Abstract

BackgroundImmunomodulatory therapies have been identified as interventions for secondary injury after traumatic brain injury (TBI). The cannabinoid receptor type-2 (CB2R) is proposed to play an important, endogenous role in regulating inflammation. The effects of CB2R stimulation, blockade, and deletion on the neurovascular inflammatory responses to TBI were assessed.MethodsWild-type C57BL/6 or CB2R knockout mice were randomly assigned to controlled cortical impact (CCI) injury or to craniotomy control groups. The effects of treatment with synthetic, selective CB2R agonists (0-1966 and JWH-133), a selective CB2R antagonist, or vehicle solution administered to CCI groups were assessed at 1-day after injury. Changes in TNF-α, intracellular adhesion molecule (ICAM-1), inducible nitric oxide synthase (iNOS), macrophage/microglial ionized calcium-binding adaptor molecule, and blood-brain-barrier (BBB) permeability were assessed using ELISA, quantitative RT-PCR, immunohistochemistry, and fluorometric analysis of sodium fluorescein uptake. CB2R knockouts and wild-type mice with CCI injury were treated with a CB2R agonist or vehicle treatment.ResultsTNF-α mRNA increased at 6 hours and 1 to 3 days after CCI; a CB2R antagonist and genetic knockout of the CB2R exacerbated TNF-α mRNA expression. Treatment with a CB2R agonist attenuated TNF-α protein levels indicating post-transcriptional mechanisms. Intracellular adhesion molecule (ICAM-1) mRNA was increased at 6 hours, and at 1 to 2 days after CCI, reduced in mice treated with a CB2R agonist, and increased in CB2R knockout mice with CCI. Sodium fluorescein uptake was increased in CB2R knockouts after CCI, with and without a CB2R agonist. iNOS mRNA expression peaked early (6 hours) and remained increased from 1 to 3 days after injury. Treatment with a CB2R agonist attenuated increases in iNOS mRNA expression, while genetic deletion of the CB2R resulted in substantial increases in iNOS expression. Double label immunohistochemistry confirmed that iNOS was expressed by macrophage/microglia in the injured cortex.ConclusionFindings demonstrate that the endogenous cannabinoid system and CB2R play an important role in regulating inflammation and neurovascular responses in the traumatically injured brain. CB2R stimulation with two agonists (0-1966 and JWH-133) dampened post-traumatic inflammation, while blockade or deletion of the CB2R worsened inflammation. Findings support previous evidence that modulating the CB2R alters infiltrating macrophages and activated resident microglia. Further investigation into the role of the CB2R on specific immune cell populations in the injured brain is warranted.

Highlights

  • Traumatic brain injury (TBI) affects over 1.4 million Americans annually, with many suffering fatal or permanently disabling injuries [1,2]

  • There were three study arms to determine the effects of cannabinoid receptor type-2 (CB2R) modulation on genes and proteins expression for primary vascular inflammatory markers (TNF-α, ICAM, inducible nitric oxide synthase (iNOS), and BBB permeability) which included: (1) cortical impact (CCI) injury over time compared to craniotomy, (2) CB2R agonists and CB2R antagonist compared to vehicle-treated mice, and (3) CB2R knockout (CB2R KO) CCI groups with and without JWH133 compared to wild-type CCI (Figure 1)

  • tumor necrosis factor-alpha (TNF-α) The expression of mRNA specific for the pro-inflammatory cytokine TNF-α was significantly increased by comparison with controls at each time points examined after CCI in wild-type mice including 6 hours (P < 0.05), 1 day (P < 0.001), 2 and 3 days (P < 0.001) (Figure 2A and 2C)

Read more

Summary

Introduction

Traumatic brain injury (TBI) affects over 1.4 million Americans annually, with many suffering fatal or permanently disabling injuries [1,2]. Blood-brain-barrier (BBB) disruption, a result of the post-traumatic inflammatory response, is a proposed mechanism of secondary injury and contributes to cell death or dysfunction, worsening neurologic function, and to poorer clinical outcome [3,4]. It is well-recognized, that this same inflammatory response plays an important role in the processes necessary for repair and recovery [5]. Pro-inflammatory cell phenotypes release cytokines and express enzymes such as inducible nitric oxide synthase (iNOS) that generate damaging free radicals and further disrupt BBB function. The effects of CB2R stimulation, blockade, and deletion on the neurovascular inflammatory responses to TBI were assessed

Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call