Mesenchymal stem cell transplantation alleviates experimental Sjögren's syndrome through IFN-β/IL-27 signaling axis

Genhong Yao, Jingjing Qi, Jun Liang, Bingyu Shi, Weiwei Chen, Wenchao Li, Xiaojun Tang, Dandan Wang, Liwei Lu, Wanjun Chen, Songtao Shi, Yayi Hou, Lingyun Sun, Genhong Yao, Jingjing Qi, Jun Liang, Bingyu Shi, Weiwei Chen, Wenchao Li, Xiaojun Tang, Dandan Wang, Liwei Lu, Wanjun Chen, Songtao Shi, Yayi Hou, Lingyun Sun

Abstract

Rationale: Although mesenchymal stem cell (MSC) transplantation has been proved to be an effective therapeutic approach to treat experimental Sjögren's syndrome (SS), the detailed underlying mechanisms remains unknown. IL-27 has diverse influences on the regulation of T cell differentiation and was involved in SS through modulating immune response. Here we aimed to explore whether IL-27-mediated regulation of immune cells was responsible for the beneficial effects of MSC transplantation on SS. Methods: The SS-like symptoms were evaluated in IL-27 deficient and recombinant IL-27-treated NOD mice. The MSCs were infused into NOD mice via the tail vein. The histological features of submandibular glands, saliva flow rate and serum IL-27 were examined. The effects of MSCs on the IL-27 production and Th17/Treg cell in SS patients and mice in vitro and in vivo were determined for the mechanistic study. Results: This study showed that SS patients had decreased IL-27 level and increased ratio of Th17/Treg cells. Consistently, exacerbated SS-like symptoms were observed in IL-27 deficient NOD mice, along with increased ratio of Th17/Treg cells. Importantly, MSC transplantation alleviated SS-like symptoms by elevating the level of IL-27 to restore Th17/Treg balance in NOD mice. Mechanistically, MSC-secreted interferon-β (IFN-β) promote dendritic cells to produce IL-27. Conclusions: Thus, we have revealed a previously unrecognized function of MSC-mediated IL-27 production by DCs in suppressing SS-like syndrome, which provided evidences for clinical application of MSC in patients with SS.

Keywords: IL-27; Mesenchymal stem cells; Sjögren's syndrome.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interest exists.

© The author(s).

