Multi-Omic Biomarkers for Patient Stratification in Sjogren’s Syndrome—A Review of the Literature
Abstract
:1. Introduction
2. Materials and Methods
3. Results
3.1. Multi-Omic Biomarkers for Patient Stratification
3.1.1. Tear Biomarkers
Reference | Type of Study/Samples/Methods | Number (N) of pSS Patients and HCs Age (Mean ± SD) | Disease Signature Identified | Correlations with Clinical Outcomes |
---|---|---|---|---|
BIOMARKERS IN TEARS | ||||
Cytokine profiling | ||||
Chen et al., 2019 [16] | Cross-sectional Tear strips for Schirmer I test Unstimulated (UWS) and stimulated (SWS) saliva samples Method: Cytokine 27-plex Assay | N = 29 pSS 56.8 ± 13.0 years N = 20 sicca (non-SS) controls 51.7 ± 10.6 years N = 17 HCs 45.4 ± 10.9 years | Increased IL-1ra, IL-2, IL-4, IL-17A, IFN-γ, MIP-1b, and Rantes in pSS vs. non-SS/HCs (p < 0.05). | Increased dry eye severity level and ocular surface staining correlated with increased tear cytokine levels, except for IP-10. Negative correlations between Schirmer’s test and tears IL-1ra, IL-2, IL-4, IL-8, IL-12p70, IL-17A, IFN-γ, MIP-1b, and Rantes (r = 0.26–0.61, p < 0.05). |
Willems et al., 2021 [17] | Cross-sectional Tear samples Method: LUNARIS™ BioChip | N = 12 pSS 41.7 ± 13.3 years N = 13 HCs 43.0 ± 13.8 years | Tears: Increased I FN-γ, TNF-α, IL-2, IL-4, IL-6, IL-10 and IL-12p70 (left eye) and IL-5 (right eye) in pSS compared to non-SS and HCs (p < 0.005). | Schirmer test correlated to IL-2 (r = −0.702), IL-4 (r = −0.769), IL-10 (r = −0.839) and IL-12p70 (r = −0.753) left eye levels; IL-10 directly correlated with SPEED test score (r = 0.722; p = 0.0001) as well as NIKBUT score (r = −0.705; p = 0.00002). |
Metabolomic profiling | ||||
Urbanski et al., 2021 [18] | Cross-sectional Tear strips for Schirmer I test Method: mass spectrometry/liquid chromatography | N = 40 female pSS 63 years N = 40 non pSS sicca controls 58 years | 9 metabolites (serine, aspartate; dopamine and six lipids) defined a tear pSS metabolomic signature (ROC-AUC = 0.83) PCA analysis showed that 2 PC explained 74.5% variance defined by 8/9 metabolites; the six lipids were distributed in the PC 1 and the amino acids in the second one. | The association between the metabolomic signature and the pSS status was not altered by age, sex, use of anticholinergic drugs or presence of anti-SSA antibodies |
Proteomic profiling | ||||
Das et al., 2021 [20] | Cross-sectional Tears, Tear washes Saliva Cryopreserved parotid gland biopsy samples Methods High performance liquid chromatography HPLC/mass spectrometry MS shotgun proteomics analysis Biopsy staining with anti-PRG4 mAb Bead-based immunoassay using the AlphaLISA | Tears N = 22 pSS (F:M = 10:1) 60.0 ± 16.5 years N = 20 HCs (F:M = 13:7) 31.2 ± 11.4 years Tear washes N = 14 pSS (F:M = 13:1) 59.5 ± 12.0 years N = 29 HCs (F:M = 17:12) 34.1 ± 14.2 years | Tears: 83 upregulated and 112 unique downregulated proteins in pSS vs. HCs. Enriched pathways in pSS: leukocyte trans-endothelial migration, protein-lipid complex remodelling and collagen catabolic. Enriched pathways in HCs: glycolysis/gluconeogenesis and glycolysis in senescence, amino acid metabolism and VEGFA/VEGFR2 signalling pathway. Overall, there was a loss of glycolysis and metabolism but an elevation of immune processes in pSS tears samples. PRG4 in tear washes was significantly decreased in pSS (p < 0.01). | Not explored |
3.1.2. Saliva Biomarkers
3.1.3. Potential pSS Biomarkers in Peripheral Blood
Reference | Type of Study/Samples/Methods | Number (N) of pSS Patients and HCs Age (Mean ± SD) | Disease Signature Identified | Correlations with Clinical Outcomes |
---|---|---|---|---|
BIOMARKERS IN PERIPHERAL BLOOD | ||||
Immunophenotype profiling | ||||
Mingueneau et al., 2016 [30] | Cross-sectional PBMC samples, a subset of paired SG biopsies Method: CyTOF, immunohistochemistry | N = 49 pSS 53 years N = 45 sicca (non-SS) and HCs 54 years | SG biopsies: increased activated CD8+ T cells, terminally differentiated plasma cells, and activated epithelial cells Blood: 6-cell signature: decreased CD4, memory B-cells, plasmacytoid dendritic cell, and increased activated CD4 and CD8 T cells and plasmablasts. | The blood cellular components correlated with clinical parameters clustered patients into subsets with distinct disease activity and glandular inflammation. |
Van der Kroef et al., 2020 [31] | Cross-sectional, PBMC samples Methods: Luminex, CyTOF | N = 88 SSc 54 years N = 31 SLE 43 years N = 23 pSS 56 years N = 44 HCs 50 years | pSS patients have increased HLA-DR CD4+ and CD8+ frequencies and reduced memory B cells and pDCs compared to HCs. | Not explored in pSS |
