Endogenous inflammatory factors are key to the pathogenesis of inflammatory and autoimmune disorders such as rheumatoid arthritis and atherosclerosis. Acute-phase proteins (APPs) are produced in response to infectious agents as well as non-infectious insult such as trauma, burn, surgery and malignancy. Clinical evidence suggests that APPs such as serum amyloid A (SAA) and C-reactive proteins (CRP) are major biomarkers for inflammatory diseases. However, the relationship between APP production and disease progression remains undefined. Our recent work shows that SAA has cytokine-inducing activity and is an endogenous stimulant for IL-23 expression. We also identified Toll- like receptor 2 (TLR2) as a functional receptor for SAA, and other published reports suggest that SAA activates TLR4 and the chemoattractant receptor FPR2. Given recent implication of these receptors in inflammatory and autoimmune diseases, we hypothesize that SAA is an endogenous mediator that deciphers danger signals through activation of the TLRs and chemoattractant receptors. In this application, we will establish an active role of SAA in sterile inflammation and identify the respective functions of the SAA receptors.
Aim 1 will examine the effect of global rise in SAA concentration by investigating a potential role of HDL in modulating the cytokine-inducing activity of SAA, which is present in the blood circulation as an apolipoprotein of HDL. We will determine how the presence of HDL influences the in vivo functions of SAA by comparing the activity of SAA in WT and apo A-1-/- apo E-/- double KO mice, which lack HDL. Using a mouse model recently developed in our lab, we will examine the effect of HDL on the ability of SAA to induced neutrophilia in mice.
In Aim 2, we will determine the structural and functional properties of SAA isoforms in activating the SAA receptors (TLR2, TLR4 and FPR2). Using SAA receptor reconstitution and DNA mutagenesis, we will identify structural determinants for SAA interaction with TLR2, TLR4 and FPR2, thereby defining the specific SAA isoforms and receptors with respect to their biological functions. Characterization of the mouse SAA3 will be conducted to explore mSAA3 as a non-hepatic APP that resembles locally produced SAA in human diseases.
In Aim 3, we will determine the pathophysiological functions of SAA in an inflammatory disease model. Using SAA transgenic mice, we will examine in vivo SAA expression in the development and progression of inflammatory arthritis. The involvement of the SAA receptors in joint inflammation will be determined using knockout mice lacking the individual receptors. Collectively, these studies aim to establish SAA as an important endogenous ligand and a damage-associated molecular pattern for activation of inflammatory cells through TLRs and chemoattractant receptors.

Public Health Relevance

This study aims to characterize the in vivo functions of an acute-phase protein, serum amyloid A or SAA, whose expression is associated with acute inflammatory diseases. Our goal is to understand how SAA attracts and activates inflammatory cells, what receptors it uses for these functions, and what roles it plays in the development of arthritis. We expect that the knowledge gained from this study will help to understand the functions of a major biomarker for acute inflammation and to identify novel targets for anti-inflammatory therapy.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
High Priority, Short Term Project Award (R56)
Project #
2R56AI040176-12A2
Application #
8089973
Study Section
Innate Immunity and Inflammation Study Section (III)
Program Officer
Dong, Gang
Project Start
1997-07-01
Project End
2011-07-31
Budget Start
2010-08-01
Budget End
2011-07-31
Support Year
12
Fiscal Year
2010
Total Cost
$406,330
Indirect Cost
Name
University of Illinois at Chicago
Department
Pharmacology
Type
Schools of Medicine
DUNS #
098987217
City
Chicago
State
IL
Country
United States
Zip Code
60612
Cheng, Ni; Liang, Yurong; Du, Xiaoping et al. (2018) Serum amyloid A promotes LPS clearance and suppresses LPS-induced inflammation and tissue injury. EMBO Rep 19:
Ye, Richard D; Sun, Lei (2015) Emerging functions of serum amyloid A in inflammation. J Leukoc Biol 98:923-9
Sun, Lei; Zhou, Huibin; Zhu, Ziyan et al. (2015) Ex vivo and in vitro effect of serum amyloid a in the induction of macrophage M2 markers and efferocytosis of apoptotic neutrophils. J Immunol 194:4891-900
Yan, Qian; Sun, Lei; Zhu, Ziyan et al. (2014) Jmjd3-mediated epigenetic regulation of inflammatory cytokine gene expression in serum amyloid A-stimulated macrophages. Cell Signal 26:1783-91
Chen, Mingjie; Zhou, Huibing; Cheng, Ni et al. (2014) Serum amyloid A1 isoforms display different efficacy at Toll-like receptor 2 and formyl peptide receptor 2. Immunobiology 219:916-23
Sun, Lei; Zhu, Ziyan; Cheng, Ni et al. (2014) Serum amyloid A induces interleukin-33 expression through an IRF7-dependent pathway. Eur J Immunol 44:2153-64
Hoeppner, Crystal Zoe; Cheng, Ni; Ye, Richard D (2012) Identification of a nuclear localization sequence in ?-arrestin-1 and its functional implications. J Biol Chem 287:8932-43
Gantner, Benjamin N; Jin, Huali; Qian, Feng et al. (2012) The Akt1 isoform is required for optimal IFN-? transcription through direct phosphorylation of ?-catenin. J Immunol 189:3104-11
Yang, Ming; He, Rong L; Benovic, Jeffrey L et al. (2009) beta-Arrestin1 interacts with the G-protein subunits beta1gamma2 and promotes beta1gamma2-dependent Akt signalling for NF-kappaB activation. Biochem J 417:287-96