Chemokine-driven transmigration of monocytes into the subendothelial space is a fundamental and rate- limiting process in atherogenesis. Our preliminary data show that this process is dysregulated by metabolic stress and that increased monocyte responsiveness to chemokines appears to accelerate atherosclerotic plaque development. We have now uncovered a novel thiol redox-sensitive mechanism in monocytes that upon dysregulation by metabolic disorders, """"""""primes"""""""" and transforms monocytes into a hyper-chemotactic pro- atherogenic phenotype. In addition, we have found that the recently discovered monocytic NADPH oxidase 4 is a mediator of monocyte priming. We propose that metabolic stress-induced Nox4 and the subsequent increase in H2O2 formation in monocytes promote the S-glutathionylation and inactivation of mitogen-activated protein kinase phosphatases (MPKs);MKPs are the enzymes responsible for the deactivation of both phospho-ERK and phospho-p38MAPK, the two principal MAPK pathways mediating MCP-1-induced monocyte adhesions and migration. We hypothesize that monocyte MPKs represent a novel, critical mechanistic link between oxidative stress induced by metabolic disorders and the formation of atherosclerotic lesions. Our studies support a new paradigm implicating monocyte priming and dysfunction as an early primary contributor to atherogenesis. The studies we propose here aim to test this paradigm and elucidate the underlying molecular mechanisms.
Specific Aim 1 : Determine the roles of mitogen-activated protein kinase phosphatases (MKP) in the redox regulation of monocyte adhesion, chemotaxis, and recruitment into atherosclerotic lesions.
Specific Aim 2 : Determine the roles of protein-S-glutathionylation in monocyte recruitment and the development of atherosclerotic lesions.
Specific Aim 3 : Determine the roles of monocytic Nox4 in monocyte priming and atherogenesis.

Public Health Relevance

Monocyte recruitment into the vessel wall is an essential step in the development of atherosclerotic lesions - a process initiated by the injured vasculature releasing chemokines in response to inflammatory processes. With our discovery that chronic metabolic stress dramatically enhances the responsiveness of blood monocytes to chemoattractants and increases monocyte migration, we have uncovered a completely novel mechanism by which metabolic disorders promote atherosclerosis. Our data support a new paradigm that implicates monocyte priming and dysfunction induced by metabolic disorders as an early primary contributor to atherogenesis. The studies we propose here aim to test this paradigm and elucidate the underlying molecular mechanisms. Should our paradigm prove correct, our studies may have identified the molecular basis for a causal relationship that explains the well-established associations between metabolic disorders, oxidative stress, and cardiovascular diseases.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
5R01HL115858-03
Application #
8664912
Study Section
Vascular Cell and Molecular Biology Study Section (VCMB)
Program Officer
Fleg, Jerome
Project Start
2012-07-16
Project End
2016-04-30
Budget Start
2014-05-01
Budget End
2015-04-30
Support Year
3
Fiscal Year
2014
Total Cost
$389,008
Indirect Cost
$128,802
Name
University of Texas Health Science Center San Antonio
Department
Other Health Professions
Type
Schools of Allied Health Profes
DUNS #
800772162
City
San Antonio
State
TX
Country
United States
Zip Code
78229
Tavakoli, Sina; Short, John D; Downs, Kevin et al. (2017) Differential Regulation of Macrophage Glucose Metabolism by Macrophage Colony-stimulating Factor and Granulocyte-Macrophage Colony-stimulating Factor: Implications for 18F FDG PET Imaging of Vessel Wall Inflammation. Radiology 283:87-97
Kim, Hong Seok; Asmis, Reto (2017) Mitogen-activated protein kinase phosphatase 1 (MKP-1) in macrophage biology and cardiovascular disease. A redox-regulated master controller of monocyte function and macrophage phenotype. Free Radic Biol Med 109:75-83
Kim, Hong Seok; Tavakoli, Sina; Piefer, Leigh Ann et al. (2016) Monocytic MKP-1 is a Sensor of the Metabolic Environment and Regulates Function and Phenotypic Fate of Monocyte-Derived Macrophages in Atherosclerosis. Sci Rep 6:34223
Short, John D; Downs, Kevin; Tavakoli, Sina et al. (2016) Protein Thiol Redox Signaling in Monocytes and Macrophages. Antioxid Redox Signal 25:816-835
Zamora, D A; Downs, K P; Ullevig, S L et al. (2015) Glutaredoxin 2a overexpression in macrophages promotes mitochondrial dysfunction but has little or no effect on atherogenesis in LDL-receptor null mice. Atherosclerosis 241:69-78
Ullevig, Sarah L; Kim, Hong Seok; Nguyen, Huynh Nga et al. (2014) Ursolic acid protects monocytes against metabolic stress-induced priming and dysfunction by preventing the induction of Nox4. Redox Biol 2:259-66
Kim, Hong Seok; Ullevig, Sarah L; Nguyen, Huynh Nga et al. (2014) Redox regulation of 14-3-3? controls monocyte migration. Arterioscler Thromb Vasc Biol 34:1514-21
Ullevig, Sarah; Kim, Hong Seok; Asmis, Reto (2013) S-glutathionylation in monocyte and macrophage (dys)function. Int J Mol Sci 14:15212-32
Lee, Chi Fung; Ullevig, Sarah; Kim, Hong Seok et al. (2013) Regulation of Monocyte Adhesion and Migration by Nox4. PLoS One 8:e66964
Tavakoli, Sina; Zamora, Debora; Ullevig, Sarah et al. (2013) Bioenergetic profiles diverge during macrophage polarization: implications for the interpretation of 18F-FDG PET imaging of atherosclerosis. J Nucl Med 54:1661-7

Showing the most recent 10 out of 11 publications