Szabó et al., 2021 [32] | Cross-sectional PBMCs samples Methods: flow cytometry, functional analysis and ELISA. | N = 38 pSS 54 years N = 27 HCs 46 years | pSS patients showed a significant increase in activated T follicular helper cells. Frequencies of T follicular regulatory cells were increased in autoantibody La positive patients compared to seronegative pSS. Transitional and naïve B cells increased, memory B cells decreased, | The percentage of activated T follicular helper cells showed a positive correlation with the levels of anti-La/SSB autoantibody and with serum IgA titre. Frequency of Tfh1 positive correlation with levels of serum IgG and anti-LA/SSB autoantibody. |
Martin-Gutierrez et al., 2021 [33] | Cross-sectional PBMC samples Method: Flow cytometry/ML | N = 45 pSS 59 (30–78) N = 29 SLE 48 (21–72) N = 14 SLE/SS 55 (26–56) N = 31 HCs | Patients with SS/SLE and SLE/SS shared immunological signatures. A signature comprising 5/29 immune cell subsets studied: transitional Bm2′ cells, late memory Bm5 cells, IgD-CD27-B cells, and CD8+ naïve and CD8+ Tem cells stratified patients | ESR correlated with 4 CD8+ T cell, 3 CD4+ T cell and 2 B cell subpopulations, which drove patient stratification. Hgb level correlated with % CD8+ Tcm cells. Disease damage scores across correlated with %CD8+ T cell, including CD8+CD25–CD127, CD8+ responder T cells, and CD8+ Temra cells |
Single-cell transcriptomic profile | ||||
Hong et al., 2021 [34] | Cross-sectional PBMCs Methods: scRNAseq and Flow cytometry | N = 10 pSS patients 48.8 years N= 10 HCs 33 years | Two subpopulations expanded in pSS: one expressing cytotoxicity genes (CD4+ CTLs cytotoxic T lymphocyte), and another highly expressing T cell receptor (TCR) variable gene (CD4+ TRAV13-2+ T cell). Total T cells significantly higher in pSS vs. HCs (p = 0.008). The IL-1β expression in macrophages, TCL1A in B cells, and IFN response genes in most cell subsets were upregulated in pSS. Susceptibility genes including HLA-DRB5, CTLA4, and AQP3 were highly expressed in pSS. | Correlation between the percentage of CD4+ CTLs and clinical characteristics, such as ESR), anti-SSA positive, and ESSDAI but no significant correlation was found. |
Serum proteomics | ||||
Nishikawa et al., 2016 [35] | Cross-sectional Methods: high-throughput proteomic analysis, ELISA. | Discovery cohort: N = 30 pSS 61 years N = 30 HCs 40 years Validation cohort: N = 50 pSS 60 years | A total of 82 (57 upregulated and 25 downregulated) serum proteins were differentially expressed in patients pSS vs. HCs. Enriched pathways: “extracellular region”, “chemokine signalling pathway”, “downstream of TNF-α”, “platelet activation”, and “platelet degranulation”. Nine proteins correlated with disease activity in the discovery cohort. In the validation cohort five proteins: CXCL13, TNF-R2, CD48, BAFF, and PD-L2 showed a correlation with ESSDAI, and therefore, were proposed as disease activity-associated biomarkers. | Serum concentrations of CXCL13, TNF-R2, and CD48 were positively correlated with that of immunoglobulin (Ig) G. TNF-R2 was negatively correlated with unstimulated salivary. BAFF was negatively correlated with the excretion rate in the submandibular gland. |
Padern et al., 2021 [36] | Cross-sectional Methods: BioPlex, ELISA | N = 42 pSS 62.5 years N = 28 RA 60.5 years N = 25 SLE 40 years | Eight biomarkers could statistically discriminate samples from pSS versus SLE patients. Four could statistically discriminate pSS patients from RA patients. None of the studied biomarkers could simultaneously discriminate pSS from RA and SLE. We, therefore, determined the positive predictive value (PPV), sensitivity, and specificity of different combinations of BDNF, I-TAC/CXCL11, sCD163 and Fractalkine/CX3CL1 concentrations. These biomarkers were chosen because they were those most strongly associated with distinguishing pSS from the other AIDs. | Negative correlation between pSS activity according to the ESSDAI score and serum sCD163 concentrations. |
Serum metabolomic profiling | ||||
Xu et al., 2021 [39] | Cross-sectional Serum samples Method: non-targeted GC-MS | Discovery: 90 pSS patients, M:F = 1/10) 53 years 153 HCs (male/female: 1/10) 50.4 years Validation: 119 pSS, M:F = 1/10 52.9 years 143 HCs (M:F = 1/10 50.23 years | Increased alanine, tryptophan, glycolic acid, pelargonic acid, cis-1-2-dihydro-1-2-naphthalenediol, etc., and decrease in catechol, anabasine, 3-6-anhydro-D-galactose, beta-gentiobiose and ethanolamine in pSS patients vs. HCs. Stearic acid and linoleic acid distinguished pSS from HCs (ROC−AUC = 0.97−0.98) | Inflammatory markers, autoantibodies and Ig G levels correlated with various metabolite levels. |
