Skip to main content
Top
Published in: Journal of Translational Medicine 1/2019

Open Access 01-12-2019 | Myasthenia Gravis | Research

Soluble glucocorticoid-induced tumor necrosis factor receptor regulates Helios expression in myasthenia gravis

Authors: Yi Li, Shumei Yang, Zhibin Li, Huanyu Meng, Wanling Jin, Huan Yang, Weifan Yin

Published in: Journal of Translational Medicine | Issue 1/2019

Login to get access

Abstract

Background

Helios is important for functional and phenotype stability of regulatory T cells (Tregs). However, the role of Helios in autoimmune diseases and its regulation remains unclear. This study aimed to investigate the role of Helios+ Tregs in myasthenia gravis (MG) and glucocorticoid-induced tumor necrosis factor receptor (GITR) and its ligand (GITRL) in the modulation of Helios.

Method

Multicolor flow cytometry was performed to analyze Helios+ Tregs in peripheral blood from MG patients and healthy donors (HDs). Enzyme-linked immunosorbent assay (ELISA) was used to determine the levels of soluble GITRL/GITR in plasma. Tregs were isolated via magnetic separation and treated with recombinant GITRL and GITR-Fc. Membrane GITRL on Tregs and expression of Helios and other markers (FOXP3, CD25, CD39, CTLA-4, PD-L1 and IL-10) involved in immunosuppressive activity were determined by flow cytometry.

Result

Both Helios+ Tregs and soluble GITR were decreased in generalized MG (GMG) patients (n = 14), compared with HDs (n = 14) and ocular MG (OMG) patients (n = 16). Helios+ Tregs possessed greater immunosuppressive capacity compared to Helios Tregs. Further analysis indicates soluble GITR was negatively correlated with quantitative MG score and promoted Helios expression and enhanced function of Tregs independently of membrane GITRL.

