der Wal JE, Vissink A, Kluin PM, Roodenburg JL, Bootsma H, Kallenberg CG, Spijkervet FK (2007) Parotid gland biopsy compared with labial biopsy in the diagnosis of patients with primary Sjogren’s syndrome. Rheumatology (Oxford) 46(2):335–341PubMedCrossRef Pijpe J, Kalk WW, van der Wal JE, Vissink A, Kluin PM, Roodenburg JL, Bootsma H, Kallenberg CG, Spijkervet FK (2007) Parotid gland biopsy compared with labial biopsy in the diagnosis of patients with primary Sjogren’s syndrome. Rheumatology (Oxford) 46(2):335–341PubMedCrossRef
9.
Zurück zum Zitat Costa S, Quintin-Roue I, Lesourd A, Jousse-Joulin S, Berthelot JM, Hachulla E, Hatron PY, Goeb V, Vittecoq O, Pers JO et al (2015) Reliability of histopathological salivary gland biopsy assessment in Sjogren’s syndrome: a multicentre cohort study. Rheumatology (Oxford) 54(6):1056–1064PubMedCrossRef Costa S, Quintin-Roue I, Lesourd A, Jousse-Joulin S, Berthelot JM, Hachulla E, Hatron PY, Goeb V, Vittecoq O, Pers JO et al (2015) Reliability of histopathological salivary gland biopsy assessment in Sjogren’s syndrome: a multicentre cohort study. Rheumatology (Oxford) 54(6):1056–1064PubMedCrossRef
10.
Zurück zum Zitat Ren Y, Cui G, Gao Y (2021) Research progress on inflammatory mechanism of primary Sjogren syndrome. Zhejiang Da Xue Xue Bao Yi Xue Ban 50(6):783–794PubMedPubMedCentral Ren Y, Cui G, Gao Y (2021) Research progress on inflammatory mechanism of primary Sjogren syndrome. Zhejiang Da Xue Xue Bao Yi Xue Ban 50(6):783–794PubMedPubMedCentral
11.
Zurück zum Zitat Mavragani CP (2017) Mechanisms and new strategies for primary Sjogren’s syndrome. Annu Rev Med 68:331–343PubMedCrossRef Mavragani CP (2017) Mechanisms and new strategies for primary Sjogren’s syndrome. Annu Rev Med 68:331–343PubMedCrossRef
12.
Zurück zum Zitat Ling J, Chan BC, Tsang MS, Gao X, Leung PC, Lam CW, Hu JM, Wong CK (2021) Current advances in mechanisms and treatment of dry eye disease: toward anti-inflammatory and immunomodulatory therapy and traditional Chinese medicine. Front Med (Lausanne) 8:815075PubMedCrossRef Ling J, Chan BC, Tsang MS, Gao X, Leung PC, Lam CW, Hu JM, Wong CK (2021) Current advances in mechanisms and treatment of dry eye disease: toward anti-inflammatory and immunomodulatory therapy and traditional Chinese medicine. Front Med (Lausanne) 8:815075PubMedCrossRef
13.
Zurück zum Zitat Stefanski AL, Tomiak C, Pleyer U, Dietrich T, Burmester GR, Dorner T (2017) The diagnosis and treatment of Sjogren’s syndrome. Dtsch Arztebl Int 114(20):354–361PubMedPubMedCentral Stefanski AL, Tomiak C, Pleyer U, Dietrich T, Burmester GR, Dorner T (2017) The diagnosis and treatment of Sjogren’s syndrome. Dtsch Arztebl Int 114(20):354–361PubMedPubMedCentral
14.
Zurück zum Zitat Fox RI, Fox CM, Gottenberg JE, Dorner T (2021) Treatment of Sjogren’s syndrome: current therapy and future directions. Rheumatology (Oxford) 60(5):2066–2074PubMedCrossRef Fox RI, Fox CM, Gottenberg JE, Dorner T (2021) Treatment of Sjogren’s syndrome: current therapy and future directions. Rheumatology (Oxford) 60(5):2066–2074PubMedCrossRef
15.
Zurück zum Zitat Ramos-Casals M, Brito-Zeron P, Bombardieri S, Bootsma H, De Vita S, Dorner T, Fisher BA, Gottenberg JE, Hernandez-Molina G, Kocher A et al (2020) EULAR recommendations for the management of Sjogren’s syndrome with topical and systemic therapies. Ann Rheum Dis 79(1):3–18PubMedCrossRef Ramos-Casals M, Brito-Zeron P, Bombardieri S, Bootsma H, De Vita S, Dorner T, Fisher BA, Gottenberg JE, Hernandez-Molina G, Kocher A et al (2020) EULAR recommendations for the management of Sjogren’s syndrome with topical and systemic therapies. Ann Rheum Dis 79(1):3–18PubMedCrossRef
16.
Zurück zum Zitat Yao Q, Song Z, Wang B, Qin Q, Zhang JA (2019) Identifying key genes and functionally enriched pathways in Sjogren's syndrome by weighted gene co-expression network analysis. Front Genet 10:1142 Yao Q, Song Z, Wang B, Qin Q, Zhang JA (2019) Identifying key genes and functionally enriched pathways in Sjogren's syndrome by weighted gene co-expression network analysis. Front Genet 10:1142
17.
Zurück zum Zitat Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, Smyth GK (2015) limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res 43(7):e47PubMedPubMedCentralCrossRef Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, Smyth GK (2015) limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res 43(7):e47PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Zeng Q, Wen J, Zheng L, Zeng W, Chen S, Zhao C (2022) Identification of immune-related diagnostic markers in primary Sjogren’s syndrome based on bioinformatics analysis. Ann Transl Med 10(8):487PubMedPubMedCentralCrossRef Zeng Q, Wen J, Zheng L, Zeng W, Chen S, Zhao C (2022) Identification of immune-related diagnostic markers in primary Sjogren’s syndrome based on bioinformatics analysis. Ann Transl Med 10(8):487PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Wu T, Hu E, Xu S, Chen M, Guo P, Dai Z, Feng T, Zhou L, Tang W, Zhan L et al (2021) clusterProfiler 4.0: a universal enrichment tool for interpreting omics data. Innovation (Camb) 2(3):100141PubMed Wu T, Hu E, Xu S, Chen M, Guo P, Dai Z, Feng T, Zhou L, Tang W, Zhan L et al (2021) clusterProfiler 4.0: a universal enrichment tool for interpreting omics data. Innovation (Camb) 2(3):100141PubMed
21.
22.
Zurück zum Zitat Huang ML, Hung YH, Lee WM, Li RK, Jiang BR (2014) SVM-RFE based feature selection and Taguchi parameters optimization for multiclass SVM classifier. ScientificWorldJournal 2014:795624PubMedPubMedCentralCrossRef Huang ML, Hung YH, Lee WM, Li RK, Jiang BR (2014) SVM-RFE based feature selection and Taguchi parameters optimization for multiclass SVM classifier. ScientificWorldJournal 2014:795624PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, Ideker T (2003) Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res 13(11):2498–2504PubMedPubMedCentralCrossRef Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, Ideker T (2003) Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res 13(11):2498–2504PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Livak KJ, Schmittgen TD (2001) Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 25(4):402–408PubMedCrossRef Livak KJ, Schmittgen TD (2001) Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 25(4):402–408PubMedCrossRef
26.