3.1.4. Genetic and Epigenetic Studies
Multi-omic pSS Signatures
4. Discussion
5. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Conflicts of Interest
References
- Acar-Denizli, N.; Horváth, I.F.; Mandl, T.; Priori, R.; Vissink, A.; Hernandez-Molina, G.; Armagan, B.; Praprotnik, S.; Sebastian, A.; Bartoloni, E.; et al. Systemic phenotype related to primary Sjögren’s syndrome in 279 patients carrying isolated anti-La/SSB antibodies. Clin. Exp. Rheumatol. 2020, 38, 85–94. [Google Scholar] [PubMed]
- Alani, H.; Henty, J.R.; Thompson, N.L.; Jury, E.; Ciurtin, C. Systematic review and meta-analysis of the epidemiology of polyautoimmunity in Sjögren’s syndrome (secondary Sjögren’s syndrome) focusing on autoimmune rheumatic diseases. Scand. J. Rheumatol. 2018, 47, 141–154. [Google Scholar] [CrossRef] [PubMed]
- Shiboski, C.H.; Shiboski, S.C.; Seror, R.; Criswell, L.A.; Labetoulle, M.; Lietman, T.M.; Rasmussen, A.; Scofield, H.; Vitali, C.; Bowman, S.J. American College of Rheumatology/European League Against Rheumatism classification criteria for primary Sjögren’s syndrome: A consensus and data-driven methodology involving three international patient cohorts. Arthritis Rheumatol. 2017, 69, 35–45. [Google Scholar] [CrossRef] [PubMed]
- Mavragani, C.P.; Moutsopoulos, H.M. Primary versus secondary Sjögren syndrome: Is it time to reconsider these terms? J. Rheumatol. 2019, 46, 665–666. [Google Scholar] [CrossRef]
- Basiaga, M.L.; Stern, S.M.; Mehta, J.J.; Edens, C.; Randell, R.L.; Pomorska, A.; Irga-Jaworska, N.; Ibarra, M.F.; Bracaglia, C.; Nicolai, R.; et al. Childhood Sjogren syndrome: Features of an international cohort and application of the 2016 ACR/EULAR classification criteria. Rheumatology 2021, 60, 3144–3155. [Google Scholar] [CrossRef]
- Ciurtin, C.; Cho, Y.; Al-Obaidi, M.; Jury, E.C.; Price, E.J. Barriers to translational research in Sjögren’s syndrome with childhood onset: Challenges of recognising and diagnosing an orphan rheumatic disease. Lancet Rheumatol. 2021, 3, e138–e148. [Google Scholar] [CrossRef]
- Seror, R.; Bowman, S.J.; Brito-Zeron, P.; Theander, E.; Bootsma, H.; Tzioufas, A.; Gottenberg, J.E.; Ramos-Casals, M.; Dörner, T.; Ravaud, P.; et al. EULAR Sjogren’s syndrome disease activity index (ESSDAI): A user guide. RMD Open 2015, 1, e000022. [Google Scholar] [CrossRef] [Green Version]
- Price, E.; Allen, A.; Rauz, S.; Tappuni, A.; Sutcliffe, N.; Bombardieri, M.; Carty, S.; Ciurtin, C.; Crampton, B.; Duncalfe, L.; et al. The management of Sjogren’s syndrome: British Society for Rheumatology guideline scope. Rheumatology 2021, 60, 2122–2127. [Google Scholar] [CrossRef]
- Doolan, G.; Faizal, N.M.; Foley, C.; Al-Obaidi, M.; Jury, E.C.; Price, E.; Ramanan, A.V.; Lieberman, S.M.; Ciurtin, C. Treatment strategies for Sjogren’s syndrome with childhood onset: A systematic review of the literature. Rheumatology 2022, 61, 892–912. [Google Scholar] [CrossRef]
- Ramos-Casals, M.; Brito-Zerón, P.; Bombardieri, S.; Bootsma, H.; De Vita, S.; Dörner, T.; Fisher, B.A.; Gottenberg, J.E.; Hernandez-Molina, G.; Kocher, A.; et al. EULAR recommendations for the management of Sjögren’s syndrome with topical and systemic therapies. Ann. Rheum. Dis. 2020, 79, 3–18. [Google Scholar] [CrossRef] [Green Version]
- Sandhya, P.; Theyilamannil Kurien, B.; Danda, D.; Hal Scofield, R. Update on pathogenesis of Sjogren’s syndrome. Curr. Rheumatol. Rev. 2017, 13, 5–22. [Google Scholar] [CrossRef] [PubMed]
- Ng, W.F. Primary Sjögren’s syndrome research and therapy: Has a new dawn arrived? Curr. Pharm. Biotechnol. 2012, 13, 1987–1988. [Google Scholar] [CrossRef] [PubMed]
- Mariette, X.; Seror, R.; Quartuccio, L.; Baron, G.; Salvin, S.; Fabris, M.; Desmoulins, F.; Nocturne, G.; Ravaud, P.; De Vita, S. Efficacy and safety of belimumab in primary Sjogren’s syndrome: Results of the BELISS open-label phase II study. Ann. Rheum. Dis. 2015, 74, 526–531. [Google Scholar] [CrossRef]
- Meiners, P.M.; Vissink, A.; Kroese, F.G.M.; Spijkervet, F.K.L.; Smitt-Kamminga, N.S.; Abdulahad, W.H.; Bulthuis-Kuiper, J.; Brouwer, E.; Arends, S.; Bootsma, H. Abatacept treatment reduces disease activity in early primary Sjogren’s syndrome (open-label proof of concept ASAP study). Ann. Rheum. Dis. 2014, 73, 1393–1396. [Google Scholar] [CrossRef] [PubMed]
