Diabetes is one of the leading causes of mortality in the US and more than half of diabetic patients have cardiovascular complications. However, the mechanisms and the critical factors involved in the development of diabetic vascular complications remain poorly understood. Our recent studies found that microvessels in diabetic rats had increased basal permeability and a markedly augmented permeability response to inflammatory mediators. We also found that circulating microparticles (MPs), the small vesicles released from the cell surface upon activation, were significantly elevated in both diabetic patients and diabetic rats. Most importantly, results derived from our newly developed experiments showed that transfusion of diabetic rat plasma into the circulation of a normal rat causes immediate leukocyte adhesion and increased microvessel permeability, and that removal of MPs from the diabetic plasma abolished the effect, indicating an important role of diabetic MPs in mediating microvascular inflammation. Similar observations were replicated when isolated diabetic MPs were directly perfused into a normal individual microvessel. In addition, MP analysis of diabetic patients' plasma showed a positive correlation between the levels of MPs and their vascular complications. These novel preliminary findings led us to hypothesize that the increased levels of MPs under diabetic conditions are not simply the results of vascular cell activation and apoptosis, but are also vectors that disseminate pro- inflammatory and pro-coagulant mediators throughout the vascular system, exacerbating vascular dysfunction. This application aims to test this hypothesis with three specific aims.
Aim 1 is to characterize the cellular sources and identify the mechanisms of increased MPs under diabetic conditions.
Aim 2 is to investigate the causal relationship between the increased plasma MPs and the inflammatory manifestation of diabetic microvessels and to identify the mechanisms involved in MP- mediated leukocyte adhesion, thrombus formation, and increased microvessel permeability.
Aim 3 is to investigate the correlations between the increased MPs and the clinical status of diabetic patients. We will evaluate the potential of using MP analysis to predict the ris or severity of vascular complications, as well as the efficacy of diabetic therapies. Present knowledge about MPs is largely derived from in vitro studies. Our unique experimental approaches that combine flow cytometry analysis of MPs with single vessel perfusion, plasma transfusion, confocal imaging, electron microscopy, and quantitative permeability measurements in intact microvessels, will provide the most needed in vivo mechanisms of diabetic MP- mediated microvascular inflammation. The direct linkage between laboratory animal findings with patient disease conditions assures that the knowledge gained from the proposed studies will provide new insight into the pathogenesis of diabetes-associated vascular dysfunction and benefit targeted therapies.

Public Health Relevance

Diabetes is one of the leading causes of death in the US. Our pilot studies found that circulating microparticles (MPs), the small membrane-bound vesicles released from the cell surface upon activation, not only significantly elevated in both diabetic patients and diabetic animals, but also play essential roles in mediating inflammation and thrombus formation in microvessels. The proposed research aims to identify the factors causing increased levels of MPs and investigate the mechanisms by which MPs promote leukocyte adhesion and coagulation under diabetic conditions, and, therefore, contributes to a better understanding of the pathogenesis of diabetes-associated vascular complications and benefit the development of targeted therapies.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Research Project (R01)
Project #
5R01DK097391-05
Application #
9120245
Study Section
Hypertension and Microcirculation Study Section (HM)
Program Officer
Jones, Teresa L Z
Project Start
2013-09-01
Project End
2017-08-31
Budget Start
2016-09-01
Budget End
2017-08-31
Support Year
5
Fiscal Year
2016
Total Cost
Indirect Cost
Name
Pennsylvania State University
Department
Physiology
Type
Schools of Medicine
DUNS #
129348186
City
Hershey
State
PA
Country
United States
Zip Code
17033
Feng, Qilong; Stork, Christian J; Xu, Sulei et al. (2018) Increased circulating microparticles in streptozotocin-induced diabetes propagate inflammation contributing to microvascular dysfunction. J Physiol :
Begum, Gulnaz; Song, Shanshan; Wang, Shaoxia et al. (2018) Selective knockout of astrocytic Na+ /H+ exchanger isoform 1 reduces astrogliosis, BBB damage, infarction, and improves neurological function after ischemic stroke. Glia 66:126-144
Dang, Thanh Q; Yoon, Nanyoung; Chasiotis, Helen et al. (2017) Transendothelial movement of adiponectin is restricted by glucocorticoids. J Endocrinol 234:101-114
Xu, Sulei; Li, Xiang; LaPenna, Kyle Brian et al. (2017) New insights into shear stress-induced endothelial signalling and barrier function: cell-free fluid versus blood flow. Cardiovasc Res 113:508-518
Gao, Feng; Lucke-Wold, Brandon P; Li, Xiang et al. (2017) Reduction of Endothelial Nitric Oxide Increases the Adhesiveness of Constitutive Endothelial Membrane ICAM-1 through Src-Mediated Phosphorylation. Front Physiol 8:1124
Xu, Sulei; Li, Xiang; Liu, Yuxin et al. (2016) Development and Characterization of In Vitro Microvessel Network and Quantitative Measurements of Endothelial [Ca2+]i and Nitric Oxide Production. J Vis Exp :
Park, Kyoungmin; Mima, Akira; Li, Qian et al. (2016) Insulin decreases atherosclerosis by inducing endothelin receptor B expression. JCI Insight 1:
Li, Xiang; Xu, Sulei; He, Pingnian et al. (2015) In vitro recapitulation of functional microvessels for the study of endothelial shear response, nitric oxide and [Ca2+]i. PLoS One 10:e0126797
Yuan, Dong; Xu, Sulei; He, Pingnian (2014) Enhanced permeability responses to inflammation in streptozotocin-induced diabetic rat venules: Rho-mediated alterations of actin cytoskeleton and VE-cadherin. Am J Physiol Heart Circ Physiol 307:H44-53
Xu, Sulei; Zhou, Xueping; Yuan, Dong et al. (2013) Caveolin-1 scaffolding domain promotes leukocyte adhesion by reduced basal endothelial nitric oxide-mediated ICAM-1 phosphorylation in rat mesenteric venules. Am J Physiol Heart Circ Physiol 305:H1484-93