Zurück zum Zitat Bharaj TK, Aqrawi LA, Fromreide S, Jonsson R, Brun JG, Appel S, Skarstein K (2021) Inflammatory stratification in primary Sjogren’s syndrome reveals novel immune cell alterations in patients’ minor salivary glands. Front Immunol 12:701581PubMedPubMedCentralCrossRef Bharaj TK, Aqrawi LA, Fromreide S, Jonsson R, Brun JG, Appel S, Skarstein K (2021) Inflammatory stratification in primary Sjogren’s syndrome reveals novel immune cell alterations in patients’ minor salivary glands. Front Immunol 12:701581PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Smatti MK, Cyprian FS, Nasrallah GK, Al Thani AA, Almishal RO, Yassine HM (2019) Viruses and autoimmunity: a review on the potential interaction and molecular mechanisms. Viruses 11(8):762 Smatti MK, Cyprian FS, Nasrallah GK, Al Thani AA, Almishal RO, Yassine HM (2019) Viruses and autoimmunity: a review on the potential interaction and molecular mechanisms. Viruses 11(8):762
28.
Zurück zum Zitat Xia CQ, Peng R, Chernatynskaya AV, Yuan L, Carter C, Valentine J, Sobel E, Atkinson MA, Clare-Salzler MJ (2014) Increased IFN-alpha-producing plasmacytoid dendritic cells (pDCs) in human Th1-mediated type 1 diabetes: pDCs augment Th1 responses through IFN-alpha production. J Immunol 193(3):1024–1034PubMedCrossRef Xia CQ, Peng R, Chernatynskaya AV, Yuan L, Carter C, Valentine J, Sobel E, Atkinson MA, Clare-Salzler MJ (2014) Increased IFN-alpha-producing plasmacytoid dendritic cells (pDCs) in human Th1-mediated type 1 diabetes: pDCs augment Th1 responses through IFN-alpha production. J Immunol 193(3):1024–1034PubMedCrossRef
29.
Zurück zum Zitat Capua I, Mercalli A, Pizzuto MS, Romero-Tejeda A, Kasloff S, De Battisti C, Bonfante F, Patrono LV, Vicenzi E, Zappulli V et al (2013) Influenza A viruses grow in human pancreatic cells and cause pancreatitis and diabetes in an animal model. J Virol 87(1):597–610PubMedPubMedCentralCrossRef Capua I, Mercalli A, Pizzuto MS, Romero-Tejeda A, Kasloff S, De Battisti C, Bonfante F, Patrono LV, Vicenzi E, Zappulli V et al (2013) Influenza A viruses grow in human pancreatic cells and cause pancreatitis and diabetes in an animal model. J Virol 87(1):597–610PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Brauner S, Folkersen L, Kvarnstrom M, Meisgen S, Petersen S, Franzen-Malmros M, Mofors J, Brokstad KA, Klareskog L, Jonsson R et al (2017) H1N1 vaccination in Sjogren’s syndrome triggers polyclonal B cell activation and promotes autoantibody production. Ann Rheum Dis 76(10):1755–1763PubMedCrossRef Brauner S, Folkersen L, Kvarnstrom M, Meisgen S, Petersen S, Franzen-Malmros M, Mofors J, Brokstad KA, Klareskog L, Jonsson R et al (2017) H1N1 vaccination in Sjogren’s syndrome triggers polyclonal B cell activation and promotes autoantibody production. Ann Rheum Dis 76(10):1755–1763PubMedCrossRef
32.
Zurück zum Zitat Iwakiri D, Zhou L, Samanta M, Matsumoto M, Ebihara T, Seya T, Imai S, Fujieda M, Kawa K, Takada K (2009) Epstein-Barr virus (EBV)-encoded small RNA is released from EBV-infected cells and activates signaling from Toll-like receptor 3. J Exp Med 206(10):2091–2099PubMedPubMedCentralCrossRef Iwakiri D, Zhou L, Samanta M, Matsumoto M, Ebihara T, Seya T, Imai S, Fujieda M, Kawa K, Takada K (2009) Epstein-Barr virus (EBV)-encoded small RNA is released from EBV-infected cells and activates signaling from Toll-like receptor 3. J Exp Med 206(10):2091–2099PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Inoue H, Mishima K, Yamamoto-Yoshida S, Ushikoshi-Nakayama R, Nakagawa Y, Yamamoto K, Ryo K, Ide F, Saito I (2012) Aryl hydrocarbon receptor-mediated induction of EBV reactivation as a risk factor for Sjogren’s syndrome. J Immunol 188(9):4654–4662PubMedCrossRef Inoue H, Mishima K, Yamamoto-Yoshida S, Ushikoshi-Nakayama R, Nakagawa Y, Yamamoto K, Ryo K, Ide F, Saito I (2012) Aryl hydrocarbon receptor-mediated induction of EBV reactivation as a risk factor for Sjogren’s syndrome. J Immunol 188(9):4654–4662PubMedCrossRef
34.
Zurück zum Zitat Medzhitov R (2007) Recognition of microorganisms and activation of the immune response. Nature 449(7164):819–826ADSPubMedCrossRef Medzhitov R (2007) Recognition of microorganisms and activation of the immune response. Nature 449(7164):819–826ADSPubMedCrossRef
35.
Zurück zum Zitat Yoneyama M, Kikuchi M, Natsukawa T, Shinobu N, Imaizumi T, Miyagishi M, Taira K, Akira S, Fujita T (2004) The RNA helicase RIG-I has an essential function in double-stranded RNA-induced innate antiviral responses. Nat Immunol 5(7):730–737PubMedCrossRef Yoneyama M, Kikuchi M, Natsukawa T, Shinobu N, Imaizumi T, Miyagishi M, Taira K, Akira S, Fujita T (2004) The RNA helicase RIG-I has an essential function in double-stranded RNA-induced innate antiviral responses. Nat Immunol 5(7):730–737PubMedCrossRef
36.
Zurück zum Zitat Kawai T, Akira S (2008) Toll-like receptor and RIG-I-like receptor signaling. Ann N Y Acad Sci 1143:1–20ADSPubMedCrossRef Kawai T, Akira S (2008) Toll-like receptor and RIG-I-like receptor signaling. Ann N Y Acad Sci 1143:1–20ADSPubMedCrossRef
37.
Zurück zum Zitat Chen JQ, Szodoray P, Zeher M (2016) Toll-like receptor pathways in autoimmune diseases. Clin Rev Allergy Immunol 50(1):1–17PubMedCrossRef Chen JQ, Szodoray P, Zeher M (2016) Toll-like receptor pathways in autoimmune diseases. Clin Rev Allergy Immunol 50(1):1–17PubMedCrossRef
38.
Zurück zum Zitat Kiripolsky J, Kramer JM (2018) Current and emerging evidence for toll-like receptor activation in Sjogren’s syndrome. J Immunol Res 2018:1246818PubMedPubMedCentralCrossRef Kiripolsky J, Kramer JM (2018) Current and emerging evidence for toll-like receptor activation in Sjogren’s syndrome. J Immunol Res 2018:1246818PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Ittah M, Miceli-Richard C, Gottenberg JE, Sellam J, Eid P, Lebon P, Pallier C, Lepajolec C, Mariette X (2008) Viruses induce high expression of BAFF by salivary gland epithelial cells through TLR- and type-I IFN-dependent and -independent pathways. Eur J Immunol 38(4):1058–1064PubMedCrossRef Ittah M, Miceli-Richard C, Gottenberg JE, Sellam J, Eid P, Lebon P, Pallier C, Lepajolec C, Mariette X (2008) Viruses induce high expression of BAFF by salivary gland epithelial cells through TLR- and type-I IFN-dependent and -independent pathways. Eur J Immunol 38(4):1058–1064PubMedCrossRef
40.