- Fisher, B.A.; Everett, C.C.; Rout, J.; O’Dwyer, J.L.; Emery, P.; Pitzalis, C.; Ng, W.F.; Carr, A.; Pease, C.T.; Price, E.J.; et al. Effect of rituximab on a salivary gland ultrasound score in primary Sjogren’s syndrome: Results of the TRACTISS randomised double-blind multicentre substudy. Ann. Rheum. Dis. 2018, 77, 412–416. [Google Scholar] [CrossRef] [PubMed]
- Chen, X.; Aqrawi, L.A.; Utheim, T.P.; Tashbayev, B.; Utheim, Ø.A.; Reppe, S.; Hove, L.H.; Herlofson, B.B.; Singh, P.B.; Palm, Ø.; et al. Elevated cytokine levels in tears and saliva of patients with primary Sjogren’s syndrome correlate with clinical ocular and oral manifestations. Sci. Rep. 2019, 9, 7319. [Google Scholar] [CrossRef]
- Willems, B.; Tong, L.; Minh, T.D.T.; Pham, N.D.; Nguyen, X.H.; Zumbansen, M. Novel cytokine multiplex assay for tear fluid analysis in Sjogren’s syndrome. Ocul. Immunol. Inflamm. 2021, 29, 1639–1644. [Google Scholar] [CrossRef]
- Urbanski, G.; Assad, S.; Chabrun, F.; de la Barca, J.M.C.; Blanchet, O.; Simard, G.; Lenaers, G.; Prunier-Mirebeau, D.; Gohier, P.; Lavigne, C.; et al. Tear metabolomics highlights new potential biomarkers for differentiating between Sjogren’s syndrome and other causes of dry eye. Ocul. Surf. 2021, 22, 110–116. [Google Scholar] [CrossRef]
- Knuplez, E.; Marsche, G. An updated review of pro-and anti-inflammatory properties of plasma lysophosphatidylcholines in the vascular system. Int. J. Mol. Sci. 2020, 21, 4501. [Google Scholar] [CrossRef]
- Das, N.; Menon, N.G.; De Almeida, L.G.; Woods, P.S.; Heynen, M.L.; Jay, G.D.; Caffery, B.; Jones, L.; Krawetz, R.; Schmidt, T.A.; et al. Proteomics analysis of tears and saliva from Sjogren’s syndrome patients. Front. Pharmacol. 2021, 12, 787193. [Google Scholar] [CrossRef]
- Ramos-Casals, M.; Brito-Zerón, P.; Sisó-Almirall, A.; Bosch, X. Primary Sjogren syndrome. BMJ 2012, 344, e3821. [Google Scholar] [CrossRef] [Green Version]
- Brito-Zeron, P.; Baldini, C.; Bootsma, H.; Bowman, S.J.; Jonsson, R.; Mariette, X.; Sivils, K.; Theander, E.; Tzioufas, A.; Ramos-Casals, M. Sjögren syndrome. Nat. Rev. Dis. Primers 2016, 2, 16047. [Google Scholar] [CrossRef]
- Olsson, P.; Ekblad, F.; Hassler, A.; Bengtsson, M.; Warfvinge, G.; Mandl, T.; Kvarnström, M. Complications after minor salivary gland biopsy: A retrospective study of 630 patients from two Swedish centres. Scand. J. Rheumatol. 2022, 1–9. [Google Scholar] [CrossRef] [PubMed]
- Kang, E.H.; Lee, Y.J.; Hyon, J.Y.; Yun, P.Y.; Song, Y.W. Salivary cytokine profiles in primary Sjogren’s syndrome differ from those in non-Sjogren sicca in terms of TNF-alpha levels and Th-1/Th-2 ratios. Clin. Exp. Rheumatol. 2011, 29, 970–976. [Google Scholar]
- Kageyama, G.; Saegusa, J.; Irino, Y.; Tanaka, S.; Tsuda, K.; Takahashi, S.; Sendo, S.; Morinobu, A. Metabolomics analysis of saliva from patients with primary Sjogren’s syndrome. Clin. Exp. Immunol. 2015, 182, 149–153. [Google Scholar] [CrossRef] [Green Version]
- Herrala, M.; Mikkonen, J.J.; Pesonen, P.; Lappalainen, R.; Tjäderhane, L.; Niemelä, R.K.; Seitsalo, H.; Salo, T.; Myllymaa, S.; Kullaa, A.M. Variability of salivary metabolite levels in patients with Sjogren’s syndrome. J. Oral. Sci. 2020, 63, 22–26. [Google Scholar] [CrossRef] [PubMed]
- Delaleu, N.; Mydel, P.; Kwee, I.; Brun, J.G.; Jonsson, M.V.; Jonsson, R. High fidelity between saliva proteomics and the biologic state of salivary glands defines biomarker signatures for primary Sjogren’s syndrome. Arthritis Rheumatol. 2015, 67, 1084–1095. [Google Scholar] [CrossRef]
- Verstappen, G.M.; Gao, L.; Pringle, S.; Haacke, E.A.; van der Vegt, B.; Liefers, S.C.; Patel, V.; Hu, Y.; Mukherjee, S.; Carman, J.; et al. The transcriptome of paired major and minor salivary gland tissue in patients with primary Sjögren’s Syndrome. Front. Immunol. 2021, 12, 681941. [Google Scholar] [CrossRef]
- Yamamoto, K. Pathogenesis of Sjogren’s syndrome. Autoimmun. Rev. 2003, 2, 13–18. [Google Scholar] [CrossRef]
- Mingueneau, M.; Boudaoud, S.; Haskett, S.; Reynolds, T.L.; Nocturne, G.; Norton, E.; Zhang, X.; Constant, M.; Park, D.; Wang, W.; et al. Cytometry by time-of-flight immunophenotyping identifies a blood Sjogren’s signature correlating with disease activity and glandular inflammation. J. Allergy Clin. Immunol. 2016, 137, 1809–1821. [Google Scholar] [CrossRef]
- van der Kroef, M.; van den Hoogen, L.L.; Mertens, J.S.; Blokland, S.L.; Haskett, S.; Devaprasad, A.; Carvalheiro, T.; Chouri, E.; Vazirpanah, N.; Cossu, M.; et al. Cytometry by time of flight identifies distinct signatures in patients with systemic sclerosis, systemic lupus erythematosus and Sjögrens syndrome. Eur. J. Immunol. 2020, 50, 119–129. [Google Scholar] [CrossRef] [PubMed]
