Nearly 20% of the US population suffers from autoimmune diseases, which are among the top ten causes of death by disease for women under 65 and the 4th leading cause of disability. This also imposes a severe economic burden measured in billions of dollars. Therapies for many autoimmune disorders rely on glucocorticoids (GC) to inhibit the production of cytokines, such as interferons (IFN)a/?, which are intimately linked to the pathogenesis of lupus, autoimmune diabetes and thyroiditis. Yet, serious side effects associated with GC use can outweigh the benefits of these otherwise effective and affordable drugs. Our long-term goal is to identify the key viewers and mechanisms of GC-mediated immunomodulation in health and disease, which is essential for developing more specific therapies for autoimmunity. GC signal through a ligand-dependent GC receptor, GR, which recruits accessory cofactors to regulate transcription. Specifically, GR-dependent repression of AP1- and NF-?B -responsive genes accounts for certain anti-inflammatory activities of GC. While additional regulators must mediate the profound GC-induced immunosuppression, their identity and mechanisms of action are obscure. We established a nuclear receptor cofactor, GR-interacting protein (GRIP)1 as a corepressor for GR: NF-?B and GR: AP1 repression complexes. Unexpectedly, a yeast 2-hybrid screen for GRIP1 Repression Domain interactors yielded Interferon Regulatory Factor (IRF)3, a key effector of innate immune responses downstream of Toll- like receptors (TLR)3/4 and an essential transactivator for IFN?. The objective of this application is to decipher the functional interaction between GC and IRF3. We hypothesize that GRIP1 is an IRF3 cofactor with a role in IFN production and autoimmunity. We further propose that GR and IRF3 compete for GRIP1 such that hormone-activated GR, in addition to directly repressing AP1/NF-?B, sequesters GRIP1 away from IRF3; the resulting inhibition of IRF3-dependent transcription is potentially a critical yet unexplored component of GC immunosuppression. Conversely, in response to bacterial or viral TLR3/4 ligands, IRF3 not only enhances cytokine transcription directly, but also depletes GRIP1 from GR:AP1 and GR: NF-?B repression complexes thereby promoting an effective immune response.
Our Specific Aims are to: (1) dissect the GRIP1:IRF3 interaction in vitro, in cells and in IRF3 complexes bound at the IFN-Stimulated Response Elements; (2) corroborate the relevance of this interaction by examining the cross-talk between GR and IRF3 in primary macrophages; (3) identify the role of GRIP1 in IRF3 transcription and, using GRIP1 KO mice, in innate immune responses to virus, or to dsRNA and bacterial LPS in vivo. The GRIP1:IRF3 interaction uncovered in an unbiased screen may have a broad physiological significance. We will explore a role of GRIP1 in the immune system and decipher associated transcriptional mechanisms operative at IRF3 target genes. This work should provide a molecular basis for a functional antagonism between GC and IFN, and help define IRF3 as a novel target for GR-mediated immunosuppression, ultimately aiding in the design of more specific drugs for autoimmunity. ? ? ?

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Research Project (R01)
Project #
1R01AI068820-01A1
Application #
7192223
Study Section
Cellular and Molecular Immunology - A Study Section (CMIA)
Program Officer
Palker, Thomas J
Project Start
2006-12-15
Project End
2011-11-30
Budget Start
2006-12-15
Budget End
2007-11-30
Support Year
1
Fiscal Year
2007
Total Cost
$391,369
Indirect Cost
Name
Hospital for Special Surgery
Department
Type
DUNS #
622146454
City
New York
State
NY
Country
United States
Zip Code
10021
Chinenov, Yurii; Gupte, Rebecca; Rogatsky, Inez (2013) Nuclear receptors in inflammation control: repression by GR and beyond. Mol Cell Endocrinol 380:55-64
Bhandare, Reena; Damera, Gautam; Banerjee, Audreesh et al. (2010) Glucocorticoid receptor interacting protein-1 restores glucocorticoid responsiveness in steroid-resistant airway structural cells. Am J Respir Cell Mol Biol 42:9-15
Adelman, Karen; Rogatsky, Inez (2010) RNA polymerase II stalling mediates cytokine gene expression. Cell Cycle 9:630-1
Flammer, Jamie R; Dobrovolna, Jana; Kennedy, Megan A et al. (2010) The type I interferon signaling pathway is a target for glucocorticoid inhibition. Mol Cell Biol 30:4564-74
Adelman, Karen; Kennedy, Megan A; Nechaev, Sergei et al. (2009) Immediate mediators of the inflammatory response are poised for gene activation through RNA polymerase II stalling. Proc Natl Acad Sci U S A 106:18207-12
Chinenov, Yurii; Sacta, Maria A; Cruz, Anna R et al. (2008) GRIP1-associated SET-domain methyltransferase in glucocorticoid receptor target gene expression. Proc Natl Acad Sci U S A 105:20185-90