Figures

Figure 1
Figure 1
Serum IL-27 negatively correlates with disease activity in patients with Sjögren's syndrome. (A, B) IL-27 mRNA in PBMCs (A) and serum IL-27 (B) in patients with Sjögren's syndrome (SS) (n=30) compared with those of healthy controls (HC) (n=30). (C,D) Expression of IL-27 receptors, gp130 mRNA (C) and IL-27RαmRNA (D), were detected in PBMC from SS patients (n=5) and HC. (n=5) (E) Serum IL-27 was assessed according the European League Against Rheumatism (EULAR) Sjögren's syndrome Disease Activity Index (ESSDAI) scores. (F) Serum IL-27 was compared between SS patients with (n=14) and without anti-SSA antibody (n=15). (G) Correlation of serum IL-27 and IgG was analyzed. (H) Percentages of Th17 and Treg cells in SS patients (n=15) and HC (n=15) were shown. (I, J) Serum TGF-β (i) and IL-17A (j) in SS patients and HC were detected. (K, L) Correlations of IL-27 and Treg and Th17 cells were evaluated. Data were based on three independent experiments. Data are presented as mean±SEM. *, p<0.05, **, p<0.01, ***, p<0.001.
Figure 2
Figure 2
IL-27 deficiency exacerbates SS-like symptoms in NOD mice. (A) Serum IL-27 was compared between NOD (n=5) (12-week old) and control ICR mice (n=5) (12-week old). (B) Salivary flow rates were measured in wild type (WT) (n=5) (12-week old) and IL-27-/- NOD mice(n=5) (12-week old). (C) Serum IgG in WT and IL-27-/- NOD mice was compared (n=5) (12-week old). (D) Representative images of H&E staining of salivary glands in WT and IL-27-/- NOD mice. Original magnification, ×40. Scale bars, 64 μm. Yellow arrows indicate infiltrating lymphocytes. (E) Lymphocyte infiltrations in salivary glands of mice were evaluated for histological scores. (F and G) Percentages of Treg and Th17 cells in WT (n=5) (12-week old) and IL-27-/- NOD mice (n=5) (12-week old) were shown. Data were based on three independent experiments. Data are presented as mean±SEM. *,p<0.05, **, p<0.01.
Figure 3
Figure 3
Mesenchymal stem cell (MSC) transplantation attenuates SS-like diseases in NOD mice. (A) Representative images of H&E staining of salivary glands in PBS (A) and MSC treated NOD mice. Yellow arrows indicate infiltrating lymphocytes. (B) Lymphocyte infiltrations in salivary glands of mice were evaluated for histological scores. (C) Salivary flow rates in PBS (n=5) (12-week old) and MSC treatment NOD mice (n=5) (12-week old). (D)Serum IL-27 was compared between PBS (n=5) (12-week old) and MSC treatment groups (n=5) (12-week old). (E-H) Representative FACS analysis of Treg and Th17 cells of spleens (E, F) and cervical lymph nodes (G, H)(n=5). Data were based on three independent experiments. Data are presented as mean±SEM. *,p<0.05, **, p<0.01.
Figure 4
Figure 4
IL-27 modulates the proliferation and differentiation of Th17 and Treg cells in PBMCs and naïve CD4+ T cells. (A, B) Treg and Th17 cells in human PBMCs with and without IL-27 treatment. (A) Fluorescence staining of CD4, Foxp-3 and IL-17 in human PBMCs. (B) Summary of the frequencies of Treg and Th17 cells (n=5). (C, D) Flow cytometric analysis of Treg and Th17 cells, which were differentiated from mouse naïve CD4+ T cells in the presence or absence of IL-27 (n=5). Data are presented as mean±SEM. Data were based on three independent experiments. *, p<0.05.
Figure 5
Figure 5
MSCs increase serum IL-27 and restore Th17/Treg balance in SS patients. (A, B, C) Serum IL-27 (A), IL-17A (B) and TGF-β (C) were determined in SS patients before and 1 week after MSC transplantation (n= 11). (D, E, F) Fluorescence staining of CD4, Foxp-3 and IL-17 in human PBMCs (D, F). Summary of Treg and Th17 cell frequencies in SS patients with MSC transplantation (E, G) (n=10). Data are presented as mean±SEM. Data were based on three independent experiments. *, p<0.05, **, p<0.01.
Figure 6
Figure 6
MSCs promote IL-27 production of dendritic cells through secreting IFN-β. (A) IL-27 produced by monocyte-derived DCs from SS patients (n=5) and HC (n=5). (B, C) IL-27 mRNA (B) and protein (C) of monocyte-derived DCs from HC with or without MSC treatment in the presence of 100ng/mL LPS was evaluated (n=5). (D) Genes related to IL-27 expression of DCs were screened and compared in MSCs co-cultured with and without monocyte-derived DCs from HC (n=4) in the presence of 100ng/mL LPS. (E) IL-27 mRNA expressions of monocyte-derived DCs with different concentrations of IFN-β treatment in the presence of 100ng/mL LPS (n=3). (F) IL-27 mRNA expressions of monocyte-derived DCs from HC co-cultured with MSC with and without IFN-β mRNA interference in the presence of 100ng/mL LPS (n=4). αMSC means siRNA silencing of IFN-β in MSCs. Data are presented as mean±SEM. Data were based on three independent experiments. *, p<0.05, **, p<0.01, ***, p<0.001.

