Although it has been demonstrated that iNOS plays an important role in host defense against microbial pathogens, the exact function of iNOS in T cells and chronic inflammatory diseases has not been defined. TH17 cells, which secrete interleukin 17 (IL-17) and IL-22 comprise a recently identified subset of CD4+ T cells distinct from the TH1 and TH2 subsets and RORgt has been identified as a key transcription factor for TH17 cell differentiation. Increasing evidence indicates that TH17 immune responses are involved in the pathogenesis of various autoimmune/inflammatory diseases. Thus, blocking TH17 cell activation could lead to the development of novel strategies for the treatment of chronic inflammatory diseases. We show that iNOS knock out mice display more robust TH17 cell differentiation without major effects on either TH1 or TH2 cell lineages. We demonstrated that iNOS protein was induced in activated CD4+ T cells and the use of an iNOS selective inhibitor L-NIL significantly increased the percentage of IL-17-producing CD4+ T cells in WT cell cultures, while, an NO donor, SNAP, dose-dependently suppressed IL-17 production in WT and iNOS-/- T cell cultures. In addition, tyrosine residues of RORgt protein were nitrated resulting in the inhibition of RORgt-mediated IL-17 promoter activation. Finally, transfer of iNOS-/- CD4+CD45Rbhi cells into RAG-/- mice induces more severe colitis compared to control CD4+CD45Rbhi cells and mice reconstituted with iNOS-/- cells had a significantly higher percentage of IL-17-producing cells than control mice. The results suggest that T cell-derived iNOS negatively regulates the development of TH17 immune responses resulting in the control of inflammation. This proposal is structured around three aims: 1) We will characterize the molecular mechanisms involved in the regulation of TH17 cell differentiation by T cell-derived iNOS. 2) We will characterize the function of T cell- derived-iNOS on human TH17, TH1, TH2, and Treg cell development. 3) We will analyze the roles of iNOS in different cell compartments including macrophages, dendritic cells, and T cells in the development of colitis. The proposed studies will define a novel transcriptional inhibitor of TH17 cell differentiation and highlight th importance of T cell-derived iNOS as a novel therapeutic target for the treatment of chronic inflammatory diseases.

Public Health Relevance

Crohn's disease (CD) and ulcerative colitis (UC) are the two main forms of inflammatory bowel diseases (IBD). Accumulating evidence indicates that TH17 immune responses are involved in the development of IBD. This research will shed light on T cell-derived iNOS as a potential therapeutic target for therapy in inflammatory bowel diseases.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Research Project (R01)
Project #
5R01AI104688-02
Application #
8722431
Study Section
Gastrointestinal Mucosal Pathobiology Study Section (GMPB)
Program Officer
Rothermel, Annette L
Project Start
2013-09-01
Project End
2017-08-31
Budget Start
2014-09-01
Budget End
2015-08-31
Support Year
2
Fiscal Year
2014
Total Cost
Indirect Cost
Name
Icahn School of Medicine at Mount Sinai
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
City
New York
State
NY
Country
United States
Zip Code
10029
Wang, Juan; Peng, Liang; Zhang, Ruihua et al. (2016) 5-Fluorouracil targets thymidylate synthase in the selective suppression of TH17 cell differentiation. Oncotarget 7:19312-26
Peng, Liang; Zhang, Hui; Hao, Yuanyuan et al. (2016) Reprogramming macrophage orientation by microRNA 146b targeting transcription factor IRF5. EBioMedicine 14:83-96
Geng, Shuo; Chen, Keqiang; Yuan, Ruoxi et al. (2016) The persistence of low-grade inflammatory monocytes contributes to aggravated atherosclerosis. Nat Commun 7:13436
Herbin, Olivier; Bonito, Anthony J; Jeong, Seihwan et al. (2016) Medullary thymic epithelial cells and CD8?+ dendritic cells coordinately regulate central tolerance but CD8?+ cells are dispensable for thymic regulatory T cell production. J Autoimmun 75:141-149
Lu, Geming; Zhang, Ruihua; Geng, Shuo et al. (2015) Myeloid cell-derived inducible nitric oxide synthase suppresses M1 macrophage polarization. Nat Commun 6:6676
Paschall, Amy V; Zhang, Ruihua; Qi, Chen-Feng et al. (2015) IFN regulatory factor 8 represses GM-CSF expression in T cells to affect myeloid cell lineage differentiation. J Immunol 194:2369-79
Gupta, Monica; Shin, Dong-Mi; Ramakrishna, Lakshmi et al. (2015) IRF8 directs stress-induced autophagy in macrophages and promotes clearance of Listeria monocytogenes. Nat Commun 6:6379
Jianjun Yang; Zhang, Ruihua; Lu, Geming et al. (2013) T cell–derived inducible nitric oxide synthase switches off Th17 cell differentiation. J Exp Med 210:1447-62
Zhang, Ruihua; Li, Qin; Chuang, Peter Y et al. (2013) Regulation of pathogenic Th17 cell differentiation by IL-10 in the development of glomerulonephritis. Am J Pathol 183:402-12