Microvascular hyperpermeability represents an end-point cellular process underlying the development of ischemic injury, diabetes, atherosclerosis, and various inflammatory diseases. The abnormality is largely attributed to neutrophil (PMN) activation and secretion of inflammatory mediators that are capable of altering the structural and functional integrity of the endothelial barrier. Currently our knowledge of the molecular control of endothelial barrier function is limited. Even less is known about PMN-induced endothelial changes in intact exchange microvessels. Among the reasons given for the lack of such information is the paucity of suitable experimental models that directly link subcellular events to physiological function. In this regard, one of our accomplishments over the prior funding period is the development of new techniques that facilitate correlative analyses of molecular reactions and microvascular function. Furthermore, the studies have produced a large body of experimental data that forms a signaling paradigm for venular hyperpermeability in inflammation. Three important endothelial structures, namely the contractile cytoskeleton, adherens junction, and focal adhesion, have been found to mediate barrier dysfunction. The current proposal is to extend these original investigations to a detailed mechanism of endothelial response to PMN activation. Our working hypothesis centers on biochemical and conformational modulations characterized by myosin light chain phosphorylation-dependent endothelial cell contraction, VE-cadherin/b-catenin sequestration-induced junctional weakening, and focal adhesion kinase-signaled focal adhesion rearrangement. The hypothesis will be tested by an approach that integrates physiological experiments with biochemical, biophysical, and molecular biology analyses at both the intact microvascular and subcellular levels. The newly developed microvessel transfection technique will be incorporated into a comprehensive evaluation of the cause-effect relationship between molecular reactions and venular barrier dysfunction in PMN activation. The study should provide novel insights into the molecular basis of microvascular leakage in inflammation. Identification and characterization of the key molecules that serve as ultimate effectors in the hyperpermeability reaction is fundamental to the development of therapeutic strategies directed against the injury.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
2R01HL061507-05
Application #
6580969
Study Section
Experimental Cardiovascular Sciences Study Section (ECS)
Program Officer
Massicot-Fisher, Judith
Project Start
1999-01-01
Project End
2006-12-31
Budget Start
2003-02-15
Budget End
2003-12-31
Support Year
5
Fiscal Year
2003
Total Cost
$312,539
Indirect Cost
Name
Texas A&M University
Department
Physiology
Type
Schools of Medicine
DUNS #
141582986
City
College Station
State
TX
Country
United States
Zip Code
77845
Beard Jr, Richard S; Haines, Ricci J; Wu, Kevin Y et al. (2014) Non-muscle Mlck is required for ?-catenin- and FoxO1-dependent downregulation of Cldn5 in IL-1?-mediated barrier dysfunction in brain endothelial cells. J Cell Sci 127:1840-53
Sun, Chongxiu; Beard Jr, Richard S; McLean, Danielle L et al. (2013) ADAM15 deficiency attenuates pulmonary hyperpermeability and acute lung injury in lipopolysaccharide-treated mice. Am J Physiol Lung Cell Mol Physiol 304:L135-42
Rigor, Robert R; Shen, Qiang; Pivetti, Christopher D et al. (2013) Myosin light chain kinase signaling in endothelial barrier dysfunction. Med Res Rev 33:911-33
Yuan, Sarah Y; Shen, Qiang; Rigor, Robert R et al. (2012) Neutrophil transmigration, focal adhesion kinase and endothelial barrier function. Microvasc Res 83:82-8
Lee, Eugene S; Van Spyk, Elyse N; Chun, Kevin C et al. (2012) Monocytic adhesion molecule expression and monocyte-endothelial cell dysfunction are increased in patients with peripheral vascular disease versus patients with abdominal aortic aneurysms. J Surg Res 177:373-81
Guo, Mingzhang; Yuan, Sarah Y; Frederich, Bert J et al. (2012) Role of non-muscle myosin light chain kinase in neutrophil-mediated intestinal barrier dysfunction during thermal injury. Shock 38:436-43
Sun, Chongxiu; Wu, Mack H; Lee, Eugene S et al. (2012) A disintegrin and metalloproteinase 15 contributes to atherosclerosis by mediating endothelial barrier dysfunction via Src family kinase activity. Arterioscler Thromb Vasc Biol 32:2444-51
Rigor, Robert R; Beard Jr, Richard S; Litovka, Olesya P et al. (2012) Interleukin-1?-induced barrier dysfunction is signaled through PKC-? in human brain microvascular endothelium. Am J Physiol Cell Physiol 302:C1513-22
Lee, Eugene S; Shen, Qiang; Pitts, Robert L et al. (2011) Serum metalloproteinases MMP-2, MMP-9, and metalloproteinase tissue inhibitors in patients are associated with arteriovenous fistula maturation. J Vasc Surg 54:454-9; discussion 459-60
Sun, Chongxiu; Wu, Mack H; Yuan, Sarah Y (2011) Nonmuscle myosin light-chain kinase deficiency attenuates atherosclerosis in apolipoprotein E-deficient mice via reduced endothelial barrier dysfunction and monocyte migration. Circulation 124:48-57

Showing the most recent 10 out of 44 publications