Cyclooxygenase-2 (COX-2)-dependent prostaglandin E2 (PGE2) synthesis exacerbates occlusive and aneurysmal vascular disease by inducing macrophage proteinase and inflammatory cytokine expression. Despite these observations, administration of selective COX-2 inhibitors is associated with an increased frequency of adverse cardiovascular events. This paradoxical effect is not fully understood;however evidence suggests that suppression of COX-2-dependent PGI2 synthesis by endothelium renders the vascular wall more sensitive to thrombotic stimuli. Nonetheless, blocking the pathophysiologic actions of PGE2 remains an important therapeutic goal. The overall hypothesis to be tested in this application is that blocking PGE2 binding to the EP4 prostanoid receptor can inhibit vascular MMP-9 and IL-6 expression, atherosclerotic lesion development and aneurysmal dilation without altering levels of vascular PGI2. In support of this hypothesis, we report that an EP4 antagonist or EP4 knockdown is as effective as COX-2 inhibition in blocking macrophage MMP-9 expression. Moreover, EP4-dependent signaling stimulates macrophage expression of IL-6, which is linked to COX-2 and MMP-9 expression by a positive feedback loop. Also, EP4-dependent signaling stimulates secretion of PGE2 by inducing expression of microsomal PGE synthase-1 and inhibiting expression of the degradative enzyme 15-hydoxyprostaglandin dehydrogenase. Finally, inhibiting PGE2 binding to EP4 has little impact on the generation of thromboprotective PGI2 by macrophages and endothelial cells in vitro. To test the validity of our hypothesis, we propose the following specific aims: [1] Determine the role of EP4- dependent signaling on the regulation of enzymes downstream of COX-2 that regulate PGE2 synthesis and degradation by macrophages;[2] Determine the role of COX-2 and EP4-dependent signaling on the regulation of smooth muscle cell MMP-9 expression;[3] Determine whether blocking PGE2 binding to EP4 attenuates atherosclerotic lesion development in ApoE-/- mice without altering vascular prostanoid profiles;and [4] Determine whether blocking PGE2 binding to EP4 attenuates the development of angiotensin II-induced aneurysms in ApoE-/- mice without altering vascular prostanoid profiles. If successful, these studies will provide the basis for a novel therapeutic strategy to modulate occlusive and aneurysmal disease.

Public Health Relevance

Complications of atherosclerotic and aneurysmal vascular diseases remain a major cause of morbidity and mortality in the United States. Prostaglandin E2 (PGE2), secreted by inflammatory cells recruited to the arterial wall, triggers processes that weaken the wall and lead to rupture and thrombosis. Cyclooxygenase-2 (COX-2) is an important regulator of PGE2 synthesis. It was thought that selective COX-2 inhibitors would attenuate vascular lesion development by inhibiting PGE2 synthesis. However, administration of COX-2 inhibitors surprisingly caused an increased frequency of cardiovascular complications. This unexpected effect is not fully understood;however evidence suggests that COX-2 inhibitors also block PGI2 synthesis by cells lining blood vessels, which renders the artery more sensitive to injury. Nonetheless, blocking PGE2-induced weakening of the arterial wall remains an important therapeutic goal. In proposed studies, we are attempting to bypass the unwanted effect of COX-2 inhibition on PGI2 synthesis by blocking the prostanoid receptor that mediates the pathophysiologic actions of PGE2 rather than inhibiting COX-2. If successful, these studies will provide the basis for novel therapeutic strategies to reduce the complications of atherosclerotic and aneurysmal vascular diseases.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
1R01HL093331-01A2
Application #
7782971
Study Section
Atherosclerosis and Inflammation of the Cardiovascular System Study Section (AICS)
Program Officer
Tolunay, Eser
Project Start
2010-01-01
Project End
2013-12-31
Budget Start
2010-01-01
Budget End
2010-12-31
Support Year
1
Fiscal Year
2010
Total Cost
$422,500
Indirect Cost
Name
Weill Medical College of Cornell University
Department
Pathology
Type
Schools of Medicine
DUNS #
060217502
City
New York
State
NY
Country
United States
Zip Code
10065
Haka, Abigail S; Barbosa-Lorenzi, Valéria C; Lee, Hyuek Jong et al. (2016) Exocytosis of macrophage lysosomes leads to digestion of apoptotic adipocytes and foam cell formation. J Lipid Res 57:980-92
Kothari, Poonam; Pestana, Roberto; Mesraoua, Rim et al. (2014) IL-6-mediated induction of matrix metalloproteinase-9 is modulated by JAK-dependent IL-10 expression in macrophages. J Immunol 192:349-57
Siao, Chia-Jen; Lorentz, Christina U; Kermani, Pouneh et al. (2012) ProNGF, a cytokine induced after myocardial infarction in humans, targets pericytes to promote microvascular damage and activation. J Exp Med 209:2291-305
Khan, K M Faisal; Kothari, Poonam; Du, Baoheng et al. (2012) Matrix metalloproteinase-dependent microsomal prostaglandin E synthase-1 expression in macrophages: role of TNF-? and the EP4 prostanoid receptor. J Immunol 188:1970-80
Hou, Zhe; Falcone, Domenick J; Subbaramaiah, Kotha et al. (2011) Macrophages induce COX-2 expression in breast cancer cells: role of IL-1* autoamplification. Carcinogenesis 32:695-702
Steenport, Michel; Khan, K M Faisal; Du, Baoheng et al. (2009) Matrix metalloproteinase (MMP)-1 and MMP-3 induce macrophage MMP-9: evidence for the role of TNF-alpha and cyclooxygenase-2. J Immunol 183:8119-27