- Szabo, K.; Jambor, I.; Szanto, A.; Horvath, I.F.; Tarr, T.; Nakken, B.; Szodoray, P.; Papp, G. The imbalance of circulating follicular T helper cell subsets in primary Sjögren’s syndrome associates with serological alterations and abnormal B-cell distribution. Front. Immunol. 2021, 12, 639975. [Google Scholar] [CrossRef] [PubMed]
- Martin-Gutierrez, L.; Peng, J.; Thompson, N.L.; Robinson, G.A.; Naja, M.; Peckham, H.; Wu, W.; J’bari, H.; Ahwireng, N.; Waddington, K.E.; et al. Stratification of patients with Sjögren’s syndrome and patients with systemic lupus erythematosus according to two shared immune cell signatures, with potential therapeutic implications. Arthritis Rheumatol. 2021, 73, 1626–1637. [Google Scholar] [CrossRef] [PubMed]
- Hong, X.; Meng, S.; Tang, D.; Wang, T.; Ding, L.; Yu, H.; Li, H.; Liu, D.; Dai, Y.; Yang, M. Single-cell RNA sequencing reveals the expansion of cytotoxic CD4+ T lymphocytes and a landscape of immune cells in primary Sjögren’s syndrome. Front. Immunol. 2020, 11, 594658. [Google Scholar] [CrossRef]
- Nishikawa, A.; Suzuki, K.; Kassai, Y.; Gotou, Y.; Takiguchi, M.; Miyazaki, T.; Yoshimoto, K.; Yasuoka, H.; Yamaoka, K.; Morita, R.; et al. Identification of definitive serum biomarkers associated with disease activity in primary Sjogren’s syndrome. Arthritis Res. Ther. 2016, 18, 106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Padern, G.; Duflos, C.; Ferreira, R.; Assou, S.; Guilpain, P.; Maria, A.T.J.; Goulabchand, R.; Galea, P.; Jurtela, M.; Jorgensen, C.; et al. Identification of a novel serum proteomic signature for primary Sjögren’s syndrome. Front. Immunol. 2021, 12, 631539. [Google Scholar] [CrossRef]
- Fernández-Ochoa, Á.; Borrás-Linares, I.; Quirantes-Piné, R.; Alarcón-Riquelme, M.E.; Beretta, L.; Segura-Carretero, A.; Consortium, P.C. Discovering new metabolite alterations in primary sjögren’s syndrome in urinary and plasma samples using an HPLC-ESI-QTOF-MS methodology. J. Pharm. Biomed. Anal. 2020, 179, 112999. [Google Scholar] [CrossRef]
- Bengtsson, A.A.; Trygg, J.; Wuttge, D.M.; Sturfelt, G.; Theander, E.; Donten, M.; Moritz, T.; Sennbro, C.J.; Torell, F.; Lood, C.; et al. Metabolic profiling of systemic lupus erythematosus and comparison with primary Sjögren’s syndrome and systemic sclerosis. PLoS ONE 2016, 11, e0159384. [Google Scholar] [CrossRef]
- Xu, T.; Guo, Y.; Lu, J.; Shan, J.; Lin, L.; Qian, W.; Chen, W.; Wang, J.; Lv, X.; Ke, M.; et al. Untargeted serum metabolomics and potential biomarkers for Sjogren’s syndrome. Clin. Exp. Rheumatol. 2021, 39, 23–29. [Google Scholar] [CrossRef]
- Bordron, A.; Devauchelle-Pensec, V.; Le Dantec, C.; Capdeville, A.; Brooks, W.H.; Renaudineau, Y. Epigenetics in primary Sjögren’s syndrome. Adv. Exp. Med. Biol. 2020, 1253, 285–308. [Google Scholar]
- Nezos, A.; Mavragani, C.P. Contribution of genetic factors to Sjögren’s syndrome and Sjögren’s syndrome related lymphomagenesis. J. Immunol. Res. 2015, 2015, 754825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Imgenberg-Kreuz, J.; Rasmussen, A.; Sivils, K.; Nordmark, G. Genetics and epigenetics in primary Sjogren’s syndrome. Rheumatology 2021, 60, 2085–2098. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Teruel, M.; Barturen, G.; Martínez-Bueno, M.; Castellini-Pérez, O.; Barroso-Gil, M.; Povedano, E.; Kerick, M.; Català-Moll, F.; Makowska, Z.; Buttgereit, A.; et al. Integrative epigenomics in Sjögren´s syndrome reveals novel pathways and a strong interaction between the HLA, autoantibodies and the interferon signature. Sci. Rep. 2021, 11, 23292. [Google Scholar] [CrossRef] [PubMed]
- Tasaki, S.; Suzuki, K.; Nishikawa, A.; Kassai, Y.; Takiguchi, M.; Kurisu, R.; Okuzono, Y.; Miyazaki, T.; Takeshita, M.; Yoshimoto, K.; et al. Multiomic disease signatures converge to cytotoxic CD8 T cells in primary Sjögren’s syndrome. Ann. Rheum. Dis. 2017, 76, 1458–1466. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- James, J.A.; Guthridge, J.M.; Chen, H.; Lu, R.; Bourn, R.L.; Bean, K.; Munroe, M.E.; Smith, M.; Chakravarty, E.; Baer, A.N.; et al. Unique Sjögren’s syndrome patient subsets defined by molecular features. Rheumatology 2020, 59, 860–868. [Google Scholar] [CrossRef] [PubMed]
- Soret, P.; Le Dantec, C.; Desvaux, E.; Foulquier, N.; Chassagnol, B.; Hubert, S.; Jamin, C.; Barturen, G.; Desachy, G.; Devauchelle-Pensec, V.; et al. A new molecular classification to drive precision treatment strategies in primary Sjogren’s syndrome. Nat. Commun. 2021, 12, 3523. [Google Scholar] [CrossRef]