Zurück zum Zitat Kwok SK, Cho ML, Her YM, Oh HJ, Park MK, Lee SY, Woo YJ, Ju JH, Park KS, Kim HY et al (2012) TLR2 ligation induces the production of IL-23/IL-17 via IL-6, STAT3 and NF-kB pathway in patients with primary Sjogren’s syndrome. Arthritis Res Ther 14(2):R64PubMedPubMedCentralCrossRef Kwok SK, Cho ML, Her YM, Oh HJ, Park MK, Lee SY, Woo YJ, Ju JH, Park KS, Kim HY et al (2012) TLR2 ligation induces the production of IL-23/IL-17 via IL-6, STAT3 and NF-kB pathway in patients with primary Sjogren’s syndrome. Arthritis Res Ther 14(2):R64PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Piccinini AM, Midwood KS (2010) DAMPening inflammation by modulating TLR signalling. Mediators Inflamm 2010:672395 Piccinini AM, Midwood KS (2010) DAMPening inflammation by modulating TLR signalling. Mediators Inflamm 2010:672395
42.
Zurück zum Zitat Schenke-Layland K, Xie J, Angelis E, Starcher B, Wu K, Riemann I, MacLellan WR, Hamm-Alvarez SF (2008) Increased degradation of extracellular matrix structures of lacrimal glands implicated in the pathogenesis of Sjogren’s syndrome. Matrix Biol 27(1):53–66PubMedCrossRef Schenke-Layland K, Xie J, Angelis E, Starcher B, Wu K, Riemann I, MacLellan WR, Hamm-Alvarez SF (2008) Increased degradation of extracellular matrix structures of lacrimal glands implicated in the pathogenesis of Sjogren’s syndrome. Matrix Biol 27(1):53–66PubMedCrossRef
43.
Zurück zum Zitat Kiripolsky J, Romano RA, Kasperek EM, Yu G, Kramer JM (2019) Activation of Myd88-dependent TLRs mediates local and systemic inflammation in a mouse model of primary Sjogren’s syndrome. Front Immunol 10:2963PubMedCrossRef Kiripolsky J, Romano RA, Kasperek EM, Yu G, Kramer JM (2019) Activation of Myd88-dependent TLRs mediates local and systemic inflammation in a mouse model of primary Sjogren’s syndrome. Front Immunol 10:2963PubMedCrossRef
44.
Zurück zum Zitat Jiang H, Gong T, Zhou R (2020) The strategies of targeting the NLRP3 inflammasome to treat inflammatory diseases. Adv Immunol 145:55–93PubMed Jiang H, Gong T, Zhou R (2020) The strategies of targeting the NLRP3 inflammasome to treat inflammatory diseases. Adv Immunol 145:55–93PubMed
45.
Zurück zum Zitat Kim SK, Choe JY, Lee GH (2017) Enhanced expression of NLRP3 inflammasome-related inflammation in peripheral blood mononuclear cells in Sjogren’s syndrome. Clin Chim Acta 474:147–154PubMedCrossRef Kim SK, Choe JY, Lee GH (2017) Enhanced expression of NLRP3 inflammasome-related inflammation in peripheral blood mononuclear cells in Sjogren’s syndrome. Clin Chim Acta 474:147–154PubMedCrossRef
46.
Zurück zum Zitat Baldini C, Santini E, Rossi C, Donati V, Solini A (2017) The P2X7 receptor-NLRP3 inflammasome complex predicts the development of non-Hodgkin’s lymphoma in Sjogren’s syndrome: a prospective, observational, single-centre study. J Intern Med 282(2):175–186PubMedCrossRef Baldini C, Santini E, Rossi C, Donati V, Solini A (2017) The P2X7 receptor-NLRP3 inflammasome complex predicts the development of non-Hodgkin’s lymphoma in Sjogren’s syndrome: a prospective, observational, single-centre study. J Intern Med 282(2):175–186PubMedCrossRef
47.
Zurück zum Zitat Song X, He X, Li X, Qian Y (2016) The roles and functional mechanisms of interleukin-17 family cytokines in mucosal immunity. Cell Mol Immunol 13(4):418–431PubMedPubMedCentralCrossRef Song X, He X, Li X, Qian Y (2016) The roles and functional mechanisms of interleukin-17 family cytokines in mucosal immunity. Cell Mol Immunol 13(4):418–431PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Alunno A, Carubbi F, Bartoloni E, Bistoni O, Caterbi S, Cipriani P, Giacomelli R, Gerli R (2014) Unmasking the pathogenic role of IL-17 axis in primary Sjogren’s syndrome: a new era for therapeutic targeting? Autoimmun Rev 13(12):1167–1173PubMedCrossRef Alunno A, Carubbi F, Bartoloni E, Bistoni O, Caterbi S, Cipriani P, Giacomelli R, Gerli R (2014) Unmasking the pathogenic role of IL-17 axis in primary Sjogren’s syndrome: a new era for therapeutic targeting? Autoimmun Rev 13(12):1167–1173PubMedCrossRef
49.
Zurück zum Zitat Hemdan NY, Birkenmeier G, Wichmann G, Abu El-Saad AM, Krieger T, Conrad K, Sack U (2010) Interleukin-17-producing T helper cells in autoimmunity. Autoimmun Rev 9(11):785–792PubMedCrossRef Hemdan NY, Birkenmeier G, Wichmann G, Abu El-Saad AM, Krieger T, Conrad K, Sack U (2010) Interleukin-17-producing T helper cells in autoimmunity. Autoimmun Rev 9(11):785–792PubMedCrossRef
50.
Zurück zum Zitat Ciccia F, Guggino G, Rizzo A, Alessandro R, Carubbi F, Giardina A, Cipriani P, Ferrante A, Cannizzaro A, Giacomelli R et al (2014) Rituximab modulates IL-17 expression in the salivary glands of patients with primary Sjogren’s syndrome. Rheumatology (Oxford) 53(7):1313–1320PubMedCrossRef Ciccia F, Guggino G, Rizzo A, Alessandro R, Carubbi F, Giardina A, Cipriani P, Ferrante A, Cannizzaro A, Giacomelli R et al (2014) Rituximab modulates IL-17 expression in the salivary glands of patients with primary Sjogren’s syndrome. Rheumatology (Oxford) 53(7):1313–1320PubMedCrossRef
51.
Zurück zum Zitat Miletic M, Stojanovic R, Pajic O, Bugarski D, Mojsilovic S, Cokic V, Milenkovic P (2012) Serum interleukin-17 & nitric oxide levels in patients with primary Sjogren’s syndrome. Indian J Med Res 135(4):513–519PubMedPubMedCentral Miletic M, Stojanovic R, Pajic O, Bugarski D, Mojsilovic S, Cokic V, Milenkovic P (2012) Serum interleukin-17 & nitric oxide levels in patients with primary Sjogren’s syndrome. Indian J Med Res 135(4):513–519PubMedPubMedCentral
52.
