Necrosis is a major contributor to human disease, however little effort has been made to develop specific therapies targeting pathologic necrosis due it its perceived uncontrolled nature. This notion has recently been challenged by the discovery that intrinsic death pathways down stream of tumor necrosis factor receptor family members can initiate necrotic death, suggesting that necrosis can be specifically targeted for inhibition. We have recently developed necrostatins, potent and selective small molecule inhibitors of """"""""necroptosis"""""""", a form of programmed necrosis initiated by TNF alpha and Fas receptor. Necrostatin-1 was used to directly establish necroptosis as a major component of neuronal cell death after ischemic brain injury in vivo. Traumatic brain injury is a leading cause of death and life-long disability in young adults and children, however mechanisms of traumatic brain cell death and associated neurological dysfunction are not well characterized. We have shown that TNF and Fas play a key role in histopathological and functional outcome after controlled cortical impact (CCI) in mice. Since TNF and Fas are well established inducers of necroptosis, we investigated whether necroptosis contributes to outcome after TBI. We found that necrostatin-1 reduced tissue damage and markedly improved functional outcome following CCI in mice. These results suggest that the target of necrostatin-1, receptor interacting protein kinase 1 (RIPK1), may represent an exciting new target for TBI therapy. To further investigate the role of RIPK1 in TBI, we propose three Specific Aims: 1) Establish the role of RIPK1 in neuronal cell death in a variety of in vitro neuronal death paradigms related to TBI using knockdown (RIPK1 shRNA) and dominant negative (RIPK1 K45M) strategies;2) Demonstrate a key contribution of RIPK1 to brain trauma in vivo, using AAV-8 viral vector transduction of neurons in vivo with shRNA RIPK1 or RIPK1 K45M constructs;and 3) Determine RIPK1-related signaling pathways operative during necroptosis in cultured neurons and in injured brain after TBI in vivo.

Public Health Relevance

Work in the proposed project could render a new treatment for patients with traumatic brain injury. The proposed drug therapy might reduce brain cell death as well as improve learning and memory in survivors, and thereby improve their quality of life.

Agency
National Institute of Health (NIH)
Institute
National Institute of Neurological Disorders and Stroke (NINDS)
Type
Research Project (R01)
Project #
5R01NS064545-05
Application #
8451465
Study Section
Brain Injury and Neurovascular Pathologies Study Section (BINP)
Program Officer
Hicks, Ramona R
Project Start
2009-05-15
Project End
2014-04-30
Budget Start
2013-05-01
Budget End
2014-04-30
Support Year
5
Fiscal Year
2013
Total Cost
$399,149
Indirect Cost
$125,044
Name
Massachusetts General Hospital
Department
Type
DUNS #
073130411
City
Boston
State
MA
Country
United States
Zip Code
02199
Whalen, Michael J (2014) Cerebrovascular autoregulation in diabetic ketoacidosis: time to go with the (microvascular cerebral blood) flow! Pediatr Crit Care Med 15:779-80
Dapul, Heda R; Park, Juyeon; Zhang, Jimmy et al. (2013) Concussive injury before or after controlled cortical impact exacerbates histopathology and functional outcome in a mixed traumatic brain injury model in mice. J Neurotrauma 30:382-91
Mbye, Lamin H; Keles, Eyup; Tao, Luyang et al. (2012) Kollidon VA64, a membrane-resealing agent, reduces histopathology and improves functional outcome after controlled cortical impact in mice. J Cereb Blood Flow Metab 32:515-24
Zhu, Xiaoxia; Tao, Luyang; Tejima-Mandeville, Emiri et al. (2012) Plasmalemma permeability and necrotic cell death phenotypes after intracerebral hemorrhage in mice. Stroke 43:524-31
Park, Juyeon; Zhang, Jimmy; Qiu, Jianhua et al. (2012) Combination therapy targeting Akt and mammalian target of rapamycin improves functional outcome after controlled cortical impact in mice. J Cereb Blood Flow Metab 32:330-40
Khuman, Jugta; Meehan 3rd, William P; Zhu, Xiaoxia et al. (2011) Tumor necrosis factor alpha and Fas receptor contribute to cognitive deficits independent of cell death after concussive traumatic brain injury in mice. J Cereb Blood Flow Metab 31:778-89
Krajewska, Maryla; You, Zerong; Rong, Juan et al. (2011) Neuronal deletion of caspase 8 protects against brain injury in mouse models of controlled cortical impact and kainic acid-induced excitotoxicity. PLoS One 6:e24341
Liu, Christina H; You, Zerong; Liu, Charng-Ming et al. (2009) Diffusion-weighted magnetic resonance imaging reversal by gene knockdown of matrix metalloproteinase-9 activities in live animal brains. J Neurosci 29:3508-17