In rheumatoid arthritis (RA) inflammatory cytokines produced by synovial (joint) cells drive disease pathogenesis by activating cells in inflamed joints and inducing recruitment of inflammatory cells. Key pathogenic roles for the cytokines tumor necrosis factor (TNF) and interleukin-6 (IL-6) in human RA have been demonstrated by the efficacy of therapies that specifically target these cytokines using blocking antibodies or soluble receptors. The long term goals of this project are to understand how cytokines activate synovial cells to drive disease pathogenesis and cause morbidity, and to elucidate mechanisms that regulate production of pathogenic cytokines in the context of RA synovitis. An associated goal is to use this knowledge to develop safer and more effective therapies that selectively target disease-related mechanisms of cytokine production and function, while preserving aspects of host defense. This project has focused on two key synovial cell types important in RA pathogenesis, fibroblast-like synoviocytes (FLS) and synovial macrophages (previously termed type A synoviocytes). In the previous project period, we found that TNF induces a sustained inflammatory response in FLS, including prolonged activation of NF-kB and transcription of genes that encode IL-6, IL-8 and MMPs. We propose that lack of effective deactivation of inflammatory responses in FLS, which contrasts with homeostatic feed-back inhibition (`tolerization') that deactivates hematopoietic cells, contributes to unremitting inflammation in chronic RA. Two complementary mechanisms underlying prolonged TNF-induced activation and inflammatory gene expression in FLS are ineffective termination of TNF and NF-kB signaling, and ineffective silencing of inflammatory gene loci by epigenetic chromatin-mediated mechanisms. Thus, ineffective engagement of homeostatic signaling and epigenetic mechanisms that terminate inflammatory responses in other cell types (including macrophages) leads to sustained inflammatory responses in FLS. Based on our overarching hypothesis that augmenting homeostatic mechanisms represents an effective therapeutic approach to suppress inflammation, we further investigated mechanisms underlying prolonged TNF-induced signaling and inflammatory gene expression. A mechanism underlying prolonged inflammatory gene expression is sustained opening of chromatin and hyperacetylation of histones, which results in sensitivity to inhibition by I-BET, a small molecule that blocks interactions of transcriptional co- activators with acetylated histones. A functional consequence of prolonged FLS activation by TNF is production of soluble factors that alter macrophage polarization and inflammatory mediator production, thus identifying a new function of FLS. In this project, we will investigate epigenetic mechanisms that regulate inflammatory activation of FLS, and the functional consequences of prolonged FLS activation on macrophage phenotype and in vivo arthritis models.

Public Health Relevance

In rheumatoid arthritis (RA) cytokines activate cells (synoviocytes) in inflamed joints to drive the disease process. In this project, we will investigat epigenetic mechanisms that regulate inflammatory activation and function of synoviocytes. We anticipate that our studies will provide knowledge that can be used to develop RA therapies based on a strategy of augmenting mechanisms that deactivate inflammation.

Agency
National Institute of Health (NIH)
Institute
National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS)
Type
Research Project (R01)
Project #
5R01AR046713-19
Application #
9529244
Study Section
Arthritis, Connective Tissue and Skin Study Section (ACTS)
Program Officer
Mao, Su-Yau
Project Start
1999-09-27
Project End
2020-05-31
Budget Start
2018-06-01
Budget End
2019-05-31
Support Year
19
Fiscal Year
2018
Total Cost
Indirect Cost
Name
Hospital for Special Surgery
Department
Type
DUNS #
City
New York
State
NY
Country
United States
Zip Code
10021
Stephenson, William; Donlin, Laura T; Butler, Andrew et al. (2018) Single-cell RNA-seq of rheumatoid arthritis synovial tissue using low-cost microfluidic instrumentation. Nat Commun 9:791
Mizoguchi, Fumitaka; Slowikowski, Kamil; Wei, Kevin et al. (2018) Functionally distinct disease-associated fibroblast subsets in rheumatoid arthritis. Nat Commun 9:789
Sokhi, Upneet K; Liber, Mark P; Frye, Laura et al. (2018) Dissection and function of autoimmunity-associated TNFAIP3 (A20) gene enhancers in humanized mouse models. Nat Commun 9:658
Rao, Deepak A; Gurish, Michael F; Marshall, Jennifer L et al. (2017) Pathologically expanded peripheral T helper cell subset drives B cells in rheumatoid arthritis. Nature 542:110-114
Loupasakis, Konstantinos; Kuo, David; Sokhi, Upneet K et al. (2017) Tumor Necrosis Factor dynamically regulates the mRNA stabilome in rheumatoid arthritis fibroblast-like synoviocytes. PLoS One 12:e0179762
Park, Sung Ho; Kang, Kyuho; Giannopoulou, Eugenia et al. (2017) Type I interferons and the cytokine TNF cooperatively reprogram the macrophage epigenome to promote inflammatory activation. Nat Immunol 18:1104-1116
Kang, Kyuho; Park, Sung Ho; Chen, Janice et al. (2017) Interferon-? Represses M2 Gene Expression in Human Macrophages by Disassembling Enhancers Bound by the Transcription Factor MAF. Immunity 47:235-250.e4
Ivashkiv, Lionel B; Park, Sung Ho (2016) Epigenetic Regulation of Myeloid Cells. Microbiol Spectr 4:
Fang, Celestia; Qiao, Yu; Mun, Se Hwan et al. (2016) Cutting Edge: EZH2 Promotes Osteoclastogenesis by Epigenetic Silencing of the Negative Regulator IRF8. J Immunol 196:4452-4456
Lee, Min Joon; Lim, Elisha; Mun, Sehwan et al. (2016) Intravenous Immunoglobulin (IVIG) Attenuates TNF-Induced Pathologic Bone Resorption and Suppresses Osteoclastogenesis by Inducing A20 Expression. J Cell Physiol 231:449-458

Showing the most recent 10 out of 56 publications