Zurück zum Zitat Katsifis GE, Rekka S, Moutsopoulos NM, Pillemer S, Wahl SM (2009) Systemic and local interleukin-17 and linked cytokines associated with Sjogren’s syndrome immunopathogenesis. Am J Pathol 175(3):1167–1177PubMedPubMedCentralCrossRef Katsifis GE, Rekka S, Moutsopoulos NM, Pillemer S, Wahl SM (2009) Systemic and local interleukin-17 and linked cytokines associated with Sjogren’s syndrome immunopathogenesis. Am J Pathol 175(3):1167–1177PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Liu R, Gao C, Chen H, Li Y, Jin Y, Qi H (2017) Analysis of Th17-associated cytokines and clinical correlations in patients with dry eye disease. PLoS ONE 12(4):e0173301PubMedPubMedCentralCrossRef Liu R, Gao C, Chen H, Li Y, Jin Y, Qi H (2017) Analysis of Th17-associated cytokines and clinical correlations in patients with dry eye disease. PLoS ONE 12(4):e0173301PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Nguyen CQ, Hu MH, Li Y, Stewart C, Peck AB (2008) Salivary gland tissue expression of interleukin-23 and interleukin-17 in Sjogren’s syndrome: findings in humans and mice. Arthritis Rheum 58(3):734–743PubMedPubMedCentralCrossRef Nguyen CQ, Hu MH, Li Y, Stewart C, Peck AB (2008) Salivary gland tissue expression of interleukin-23 and interleukin-17 in Sjogren’s syndrome: findings in humans and mice. Arthritis Rheum 58(3):734–743PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Gan Y, Zhao X, He J, Liu X, Li Y, Sun X, Li Z (2017) Increased Interleukin-17F is associated with elevated autoantibody levels and more clinically relevant than interleukin-17A in primary Sjogren’s syndrome. J Immunol Res 2017:4768408PubMedPubMedCentralCrossRef Gan Y, Zhao X, He J, Liu X, Li Y, Sun X, Li Z (2017) Increased Interleukin-17F is associated with elevated autoantibody levels and more clinically relevant than interleukin-17A in primary Sjogren’s syndrome. J Immunol Res 2017:4768408PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Yu J, Liu SL (2019) Emerging role of LY6E in virus-host interactions. Viruses 11(11):1020 Yu J, Liu SL (2019) Emerging role of LY6E in virus-host interactions. Viruses 11(11):1020
57.
Zurück zum Zitat Yao Q, Song Z, Wang B, Qin Q, Zhang JA (2019) Identifying key genes and functionally enriched pathways in Sjogren’s syndrome by weighted gene co-expression network analysis. Front Genet 10:1142PubMedPubMedCentralCrossRef Yao Q, Song Z, Wang B, Qin Q, Zhang JA (2019) Identifying key genes and functionally enriched pathways in Sjogren’s syndrome by weighted gene co-expression network analysis. Front Genet 10:1142PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Maria NI, Brkic Z, Waris M, van Helden-Meeuwsen CG, Heezen K, van de Merwe JP, van Daele PL, Dalm VA, Drexhage HA, Versnel MA (2014) MxA as a clinically applicable biomarker for identifying systemic interferon type I in primary Sjogren’s syndrome. Ann Rheum Dis 73(6):1052–1059PubMedCrossRef Maria NI, Brkic Z, Waris M, van Helden-Meeuwsen CG, Heezen K, van de Merwe JP, van Daele PL, Dalm VA, Drexhage HA, Versnel MA (2014) MxA as a clinically applicable biomarker for identifying systemic interferon type I in primary Sjogren’s syndrome. Ann Rheum Dis 73(6):1052–1059PubMedCrossRef
59.
Zurück zum Zitat Brkic Z, Maria NI, van Helden-Meeuwsen CG, van de Merwe JP, van Daele PL, Dalm VA, Wildenberg ME, Beumer W, Drexhage HA, Versnel MA (2013) Prevalence of interferon type I signature in CD14 monocytes of patients with Sjogren’s syndrome and association with disease activity and BAFF gene expression. Ann Rheum Dis 72(5):728–735PubMedCrossRef Brkic Z, Maria NI, van Helden-Meeuwsen CG, van de Merwe JP, van Daele PL, Dalm VA, Wildenberg ME, Beumer W, Drexhage HA, Versnel MA (2013) Prevalence of interferon type I signature in CD14 monocytes of patients with Sjogren’s syndrome and association with disease activity and BAFF gene expression. Ann Rheum Dis 72(5):728–735PubMedCrossRef
60.
Zurück zum Zitat Villarroya-Beltri C, Guerra S, Sanchez-Madrid F (2017) ISGylation - a key to lock the cell gates for preventing the spread of threats. J Cell Sci 130(18):2961–2969PubMed Villarroya-Beltri C, Guerra S, Sanchez-Madrid F (2017) ISGylation - a key to lock the cell gates for preventing the spread of threats. J Cell Sci 130(18):2961–2969PubMed
61.
Zurück zum Zitat Ittah M, Miceli-Richard C, Gottenberg JE, Sellam J, Lepajolec C, Mariette X (2009) B-cell-activating factor expressions in salivary epithelial cells after dsRNA virus infection depends on RNA-activated protein kinase activation. Eur J Immunol 39(5):1271–1279PubMedCrossRef Ittah M, Miceli-Richard C, Gottenberg JE, Sellam J, Lepajolec C, Mariette X (2009) B-cell-activating factor expressions in salivary epithelial cells after dsRNA virus infection depends on RNA-activated protein kinase activation. Eur J Immunol 39(5):1271–1279PubMedCrossRef
62.
Zurück zum Zitat Ittah M, Miceli-Richard C, Eric Gottenberg J, Lavie F, Lazure T, Ba N, Sellam J, Lepajolec C, Mariette X (2006) B cell-activating factor of the tumor necrosis factor family (BAFF) is expressed under stimulation by interferon in salivary gland epithelial cells in primary Sjogren’s syndrome. Arthritis Res Ther 8(2):R51PubMedPubMedCentralCrossRef Ittah M, Miceli-Richard C, Eric Gottenberg J, Lavie F, Lazure T, Ba N, Sellam J, Lepajolec C, Mariette X (2006) B cell-activating factor of the tumor necrosis factor family (BAFF) is expressed under stimulation by interferon in salivary gland epithelial cells in primary Sjogren’s syndrome. Arthritis Res Ther 8(2):R51PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Kurowska W, Przygodzka M, Jakubaszek M, Kwiatkowska B, Maslinski W (2020) Interleukin-15 as a biomarker candidate of rheumatoid arthritis development. J Clin Med 9(5):1555 Kurowska W, Przygodzka M, Jakubaszek M, Kwiatkowska B, Maslinski W (2020) Interleukin-15 as a biomarker candidate of rheumatoid arthritis development. J Clin Med 9(5):1555
64.
Zurück zum Zitat Sakai T, Kusugami K, Nishimura H, Ando T, Yamaguchi T, Ohsuga M, Ina K, Enomoto A, Kimura Y, Yoshikai Y (1998) Interleukin 15 activity in the rectal mucosa of inflammatory bowel disease. Gastroenterology 114(6):1237–1243PubMedCrossRef Sakai T, Kusugami K, Nishimura H, Ando T, Yamaguchi T, Ohsuga M, Ina K, Enomoto A, Kimura Y, Yoshikai Y (1998) Interleukin 15 activity in the rectal mucosa of inflammatory bowel disease. Gastroenterology 114(6):1237–1243PubMedCrossRef
65.
