Each year in the United States ~600,000 people suffer from cardiac arrest (CA) and receive cardiopulmonary resuscitation (CPR), resulting in hypoxia-ischemia (HI) of the brain and consequently in severe neurological deficits in most of the survivors. No pharmacological treatment is available to improve survival or long-term neurological outcome. Ischemic damage in the brain following cerebral ischemia induced by CA is in large part due to glutamate excitotoxicity triggered by a pathological flood calcium through NMDA-type glutamate receptors (NMDAr), ultimately resulting in neuronal cell death. While the pathology and cascade of events leading to injury following cerebral ischemia is complex, overstimulation of NMDAr is considered the major triggering spark for excitotoxicity and ischemic neuronal damage. Excitotoxic stimulation of NMDAr triggers a series of Ca2+-dependent signaling pathways, including activation of Ca2+/calmodulin-dependent protein kinase II (CaMKII). Our recent findings, and preliminary data, demonstrate that inhibition of CaMKII is a novel approach to minimizing downstream effects of excessive glutamatergic stimulation (excitotoxicity). CaMKII is well established as a major mediator of physiological glutamate signaling involved in synaptic plasticity, particularly synaptic potentiation in CA1 neurons, likel contributing to hippocampal learning. Two major forms of CaMKII regulation have been described, stimulated and autonomous CaMKII activity. Stimulated activity is induced by Ca2+/CaM to CaMKII, and prolonged autonomous (Ca2+-independent) activity is induced by autophosphorylation of residue T286 and/or direct binding to NMDAr subunit NR2B at the synapse (which also mediates CaMKII accumulation at the synapse). Additionally, our preliminary studies reveal a novel link between CaMKII autonomy and nitric oxide (NO), which is produced during excitotoxicity following cerebral ischemia and contributes to oxidative stress and neuronal damage. Our preliminary data indicate that NO-induced nitrosylation/oxidation of CaMKII promotes autonomous CaMKII activity, directly and by protecting T286 from de-phosphorylation. We recently demonstrated that our new CaMKII inhibitor tatCN21 (which blocks both stimulated and autonomous activity) provides robust neuroprotection both in vitro and following experimental stroke. In contrast, traditional CaMKII inhibitors (which block only stimulated activity) do not provide post-insult neuroprotection. This indicates that inhibition of autonomous, but not stimulated CaMKII activity, is a relevant drug target for post-insult neuroprotection. [Importantly, our preliminary results demonstrate that administration of tatCN21 after cardiac arrest results in significant neuroprotection.] The current proposal will utilize our novel mouse CA/CPR model to take advantage of several mutant mouse strains deficient in each form of autonomous CaMKII activity to unravel the complex interactions between these forms of CaMKII activity and their relative contribution to ischemic neuronal cell death. We hypothesize that (i) each autonomy mechanism contributes to neuronal cell death, and that (ii) T286-autophosphorylation mediated CaMKII autonomy is of most direct importance, that (iii) the novel nitrosylation mechanisms contributes to ischemic damage by prolonging T286 phosphorylation, and that (iv) NR2B-binding enables efficient nitrosylation via localizing CaMKII near nNOS.

Public Health Relevance

Heart attack and consequent cerebral ischemia is one of the leading causes of death and disability in the United States. Unfortunately, there are currently no drugs available to improve outcome and quality of life following severe heart attack. The proposed studies will demonstrate for the first time that autonomous CaMKII activity contributes to ischemia-induced neuronal cell death and suggest a novel interventional strategy to protect brain following heart attack.

Agency
National Institute of Health (NIH)
Institute
National Institute of Neurological Disorders and Stroke (NINDS)
Type
Research Project (R01)
Project #
5R01NS080851-02
Application #
8653999
Study Section
Brain Injury and Neurovascular Pathologies Study Section (BINP)
Program Officer
Koenig, James I
Project Start
2013-05-01
Project End
2018-04-30
Budget Start
2014-05-01
Budget End
2015-04-30
Support Year
2
Fiscal Year
2014
Total Cost
Indirect Cost
Name
University of Colorado Denver
Department
Anesthesiology
Type
Schools of Medicine
DUNS #
City
Aurora
State
CO
Country
United States
Zip Code
80045
Opazo, Patricio; Viana da Silva, Silvia; Carta, Mario et al. (2018) CaMKII Metaplasticity Drives A? Oligomer-Mediated Synaptotoxicity. Cell Rep 23:3137-3145
Woolfrey, Kevin M; O'Leary, Heather; Goodell, Dayton J et al. (2018) CaMKII regulates the depalmitoylation and synaptic removal of the scaffold protein AKAP79/150 to mediate structural long-term depression. J Biol Chem 293:1551-1567
Orfila, James E; McKinnon, Nicole; Moreno, Myriam et al. (2018) Cardiac Arrest Induces Ischemic Long-Term Potentiation of Hippocampal CA1 Neurons That Occludes Physiological Long-Term Potentiation. Neural Plast 2018:9275239
Cook, Sarah G; Bourke, Ashley M; O'Leary, Heather et al. (2018) Analysis of the CaMKII? and ? splice-variant distribution among brain regions reveals isoform-specific differences in holoenzyme formation. Sci Rep 8:5448
Goodell, Dayton J; Zaegel, Vincent; Coultrap, Steven J et al. (2017) DAPK1 Mediates LTD by Making CaMKII/GluN2B Binding LTP Specific. Cell Rep 19:2231-2243
Deng, Guiying; Orfila, James E; Dietz, Robert M et al. (2017) Autonomous CaMKII Activity as a Drug Target for Histological and Functional Neuroprotection after Resuscitation from Cardiac Arrest. Cell Rep 18:1109-1117
Quillinan, Nidia; Herson, Paco S; Traystman, Richard J (2016) Neuropathophysiology of Brain Injury. Anesthesiol Clin 34:453-64
Kelly, Kathleen K; MacPherson, Amber M; Grewal, Himmat et al. (2016) Col1a1+ perivascular cells in the brain are a source of retinoic acid following stroke. BMC Neurosci 17:49
Shimizu, Kaori; Quillinan, Nidia; Orfila, James E et al. (2016) Sirtuin-2 mediates male specific neuronal injury following experimental cardiac arrest through activation of TRPM2 ion channels. Exp Neurol 275 Pt 1:78-83
Barcomb, Kelsey; Hell, Johannes W; Benke, Tim A et al. (2016) The CaMKII/GluN2B Protein Interaction Maintains Synaptic Strength. J Biol Chem 291:16082-9

Showing the most recent 10 out of 25 publications