- Barturen, G.; Babaei, S.; Català-Moll, F.; Martínez-Bueno, M.; Makowska, Z.; Martorell-Marugán, J.; Carmona-Sáez, P.; Toro-Domínguez, D.; Carnero-Montoro, E.; Teruel, M.; et al. Integrative analysis reveals a molecular stratification of systemic autoimmune diseases. Arthritis Rheumatol. 2021, 73, 1073–1085. [Google Scholar] [CrossRef]
- Tsang-A-Sjoe, M.W.; Bultink, I.E. New developments in systemic lupus erythematosus. Rheumatology 2021, 60, vi21–vi28. [Google Scholar] [CrossRef]
- Shah, N.R.; Noll, B.D.; Stevens, C.B.; Brennan, M.T.; Mougeot, F.B.; Mougeot, J.L.C. Biosemantics guided gene expression profiling of Sjogren’s syndrome: A comparative analysis with systemic lupus erythematosus and rheumatoid arthritis. Arthritis Res. Ther. 2017, 19, 192. [Google Scholar] [CrossRef] [Green Version]
- Sada, P.R.; Isenberg, D.; Ciurtin, C. Biologic treatment in Sjogren’s syndrome. Rheumatology 2015, 54, 219–230. [Google Scholar] [CrossRef] [Green Version]
- Fox, R.I.; Fox, C.M.; Gottenberg, J.E.; Dörner, T. Treatment of Sjogren’s syndrome: Current therapy and future directions. Rheumatology 2021, 60, 2066–2074. [Google Scholar] [CrossRef]
- Seror, R.; Baron, G.; Camus, M.; Cornec, D.; Perrodeau, E.; Bowman, S.J.; Bombardieri, M.; Bootsma, H.; Gottenberg, J.E.; Fisher, B.; et al. Development and preliminary validation of the Sjogren’s Tool for Assessing Response (STAR): A consensual composite score for assessing treatment effect in primary Sjogren’s syndrome. Ann. Rheum. Dis. 2022, 81, 979–989. [Google Scholar] [CrossRef] [PubMed]
- Lessard, C.J.; Li, H.; Adrianto, I.; Ice, J.A.; Rasmussen, A.; Grundahl, K.M.; Kelly, J.A.; Dozmorov, M.G.; Miceli-Richard, C.; Bowman, S.; et al. Variants at multiple loci implicated in both innate and adaptive immune responses are associated with Sjogren’s syndrome. Nat. Genet. 2013, 45, 1284–1292. [Google Scholar] [CrossRef] [PubMed]
- Cruz-Tapias, P.; Rojas-Villarraga, A.; Maier-Moore, S.; Anaya, J.M. HLA and Sjogren’s syndrome susceptibility. A meta-analysis of worldwide studies. Autoimmun. Rev. 2012, 11, 281–287. [Google Scholar] [CrossRef] [PubMed]
- Lucchesi, D.; Pitzalis, C.; Bombardieri, M. EBV and other viruses as triggers of tertiary lymphoid structures in primary Sjogren’s syndrome. Expert Rev. Clin. Immunol. 2014, 10, 445–455. [Google Scholar] [CrossRef] [PubMed]
- Nezos, A.; Gravani, F.; Tassidou, A.; Kapsogeorgou, E.K.; Voulgarelis, M.; Koutsilieris, M.; Crow, M.K.; Mavragani, C.P. Type I and II interferon signatures in Sjogren’s syndrome pathogenesis: Contributions in distinct clinical phenotypes and Sjogren’s related lymphomagenesis. J. Autoimmun. 2015, 63, 47–58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Reference | Type of Study/Samples/Methods | Number (N) of pSS Patients and HCs Age (Mean ± SD) | Disease Signature Identified | Correlations with Clinical Outcomes |
---|---|---|---|---|
BIOMARKERS IN SALIVA/SALIVARY GLANDS | ||||
Cytokine profiling | ||||
Kang et al., 2011 [24] | Cross-sectional Unstimulated saliva samples Method: Luminex® bead-based assay | N = 30 pSS 49.9 ± 9.0 years N = 30 sicca (non-SS) 51.5 ± 10.0 years N = 25 HCs 49.4 ± 9.5 years | Saliva: Increased IFN-γ, IL-1, IL-4, IL-10, IL-12p40, IL-17, and TNF-α levels in pSS vs. non-SS and HCs (p < 0.005). IL-6 levels higher in pSS vs. HCs (p = 0.011). INF-γ/IL-4; TNF-α/IL-4 higher in pSS vs. HCs (p = 0.028, p = 0.038, respectively). | No correlations were found between any salivary cytokine levels and clinical parameters. Unstimulated saliva flow rate correlated with INF-γ/IL-4 ratio (r = 0.411 p = 0.024) and focus score correlated with TNF-α/IL-4 ratio (r = 0.581, p = 0.023) in pSS, suggesting a predominant Th1 saliva signature. |
Chen et al., 2019 [16] | Cross-sectional Tear strips for Schirmer I test Unstimulated (UWS) and stimulated (SWS) saliva samples Method: Cytokine 27-plex Assay | N = 29 pSS 56.8 ± 13.0 years N = 20 sicca (non-SS) controls 51.7 ± 10.6 years N = 17 HCs 45.4 ± 10.9 years | Saliva: increased IP-10 in pSS vs. non-SS/HCs. Both pSS and non-SS subjects had higher MIP-1α levels than HCs (p < 0.05). | UWS and SWS correlated negatively with MIP-1a saliva level (r = −0.276, p = 0.046 and r = −0.282, p = 0.040, respectively). |
Metabolomic profiling | ||||
Kageyama et al., 2015 [25] | Cross-sectional Unstimulated saliva samples Method: Gas chromatography mass spectrometry (GC-MS) analysis | N = 14 female pSS 44.2 ± 13.01 years N = 21 HCs 46.7± 10.24 years | 41 of the metabolites were reduced in pSS patients compared to HCs (p < 0.05). Decreased glycine, tyrosine, uric acid and fucose in pSS vs. HCs in PCA analysis. | Patient stratification based on saliva metabolome depicted two groups: one younger (p = 0.082) and with a lower SG biopsy score (p = 0.014). Glycine levels differentiate between the two groups. |