Zurück zum Zitat Bikker A, van Woerkom JM, Kruize AA, Wenting-van Wijk M, de Jager W, Bijlsma JW, Lafeber FP, van Roon JA (2010) Increased expression of interleukin-7 in labial salivary glands of patients with primary Sjogren’s syndrome correlates with increased inflammation. Arthritis Rheum 62(4):969–977PubMedCrossRef Bikker A, van Woerkom JM, Kruize AA, Wenting-van Wijk M, de Jager W, Bijlsma JW, Lafeber FP, van Roon JA (2010) Increased expression of interleukin-7 in labial salivary glands of patients with primary Sjogren’s syndrome correlates with increased inflammation. Arthritis Rheum 62(4):969–977PubMedCrossRef
66.
Zurück zum Zitat Aqrawi LA, Jensen JL, Oijordsbakken G, Ruus AK, Nygard S, Holden M, Jonsson R, Galtung HK, Skarstein K (2018) Signalling pathways identified in salivary glands from primary Sjogren’s syndrome patients reveal enhanced adipose tissue development. Autoimmunity 51(3):135–146PubMedCrossRef Aqrawi LA, Jensen JL, Oijordsbakken G, Ruus AK, Nygard S, Holden M, Jonsson R, Galtung HK, Skarstein K (2018) Signalling pathways identified in salivary glands from primary Sjogren’s syndrome patients reveal enhanced adipose tissue development. Autoimmunity 51(3):135–146PubMedCrossRef
67.
Zurück zum Zitat Reksten TR, Jonsson MV, Szyszko EA, Brun JG, Jonsson R, Brokstad KA (2009) Cytokine and autoantibody profiling related to histopathological features in primary Sjogren’s syndrome. Rheumatology (Oxford) 48(9):1102–1106PubMedCrossRef Reksten TR, Jonsson MV, Szyszko EA, Brun JG, Jonsson R, Brokstad KA (2009) Cytokine and autoantibody profiling related to histopathological features in primary Sjogren’s syndrome. Rheumatology (Oxford) 48(9):1102–1106PubMedCrossRef
68.
Zurück zum Zitat Sisto M, Lorusso L, Lisi S (2016) Interleukin-15 as a potential new target in Sjogren’s syndrome-associated inflammation. Pathology 48(6):602–607PubMedCrossRef Sisto M, Lorusso L, Lisi S (2016) Interleukin-15 as a potential new target in Sjogren’s syndrome-associated inflammation. Pathology 48(6):602–607PubMedCrossRef
69.
Zurück zum Zitat Sisto M, Lorusso L, Lisi S (2017) TLR2 signals via NF-kappaB to drive IL-15 production in salivary gland epithelial cells derived from patients with primary Sjogren’s syndrome. Clin Exp Med 17(3):341–350PubMedCrossRef Sisto M, Lorusso L, Lisi S (2017) TLR2 signals via NF-kappaB to drive IL-15 production in salivary gland epithelial cells derived from patients with primary Sjogren’s syndrome. Clin Exp Med 17(3):341–350PubMedCrossRef
70.
Zurück zum Zitat Antonelli A, Ferrari SM, Giuggioli D, Ferrannini E, Ferri C, Fallahi P (2014) Chemokine (C-X-C motif) ligand (CXCL)10 in autoimmune diseases. Autoimmun Rev 13(3):272–280PubMedCrossRef Antonelli A, Ferrari SM, Giuggioli D, Ferrannini E, Ferri C, Fallahi P (2014) Chemokine (C-X-C motif) ligand (CXCL)10 in autoimmune diseases. Autoimmun Rev 13(3):272–280PubMedCrossRef
71.
Zurück zum Zitat Fallahi P, Ferrari SM, Ragusa F, Ruffilli I, Elia G, Paparo SR, Antonelli A (2020) Th1 chemokines in autoimmune endocrine disorders. J Clin Endocrinol Metab 105(4):dgz289 Fallahi P, Ferrari SM, Ragusa F, Ruffilli I, Elia G, Paparo SR, Antonelli A (2020) Th1 chemokines in autoimmune endocrine disorders. J Clin Endocrinol Metab 105(4):dgz289
72.
Zurück zum Zitat Mazzi V, Ferrari SM, Giuggioli D, Antonelli A, Ferri C, Fallahi P (2015) Role of CXCL10 in cryoglobulinemia. Clin Exp Rheumatol 33(3):433–436PubMed Mazzi V, Ferrari SM, Giuggioli D, Antonelli A, Ferri C, Fallahi P (2015) Role of CXCL10 in cryoglobulinemia. Clin Exp Rheumatol 33(3):433–436PubMed
73.
Zurück zum Zitat Klein RS (2004) Regulation of neuroinflammation: the role of CXCL10 in lymphocyte infiltration during autoimmune encephalomyelitis. J Cell Biochem 92(2):213–222PubMedCrossRef Klein RS (2004) Regulation of neuroinflammation: the role of CXCL10 in lymphocyte infiltration during autoimmune encephalomyelitis. J Cell Biochem 92(2):213–222PubMedCrossRef
74.
Zurück zum Zitat Schmidt A, Farine H, Keller MP, Sebastian A, Kozera L, Welford RWD, Strasser DS (2020) Immunoaffinity targeted mass spectrometry analysis of human plasma samples reveals an imbalance of active and inactive CXCL10 in primary Sjogren’s syndrome disease patients. J Proteome Res 19(10):4196–4209PubMedCrossRef Schmidt A, Farine H, Keller MP, Sebastian A, Kozera L, Welford RWD, Strasser DS (2020) Immunoaffinity targeted mass spectrometry analysis of human plasma samples reveals an imbalance of active and inactive CXCL10 in primary Sjogren’s syndrome disease patients. J Proteome Res 19(10):4196–4209PubMedCrossRef
75.
Zurück zum Zitat Blokland SLM, Kislat A, Homey B, Smithson GM, Kruize AA, Radstake T, van Roon JAG (2020) Decreased circulating CXCR3 + CCR9+T helper cells are associated with elevated levels of their ligands CXCL10 and CCL25 in the salivary gland of patients with Sjogren’s syndrome to facilitate their concerted migration. Scand J Immunol 91(3):e12852PubMedCrossRef Blokland SLM, Kislat A, Homey B, Smithson GM, Kruize AA, Radstake T, van Roon JAG (2020) Decreased circulating CXCR3 + CCR9+T helper cells are associated with elevated levels of their ligands CXCL10 and CCL25 in the salivary gland of patients with Sjogren’s syndrome to facilitate their concerted migration. Scand J Immunol 91(3):e12852PubMedCrossRef
76.
Zurück zum Zitat Psianou K, Panagoulias I, Papanastasiou AD, de Lastic AL, Rodi M, Spantidea PI, Degn SE, Georgiou P, Mouzaki A (2018) Clinical and immunological parameters of Sjogren’s syndrome. Autoimmun Rev 17(10):1053–1064PubMedCrossRef Psianou K, Panagoulias I, Papanastasiou AD, de Lastic AL, Rodi M, Spantidea PI, Degn SE, Georgiou P, Mouzaki A (2018) Clinical and immunological parameters of Sjogren’s syndrome. Autoimmun Rev 17(10):1053–1064PubMedCrossRef
77.
