Resolvins AT-D1 and E1 differentially impact functional outcome, post-traumatic sleep, and microglial activation following diffuse brain injury in the mouse

JL Harrison, RK Rowe, TW Ellis, NS Yee… - Brain, behavior, and …, 2015 - Elsevier
JL Harrison, RK Rowe, TW Ellis, NS Yee, BF O'Hara, PD Adelson, J Lifshitz
Brain, behavior, and immunity, 2015Elsevier
Traumatic brain injury (TBI) is induced by mechanical forces which initiate a cascade of
secondary injury processes, including inflammation. Therapies which resolve the
inflammatory response may promote neural repair without exacerbating the primary injury.
Specific derivatives of omega-3 fatty acids loosely grouped as specialized pro-resolving lipid
mediators (SPMs) and termed resolvins promote the active resolution of inflammation. In the
current study, we investigate the effect of two resolvin molecules, RvE1 and AT-RvD1, on …
Abstract
Traumatic brain injury (TBI) is induced by mechanical forces which initiate a cascade of secondary injury processes, including inflammation. Therapies which resolve the inflammatory response may promote neural repair without exacerbating the primary injury. Specific derivatives of omega-3 fatty acids loosely grouped as specialized pro-resolving lipid mediators (SPMs) and termed resolvins promote the active resolution of inflammation. In the current study, we investigate the effect of two resolvin molecules, RvE1 and AT-RvD1, on post-traumatic sleep and functional outcome following diffuse TBI through modulation of the inflammatory response.
Adult, male C57BL/6 mice were injured using a midline fluid percussion injury (mFPI) model (6–10 min righting reflex time for brain-injured mice). Experimental groups included mFPI administered RvE1 (100 ng daily), AT-RvD1 (100 ng daily), or vehicle (sterile saline) and counterbalanced with uninjured sham mice. Resolvins or saline were administered daily for seven consecutive days beginning 3 days prior to TBI to evaluate proof-of-principle to improve outcome. Immediately following diffuse TBI, post-traumatic sleep was recorded for 24 h post-injury. For days 1–7 post-injury, motor outcome was assessed by rotarod. Cognitive function was measured at 6 days post-injury using novel object recognition (NOR). At 7 days post-injury, microglial activation was quantified using immunohistochemistry for Iba-1.
In the diffuse brain-injured mouse, AT-RvD1 treatment, but not RvE1, mitigated motor and cognitive deficits. RvE1 treatment significantly increased post-traumatic sleep in brain-injured mice compared to all other groups. RvE1 treated mice displayed a higher proportion of ramified microglia and lower proportion of activated rod microglia in the cortex compared to saline or AT-RvD1 treated brain-injured mice. Thus, RvE1 treatment modulated post-traumatic sleep and the inflammatory response to TBI, albeit independently of improvement in motor and cognitive outcome as seen in AT-RvD1-treated mice. This suggests AT-RvD1 may impart functional benefit through mechanisms other than resolution of inflammation alone.
Elsevier