×
Home Current Archive Editorial board
News Contact
Review paper

Hypoxic mesenchymal stem cells (MSCS)-induced interleukin (IL)-10 alleviate systemic lupus erythematosus (SLE) infl ammation through inhibiting interferon (IFN)-gamma production

By
Yan Wisnu Prajoko Orcid logo ,
Yan Wisnu Prajoko
Contact Yan Wisnu Prajoko

Department of Surgery, Faculty of Medicine, Universitas Diponegoro, Semarang, Indonesia

Agung Putra ,
Agung Putra

Stem Cell and Cancer Research, Semarang, Indonesia

Department of Postgraduate Biomedical Science, Faculty of Medicine, Universitas Islam Sultan Agung, Semarang, Indonesia

Department of Pathology Anatomy, Faculty of Medicine, Universitas Islam Sultan Agung, Semarang, Indonesia

Ardi Prasetio ,
Ardi Prasetio

Stem Cell and Cancer Research, Semarang, Indonesia

Nur Dina Amalina
Nur Dina Amalina

Stem Cell and Cancer Research, Semarang, Indonesia

Pharmaceutical Sciences Department, Faculty of Medicine, Universitas Negeri Semarang, Semarang, Indonesia

Abstract

Aim
To determine the eff ect of hypoxic mesenchymal stem cells (MSCs) on the interleukin (IL)-10 and interferon (IFN)-gamma in peripheral blood mononuclear cells (PBMCs) from systemic lupus erythematosus (SLE) patients.
Methods
This study used a post-test control group design. Hypoxic MSCs were isolated and characterized according to their surface marker expression and diff erentiation capacities. PBMCs isolated from SLE patients were divided into three groups: control and two treatment groups. The treatment groups were treated by co-culturing MSCs to PBMCs with a ratio of 1:10 (T1) and 1:1 (T2) for 48 h incubation. Furthermore, IFN-gamma and IL-10 levels were determined by cytometric bead array (CBA) fl ow cytometry.
Results
Hypoxic MSCs signifi cantly decreased the IFN-gamma levels and increased the IL-10 levels in dose-dependent manner
compared to the control group. The highest activity of hypoxic MSCs was noticed in T2 group.
Conclusion
Hypoxic MSCs- induced IL-10 are important in the control of anti-infl ammatory eff ect on SLE through inhibiting IFN-gamma. 