Zurück zum Zitat Christodoulou MI, Kapsogeorgou EK, Moutsopoulos NM, Moutsopoulos HM (2008) Foxp3+ T-regulatory cells in Sjogren’s syndrome: correlation with the grade of the autoimmune lesion and certain adverse prognostic factors. Am J Pathol 173(5):1389–1396PubMedPubMedCentralCrossRef Christodoulou MI, Kapsogeorgou EK, Moutsopoulos NM, Moutsopoulos HM (2008) Foxp3+ T-regulatory cells in Sjogren’s syndrome: correlation with the grade of the autoimmune lesion and certain adverse prognostic factors. Am J Pathol 173(5):1389–1396PubMedPubMedCentralCrossRef
78.
Zurück zum Zitat Both T, Dalm VA, van Hagen PM, van Daele PL (2017) Reviewing primary Sjogren’s syndrome: beyond the dryness - from pathophysiology to diagnosis and treatment. Int J Med Sci 14(3):191–200PubMedPubMedCentralCrossRef Both T, Dalm VA, van Hagen PM, van Daele PL (2017) Reviewing primary Sjogren’s syndrome: beyond the dryness - from pathophysiology to diagnosis and treatment. Int J Med Sci 14(3):191–200PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Chivasso C, Sarrand J, Perret J, Delporte C, Soyfoo MS (2021) The involvement of innate and adaptive immunity in the initiation and perpetuation of Sjogren's syndrome. Int J Mol Sci 22(2):658 Chivasso C, Sarrand J, Perret J, Delporte C, Soyfoo MS (2021) The involvement of innate and adaptive immunity in the initiation and perpetuation of Sjogren's syndrome. Int J Mol Sci 22(2):658
80.
Zurück zum Zitat Saito M, Otsuka K, Ushio A, Yamada A, Arakaki R, Kudo Y, Ishimaru N (2018) Unique phenotypes and functions of follicular helper T cells and regulatory T cells in Sjogren’s syndrome. Curr Rheumatol Rev 14(3):239–245PubMedPubMedCentralCrossRef Saito M, Otsuka K, Ushio A, Yamada A, Arakaki R, Kudo Y, Ishimaru N (2018) Unique phenotypes and functions of follicular helper T cells and regulatory T cells in Sjogren’s syndrome. Curr Rheumatol Rev 14(3):239–245PubMedPubMedCentralCrossRef
81.
Zurück zum Zitat Li X, Li X, Qian L, Wang G, Zhang H, Wang X, Chen K, Zhai Z, Li Q, Wang Y et al (2007) T regulatory cells are markedly diminished in diseased salivary glands of patients with primary Sjogren’s syndrome. J Rheumatol 34(12):2438–2445PubMed Li X, Li X, Qian L, Wang G, Zhang H, Wang X, Chen K, Zhai Z, Li Q, Wang Y et al (2007) T regulatory cells are markedly diminished in diseased salivary glands of patients with primary Sjogren’s syndrome. J Rheumatol 34(12):2438–2445PubMed
82.
Zurück zum Zitat Sylvester M, Son A, Schwartz DM (2022) The interactions between autoinflammation and type 2 immunity: from mechanistic studies to epidemiologic associations. Front Immunol 13:818039PubMedPubMedCentralCrossRef Sylvester M, Son A, Schwartz DM (2022) The interactions between autoinflammation and type 2 immunity: from mechanistic studies to epidemiologic associations. Front Immunol 13:818039PubMedPubMedCentralCrossRef
83.
Zurück zum Zitat Charles N, Hardwick D, Daugas E, Illei GG, Rivera J (2010) Basophils and the T helper 2 environment can promote the development of lupus nephritis. Nat Med 16(6):701–707PubMedPubMedCentralCrossRef Charles N, Hardwick D, Daugas E, Illei GG, Rivera J (2010) Basophils and the T helper 2 environment can promote the development of lupus nephritis. Nat Med 16(6):701–707PubMedPubMedCentralCrossRef
84.
Zurück zum Zitat Voulgarelis M, Tzioufas AG (2010) Pathogenetic mechanisms in the initiation and perpetuation of Sjogren’s syndrome. Nat Rev Rheumatol 6(9):529–537PubMedCrossRef Voulgarelis M, Tzioufas AG (2010) Pathogenetic mechanisms in the initiation and perpetuation of Sjogren’s syndrome. Nat Rev Rheumatol 6(9):529–537PubMedCrossRef
85.
Zurück zum Zitat Balkan E, Bilge N (2021) Expression levels of IL-17/IL-23 cytokine-targeting microRNAs 20, 21, 26, 155, and Let-7 in patients with relapsing-remitting multiple sclerosis. Neurol Res 43(9):778–783PubMedCrossRef Balkan E, Bilge N (2021) Expression levels of IL-17/IL-23 cytokine-targeting microRNAs 20, 21, 26, 155, and Let-7 in patients with relapsing-remitting multiple sclerosis. Neurol Res 43(9):778–783PubMedCrossRef
86.
Zurück zum Zitat Yildirim T, Yesilada E, Eren F, Apaydin H, Gulbay G (2021) Assessment of plasma microRNA potentials as a non-invasive biomarker in patients with axial spondyloarthropathy. Eur Rev Med Pharmacol Sci 25(2):620–625PubMed Yildirim T, Yesilada E, Eren F, Apaydin H, Gulbay G (2021) Assessment of plasma microRNA potentials as a non-invasive biomarker in patients with axial spondyloarthropathy. Eur Rev Med Pharmacol Sci 25(2):620–625PubMed
87.
Zurück zum Zitat Beretta F, Huang YF, Punga AR (2022) Towards personalized medicine in myasthenia gravis: role of circulating microRNAs miR-30e-5p, miR-150–5p and miR-21–5p. Cells 11(4):740 Beretta F, Huang YF, Punga AR (2022) Towards personalized medicine in myasthenia gravis: role of circulating microRNAs miR-30e-5p, miR-150–5p and miR-21–5p. Cells 11(4):740
88.
Zurück zum Zitat Punga AR, Punga T (2018) Circulating microRNAs as potential biomarkers in myasthenia gravis patients. Ann N Y Acad Sci 1412(1):33–40ADSPubMedCrossRef Punga AR, Punga T (2018) Circulating microRNAs as potential biomarkers in myasthenia gravis patients. Ann N Y Acad Sci 1412(1):33–40ADSPubMedCrossRef
89.
Zurück zum Zitat Sabre L, Punga T, Punga AR (2020) Circulating miRNAs as potential biomarkers in myasthenia gravis: tools for personalized medicine. Front Immunol 11:213PubMedPubMedCentralCrossRef Sabre L, Punga T, Punga AR (2020) Circulating miRNAs as potential biomarkers in myasthenia gravis: tools for personalized medicine. Front Immunol 11:213PubMedPubMedCentralCrossRef
90.
Zurück zum Zitat Ghafouri-Fard S, Azimi T, Hussen BM, Taheri M, Jalili Khoshnoud R (2021) A review on the role of non-coding RNAs in the pathogenesis of myasthenia gravis. Int J Mol Sci 22(23):12964 Ghafouri-Fard S, Azimi T, Hussen BM, Taheri M, Jalili Khoshnoud R (2021) A review on the role of non-coding RNAs in the pathogenesis of myasthenia gravis. Int J Mol Sci 22(23):12964
91.