Conclusion

This work demonstrates abnormal changes in Helios+ Tregs and soluble GITR in MG, as well as direct regulation of Helios by GITR in the context of Tregs. This work provides new insight into the role of GITR in the regulatory pathway of Helios and pathogenesis of MG.
Literature
1.
go back to reference Gilhus NE, Verschuuren JJ. Myasthenia gravis: subgroup classification and therapeutic strategies. Lancet Neurol. 2015;14:1023–36.CrossRef Gilhus NE, Verschuuren JJ. Myasthenia gravis: subgroup classification and therapeutic strategies. Lancet Neurol. 2015;14:1023–36.CrossRef
2.
go back to reference Grant CR, Liberal R, Mieli-Vergani G, Vergani D, Longhi MS. Regulatory T-cells in autoimmune diseases: challenges, controversies and—yet—unanswered questions. Autoimmun Rev. 2015;14:105–16.CrossRef Grant CR, Liberal R, Mieli-Vergani G, Vergani D, Longhi MS. Regulatory T-cells in autoimmune diseases: challenges, controversies and—yet—unanswered questions. Autoimmun Rev. 2015;14:105–16.CrossRef
3.
go back to reference Masuda M, Matsumoto M, Tanaka S, Nakajima K, Yamada N, Ido N, et al. Clinical implication of peripheral CD4+ CD25+ regulatory T cells and Th17 cells in myasthenia gravis patients. J Neuroimmunol. 2010;225:123–31.CrossRef Masuda M, Matsumoto M, Tanaka S, Nakajima K, Yamada N, Ido N, et al. Clinical implication of peripheral CD4+ CD25+ regulatory T cells and Th17 cells in myasthenia gravis patients. J Neuroimmunol. 2010;225:123–31.CrossRef
4.
go back to reference Xu WH, Zhang AM, Ren MS, Zhang XD, Wang F, Xu XC, et al. Changes of Treg-associated molecules on CD4+ CD25+ Treg cells in myasthenia gravis and effects of immunosuppressants. J Clin Immunol. 2012;32:975–83.CrossRef Xu WH, Zhang AM, Ren MS, Zhang XD, Wang F, Xu XC, et al. Changes of Treg-associated molecules on CD4+ CD25+ Treg cells in myasthenia gravis and effects of immunosuppressants. J Clin Immunol. 2012;32:975–83.CrossRef
5.
go back to reference Shevach EM, Thornton AM. tTregs, pTregs, and iTregs: similarities and differences. Immunol Rev. 2014;259:88–102.CrossRef Shevach EM, Thornton AM. tTregs, pTregs, and iTregs: similarities and differences. Immunol Rev. 2014;259:88–102.CrossRef
6.
go back to reference Singh K, Hjort M, Thorvaldson L, Sandler S. Concomitant analysis of Helios and Neuropilin-1 as a marker to detect thymic derived regulatory T cells in naïve mice. Sci Rep. 2015;5:7767.CrossRef Singh K, Hjort M, Thorvaldson L, Sandler S. Concomitant analysis of Helios and Neuropilin-1 as a marker to detect thymic derived regulatory T cells in naïve mice. Sci Rep. 2015;5:7767.CrossRef
7.
go back to reference Takatori H, Kawashima H, Matsuki A, Meguro K, Tanaka S, Iwamoto T, et al. Helios enhances Treg cell function in cooperation with FoxP3. Arthritis Rheumatol. 2015;67:1491–502.CrossRef Takatori H, Kawashima H, Matsuki A, Meguro K, Tanaka S, Iwamoto T, et al. Helios enhances Treg cell function in cooperation with FoxP3. Arthritis Rheumatol. 2015;67:1491–502.CrossRef
8.
go back to reference Nakagawa H, Sido JM, Reyes EE, Kiers V, Cantor H, Kim HJ. Instability of Helios-deficient Tregs is associated with conversion to a T-effector phenotype and enhanced antitumor immunity. Proc Natl Acad Sci USA. 2016;113:6248–53.CrossRef Nakagawa H, Sido JM, Reyes EE, Kiers V, Cantor H, Kim HJ. Instability of Helios-deficient Tregs is associated with conversion to a T-effector phenotype and enhanced antitumor immunity. Proc Natl Acad Sci USA. 2016;113:6248–53.CrossRef
9.
go back to reference Petrillo MG, Ronchetti S, Ricci E, Alunno A, Gerli R, Nocentini G, et al. GITR+ regulatory T cells in the treatment of autoimmune diseases. Autoimmun Rev. 2015;14:117–26.CrossRef Petrillo MG, Ronchetti S, Ricci E, Alunno A, Gerli R, Nocentini G, et al. GITR+ regulatory T cells in the treatment of autoimmune diseases. Autoimmun Rev. 2015;14:117–26.CrossRef