References

1.
Choi J, Kim ST, Craft J. The pathogenesis of SLE - an update. Vol. 24, Curr Opin Immunol. 2013. p. 651–7.
2.
Barbado J, Tabera S, Sánchez A, García-Sancho J. Therapeutic potential of allogeneic mesenchymal stromal cells transplantation for lupus nephritis. Vol. 27, Lupus. 2018. p. 2161–5.
3.
Gottschalk TA, Tsantikos E, Hibbs ML. Pathogenic inflammation and its therapeutic targeting in systemic lupus erythematosus. Vol. 6, Front Immunol. 2015. p. 1–10.
4.
Yap DYH, Chan TM. B cell abnormalities in systemic lupus erythematosus and lupus nephritis—role in pathogenesis and effect of immunosuppressive treatments. Vol. 20, Int J Mol Sci. 2019. p. 1–18.
5.
Leffers HCB, Lange T, Collins C, Ulff-Møller CJ, Jacobsen S. The study of interactions between genome and exposome in the development of systemic lupus erythematosus. Vol. 18, Autoimmun Rev. 2019. p. 382–92.
6.
Weiss ARR, Dahlke MH. Immunomodulation by mesenchymal stem cells (MSCs): mechanisms of action of living, apoptotic, and dead MSCs. Vol. 10, Front Immunol. 2019. p. 1–10.
7.
Noronha ND, Mizukami A, Caliári-Oliveira C, Cominal JG, Rocha JL, Covas DT, et al. Priming approaches to improve the efficacy of mesenchymal stromal cell-based therapies. Vol. 10, Stem Cell Research and Therapy. 2019. p. 1–21.
8.
Darlan DM, Munir D, Putra A, Alif I, Amalina ND, Jusuf NK, et al. Revealing the decrease of indoleamine 2,3-dioxygenase as a major constituent for B cells survival post-mesenchymal stem cells co-cultured with peripheral blood mononuclear cell (PBMC) of systemic lupus erythematosus (SLE) patients.
9.
Darlan DM, Munir D, Putra A, Jusuf NK. MSCs-released TGFβ1 generate CD4+CD25+Foxp3+ in T-reg cells of human SLE PBMC. Vol. 120, J Formos Med Assoc. 2021. p. 602–8.
10.
Restimulia L, Ilyas S, Munir D, Madiadipoera T, Farhat F, Sembiring RJ, et al. Rats’ umbilical-cord mesenchymal stem cells ameliorate mast cells and Hsp70 on ovalbumin-induced allergic rhinitis rats.
11.
Munir D, Lubis RR, Darlan DM, Putra A, Alif I. The role of mesenchymal stem cells in decreasing interleukin-12 human systemic lupus erythematosus. Vol. 8, Open Access Maced J Med Sci. 2020. p. 787–92.
12.
Prajoko YW, Putra A, Dirja BT, Muhar AM, Amalina ND. The ameliorating effects of MSCs in controlling Treg-mediated B-Cell depletion by indoleamine 2, 3-dioxygenase Induction in PBMC of SLE patients. Vol. 10, Open Access Maced J Med Sci. 2022. p. 6–11.
13.
Eisenstein ELIM, Williams CB. The Treg/Th17 Cell Balance: A new paradigm for autoimmunity. Vol. 65, Pediatr Res. 2009. p. 26–31.
14.
Wang D, Huang S, Yuan X, Liang J, Xu R, Yao G, et al. The regulation of the Treg/Th17 balance by mesenchymal stem cells in human systemic lupus erythematosus. Vol. 14, Cell Mol Immunol. 2017. p. 423–31.
15.
Carvalho AÉS, Sousa MRR, Alencar-Silva T, Carvalho JL, Saldanha-Araujo F. Mesenchymal stem cells immunomodulation: The road to IFN-γ licensing and the path ahead. Vol. 47, Cytokine Growth Factor Rev. 2019. p. 32–42.
16.
Wu L, Rong C, Zhou Q, Zhao X, Zhuansun XM, Wan S, et al. Bone marrow mesenchymal stem cells ameliorate cisplatin-induced renal fibrosis via miR-146a-5p/Tfdp2 axis in renal tubular epithelial cells. Vol. 11, Front Immunol. 2021.
17.
Calzada D, Baos S, Cremades-Jimeno L, Cárdaba B. Immunological mechanisms in allergic diseases and allergen tolerance: The role of Treg cells. J Immunol Res. 2018.
18.
Wang D, Huang S, Yuan X, Liang J, Xu R, Yao G, et al. The regulation of the Treg / Th17 balance by mesenchymal stem cells in human systemic lupus erythematosus. Vol. 14. 2017. p. 423–31.
19.
Hartanto MM, Prajoko YW, Putra A, Amalina ND. The combination of mesenchymal stem cells and bovine colostrum in reducing α-SMA expression and NLR levels in wistar rats after 50% fibrotic liver resection. Vol. 10, Open Access Maced J Med Sci. 2022. p. 1634–9.
20.
Hamra NF, Putra A, Tjipta A, Amalina ND, Nasihun T. Hypoxia mesenchymal stem cells accelerate wound closure improvement by controlling α-smooth muscle actin expression in the full-thickness animal model. Vol. 9, Open Access Maced J Med Sci. 2021. p. 35–41.