References

    1. Both T, Dalm VA, van Hagen PM, van Daele PL. Reviewing primary Sjögren's syndrome: beyond the dryness-from pathophysiology to diagnosis and treatment. Int J Med Sci. 2017;14:191–200.
    1. Ferro F, Marcucci E, Orlandi M, Baldini C, Bartoloni-Bocci E. One year in review 2017: primary Sjögren's syndrome. Clin Exp Rheumatol. 2017;35:179–91.
    1. Brito-Zerón P, Retamozo S, Gheitasi H, Ramos-Casals M. Treating the underlying pathophysiology of primary Sjögren syndrome: recent advances and future prospects. Drugs. 2016;76:1601–23.
    1. Shang F, Liu S, Ming L, Tian R, Jin F, Ding Y. et al. Human umbilical cord MSCs as new cell sources for promoting periodontal regeneration in inflammatory periodontal defect. Theranostics. 2017;7:4370–82.
    1. Li H, Wang C, He T, Zhao T, Chen YY, Shen YL. et al. Mitochondrial transfer from bone marrow mesenchymal stem cells to motor neurons in spinal cord injury rats via gap junction. Theranostics. 2019;9:2017–35.
    1. Qi M, Zhang L, Ma Y, Shuai Y, Li L, Luo K. et al. Autophagy maintains the function of bone marrow mesenchymal stem cells to prevent estrogen deficiency-induced osteoporosis. Theranostics. 2017;7:4498–516.
    1. Wang X, Jiang B, Sun H, Zheng D, Zhang Z, Yan L. et al. Noninvasive application of mesenchymal stem cell spheres derived from hESC accelerates wound healing in a CXCL12-CXCR4 axis-dependent manner. Theranostics. 2019;9:6112–28.
    1. Detry O, Vandermeulen M, Delbouille MH, Somja J, Bletard N, Briquet A. et al. Infusion of mesenchymal stromal cells after deceased liver transplantation: A phase I-II, open-label, clinical study. J Hepatol. 2017;67:47–55.
    1. Huang Y, Wang J, Cai J, Qiu Y, Zheng H, Lai X. et al. Targeted homing of CCR2-overexpressing mesenchymal stromal cells to ischemic brain enhances post-stroke recovery partially through PRDX4-mediated blood-brain barrier preservation. Theranostics. 2018;8:5929–44.
    1. Ryu CM, Yu HY, Lee HY, Shin JH, Lee S, Ju H. et al. Longitudinal intravital imaging of transplanted mesenchymal stem cells elucidates their functional integration and therapeutic potency in an animal model of interstitial cystitis/bladder pain syndrome. Theranostics. 2018;8:5610–24.
    1. Khalili S, Liu Y, Kornete M, Roescher N, Kodama S, Peterson A. et al. Mesenchymal stromal cells improve salivary function and reduce lymphocytic infiltrates in mice with Sjögren's-like disease. PLoS One. 2012;7:e38615.
    1. Xu J, Wang D, Liu D, Fan Z, Zhang H, Liu O. et al. Allogeneic mesenchymal stem cell treatment alleviates experimental and clinical Sjögren syndrome. Blood. 2012;120:3142–51.
    1. Nagy G, Huszthy PC, Fossum E, Konttinen Y, Nakken B, Szodoray P. Selected aspects in the pathogenesis of autoimmune diseases. Mediators Inflamm. 2015;2015:351732.
    1. Handono K, Firdausi SN, Pratama MZ, Endhart AT, Kalim H. Vitamin A improve Th17 and Treg regulation in systemic lupus erythematosus. Clin Rheumatol. 2016;35:631–8.
    1. Meka RR, Venkatesha SH, Dudics S, Acharya B, Moudgil KD. IL-27-induced modulation of autoimmunity and its therapeutic potential. Autoimmun Rev. 2015;14:1131–41.
    1. Vignali DA, Kuchroo -12 family cytokines. immunological playmakers. Nat Immunol. 2012;13:722–8.
    1. Hall AO, Silver JS, Hunter CA. The immunobiology of IL-27. Adv Immunol. 2012;115:1–44.
    1. Aparicio-Siegmund S, Garbers C. The biology of interleukin-27 reveals unique pro- and anti-inflammatory functions in immunity. Cytokine Growth Factor Rev. 2015;26:579–86.
    1. Favaro E, Carpanetto A, Caorsi C, Giovarelli M, Angelini C, Cavallo-Perin P. et al. Human mesenchymal stem cells and derived extracellular vesicles induce regulatory dendritic cells in type 1 diabetic patients. Diabetologia. 2016;59:325–33.
    1. Mohammadpour H, Pourfathollah AA, Zarif MN, Tahoori MT. TNF-α modulates the immunosuppressive effects of MSCs on dendritic cells and T cells. Int Immunopharmacol. 2015;28:1009–17.
    1. Vitali C, Bombardieri S, Jonsson R, Moutosopoulos HM, Alexansder EL, Carsons SE. et al. European Study Group on Classification criteria for Sjögren's syndrome: a revised version of the European criteria proposed by the American-European Consensus Group. Ann Rheum Dis. 2002;61:554–8.
    1. Peck AB, Nguyen CQ. What can Sjögren's syndrome-like disease in mice contribute to human Sjögren's syndrome? Clin Immunol. 2017;182:14–23.
    1. Wang D, Feng X, Lu L, Konkel JE, Zhang H, Chen Z. et al. A CD8 T cell/indoleamine 2,3-dioxygenase axis is required for mesenchymal stem cell suppression of human systemic lupus erythematosus. Arthritis Rheumatol. 2014;66:2234–45.