10.
go back to reference Kim JD, Choi BK, Bae JS, Lee UH, Han IS, Lee HW, et al. Cloning and characterization of GITR ligand. Genes Immun. 2003;4:564–9.CrossRef Kim JD, Choi BK, Bae JS, Lee UH, Han IS, Lee HW, et al. Cloning and characterization of GITR ligand. Genes Immun. 2003;4:564–9.CrossRef
11.
go back to reference Tone M, Tone Y, Adams E, Yates SF, Frewin MR, Cobbold SP, et al. Mouse glucocorticoid-induced tumor necrosis factor receptor ligand is costimulatory for T cells. Proc Natl Acad Sci USA. 2003;100:15059–64.CrossRef Tone M, Tone Y, Adams E, Yates SF, Frewin MR, Cobbold SP, et al. Mouse glucocorticoid-induced tumor necrosis factor receptor ligand is costimulatory for T cells. Proc Natl Acad Sci USA. 2003;100:15059–64.CrossRef
12.
go back to reference Nocentini G, Ronchetti S, Petrillo MG, Riccardi C. Pharmacological modulation of GITRL/GITR system: therapeutic perspectives. Br J Pharmacol. 2012;165:2089–99.CrossRef Nocentini G, Ronchetti S, Petrillo MG, Riccardi C. Pharmacological modulation of GITRL/GITR system: therapeutic perspectives. Br J Pharmacol. 2012;165:2089–99.CrossRef
13.
go back to reference Joetham A, Ohnishi H, Okamoto M, Takeda K, Schedel M, Domenico J, et al. Loss of T regulatory cell suppression following signaling through glucocorticoid-induced tumor necrosis receptor (GITR) is dependent on c-Jun N-terminal kinase activation. J Biol Chem. 2012;287:17100–8.CrossRef Joetham A, Ohnishi H, Okamoto M, Takeda K, Schedel M, Domenico J, et al. Loss of T regulatory cell suppression following signaling through glucocorticoid-induced tumor necrosis receptor (GITR) is dependent on c-Jun N-terminal kinase activation. J Biol Chem. 2012;287:17100–8.CrossRef
14.
go back to reference Kim YH, Shin SM, Choi BK, Oh HS, Kim CH, Lee SJ, et al. Authentic GITR signaling fails to induce tumor regression unless Foxp3+ regulatory T cells are depleted. J Immunol. 2015;195:4721–9.CrossRef Kim YH, Shin SM, Choi BK, Oh HS, Kim CH, Lee SJ, et al. Authentic GITR signaling fails to induce tumor regression unless Foxp3+ regulatory T cells are depleted. J Immunol. 2015;195:4721–9.CrossRef
15.
go back to reference Schaer DA, Budhu S, Liu C, Bryson C, Malandro N, Cohen A, et al. GITR pathway activation abrogates tumor immune suppression through loss of regulatory T cell lineage stability. Cancer Immunol Res. 2013;1:320–31.CrossRef Schaer DA, Budhu S, Liu C, Bryson C, Malandro N, Cohen A, et al. GITR pathway activation abrogates tumor immune suppression through loss of regulatory T cell lineage stability. Cancer Immunol Res. 2013;1:320–31.CrossRef
16.
go back to reference Knee DA, Hewes B, Brogdon JL. Rationale for anti-GITR cancer immunotherapy. Eur J Cancer. 2016;67:1–10.CrossRef Knee DA, Hewes B, Brogdon JL. Rationale for anti-GITR cancer immunotherapy. Eur J Cancer. 2016;67:1–10.CrossRef
17.
go back to reference Gan X, Feng X, Gu L, Tan W, Sun X, Lv C, et al. Correlation of increased blood levels of GITR and GITRL with disease severity in patients with primary Sjögren’s syndrome. Clin Dev Immunol. 2013;2013:340751.CrossRef Gan X, Feng X, Gu L, Tan W, Sun X, Lv C, et al. Correlation of increased blood levels of GITR and GITRL with disease severity in patients with primary Sjögren’s syndrome. Clin Dev Immunol. 2013;2013:340751.CrossRef
18.
go back to reference Liu Y, Tang X, Tian J, Zhu C, Peng H, Rui K, et al. Th17/Treg cells imbalance and GITRL profile in patients with Hashimoto’s thyroiditis. Int J Mol Sci. 2014;15:21674–86.CrossRef Liu Y, Tang X, Tian J, Zhu C, Peng H, Rui K, et al. Th17/Treg cells imbalance and GITRL profile in patients with Hashimoto’s thyroiditis. Int J Mol Sci. 2014;15:21674–86.CrossRef
19.
