verbatim): An increase in skeletal muscle mass is accompanied by the proliferation of endothelial cells and growth of arterioles. This vessel growth is triggered by the production of endothelial cell trophic factors by differentiating myofibers, such as vascular endothelial growth factor (VEGF), leading to the induction of angiogenesis. Recent data in cell culture systems demonstrate that Akt signaling is both essential and sufficient for endothelial cell survival, NO production and migration in response to angiogenic growth factor stimulation. Since these cellular responses are believed to be features of the angiogenic process, Akt may be uniquely situated within the endothelial cell signaling cascade to function as a key regulator of blood vessel growth. The experiments proposed here will extend the in vitro observations by testing the role of Akt signaling in blood vessel growth and vascular homeostasis in animal models. The proposed gene transfer studies will provide a better understanding of the signaling pathways that mediate angiogenesis within skeletal muscle and may lead to the development of new molecular therapeutic strategies to control blood vessel growth. Therefore, we propose to: 1) Assess the role of Akt signaling in vascular permeability using the Miles assay, 2) Examine the role of Akt signaling on blood vessel formation in matrigel plugs in mice, 3) Assess the consequences of enhanced endothelial cell Akt signaling in transgenic mice, and 4) Determine the role of endothelial Akt signaling in the revascularization of skeletal muscle in ischemic rabbit hindlimb.

Agency
National Institute of Health (NIH)
Institute
National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS)
Type
Research Project (R01)
Project #
5R01AR040197-15
Application #
6847855
Study Section
Cardiovascular and Pulmonary Research A Study Section (CVA)
Program Officer
Nuckolls, Glen H
Project Start
1991-01-15
Project End
2006-02-28
Budget Start
2005-03-01
Budget End
2006-02-28
Support Year
15
Fiscal Year
2005
Total Cost
$270,988
Indirect Cost
Name
Boston University
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
604483045
City
Boston
State
MA
Country
United States
Zip Code
02118
Ouchi, Noriyuki; Walsh, Kenneth (2007) Adiponectin as an anti-inflammatory factor. Clin Chim Acta 380:24-30
Galasso, Gennaro; Schiekofer, Stephan; Sato, Kaori et al. (2006) Impaired angiogenesis in glutathione peroxidase-1-deficient mice is associated with endothelial progenitor cell dysfunction. Circ Res 98:254-61
Schiekofer, Stephan; Shiojima, Ichiro; Sato, Kaori et al. (2006) Microarray analysis of Akt1 activation in transgenic mouse hearts reveals transcript expression profiles associated with compensatory hypertrophy and failure. Physiol Genomics 27:156-70
Ouchi, Noriyuki; Shibata, Rei; Walsh, Kenneth (2006) Cardioprotection by adiponectin. Trends Cardiovasc Med 16:141-6
Aprahamian, Tamar; Bonegio, Ramon; Rizzo, Jennifer et al. (2006) Simvastatin treatment ameliorates autoimmune disease associated with accelerated atherosclerosis in a murine lupus model. J Immunol 177:3028-34
Ouchi, Noriyuki; Shibata, Rei; Walsh, Kenneth (2006) Targeting adiponectin for cardioprotection. Expert Opin Ther Targets 10:573-81
Schiekofer, Stephan; Galasso, Gennaro; Sato, Kaori et al. (2005) Impaired revascularization in a mouse model of type 2 diabetes is associated with dysregulation of a complex angiogenic-regulatory network. Arterioscler Thromb Vasc Biol 25:1603-9
Shibata, Rei; Sato, Kaori; Pimentel, David R et al. (2005) Adiponectin protects against myocardial ischemia-reperfusion injury through AMPK- and COX-2-dependent mechanisms. Nat Med 11:1096-103
Shibata, Rei; Ouchi, Noriyuki; Ito, Masahiro et al. (2004) Adiponectin-mediated modulation of hypertrophic signals in the heart. Nat Med 10:1384-9
Mogi, Masaki; Yang, Jiang; Lambert, Jean-Francois et al. (2003) Akt signaling regulates side population cell phenotype via Bcrp1 translocation. J Biol Chem 278:39068-75

Showing the most recent 10 out of 27 publications