Herrala et al., 2020 [26] | Longitudinal study Stimulated saliva samples Method: proton nuclear magnetic resonance (1 H-NMR) spectroscopy | 56 samples from N = 14 female pSS patients during four laboratory visits within 20 weeks. 48.6 years N = 15 HCs (mean age 49.8 years) | Increased choline in pSS patients at each time point (p ≤ 0.015), taurine at the last three time points (p ≤ 0.013), alanine at the last two time points (p ≤ 0.007) and glycine at the last time point (p = 0.005). Inter-individual variation observed for glycine (p ≤ 0.007, all time points), choline (p ≤ 0.033, three last time points) and alanine (p = 0.028, baseline). | Not explored |
Proteomic profiling | ||||
Delaleu et al., 2015 [27] | Cross-sectional Unstimulated whole saliva Methods: 187-plex capture antibody-based assay | Saliva N = 48 pSS (females) 47 years N = 24 non-SS cohort (12 RA patients + 12 HCs) 51 years | Significant differences in 61 biomarkers in pSS vs. controls (p < 0.001). FGF-4 levels lower in pSS (the only decreased protein). A biomarker signature comprising clusterin, IL-5, FGF-4, and IL-4 yielded accurate group prediction for 93.8% of pSS and 100% of non-SS controls classification. | No biomarkers correlated with salivary flow rates |
Das et al., 2021 [20] | Cross-sectional Tears, Tear washes Saliva Cryopreserved parotid gland biopsy samples Methods High performance liquid chromatography HPLC/mass spectrometry MS shotgun proteomics analysis Biopsy staining with anti-PRG4 mAb Bead-based immunoassay using the AlphaLISA | Saliva N = 30 pSS (F:M = 22:8) 45.2 ± 14.6 years N = 10 HCs (F:M =5:5) 46.8 ± 14.5 years | Saliva: 104 upregulated and 42 downregulated proteins in pSS vs. HCs. Enriched pathways in pSS: JAK-STAT signalling after IL-12 stimulation, superoxide metabolic process and phagocytosis. Enriched pathways in HCs: neutrophil degranulation, negative regulation of peptidase activity; 2.3-fold increase in PRG4 in pSS (p < 0.05). PRG4 expression was found in both the serous acini and the striated duct on parotid gland biopsies (N = 4). | Not explored |
Salivary gland tissue transcriptomic profiling | ||||
Vertstappen et al., 2021 [28] | Cross-sectional Parotid and labial gland biopsy Methods: RNAseq—HiSeq 2500 System (Illumina). Multiplexed bead-based immunoassays for cytokine profiling Assessment of CD45-positive infiltrates on SG biopsies | N = 34 pSS with 51 paired (parotid and labial) biopsies 21 biopsy positive 13 biopsy negative 52 years N = 20 non SS sicca controls 17 biopsy negative 50 years | Parotid glands: 1041 up-regulated and 194 down-regulated DEG and labial glands: 581 and 43, respectively, between biopsy positive pSS and controls. The top 20 up-regulated genes in both tissues were mostly B-cell or T cell related. No significant differences between biopsy negative pSS and controls. Transcript expression levels correlated between parotid and labial glands (R2 = 0.86, p < 0.0001). Signatures enriched in biopsy-positive pSS compared with either biopsy-negative pSS or controls: IFN-α signalling, IL-12/IL-18 signalling, CD3/CD28 T cell activation, CD40 signalling in B-cells, double negative type-2 B-cells, and FcRL4+ B-cells. Strong correlation between the IFN-α score in PBMCs and SGs. | No difference in ESSDAI, unstimulated salivary flow or ESSPRI in patient DEG clusters. |
Reference | Type of Study/Samples/Methods | Number (N) of pSS Patients and HCs Age (Mean ± SD) | Disease Signature Identified | Correlations with Clinical Outcomes |
---|---|---|---|---|
MULTIOMIC SIGNATURES | ||||
Tasaki et al., 2017 [44] | Cross-sectional Methods: whole blood Transcriptomes microarrays, serum proteomes and peripheral immunophenotyping | N = 36 pSS patients 61 years N = 36 HCs 39 years | pSS gene signature predominantly involves the interferon signature including HERC5, EPSTI1 and CMPK2and ADAMs substrates. SGS was significantly overlapped with SS-causing genes indicated by a genome-wide association study as the regions that code genes in the SS gene signature were hypomethylated. Combining the molecular signatures with immunophenotypic profiles revealed that cytotoxic CD8 T cells were associated with SGS. | SGS positively correlated with the levels of autoantibodies, including anti-Ro/SSA and anti-La/SSB antigen–antibodies and serum IgG levels. Most ADAM substrates showed significant positive correlations with ESSDAI. |