    1. Shi B, Qi J, Yao G, Feng R, Zhang Z, Wang D. et al. Mesenchymal stem cell transplantation ameliorates Sjogren's syndrome via suppressing IL-12 production by dendritic cells. Stem Cell Res Ther. 2018;9:308.
    1. Yao G, Yang L, Hou Y. Phenotype and functions of spleen dendritic cells in RICK-knockout mice. IntImmunopharmacol. 2010;10:130–3.
    1. Zundler S, Neurath MF.Interleukin-12. Functional activities and implications for disease. Cytokine Growth Factor Rev. 2015;26:559–68.
    1. Gee K, Guzzo C, Che Mat NF, Ma W, Kumar A. The IL-12 family of cytokines in infection, inflammation and autoimmune disorders. Inflamm Allergy Drug Targets. 2009;8:40–52.
    1. Macedo C, Turnquist HR, Castillo-Rama M, Zahorchak AF, Shapiro R, Thomson AW. et al. Rapamycin augments human DC IL-12p70 and IL-27 secretion to promote allogeneic Type 1 polarization modulated by NK cells. Am J Transplant. 2013;13:2322–33.
    1. Yoshida H, Hunter CA. The immunobiology of interleukin-27. Annu Rev Immunol. 2015;33:417–43.
    1. Lee BH, Carcamo WC, Chiorini JA, Peck AB, Nguyen CQ. Gene therapy using IL-27 ameliorates Sjögren's syndrome-like autoimmune exocrinopathy. Arthritis Res Ther. 2012;14:R172.
    1. Xia L, Shen H, Zhao L, Lu J. Elevated levels of interleukin-27 in patients with Sjögren's syndrome. Scand J Rheumatol. 2012;41:73–74.
    1. Lin X, Rui K, Deng J, Tian J, Wang X, Wang S. et al. Th17 cells play a critical role in the development of experimental Sjögren's syndrome. Ann Rheum Dis. 2015;74:1302–10.
    1. Liu MF, Lin LH, Weng CT, Weng MY. Decreased CD4+CD25+bright T cells in peripheral blood of patients with primary Sjogren's syndrome. Lupus. 2008;17:34–9.
    1. Szodoray P, Papp G, Horvath IF, Barath S, Sipka S, Nakken B. et al. Cells with regulatory function of the innate and adaptive immune system in primary Sjögren's syndrome. Clin Exp Immunol. 2009;157:343–49.
    1. Wang Q, Liu J. Regulation and immune function of IL-27. Adv Exp Med Biol. 2016;941:191–211.
    1. Belle L, Agle K, Zhou V, Yin-Yuan C, Komorowski R, Eastwood D. et al. Blockade of interleukin-27 signaling reduces GVHD in mice by augmenting Treg reconstitution and stabilizing Foxp3 expression. Blood. 2016;128:2068–82.
    1. Tyndall A. Hematopoietic stem cell transplantation for systemic sclerosis: review of current status. BioDurgs. 2019;33:401–9.
    1. Ferraccioli G, Damato R, De Vita S, Fanin R, Damiani D, Baccarani M. Hematopoietic stem cell transplantation (HSCT) in a patient with Sjögren's syndrome and lung malt lymphoma cured lymphoma not the autoimmune disease. Ann Rheum Dis. 2001;60:174–6.
    1. Jensen DH, Oliveri RS, Trojahn Kølle SF, Fischer-Nielsen A, Specht L, Bardow A. et al. Mesenchymal stem cell therapy for salivary gland dysfunction and xerostomia: a systematic review of preclinical studies. Oral Surg Oral Med Oral Pathol Oral Radiol. 2014;117:335–42.
    1. Gu F, Wang D, Zhang H, Feng X, Gilkeson GS, Shi S. et al. Allogeneic mesenchymal stem cell transplantation for lupus nephritis patients refractory to conventional therapy. Clin Rheumatol. 2014;33:1611–9.
    1. Wang D, Akiyama K, Zhang H, Yamaza T, Li X, Feng X. et al. Double allogenic mesenchymal stem cells transplantations could not enhance therapeutic effect compared with single transplantation in systemic lupus erythematosus. Clin Dev Immunol. 2012;2012:273291.
    1. Lim JY, Yi T, Choi JS, Jang YH, Lee S, Kim HJ. et al. Intraglandular transplantation of bone marrow-derived clonal mesenchymal stem cells for amelioration of post-irradiation salivary gland damage. Oral Oncol. 2013;49:136–43.
    1. Ruan GF, Zheng L, Huang JS, Huang WX, Gong BD, Fang XX. et al. Effect of mesenchymal stem cells on Sjögren-like mice and the microRNA expression profiles of splenic CD4+ T cells. Exp Ther Med. 2017;13:2828–38.
    1. Theofilopoulos AN, Baccala R, Beutler B, Kono DH. Type I interferons (alpha/beta) in immunity and autoimmunity. Ann Rev Immunol. 2005;23:307–36.
    1. Bodewes ILA, Versnel MA. Interferon activation in primary Sjögren's syndrome: recent insights and future perspective as novel treatment target. Expert Rev Clin Immunol. 2018;14:817–29.
    1. Cummins MJ, Papas A, Kammer GM, Fox PC. Treatment of primary Sjögren's syndrome with low-dose human interferon alfa administered by the oromucosal route: combined phase III results. Arthritis Rheum. 2003;49:585–93.
    1. Benchabane S, Belkhelfa M, Belguendouz H, Zidi S, Boudjelida A, Youinou P. et al. Interferon-β inhibits inflammatory responses mediators via suppression of iNOS signaling pathway in PBMCs from patients with primary Sjögren's syndrome. Inflammapharmacology. 2018;26:1165–74.

Source: PubMed

3
Subscribe