go back to reference Li L, Wen W, Jia R, Li Y, Liu X, Sun X, et al. GITRL is associated with increased autoantibody production in patients with rheumatoid arthritis. Clin Rheumatol. 2016;35:2195–202.CrossRef Li L, Wen W, Jia R, Li Y, Liu X, Sun X, et al. GITRL is associated with increased autoantibody production in patients with rheumatoid arthritis. Clin Rheumatol. 2016;35:2195–202.CrossRef
20.
go back to reference Gu L, Xu L, Zhang X, Tan W, Wang H, Zhang M. Correlation of circulating glucocorticoid-induced TNFR-related protein ligand levels with disease activity in patients with systemic lupus erythematosus. Clin Dev Immunol. 2012;2012:265868.CrossRef Gu L, Xu L, Zhang X, Tan W, Wang H, Zhang M. Correlation of circulating glucocorticoid-induced TNFR-related protein ligand levels with disease activity in patients with systemic lupus erythematosus. Clin Dev Immunol. 2012;2012:265868.CrossRef
21.
go back to reference Grohmann U, Volpi C, Fallarino F, Bozza S, Bianchi R, Vacca C, et al. Reverse signaling through GITR ligand enables dexamethasone to activate IDO in allergy. Nat Med. 2007;13:579–86.CrossRef Grohmann U, Volpi C, Fallarino F, Bozza S, Bianchi R, Vacca C, et al. Reverse signaling through GITR ligand enables dexamethasone to activate IDO in allergy. Nat Med. 2007;13:579–86.CrossRef
22.
go back to reference Bae EM, Kim WJ, Suk K, Kang YM, Park JE, Kim WY, et al. Reverse signaling initiated from GITRL induces NF-kappaB activation through ERK in the inflammatory activation of macrophages. Mol Immunol. 2008;45:523–33.CrossRef Bae EM, Kim WJ, Suk K, Kang YM, Park JE, Kim WY, et al. Reverse signaling initiated from GITRL induces NF-kappaB activation through ERK in the inflammatory activation of macrophages. Mol Immunol. 2008;45:523–33.CrossRef
23.
go back to reference Kim HJ, Barnitz RA, Kreslavsky T, Brown FD, Moffett H, Lemieux ME, et al. Stable inhibitory activity of regulatory T cells requires the transcription factor Helios. Science. 2015;350:334–9.CrossRef Kim HJ, Barnitz RA, Kreslavsky T, Brown FD, Moffett H, Lemieux ME, et al. Stable inhibitory activity of regulatory T cells requires the transcription factor Helios. Science. 2015;350:334–9.CrossRef
24.
go back to reference Alexander T, Sattler A, Templin L, Kohler S, Groß C, Meisel A, et al. Foxp3+ Helios+ regulatory T cells are expanded in active systemic lupus erythematosus. Ann Rheum Dis. 2013;72:1549–58.CrossRef Alexander T, Sattler A, Templin L, Kohler S, Groß C, Meisel A, et al. Foxp3+ Helios+ regulatory T cells are expanded in active systemic lupus erythematosus. Ann Rheum Dis. 2013;72:1549–58.CrossRef
25.
go back to reference Nocentini G, Riccardi C. GITR: a modulator of immune response and inflammation. Adv Exp Med Biol. 2009;647:156–73.CrossRef Nocentini G, Riccardi C. GITR: a modulator of immune response and inflammation. Adv Exp Med Biol. 2009;647:156–73.CrossRef
26.
go back to reference Galuppo M, Nocentini G, Mazzon E, Ronchetti S, Esposito E, Riccardi L, et al. GITR gene deletion and GITR-FC soluble protein administration inhibit multiple organ failure induced by zymosan. Shock. 2011;36:263–71.CrossRef Galuppo M, Nocentini G, Mazzon E, Ronchetti S, Esposito E, Riccardi L, et al. GITR gene deletion and GITR-FC soluble protein administration inhibit multiple organ failure induced by zymosan. Shock. 2011;36:263–71.CrossRef
Metadata
Title
Soluble glucocorticoid-induced tumor necrosis factor receptor regulates Helios expression in myasthenia gravis
Authors
Yi Li
Shumei Yang
Zhibin Li
Huanyu Meng
Wanling Jin
Huan Yang
Weifan Yin
Publication date
01-12-2019
Publisher
BioMed Central
Published in
Journal of Translational Medicine / Issue 1/2019
Electronic ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-019-1916-1

Other articles of this Issue 1/2019

Journal of Translational Medicine 1/2019 Go to the issue