Dexamethasone-Induced Myeloid-Derived Suppressor Cells Prolong Allo Cardiac Graft Survival through iNOS- and Glucocorticoid Receptor-Dependent Mechanism

How to induce immune tolerance without long-term need for immunosuppressive drugs has always been a central problem in solid organ transplantation. Modulating immunoregulatory cells represents a potential target to resolve this problem. Myeloid-derived suppressor cells (MDSCs) are novel key immunore...

Full description

Bibliographic Details
Main Authors: Yang Zhao, Xiao-Fei Shen, Ke Cao, Jie Ding, Xing Kang, Wen-xian Guan, Yi-tao Ding, Bao-rui Liu, Jun-Feng Du
Format: Article
Language:English
Published: Frontiers Media S.A. 2018-02-01
Series:Frontiers in Immunology
Subjects:
Online Access:http://journal.frontiersin.org/article/10.3389/fimmu.2018.00282/full
_version_ 1811194226644353024
author Yang Zhao
Xiao-Fei Shen
Ke Cao
Jie Ding
Xing Kang
Wen-xian Guan
Yi-tao Ding
Bao-rui Liu
Jun-Feng Du
author_facet Yang Zhao
Xiao-Fei Shen
Ke Cao
Jie Ding
Xing Kang
Wen-xian Guan
Yi-tao Ding
Bao-rui Liu
Jun-Feng Du
author_sort Yang Zhao
collection DOAJ
description How to induce immune tolerance without long-term need for immunosuppressive drugs has always been a central problem in solid organ transplantation. Modulating immunoregulatory cells represents a potential target to resolve this problem. Myeloid-derived suppressor cells (MDSCs) are novel key immunoregulatory cells in the context of tumor development or transplantation, and can be generated in vitro. However, none of current systems for in vitro differentiation of MDSCs have successfully achieved long-term immune tolerance. Herein, we combined dexamethasone (Dex), which is a classic immune regulatory drug in the clinic, with common MDSCs inducing cytokine granulocyte macrophage colony stimulating factor (GM-CSF) to generate MDSCs in vitro. Addition of Dex into GM-CSF system specifically increased the number of CD11b+ Gr-1int/low MDSCs with an enhanced immunosuppressive function in vitro. Adoptive transfer of these MDSCs significantly prolonged heart allograft survival and also favored the expansion of regulatory T cells in vivo. Mechanistic studies showed that inducible nitric oxide sythase (iNOS) signaling was required for MDSCs in the control of T-cell response and glucocorticoid receptor (GR) signaling played a critical role in the recruitment of transferred MDSCs into allograft through upregulating CXCR2 expression on MDSCs. Blockade of GR signaling with its specific inhibitor or genetic deletion of iNOS reversed the protective effect of Dex-induced MDSCs on allograft rejection. Together, our results indicated that co-application of Dex and GM-CSF may be a new and important strategy for the induction of potent MDSCs to achieve immune tolerance in organ transplantation.
first_indexed 2024-04-12T00:22:23Z
format Article
id doaj.art-9c28a364407f455d865341105bf0e1ec
institution Directory Open Access Journal
issn 1664-3224
language English
last_indexed 2024-04-12T00:22:23Z
publishDate 2018-02-01
publisher Frontiers Media S.A.
record_format Article
series Frontiers in Immunology
spelling doaj.art-9c28a364407f455d865341105bf0e1ec2022-12-22T03:55:40ZengFrontiers Media S.A.Frontiers in Immunology1664-32242018-02-01910.3389/fimmu.2018.00282308942Dexamethasone-Induced Myeloid-Derived Suppressor Cells Prolong Allo Cardiac Graft Survival through iNOS- and Glucocorticoid Receptor-Dependent MechanismYang Zhao0Xiao-Fei Shen1Ke Cao2Jie Ding3Xing Kang4Wen-xian Guan5Yi-tao Ding6Bao-rui Liu7Jun-Feng Du8The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, ChinaDepartment of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, ChinaDepartment of Critical Care Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, ChinaDepartment of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, ChinaDepartment of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, ChinaDepartment of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, ChinaDepartment of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, ChinaThe Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, ChinaDepartment of General Surgery, PLA Army General Hospital, Beijing, ChinaHow to induce immune tolerance without long-term need for immunosuppressive drugs has always been a central problem in solid organ transplantation. Modulating immunoregulatory cells represents a potential target to resolve this problem. Myeloid-derived suppressor cells (MDSCs) are novel key immunoregulatory cells in the context of tumor development or transplantation, and can be generated in vitro. However, none of current systems for in vitro differentiation of MDSCs have successfully achieved long-term immune tolerance. Herein, we combined dexamethasone (Dex), which is a classic immune regulatory drug in the clinic, with common MDSCs inducing cytokine granulocyte macrophage colony stimulating factor (GM-CSF) to generate MDSCs in vitro. Addition of Dex into GM-CSF system specifically increased the number of CD11b+ Gr-1int/low MDSCs with an enhanced immunosuppressive function in vitro. Adoptive transfer of these MDSCs significantly prolonged heart allograft survival and also favored the expansion of regulatory T cells in vivo. Mechanistic studies showed that inducible nitric oxide sythase (iNOS) signaling was required for MDSCs in the control of T-cell response and glucocorticoid receptor (GR) signaling played a critical role in the recruitment of transferred MDSCs into allograft through upregulating CXCR2 expression on MDSCs. Blockade of GR signaling with its specific inhibitor or genetic deletion of iNOS reversed the protective effect of Dex-induced MDSCs on allograft rejection. Together, our results indicated that co-application of Dex and GM-CSF may be a new and important strategy for the induction of potent MDSCs to achieve immune tolerance in organ transplantation.http://journal.frontiersin.org/article/10.3389/fimmu.2018.00282/fullmyeloid-derived suppressor cellsimmune tolerancedexamethasoneiNOSglucocorticoid receptor
spellingShingle Yang Zhao
Xiao-Fei Shen
Ke Cao
Jie Ding
Xing Kang
Wen-xian Guan
Yi-tao Ding
Bao-rui Liu
Jun-Feng Du
Dexamethasone-Induced Myeloid-Derived Suppressor Cells Prolong Allo Cardiac Graft Survival through iNOS- and Glucocorticoid Receptor-Dependent Mechanism
Frontiers in Immunology
myeloid-derived suppressor cells
immune tolerance
dexamethasone
iNOS
glucocorticoid receptor
title Dexamethasone-Induced Myeloid-Derived Suppressor Cells Prolong Allo Cardiac Graft Survival through iNOS- and Glucocorticoid Receptor-Dependent Mechanism
title_full Dexamethasone-Induced Myeloid-Derived Suppressor Cells Prolong Allo Cardiac Graft Survival through iNOS- and Glucocorticoid Receptor-Dependent Mechanism
title_fullStr Dexamethasone-Induced Myeloid-Derived Suppressor Cells Prolong Allo Cardiac Graft Survival through iNOS- and Glucocorticoid Receptor-Dependent Mechanism
title_full_unstemmed Dexamethasone-Induced Myeloid-Derived Suppressor Cells Prolong Allo Cardiac Graft Survival through iNOS- and Glucocorticoid Receptor-Dependent Mechanism
title_short Dexamethasone-Induced Myeloid-Derived Suppressor Cells Prolong Allo Cardiac Graft Survival through iNOS- and Glucocorticoid Receptor-Dependent Mechanism
title_sort dexamethasone induced myeloid derived suppressor cells prolong allo cardiac graft survival through inos and glucocorticoid receptor dependent mechanism
topic myeloid-derived suppressor cells
immune tolerance
dexamethasone
iNOS
glucocorticoid receptor
url http://journal.frontiersin.org/article/10.3389/fimmu.2018.00282/full
work_keys_str_mv AT yangzhao dexamethasoneinducedmyeloidderivedsuppressorcellsprolongallocardiacgraftsurvivalthroughinosandglucocorticoidreceptordependentmechanism
AT xiaofeishen dexamethasoneinducedmyeloidderivedsuppressorcellsprolongallocardiacgraftsurvivalthroughinosandglucocorticoidreceptordependentmechanism
AT kecao dexamethasoneinducedmyeloidderivedsuppressorcellsprolongallocardiacgraftsurvivalthroughinosandglucocorticoidreceptordependentmechanism
AT jieding dexamethasoneinducedmyeloidderivedsuppressorcellsprolongallocardiacgraftsurvivalthroughinosandglucocorticoidreceptordependentmechanism
AT xingkang dexamethasoneinducedmyeloidderivedsuppressorcellsprolongallocardiacgraftsurvivalthroughinosandglucocorticoidreceptordependentmechanism
AT wenxianguan dexamethasoneinducedmyeloidderivedsuppressorcellsprolongallocardiacgraftsurvivalthroughinosandglucocorticoidreceptordependentmechanism
AT yitaoding dexamethasoneinducedmyeloidderivedsuppressorcellsprolongallocardiacgraftsurvivalthroughinosandglucocorticoidreceptordependentmechanism
AT baoruiliu dexamethasoneinducedmyeloidderivedsuppressorcellsprolongallocardiacgraftsurvivalthroughinosandglucocorticoidreceptordependentmechanism
AT junfengdu dexamethasoneinducedmyeloidderivedsuppressorcellsprolongallocardiacgraftsurvivalthroughinosandglucocorticoidreceptordependentmechanism