Endocannabinoid degradation inhibition improves neurobehavioral function, blood-brain barrier integrity, and neuroinflammation following mild traumatic brain injury

J Neurotrauma. 2015 Mar 1;32(5):297-306. doi: 10.1089/neu.2014.3508. Epub 2014 Dec 19.

Abstract

Traumatic brain injury (TBI) is an increasingly frequent and poorly understood condition lacking effective therapeutic strategies. Inflammation and oxidative stress (OS) are critical components of injury, and targeted interventions to reduce their contribution to injury should improve neurobehavioral recovery and outcomes. Recent evidence reveals potential protective, yet short-lived, effects of the endocannabinoids (ECs), 2-arachidonoyl glycerol (2-AG) and N-arachidonoyl-ethanolamine (AEA), on neuroinflammatory and OS processes after TBI. The aim of this study was to determine whether EC degradation inhibition after TBI would improve neurobehavioral recovery by reducing inflammatory and oxidative damage. Adult male Sprague-Dawley rats underwent a 5-mm left lateral craniotomy, and TBI was induced by lateral fluid percussion. TBI produced apnea (17±5 sec) and a delayed righting reflex (479±21 sec). Thirty minutes post-TBI, rats were randomized to receive intraperitoneal injections of vehicle (alcohol, emulphor, and saline; 1:1:18) or a selective inhibitor of 2-AG (JZL184, 16 mg/kg) or AEA (URB597, 0.3 mg/kg) degradation. At 24 h post-TBI, animals showed significant neurological and -behavioral impairment as well as disruption of blood-brain barrier (BBB) integrity. Improved neurological and -behavioral function was observed in JZL184-treated animals. BBB integrity was protected in both JZL184- and URB597-treated animals. No significant differences in ipsilateral cortex messenger RNA expression of interleukin (IL)-1β, IL-6, chemokine (C-C motif) ligand 2, tumor necrosis factor alpha, cyclooxygenase 2 (COX2), or nicotinamide adenine dinucleotide phosphate oxidase (NOX2) and protein expression of COX2 or NOX2 were observed across experimental groups. Astrocyte and microglia activation was significantly increased post-TBI, and treatment with JZL184 or URB597 blocked activation of both cell types. These findings suggest that EC degradation inhibition post-TBI exerts neuroprotective effects. Whether repeated dosing would achieve greater protection remains to be examined.

Keywords: 2-AG; AEA; TBI; endocannabinoids; neuroinflammation.

Publication types

  • Research Support, U.S. Gov't, Non-P.H.S.

MeSH terms

  • Animals
  • Arachidonic Acids / metabolism*
  • Benzamides / pharmacology
  • Benzodioxoles / pharmacology
  • Blood-Brain Barrier / drug effects*
  • Blood-Brain Barrier / pathology
  • Blotting, Western
  • Brain Injuries / pathology*
  • Carbamates / pharmacology
  • Disease Models, Animal
  • Endocannabinoids / metabolism*
  • Glycerides / metabolism*
  • Immunohistochemistry
  • Inflammation / pathology*
  • Male
  • Neuroprotective Agents / pharmacology
  • Piperidines / pharmacology
  • Polyunsaturated Alkamides
  • Rats
  • Rats, Sprague-Dawley
  • Real-Time Polymerase Chain Reaction
  • Recovery of Function / drug effects

Substances

  • Arachidonic Acids
  • Benzamides
  • Benzodioxoles
  • Carbamates
  • Endocannabinoids
  • Glycerides
  • JZL 184
  • Neuroprotective Agents
  • Piperidines
  • Polyunsaturated Alkamides
  • cyclohexyl carbamic acid 3'-carbamoylbiphenyl-3-yl ester
  • glyceryl 2-arachidonate
  • anandamide