James et al., 2019 [45] | Cross-sectional Methods: RNAseq, Bioplex, ELISA, Luminex | N = 47 pSS patients 52 years | Three clusters of patients were identified based on transcriptomic analysis. No demographic differences between clusters. C1 weakest IFN signature and minimal activity of inflammation gene modules. C2 strongest IFN signature, strongest inflammation module. Higher ESSDAI scores. More patients presented anti-Ro and anti-La antibodies and higher levels. Higher levels of cytokines, such as LIGHT and Blys. CXCL13. C3 intermediate IFN signature, low activity of the inflammation modules.c3 higher levels of IL1, IL2 IL2RA. | C2 cluster presented higher ESSDAI scores |
Soret et al., 2021 [46] | Cross-sectional Whole blood Methods: RNAseq, GWAS, Methylation and flow cytometry Serum sample Methods: Luminex, automated chemiluminescent immunoanalyzer (IDS-iSYS) | N = 304 pSS patients 58 years N = 330 HCs 53 years | Clustering of pSS samples based on transcriptomic data identified 4 different clusters (C1, C2, C3 and C4). C1 was enriched with IFN-related pathways, present an enriched up-regulated IFN signalling pathway; 35 SNPS were detected in genes associated with the immune system (HLA-DQB1, HLADQA1, HLA-DRA, HLA-C, HLA-G), signal transduction (NOTCH4), developmental biology (POU5F1), gene expression (DDX39B). Methylation in 87 genes. T cell lymphopenia. Increased in The IFNγ-induced protein (CXCL10/IP-10) as well as CCL8/MCP-2 and TNFα. IL-1 RII, was downregulated. No DEGs were noticed when comparing C2 to HCs. No SNPS were found in C2. Methylation of IFN genes MX1 and NLRC5. Frequency and the absolute number of T and B cells, monocytes, NK-like, DC, basophils, eosinophils, and neutrophils are similar to HCs. C3 was enriched with pathways related to B cell activation, and IFN signalling. SNPs detected in HLA-DQA, HLA-DRA (2 SNPs), BTNL2 and HCG23. Methylation in 56genes. Increased frequency of monocytes and lymphocytes. Increased in The IFNγ-induced protein (CXCL10/IP-10) as well as CCL8/MCP-2 and TNFα. IL-1 RII, was downregulated. C4 endotype with higher DEG including T and B activation, cytokine signalling and IL-15 production. The only SNPs identified in the intron LINC02571 gene and were previously associated with a risk for developing SLE. Methylation in 3000 genes. Decreased in B and T cells and monocytes. High percentage of neutrophils. | No statistically significant differences between the four clusters in ESSDAI or PGA mean scores. Statistically significant differences in the distribution of reported arthritis, rate of cancer history, coronary artery disease and chronic obstructive pulmonary disease were observed between the four clusters. Patients from C4 reported more severe clinical symptoms compared to the three other clusters. Some serological characteristics were significantly different across the four clusters, C1 and C3 have higher hypergammaglobulinemia, extractable nuclear antigen (ENA) antibodies, the presence of serum anti-SSA52/anti-SSA60 autoantibodies and higher circulating kappa and lambda free light chains (cFLC). |
Barturen et al., 2021 [47] | Cross-sectional Follow-up Methods: Whole blood transcriptome and methylome | N = 955 cross-sectional patients with 7 autoimmune diseases 53.4 years N = 113 follow up patients. 47 years N = 267 HCs 46 years | Four clusters were identified and validated; 3 clusters represented inflammatory, lymphoid and interferon patterns; 1 cluster with low disease activity with no specific molecular pattern. | SLEDAI and ESSDAI scores were higher in all 3 clusters compared to the undefined cluster. |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Martin-Gutierrez, L.; Wilson, R.; Castelino, M.; Jury, E.C.; Ciurtin, C. Multi-Omic Biomarkers for Patient Stratification in Sjogren’s Syndrome—A Review of the Literature. Biomedicines 2022, 10, 1773. https://doi.org/10.3390/biomedicines10081773
Martin-Gutierrez L, Wilson R, Castelino M, Jury EC, Ciurtin C. Multi-Omic Biomarkers for Patient Stratification in Sjogren’s Syndrome—A Review of the Literature. Biomedicines. 2022; 10(8):1773. https://doi.org/10.3390/biomedicines10081773
Chicago/Turabian StyleMartin-Gutierrez, Lucia, Robert Wilson, Madhura Castelino, Elizabeth C. Jury, and Coziana Ciurtin. 2022. "Multi-Omic Biomarkers for Patient Stratification in Sjogren’s Syndrome—A Review of the Literature" Biomedicines 10, no. 8: 1773. https://doi.org/10.3390/biomedicines10081773
APA StyleMartin-Gutierrez, L., Wilson, R., Castelino, M., Jury, E. C., & Ciurtin, C. (2022). Multi-Omic Biomarkers for Patient Stratification in Sjogren’s Syndrome—A Review of the Literature. Biomedicines, 10(8), 1773. https://doi.org/10.3390/biomedicines10081773