The blood-brain barrier (BBB) is a physical and metabolic barrier that separates the peripheral circulation from the central nervous system (CNS) and serves to regulate and protect the microenvironment of the brain. Disruption of the BBB occurs in a number of pathological conditions, including inflammatory regulated disorders, Alzheimer's disease, diabetes, multiple sclerosis, and ischemia/reperfusion. Hypoxia and reoxygenation correlate with changes in ischemia and reperfusion, are associated with various disease states which significantly influence the BBB, with subsequent effects in more sensitive neural tissue. Clarifying the extent and manner of BBB alterations induced by the hypoxia (H) and/or post-hypoxic reoxygenation (H/R), remains a considerable task. Examination of the regulation and activation of intracellular pathways, transcription factors, and tight junction proteins under such conditions is vital to understanding these BBB alterations. The overall goal of this submission is to determine the intracellular signaling mechanisms (protein kinase C and transcription factors) responsible for BBB structural and functional changes during hypoxia/post-hypoxic reoxygenation exposure. These studies will focus on the effects of H and H/R on the signaling molecule, protein kinase C, and the transcription factors, HIF1 and NFKappaB, and how these mediators contribute to BBB disruptions which lead to vasogenic brain edema, a major factor associated with ischemic stroke. Our hypothesis is that functional changes at the BBB occur due to alterations in the cytoarchitecture of the endothelial cell as a result of intracellular mechanisms/ mediators stimulated by hypoxia and/or post-hypoxic reoxvqenation insult. To investigate this hypothesis, our objectives and Specific Aims are to examine the effects of H and H/R insult on capillary endothelial cells of the BBB, by examining BBB permeability changes, tight junction/ cytoskeletal protein expression and localization, and changes in transcription factor expression/activation in conjunction with cellular signaling pathways/mechanisms (PKC). This study requires the use of multiple techniques and the integration of both in vitro (i.e., rat cerebral microvessel endothelial cells) and in vivo (i.e., Sprague-Dawley rat cerebral microvessel) models to allow us to differentiate and correlate cellular signaling pathways to functional and structural changes associated with BBB perturbation following H and H/R insult. Our goal is to gain a better understanding on the mechanisms involved in hypoxia associated with ischemic stroke at the BBB, leading to better treatment paradigms.

Agency
National Institute of Health (NIH)
Institute
National Institute of Neurological Disorders and Stroke (NINDS)
Type
Research Project (R01)
Project #
5R01NS039592-10
Application #
7540941
Study Section
Special Emphasis Panel (ZRG1-BDCN-D (90))
Program Officer
Jacobs, Tom P
Project Start
2000-01-01
Project End
2010-12-31
Budget Start
2009-01-01
Budget End
2010-12-31
Support Year
10
Fiscal Year
2009
Total Cost
$330,188
Indirect Cost
Name
University of Arizona
Department
Pharmacology
Type
Schools of Medicine
DUNS #
806345617
City
Tucson
State
AZ
Country
United States
Zip Code
85721
Ronaldson, Patrick T; Davis, Thomas P (2015) Targeting transporters: promoting blood-brain barrier repair in response to oxidative stress injury. Brain Res 1623:39-52
McCaffrey, Gwen; Staatz, William D; Sanchez-Covarrubias, Lucy et al. (2012) P-glycoprotein trafficking at the blood-brain barrier altered by peripheral inflammatory hyperalgesia. J Neurochem 122:962-75
McCaffrey, Gwen; Davis, Thomas P (2012) Physiology and pathophysiology of the blood-brain barrier: P-glycoprotein and occludin trafficking as therapeutic targets to optimize central nervous system drug delivery. J Investig Med 60:1131-40
Willis, Colin L; Meske, Diana S; Davis, Thomas P (2010) Protein kinase C activation modulates reversible increase in cortical blood-brain barrier permeability and tight junction protein expression during hypoxia and posthypoxic reoxygenation. J Cereb Blood Flow Metab 30:1847-59
Lochhead, Jeffrey J; McCaffrey, Gwen; Quigley, Colleen E et al. (2010) Oxidative stress increases blood-brain barrier permeability and induces alterations in occludin during hypoxia-reoxygenation. J Cereb Blood Flow Metab 30:1625-36
McCaffrey, Gwen; Willis, Colin L; Staatz, William D et al. (2009) Occludin oligomeric assemblies at tight junctions of the blood-brain barrier are altered by hypoxia and reoxygenation stress. J Neurochem 110:58-71
Wang, Yaoming; Thiyagarajan, Meenakshisundaram; Chow, Nienwen et al. (2009) Differential neuroprotection and risk for bleeding from activated protein C with varying degrees of anticoagulant activity. Stroke 40:1864-9
Campos, Christopher R; Ocheltree, Scott M; Hom, Sharon et al. (2008) Nociceptive inhibition prevents inflammatory pain induced changes in the blood-brain barrier. Brain Res 1221:6-13
McCaffrey, Gwen; Seelbach, Melissa J; Staatz, William D et al. (2008) Occludin oligomeric assembly at tight junctions of the blood-brain barrier is disrupted by peripheral inflammatory hyperalgesia. J Neurochem 106:2395-409
Witt, Ken A; Mark, Karen S; Sandoval, Karin E et al. (2008) Reoxygenation stress on blood-brain barrier paracellular permeability and edema in the rat. Microvasc Res 75:91-6

Showing the most recent 10 out of 36 publications