Zurück zum Zitat Nakamaru K, Tomiyama T, Kobayashi S, Ikemune M, Tsukuda S, Ito T, Tanaka T, Yamaguchi T, Ando Y, Ikeura T et al (2020) Extracellular vesicles microRNA analysis in type 1 autoimmune pancreatitis: increased expression of microRNA-21. Pancreatology 20(3):318–324PubMedCrossRef Nakamaru K, Tomiyama T, Kobayashi S, Ikemune M, Tsukuda S, Ito T, Tanaka T, Yamaguchi T, Ando Y, Ikeura T et al (2020) Extracellular vesicles microRNA analysis in type 1 autoimmune pancreatitis: increased expression of microRNA-21. Pancreatology 20(3):318–324PubMedCrossRef
92.
Zurück zum Zitat Wade SM, McGarry T, Wade SC, Fearon U, Veale DJ (2020) Serum MicroRNA signature as a diagnostic and therapeutic marker in patients with psoriatic arthritis. J Rheumatol 47(12):1760–1767PubMedCrossRef Wade SM, McGarry T, Wade SC, Fearon U, Veale DJ (2020) Serum MicroRNA signature as a diagnostic and therapeutic marker in patients with psoriatic arthritis. J Rheumatol 47(12):1760–1767PubMedCrossRef
93.
Zurück zum Zitat Kim YJ, Yeon Y, Lee WJ, Shin YU, Cho H, Sung YK, Kim DR, Lim HW, Kang MH (2019) Comparison of MicroRNA expression in tears of normal subjects and Sjogren syndrome patients. Invest Ophthalmol Vis Sci 60(14):4889–4895PubMedCrossRef Kim YJ, Yeon Y, Lee WJ, Shin YU, Cho H, Sung YK, Kim DR, Lim HW, Kang MH (2019) Comparison of MicroRNA expression in tears of normal subjects and Sjogren syndrome patients. Invest Ophthalmol Vis Sci 60(14):4889–4895PubMedCrossRef
94.
Zurück zum Zitat Zhao G, Luo WD, Yuan Y, Lin F, Guo LM, Ma JJ, Chen HB, Tang H, Shu J (2022) LINC02381, a sponge of miR-21, weakens osteogenic differentiation of hUC-MSCs through KLF12-mediated Wnt4 transcriptional repression. J Bone Miner Metab 40(1):66–80PubMedCrossRef Zhao G, Luo WD, Yuan Y, Lin F, Guo LM, Ma JJ, Chen HB, Tang H, Shu J (2022) LINC02381, a sponge of miR-21, weakens osteogenic differentiation of hUC-MSCs through KLF12-mediated Wnt4 transcriptional repression. J Bone Miner Metab 40(1):66–80PubMedCrossRef
95.
Zurück zum Zitat Jafarzadeh M, Soltani BM (2020) Long noncoding RNA LOC400043 (LINC02381) inhibits gastric cancer progression through regulating Wnt signaling pathway. Front Oncol 10:562253PubMedPubMedCentralCrossRef Jafarzadeh M, Soltani BM (2020) Long noncoding RNA LOC400043 (LINC02381) inhibits gastric cancer progression through regulating Wnt signaling pathway. Front Oncol 10:562253PubMedPubMedCentralCrossRef
96.
Zurück zum Zitat Yan YM, Zheng JN, Wu LW, Rao QW, Yang QR, Gao D, Wang Q (2021) Prediction of a competing endogenous RNA co-expression network by comprehensive methods in systemic sclerosis-related interstitial lung disease. Front Genet 12:633059PubMedPubMedCentralCrossRef Yan YM, Zheng JN, Wu LW, Rao QW, Yang QR, Gao D, Wang Q (2021) Prediction of a competing endogenous RNA co-expression network by comprehensive methods in systemic sclerosis-related interstitial lung disease. Front Genet 12:633059PubMedPubMedCentralCrossRef
97.
Zurück zum Zitat Ho KH, Huang TW, Shih CM, Lee YT, Liu AJ, Chen PH, Chen KC (2021) Glycolysis-associated lncRNAs identify a subgroup of cancer patients with poor prognoses and a high-infiltration immune microenvironment. BMC Med 19(1):59PubMedPubMedCentralCrossRef Ho KH, Huang TW, Shih CM, Lee YT, Liu AJ, Chen PH, Chen KC (2021) Glycolysis-associated lncRNAs identify a subgroup of cancer patients with poor prognoses and a high-infiltration immune microenvironment. BMC Med 19(1):59PubMedPubMedCentralCrossRef
98.
Zurück zum Zitat Lahdeaho ML, Scheinin M, Vuotikka P, Taavela J, Popp A, Laukkarinen J, Koffert J, Koivurova OP, Pesu M, Kivela L et al (2019) Safety and efficacy of AMG 714 in adults with coeliac disease exposed to gluten challenge: a phase 2a, randomised, double-blind, placebo-controlled study. Lancet Gastroenterol Hepatol 4(12):948–959PubMedCrossRef Lahdeaho ML, Scheinin M, Vuotikka P, Taavela J, Popp A, Laukkarinen J, Koffert J, Koivurova OP, Pesu M, Kivela L et al (2019) Safety and efficacy of AMG 714 in adults with coeliac disease exposed to gluten challenge: a phase 2a, randomised, double-blind, placebo-controlled study. Lancet Gastroenterol Hepatol 4(12):948–959PubMedCrossRef
99.
Zurück zum Zitat Balaban DV, Mihai A, Dima A, Popp A, Jinga M, Jurcut C (2020) Celiac disease and Sjogren’s syndrome: a case report and review of literature. World J Clin Cases 8(18):4151–4161PubMedPubMedCentralCrossRef Balaban DV, Mihai A, Dima A, Popp A, Jinga M, Jurcut C (2020) Celiac disease and Sjogren’s syndrome: a case report and review of literature. World J Clin Cases 8(18):4151–4161PubMedPubMedCentralCrossRef
100.
Zurück zum Zitat Salat D, Tolosa E (2013) Levodopa in the treatment of Parkinson’s disease: current status and new developments. J Parkinsons Dis 3(3):255–269PubMedCrossRef Salat D, Tolosa E (2013) Levodopa in the treatment of Parkinson’s disease: current status and new developments. J Parkinsons Dis 3(3):255–269PubMedCrossRef
101.
Zurück zum Zitat Kchaou M, Ben Ali N, Hmida I, Fray S, Jamoussi H, Jalleli M, Echebbi S, Achouri A, Belal S (2015) Parkinsonism and Sjogren’s syndrome: a fortuitous association or a shared immunopathogenesis? Case Rep Med 2015:432910PubMedPubMedCentralCrossRef Kchaou M, Ben Ali N, Hmida I, Fray S, Jamoussi H, Jalleli M, Echebbi S, Achouri A, Belal S (2015) Parkinsonism and Sjogren’s syndrome: a fortuitous association or a shared immunopathogenesis? Case Rep Med 2015:432910PubMedPubMedCentralCrossRef
102.
