Inflammatory disorders account for a significant proportion of disabling and life-threatening disease. For example, heart disease, chronic lower respiratory diseases, sepsis, and hypertension/hypertensive renal disease accounted for 28%, 5.2%, 1.4%, and 0.9%, respectively, of all deaths in the US in 2003. Dramatic changes in arginine metabolism occur in inflammation and injury as a result of changes in activity or expression of the nitric oxide synthases (NOS) and the arginases. There is considerable evidence that increases in arginase activity can contribute to the pathophysiologic consequences of inflammation. However, the precise contributions of changes in arginase activity and expression to inflammatory disease processes are not fully understood, thus limiting rational development of therapeutic strategies. The arginases are of particular interest because they can affect the synthesis not only of NO but also of polyamines and proline, which are essential for cell proliferation and collagen synthesis, respectively. The latter processes are involved not only in wound healing but also in fibrotic complications of chronic inflammation, such as vascular remodeling. Our understanding of the role and regulation of arginase activity in these processes is complicated by the existence of two arginase isozymes that are independently regulated, as well as by absence of information regarding expression of arginine transporters and other enzymes that use and produce arginine metabolites. In order to elucidate the roles of the individual arginase isozymes in vivo and in specific cell types, this proposal will use mouse strains in which expression of individual arginase isozymes has been selectively ablated or overexpressed in specific cell types. The following aims will test specific hypotheses regarding the roles of each arginase isozyme in vivo and in specific cell types during responses to inflammatory stimuli:
AIM I. Elucidate the contributions of each arginase isozyme in in vivo models of acute and chronic inflammation.
AIM II. Elucidate the metabolic roles of each arginase isozyme in macrophages and endothelial cells in inflammation.
AIM III. Elucidate mechanisms by which increased arginase activity alters expression of specific genes. OUTCOME: Results of these studies will provide a basis for development of tests for rational development of new and more effective therapeutic strategies and for monitoring efficacy of treatment in diseases in which arginases play a role.

Public Health Relevance

It is now recognized that increases in activities and expression of the arginase isoenzymes can contribute to development or progression of many inflammatory diseases. However, the mechanisms whereby the increases in arginase activity contribute to the initiation and progression of disease are not fully understood. The objective of this proposal is to elucidate the roles of the individual arginase isoenzymes in multiple models of inflammation in mice and in cultured cells. Results of these studies will provide a basis for rational development of new and more effective therapeutic strategies and for monitoring efficacy of treatment in inflammatory diseases.

Agency
National Institute of Health (NIH)
Institute
National Institute of General Medical Sciences (NIGMS)
Type
Research Project (R01)
Project #
5R01GM057384-11
Application #
7993584
Study Section
Surgery, Anesthesiology and Trauma Study Section (SAT)
Program Officer
Somers, Scott D
Project Start
1998-05-01
Project End
2012-11-30
Budget Start
2010-12-01
Budget End
2011-11-30
Support Year
11
Fiscal Year
2011
Total Cost
$356,364
Indirect Cost
Name
University of Pittsburgh
Department
Genetics
Type
Schools of Medicine
DUNS #
004514360
City
Pittsburgh
State
PA
Country
United States
Zip Code
15213
Zhu, Min; Goetsch, Sean C; Wang, Zhaoning et al. (2015) FoxO4 promotes early inflammatory response upon myocardial infarction via endothelial Arg1. Circ Res 117:967-77
Zhu, Xinmei; Pribis, John P; Rodriguez, Paulo C et al. (2014) The central role of arginine catabolism in T-cell dysfunction and increased susceptibility to infection after physical injury. Ann Surg 259:171-8
Mattila, Joshua T; Ojo, Olabisi O; Kepka-Lenhart, Diane et al. (2013) Microenvironments in tuberculous granulomas are delineated by distinct populations of macrophage subsets and expression of nitric oxide synthase and arginase isoforms. J Immunol 191:773-84
You, Hanning; Gao, Ting; Cooper, Timothy K et al. (2013) Arginase inhibition mediates renal tissue protection in diabetic nephropathy by a nitric oxide synthase 3-dependent mechanism. Kidney Int 84:1189-97
Chang, Jinsam; Thangamani, Shankar; Kim, Myung H et al. (2013) Retinoic acid promotes the development of Arg1-expressing dendritic cells for the regulation of T-cell differentiation. Eur J Immunol 43:967-78
Sheldon, Kathryn E; Shandilya, Harish; Kepka-Lenhart, Diane et al. (2013) Shaping the murine macrophage phenotype: IL-4 and cyclic AMP synergistically activate the arginase I promoter. J Immunol 191:2290-8
Vaisman, Boris L; Andrews, Karen L; Khong, Sacha M L et al. (2012) Selective endothelial overexpression of arginase II induces endothelial dysfunction and hypertension and enhances atherosclerosis in mice. PLoS One 7:e39487
Morris Jr, Sidney M (2012) Arginases and arginine deficiency syndromes. Curr Opin Clin Nutr Metab Care 15:64-70
Csóka, Balázs; Selmeczy, Zsolt; Koscsó, Balázs et al. (2012) Adenosine promotes alternative macrophage activation via A2A and A2B receptors. FASEB J 26:376-86
Morris Jr, Sidney M; Gao, Ting; Cooper, Timothy K et al. (2011) Arginase-2 mediates diabetic renal injury. Diabetes 60:3015-22

Showing the most recent 10 out of 34 publications