Vascular remodeling is the ability of the vessel wall to reorganize its cellular and extracellular components in response to a chronic stimulus. Work published during the tenure of this grant demonstrated that in response to a chronic change in blood flow, eNOS knockout mice exhibited a paradoxical increase in wall thickness accompanied by an increase in smooth muscle nuclei and proliferation, reminiscent of the changes seen in hypertension and atherosclerosis. New preliminary data show that there is marked upregulation of PDGF BB and survivin, an inhibitor of apoptosis, in abnormally remodeled vessels from eNOS (-I-) mice. In addition we show that during the ontogeny of a remodeling response in normal mice, there is a deficit in basal, but not acetylcholine stimulated endothelium-dependent responses that occurs contemporaneously with cell death in the vessel wall sensing the flow change. These data indicate that impaired endothelium-dependent relaxation of blood vessels, occurs during cellular adaptation to a prolonged remodeling stimulus. With this background in mind, we hypothesize that endothelium-derived NO coordinates hemodynamic changes in the vessel wall to long term structural changes in vessel geometry by influencing fundamental processes that govern apoptosis versus survival, extracellular matrix synthesis and degradation, vascular cell proliferation and migration. As a corollary to this hypothesis, we predict that abnormal vascular remodeling secondary to elevations in plasma cholesterol will be accentuated by the loss of eNOS derived NO. To examine the importance of eNOS derived NO in remodeling the following Specific Aims are proposed:
Aim 1. Define the relationship between eNOS and arterial remodeling in mice;
Aim 2. Examine the mechanistic links between NO, PDGF and survivin in the remodeling process and Aim 3. Elucidate the effects of elevated cholesterol levels on vascular remodeling and vasomotor function. These studies will use a multi-disciplinary approach (cellular, molecular and functional physiology) to understand the importance of eNOS derived NO in vascular remodeling. Results from these studies will assist in our understanding of mechanisms leading to abnormal remodeling in disease with endothelial dysfunction.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
5R01HL057665-08
Application #
6747656
Study Section
Pathology A Study Section (PTHA)
Program Officer
Lin, Michael
Project Start
1996-09-30
Project End
2005-06-30
Budget Start
2004-07-01
Budget End
2005-06-30
Support Year
8
Fiscal Year
2004
Total Cost
$377,000
Indirect Cost
Name
Yale University
Department
Pharmacology
Type
Schools of Medicine
DUNS #
043207562
City
New Haven
State
CT
Country
United States
Zip Code
06520
Kraehling, Jan R; Hao, Zhengrong; Lee, Monica Y et al. (2016) Uncoupling Caveolae From Intracellular Signaling In Vivo. Circ Res 118:48-55
Kraehling, Jan R; Sessa, William C (2015) Enhanced eNOS Activation as the Fountain of Youth for Vascular Disease: Is BPIFB4 What Ponce de León Was Looking For? Circ Res 117:309-10
Chidlow Jr, John H; Sessa, William C (2010) Caveolae, caveolins, and cavins: complex control of cellular signalling and inflammation. Cardiovasc Res 86:219-25
Fernández-Hernando, Carlos; József, Levente; Jenkins, Deborah et al. (2009) Absence of Akt1 reduces vascular smooth muscle cell migration and survival and induces features of plaque vulnerability and cardiac dysfunction during atherosclerosis. Arterioscler Thromb Vasc Biol 29:2033-40
Chidlow Jr, John H; Greer, Joshua J M; Anthoni, Christoph et al. (2009) Endothelial caveolin-1 regulates pathologic angiogenesis in a mouse model of colitis. Gastroenterology 136:575-84.e2
Suárez, Yajaira; Sessa, William C (2009) MicroRNAs as novel regulators of angiogenesis. Circ Res 104:442-54
Fernández-Hernando, Carlos; Yu, Jun; Suárez, Yajaira et al. (2009) Genetic evidence supporting a critical role of endothelial caveolin-1 during the progression of atherosclerosis. Cell Metab 10:48-54
Dobrucki, Lawrence W; Dione, Donald P; Kalinowski, Leszek et al. (2009) Serial noninvasive targeted imaging of peripheral angiogenesis: validation and application of a semiautomated quantitative approach. J Nucl Med 50:1356-63
Sessa, W C (2009) Molecular control of blood flow and angiogenesis: role of nitric oxide. J Thromb Haemost 7 Suppl 1:35-7
Di Lorenzo, Annarita; Fernández-Hernando, Carlos; Cirino, Giuseppe et al. (2009) Akt1 is critical for acute inflammation and histamine-mediated vascular leakage. Proc Natl Acad Sci U S A 106:14552-7

Showing the most recent 10 out of 77 publications