The primary objective of the UC Davis CounterACT Center of Excellence is to identify and advance improved medical countermeasures for rapidly terminating seizures and mitigating the delayed neurologic consequences following acute intoxication with convulsant chemical threat agents. The Center comprises three research projects: Project 1 discovers therapeutic candidates via in vitro mechanistic screens, which are tested for in vivo antiseizure and neuroprotective efficacy by Projects 2 and 3, respectively. The projects rely on three scientific cores to support drug analysis and biomarker detection (Core A), medicinal chemistry and pharmacological testing (Core B), and experimental design and data analysis (Core C). A Research Education Core supports training in countermeasure research, and an Administrative Core oversees and coordinates scientific and administrative operations. The Center focuses on the GABAA receptor antagonist tetramethylenedisulfotetramine (TETS) and the organophosphate cholinesterase inhibitor diisopropylfluorophosphate (DFP), which can trigger convulsions that progress to life threatening status epilepticus (SE). Survivors face significant, long-term morbidity, including mild-to-severe memory loss, affective disorders and recurrent seizures. Current medical countermeasures can reduce mortality in exposed individuals, but they do so with significant side effects and are maximally effective only if administered within minutes of exposure. These limitations underscore the urgent need for improved medical countermeasures. In the first project period, we developed innovative in vitro platforms for mechanism-based screening to identify candidate antiseizure and neuroprotective therapeutics, and novel preclinical models that recapitulate acute seizure activity and neurological deficits observed in humans following acute intoxication with TETS or OPs. Using these models, we discovered: (1) allopregnanolone, a GABAA receptor positive allosteric modulator, was a superior countermeasure for TETS-induced SE, and (2) combining standard-of-care with allopregnanolone and a low dose of perampanel, a potent AMPA receptor antagonist, was more effective than standard-of-care alone in terminating DFP-induced SE. We also discovered that neuropathology was mitigated by post-exposure treatment with dantrolene, a Ca2+ channel stabilizer, or a novel small molecule dual inhibitor of soluble epoxide hydrolase (sEH) and cyclooxygenase-2 (COX-2). Our goals in this second project period are to: (1) advance our antiseizure lead allopregnanolone; (2) continue development of allopregnanolone and perampanel; (3) identify adjunct neuroprotective leads, focusing initially on the dual sEH-COX-2 inhibitor and dantrolene; and (4) conduct mechanistic studies to discover new therapeutic candidates. Our milestones for the second project period are to: (i) produce data and a regulatory strategy to advance allopregnanolone for treatment of GABAAR antagonist-induced seizures; (ii) determine whether combination treatment with allopregnanolone and perampanel warrants development as a lead ?universal antidote?; and (iii) identify lead neuroprotective treatments for improving long-term outcomes.

Public Health Relevance

Current medical countermeasures for acute exposures to seizure-inducing chemical threat agents can prevent death but do not sufficiently protect the brain from chronic injury, such as chemical-induced epilepsy or cognitive deficits. The goal of the UC Davis CounterACT Center of Excellence is to identify and advance improved medical treatments to stop seizures and to mitigate their neurological impact in order to protect the general public during a chemical emergency.

Agency
National Institute of Health (NIH)
Institute
National Institute of Neurological Disorders and Stroke (NINDS)
Type
Specialized Center--Cooperative Agreements (U54)
Project #
5U54NS079202-08
Application #
9744834
Study Section
Special Emphasis Panel (ZRG1)
Program Officer
Jett, David A
Project Start
2012-09-01
Project End
2022-05-31
Budget Start
2019-06-01
Budget End
2020-05-31
Support Year
8
Fiscal Year
2019
Total Cost
Indirect Cost
Name
University of California Davis
Department
Veterinary Sciences
Type
Schools of Veterinary Medicine
DUNS #
047120084
City
Davis
State
CA
Country
United States
Zip Code
95618
Tu, Ranran; Armstrong, Jillian; Lee, Kin Sing Stephen et al. (2018) Soluble epoxide hydrolase inhibition decreases reperfusion injury after focal cerebral ischemia. Sci Rep 8:5279
Hampe, Alexander E; Li, Zidong; Sethi, Sunjay et al. (2018) A Microfluidic Platform to Study Astrocyte Adhesion on Nanoporous Gold Thin Films. Nanomaterials (Basel) 8:
Hobson, Brad A; Rowland, Douglas J; Supasai, Suangsuda et al. (2018) A magnetic resonance imaging study of early brain injury in a rat model of acute DFP intoxication. Neurotoxicology 66:170-178
Moeller, Benjamin; Espelien, Brenna; Weber, Waylon et al. (2018) The pharmacokinetics of ketamine following intramuscular injection to F344 rats. Drug Test Anal :
Pressly, Brandon; Nguyen, Hai M; Wulff, Heike (2018) GABAA receptor subtype selectivity of the proconvulsant rodenticide TETS. Arch Toxicol 92:833-844
Dhir, Ashish; Rogawski, Michael A (2018) Determination of minimal steady-state plasma level of diazepam causing seizure threshold elevation in rats. Epilepsia 59:935-944
Hobson, Brad A; Sisó, Sílvia; Rowland, Douglas J et al. (2017) From the Cover: MagneticResonance Imaging Reveals Progressive Brain Injury in Rats Acutely Intoxicated With Diisopropylfluorophosphate. Toxicol Sci 157:342-353
Brown, Brandon M; Shim, Heesung; Zhang, Miao et al. (2017) Structural Determinants for the Selectivity of the Positive KCa3.1 Gating Modulator 5-Methylnaphtho[2,1-d]oxazol-2-amine (SKA-121). Mol Pharmacol 92:469-480
Vasylieva, Natalia; Barnych, Bogdan; Rand, Amy et al. (2017) Sensitive Immunoassay for Detection and Quantification of the Neurotoxin, Tetramethylenedisulfotetramine. Anal Chem 89:5612-5619
Nguyen, Hai M; Singh, Vikrant; Pressly, Brandon et al. (2017) Structural Insights into the Atomistic Mechanisms of Action of Small Molecule Inhibitors Targeting the KCa3.1 Channel Pore. Mol Pharmacol 91:392-402

Showing the most recent 10 out of 85 publications