Despite recent advances in prevention and therapy, stroke remains the leading cause of long-term disability and the third most common cause of death in the United States. A major conceptual advance in understanding the pathophysiology of stroke was the identification of two distinct stages of disease progression: primary tissue damage in the ischemic core, and secondary tissue injury in the surrounding penumbra. The inflammatory response elicited by cerebral ischemia is a promising target for pharmaceutical intervention of stroke, because it progresses over many hours after stroke, and exacerbates tissue injury. As a feedback regulatory mechanism, an endogenous ubiquitous molecule, spermine, accumulates at sites of injury, and inhibits the release of various proinflammatory cytokines. Sperminemediated immune suppression is dependent on the negative acute phase protein, fetuin, which is significantly increased in the brains of animals subjected to experimental cerebral ischemia (middle cerebral artery occlusion, MCAO). Furthermore, peripheral administration of exogenous fetuin immediately after MCAO confers dose-dependent protection against cerebral ischemic injury at 24 hours after MCAO. Several important questions remain unanswered regarding the long-term neuroprotective efficacy of fetuin, and the mechanisms underlying ischemia-elicited elevation of brain fetuin levels, as well as the fetuin-mediated neuroprotection against cerebral ischemic injury. We will first determine the long-term effects of inhibiting (by intracerebral administration of fetuin-specific antibodies or genetic disruption of fetuin expression) or augmenting (by intravenous or intracerebroventricular administration of exogenous fetuin) cerebral fetuin levels on cerebral ischemic injury (Specific Aim 1). To delineate the mechanisms underlying ischemia-induced increase of brain fetuin levels, we will determine whether fetuin in the ischemic brain tissue is produced locally by the central nervous system, or obtained (across the blood-brain barrier) from the peripheral circulation (Specific Aim 2). Lastly, we will delineate the specific mechanisms of fetuin-mediated neuronal protection by determining whether administration of fetuin alters expression of proinflammatory cytokines (such as TNF and HMGB1), or animal physiological parameters such as blood pressure and organ perfusion (Specific Aim 3). Answers to these questions will improve our understanding of the pathophysiology of ischemic neural injury, and identify novel therapeutic agents for the treatment of stroke and related disorders.

Agency
National Institute of Health (NIH)
Institute
National Institute of General Medical Sciences (NIGMS)
Type
Research Project (R01)
Project #
5R01GM070817-04
Application #
7373505
Study Section
Surgery, Anesthesiology and Trauma Study Section (SAT)
Program Officer
Somers, Scott D
Project Start
2005-03-01
Project End
2009-08-28
Budget Start
2008-03-01
Budget End
2009-08-28
Support Year
4
Fiscal Year
2008
Total Cost
$312,900
Indirect Cost
Name
Feinstein Institute for Medical Research
Department
Type
DUNS #
110565913
City
Manhasset
State
NY
Country
United States
Zip Code
11030
Wang, H; Sama, A E (2012) Anti-inflammatory role of fetuin-A in injury and infection. Curr Mol Med 12:625-33
Li, Wei; Zhu, Shu; Li, Jianhua et al. (2011) A hepatic protein, fetuin-A, occupies a protective role in lethal systemic inflammation. PLoS One 6:e16945
Kang, R; Tang, D; Yu, Y et al. (2010) WAVE1 regulates Bcl-2 localization and phosphorylation in leukemia cells. Leukemia 24:177-86
Wang, Haichao; Li, Wei; Zhu, Shu et al. (2010) Peripheral administration of fetuin-A attenuates early cerebral ischemic injury in rats. J Cereb Blood Flow Metab 30:493-504
Zhu, Shu; Li, Wei; Ward, Mary F et al. (2010) High mobility group box 1 protein as a potential drug target for infection- and injury-elicited inflammation. Inflamm Allergy Drug Targets 9:60-72
Tang, Daolin; Kang, Rui; Xiao, Weimin et al. (2009) Quercetin prevents LPS-induced high-mobility group box 1 release and proinflammatory function. Am J Respir Cell Mol Biol 41:651-60
Zhu, Shu; Ashok, Mala; Li, Jianhua et al. (2009) Spermine protects mice against lethal sepsis partly by attenuating surrogate inflammatory markers. Mol Med 15:275-82
Lv, Ben; Wang, Haichao; Tang, Yiting et al. (2009) High-mobility group box 1 protein induces tissue factor expression in vascular endothelial cells via activation of NF-kappaB and Egr-1. Thromb Haemost 102:352-9
Wang, Haichao; Ward, Mary F; Sama, Andrew E (2009) Novel HMGB1-inhibiting therapeutic agents for experimental sepsis. Shock 32:348-57
Wang, Haichao; Zhu, Shu; Zhou, Rongrong et al. (2008) Therapeutic potential of HMGB1-targeting agents in sepsis. Expert Rev Mol Med 10:e32

Showing the most recent 10 out of 21 publications