Zurück zum Zitat Anwikar SR, Bandekar MS, Smrati B, Pazare AP, Tatke PA, Kshirsagar NA (2011) HAART induced adverse drug reactions: a retrospective analysis at a tertiary referral health care center in India. Int J Risk Saf Med 23(3):163–169PubMedCrossRef Anwikar SR, Bandekar MS, Smrati B, Pazare AP, Tatke PA, Kshirsagar NA (2011) HAART induced adverse drug reactions: a retrospective analysis at a tertiary referral health care center in India. Int J Risk Saf Med 23(3):163–169PubMedCrossRef
103.
Zurück zum Zitat Yu Z, Zhou X, Yu S, Xie H, Zheng S (2015) IL-15 is decreased upon CsA and FK506 treatment of acute rejection following heart transplantation in mice. Mol Med Rep 11(1):37–42PubMedCrossRef Yu Z, Zhou X, Yu S, Xie H, Zheng S (2015) IL-15 is decreased upon CsA and FK506 treatment of acute rejection following heart transplantation in mice. Mol Med Rep 11(1):37–42PubMedCrossRef
104.
Zurück zum Zitat Asadzadeh R, Ahmadpoor P, Nafar M, Samavat S, Nikoueinejad H, Hosseinzadeh M, Mamizadeh N, Hatami S, Masoumi E, Amirzargar A (2021) Association of IL-15 and IP-10 serum levels with Cytomegalovirus infection, CMV viral load and cyclosporine level after kidney transplantation. Rep Biochem Mol Biol 10(2):216–223PubMedPubMedCentralCrossRef Asadzadeh R, Ahmadpoor P, Nafar M, Samavat S, Nikoueinejad H, Hosseinzadeh M, Mamizadeh N, Hatami S, Masoumi E, Amirzargar A (2021) Association of IL-15 and IP-10 serum levels with Cytomegalovirus infection, CMV viral load and cyclosporine level after kidney transplantation. Rep Biochem Mol Biol 10(2):216–223PubMedPubMedCentralCrossRef
105.
Zurück zum Zitat Gay L, Melenotte C, Lopez A, Desnues B, Raoult D, Leone M, Mezouar S, Mege JL (2021) Impact of sex hormones on macrophage responses to Coxiella burnetii. Front Immunol 12:705088PubMedPubMedCentralCrossRef Gay L, Melenotte C, Lopez A, Desnues B, Raoult D, Leone M, Mezouar S, Mege JL (2021) Impact of sex hormones on macrophage responses to Coxiella burnetii. Front Immunol 12:705088PubMedPubMedCentralCrossRef
106.
Zurück zum Zitat Xiao L, Xiao W, Zhan F (2022) Integrative analyses of biomarkers and potential therapeutic drugs for rheumatoid arthritis. Ann Clin Lab Sci 52(1):141–153PubMed Xiao L, Xiao W, Zhan F (2022) Integrative analyses of biomarkers and potential therapeutic drugs for rheumatoid arthritis. Ann Clin Lab Sci 52(1):141–153PubMed
107.
Zurück zum Zitat Kedor C, Zernicke J, Hagemann A, Gamboa LM, Callhoff J, Burmester GR, Feist E (2016) A phase II investigator-initiated pilot study with low-dose cyclosporine A for the treatment of articular involvement in primary Sjogren’s syndrome. Clin Rheumatol 35(9):2203–2210PubMedCrossRef Kedor C, Zernicke J, Hagemann A, Gamboa LM, Callhoff J, Burmester GR, Feist E (2016) A phase II investigator-initiated pilot study with low-dose cyclosporine A for the treatment of articular involvement in primary Sjogren’s syndrome. Clin Rheumatol 35(9):2203–2210PubMedCrossRef
108.
Zurück zum Zitat Sullivan DA, Belanger A, Cermak JM, Berube R, Papas AS, Sullivan RM, Yamagami H, Dana MR, Labrie F (2003) Are women with Sjogren’s syndrome androgen-deficient? J Rheumatol 30(11):2413–2419PubMed Sullivan DA, Belanger A, Cermak JM, Berube R, Papas AS, Sullivan RM, Yamagami H, Dana MR, Labrie F (2003) Are women with Sjogren’s syndrome androgen-deficient? J Rheumatol 30(11):2413–2419PubMed
109.
Zurück zum Zitat Morthen MK, Tellefsen S, Richards SM, Lieberman SM, Rahimi Darabad R, Kam WR, Sullivan DA (2019) Testosterone influence on gene expression in lacrimal glands of mouse models of Sjogren syndrome. Invest Ophthalmol Vis Sci 60(6):2181–2197PubMedPubMedCentralCrossRef Morthen MK, Tellefsen S, Richards SM, Lieberman SM, Rahimi Darabad R, Kam WR, Sullivan DA (2019) Testosterone influence on gene expression in lacrimal glands of mouse models of Sjogren syndrome. Invest Ophthalmol Vis Sci 60(6):2181–2197PubMedPubMedCentralCrossRef
Metadaten
Titel
Identification and verification of inflammatory biomarkers for primary Sjögren’s syndrome
verfasst von
Xiaodan Liu
Haojie Wang
Xiao Wang
Xiaodan Jiang
Yinji Jin
Ying Han
Zhihui Zhang
Publikationsdatum
20.02.2024
Verlag
Springer International Publishing
Erschienen in
Clinical Rheumatology / Ausgabe 4/2024
Print ISSN: 0770-3198
Elektronische ISSN: 1434-9949
DOI
https://doi.org/10.1007/s10067-024-06901-y

Weitere Artikel der Ausgabe 4/2024

Clinical Rheumatology 4/2024 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

RAS-Blocker bei Hyperkaliämie möglichst nicht sofort absetzen

14.05.2024 Hyperkaliämie Nachrichten

Bei ausgeprägter Nierenfunktionsstörung steigen unter der Einnahme von Renin-Angiotensin-System(RAS)-Hemmstoffen nicht selten die Serumkaliumspiegel. Was in diesem Fall zu tun ist, erklärte Prof. Jürgen Floege beim diesjährigen Allgemeinmedizin-Update-Seminar.

Gestationsdiabetes: In der zweiten Schwangerschaft folgenreicher als in der ersten

13.05.2024 Gestationsdiabetes Nachrichten

Das Risiko, nach einem Gestationsdiabetes einen Typ-2-Diabetes zu entwickeln, hängt nicht nur von der Zahl, sondern auch von der Reihenfolge der betroffenen Schwangerschaften ab.

Labor, CT-Anthropometrie zeigen Risiko für Pankreaskrebs

13.05.2024 Pankreaskarzinom Nachrichten

Gerade bei aggressiven Malignomen wie dem duktalen Adenokarzinom des Pankreas könnte Früherkennung die Therapiechancen verbessern. Noch jedoch klafft hier eine Lücke. Ein Studienteam hat einen Weg gesucht, sie zu schließen.

Battle of Experts: Sport vs. Spritze bei Adipositas und Typ-2-Diabetes

11.05.2024 DDG-Jahrestagung 2024 Kongressbericht

Im Battle of Experts traten zwei Experten auf dem Diabeteskongress gegeneinander an: Die eine vertrat die Auffassung „Sport statt Spritze“ bei Adipositas und Typ-2-Diabetes, der andere forderte „Spritze statt Sport!“ Am Ende waren sie sich aber einig: Die Kombination aus beidem erzielt die besten Ergebnisse.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.