21.
Zukhiroh Z, Putra A, Chodidjah C, Sumarawati T, Subchan P, Trisnadi S, et al. Effect of secretome-hypoxia mesenchymal stem cells on regulating SOD and MMP-1 mrna expressions in skin hyperpigmentation rats. Vol. 10, Open Access Maced J Med Sci. 2022. p. 1–7.
22.
Drawina P, Putra A, Nasihun T, Prajoko YW, Dirja BT, Amalina ND. Increased serial levels of platelet‐ derived growth factor using hypoxic mesenchymal stem cell‐conditioned medium to promote closure acceler‐ ation in a full‐thickness wound. Vol. 27, Indones J Biotechnol. 2022. p. 36–42.
23.
Lv FJ, Tuan RS, Cheung KMC, Leung VYL. Concise review: The surface markers and identity of human mesenchymal stem cells. Vol. 32, Stem Cells. 2014. p. 1408–19.
24.
Iyer SS, Rojas M. Anti-inflammatory effects of mesenchymal stem cells: novel concept for future therapies. Expert Opin Biol Ther. 2008.
25.
Tsuchiya A, Takeuchi S, Watanabe T, Yoshida T, Nojiri S, Ogawa M, et al. Mesenchymal stem cell therapies for liver cirrhosis: MSCs as “conducting cells” for improvement of liver fibrosis and regeneration. Vol. 39, Inflamm Regen. 2019. p. 4–9.
26.
Jahandideh S, Khatami S, Eslami Far A, Kadivar M. Anti-inflammatory effects of human embryonic stem cell-derived mesenchymal stem cells secretome preconditioned with diazoxide, trimetazidine and MG-132 on LPS-induced systemic inflammation mouse model. Vol. 46, Artif Cells Nanomed Biotechnol. 2018. p. 1178–87.
27.
Yang SH, Park MJ, Yoon IH, Kim SY, Hong SH, Shin JY, et al. Soluble mediators from mesenchymal stem cells suppress T cell proliferation by inducing IL-10. Vol. 41, Exp Mol Med. 2009. p. 315–24.
28.
Muhar AM, Putra A, Warli SM, Munir D. Hypoxiamesenchymal stem cells inhibit intra-peritoneal adhesions formation by upregulation of the il-10 expression. Vol. 7, Open Access Maced J Med Sci. 2019. p. 3937–43.
29.
Palomares O, Martin-Fontecha M, Lauener R, TraidlHoffmann C, Cavkaytar O, Akdis M, et al. Regulatory T cells and immune regulation of allergic diseases: Roles of IL-10 and TGF-β. Vol. 15, Genes Immun. 2014. p. 511–20.
30.
Putra A, Ridwan FB, Putridewi AI, Kustiyah AR, Wirastuti K, Sadyah NA, et al. The role of tnf-α induced mscs on suppressive inflammation by increasing tgf-β and il-10. Vol. 6, Open Access Maced J Med Sci. 2018. p. 1779–83.
31.
Lim JY, Kim BS, bin RD, Kim TW, Park G, Min CK. The therapeutic efficacy of mesenchymal stromal cells on experimental colitis was improved by the IFN-γ and poly(I:C) priming through promoting the expression of indoleamine 2,3-dioxygenase. Vol. 12, Stem Cell Res Ther. 2021. p. 1–13.
32.
Restimulia L, Ilyas S, Munir D, Putra A, Madiadipoera T, Farhat F, et al. The CD4+CD25+FoxP3+ regulatory T cells regulated by MSCs suppress plasma cells in a mouse model of allergic rhinitis. Vol. 75, Medical Archives. 2021.
33.
Mohyuddin SG, Qamar A, Hu CY, Chen SW, Wen JY, Liu M, et al. Effect of chitosan on blood profile, inflammatory cytokines by activating TLR4/NF-κB signaling pathway in intestine of heat stressed mice. Vol. 11, Sci Rep. 2021. p. 1–13.
34.
Ran S. The role of TLR4 in chemotherapy-driven metastasis. Vol. 75, Cancer Res. 2015. p. 2405–10.
35.
OЕ MА, FA S, Y O, M S, DT O ’Hagan, L T, et al. The vaccine adjuvant alum inhibits IL-12 by promoting PI3 kinase signaling while chitosan does not inhibit IL-12 and enhances Th1 and Th17 responses. Vol. 42, Eur J Immunol. 2012. p. 2709–19.

Citation

Authors retain copyright. This work is licensed under a Creative Commons Attribution 4.0 International License. Creative Commons License

 

Article metrics

Google scholar: See link

The statements, opinions and data contained in the journal are solely those of the individual authors and contributors and not of the publisher and the editor(s). We stay neutral with regard to jurisdictional claims in published maps and institutional affiliations.