GPR110 ligands reduce chronic optic tract gliosis and visual deficit following repetitive mild traumatic brain injury in mice

Abstract Background Repetitive mild traumatic brain injury (mTBI) can result in chronic visual dysfunction. G-protein receptor 110 (GPR110, ADGRF1) is the target receptor of N-docosahexaenoylethanolamine (synaptamide) mediating the anti-neuroinflammatory function of synaptamide. In this study, we ev...

Full description

Bibliographic Details
Main Authors: Huazhen Chen, Karl Kevala, Elma Aflaki, Juan Marugan, Hee-Yong Kim
Format: Article
Language:English
Published: BMC 2021-07-01
Series:Journal of Neuroinflammation
Subjects:
Online Access:https://doi.org/10.1186/s12974-021-02195-y
id doaj-18e3ce6773844654a15a2b3071f2fe61
record_format Article
spelling doaj-18e3ce6773844654a15a2b3071f2fe612021-07-18T11:47:10ZengBMCJournal of Neuroinflammation1742-20942021-07-0118111310.1186/s12974-021-02195-yGPR110 ligands reduce chronic optic tract gliosis and visual deficit following repetitive mild traumatic brain injury in miceHuazhen Chen0Karl Kevala1Elma Aflaki2Juan Marugan3Hee-Yong Kim4Laboratory of Molecular Signaling, NIAAA, NIHLaboratory of Molecular Signaling, NIAAA, NIHLaboratory of Molecular Signaling, NIAAA, NIHDivision of Pre-Clinical Innovation, NCATS, NIHLaboratory of Molecular Signaling, NIAAA, NIHAbstract Background Repetitive mild traumatic brain injury (mTBI) can result in chronic visual dysfunction. G-protein receptor 110 (GPR110, ADGRF1) is the target receptor of N-docosahexaenoylethanolamine (synaptamide) mediating the anti-neuroinflammatory function of synaptamide. In this study, we evaluated the effect of an endogenous and a synthetic ligand of GPR110, synaptamide and (4Z,7Z,10Z,13Z,16Z,19Z)-N-(2-hydroxy-2-methylpropyl) docosa-4,7,10,13,16,19-hexaenamide (dimethylsynaptamide, A8), on the mTBI-induced long-term optic tract histopathology and visual dysfunction using Closed-Head Impact Model of Engineered Rotational Acceleration (CHIMERA), a clinically relevant model of mTBI. Methods The brain injury in wild-type (WT) and GPR110 knockout (KO) mice was induced by CHIMERA applied daily for 3 days, and GPR110 ligands were intraperitoneally injected immediately following each impact. The expression of GPR110 and proinflammatory mediator tumor necrosis factor (TNF) in the brain was measured by using real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR) in an acute phase. Chronic inflammatory responses in the optic tract and visual dysfunction were assessed by immunostaining for Iba-1 and GFAP and visual evoked potential (VEP), respectively. The effect of GPR110 ligands in vitro was evaluated by the cyclic adenosine monophosphate (cAMP) production in primary microglia isolated from adult WT or KO mouse brains. Results CHIMERA injury acutely upregulated the GPR110 and TNF gene level in mouse brain. Repetitive CHIMERA (rCHIMERA) increased the GFAP and Iba-1 immunostaining of glia cells and silver staining of degenerating axons in the optic tract with significant reduction of N1 amplitude of visual evoked potential at up to 3.5 months after injury. Both GPR110 ligands dose- and GPR110-dependently increased cAMP in cultured primary microglia with A8, a ligand with improved stability, being more effective than synaptamide. Intraperitoneal injection of A8 at 1 mg/kg or synaptamide at 5 mg/kg significantly reduced the acute expression of TNF mRNA in the brain and ameliorated chronic optic tract microgliosis, astrogliosis, and axonal degeneration as well as visual deficit caused by injury in WT but not in GPR110 KO mice. Conclusion Our data demonstrate that ligand-induced activation of the GPR110/cAMP system upregulated after injury ameliorates the long-term optic tract histopathology and visual impairment caused by rCHIMERA. Based on the anti-inflammatory nature of GPR110 activation, we suggest that GPR110 ligands may have therapeutic potential for chronic visual dysfunction associated with mTBI.https://doi.org/10.1186/s12974-021-02195-ySynaptamideA8 (4Z,7Z,10Z,13Z,16Z,19Z)-N-(2-hydroxy-2-methylpropyl) docosa-4,7,10,13,16,19-hexaenamide (dimethylsynaptamide)repetitive CHIMERAGPR110HistopathologyOptic tract
collection DOAJ
language English
format Article
sources DOAJ
author Huazhen Chen
Karl Kevala
Elma Aflaki
Juan Marugan
Hee-Yong Kim
spellingShingle Huazhen Chen
Karl Kevala
Elma Aflaki
Juan Marugan
Hee-Yong Kim
GPR110 ligands reduce chronic optic tract gliosis and visual deficit following repetitive mild traumatic brain injury in mice
Journal of Neuroinflammation
Synaptamide
A8 (4Z,7Z,10Z,13Z,16Z,19Z)-N-(2-hydroxy-2-methylpropyl) docosa-4,7,10,13,16,19-hexaenamide (dimethylsynaptamide)
repetitive CHIMERA
GPR110
Histopathology
Optic tract
author_facet Huazhen Chen
Karl Kevala
Elma Aflaki
Juan Marugan
Hee-Yong Kim
author_sort Huazhen Chen
title GPR110 ligands reduce chronic optic tract gliosis and visual deficit following repetitive mild traumatic brain injury in mice
title_short GPR110 ligands reduce chronic optic tract gliosis and visual deficit following repetitive mild traumatic brain injury in mice
title_full GPR110 ligands reduce chronic optic tract gliosis and visual deficit following repetitive mild traumatic brain injury in mice
title_fullStr GPR110 ligands reduce chronic optic tract gliosis and visual deficit following repetitive mild traumatic brain injury in mice
title_full_unstemmed GPR110 ligands reduce chronic optic tract gliosis and visual deficit following repetitive mild traumatic brain injury in mice
title_sort gpr110 ligands reduce chronic optic tract gliosis and visual deficit following repetitive mild traumatic brain injury in mice
publisher BMC
series Journal of Neuroinflammation
issn 1742-2094
publishDate 2021-07-01
description Abstract Background Repetitive mild traumatic brain injury (mTBI) can result in chronic visual dysfunction. G-protein receptor 110 (GPR110, ADGRF1) is the target receptor of N-docosahexaenoylethanolamine (synaptamide) mediating the anti-neuroinflammatory function of synaptamide. In this study, we evaluated the effect of an endogenous and a synthetic ligand of GPR110, synaptamide and (4Z,7Z,10Z,13Z,16Z,19Z)-N-(2-hydroxy-2-methylpropyl) docosa-4,7,10,13,16,19-hexaenamide (dimethylsynaptamide, A8), on the mTBI-induced long-term optic tract histopathology and visual dysfunction using Closed-Head Impact Model of Engineered Rotational Acceleration (CHIMERA), a clinically relevant model of mTBI. Methods The brain injury in wild-type (WT) and GPR110 knockout (KO) mice was induced by CHIMERA applied daily for 3 days, and GPR110 ligands were intraperitoneally injected immediately following each impact. The expression of GPR110 and proinflammatory mediator tumor necrosis factor (TNF) in the brain was measured by using real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR) in an acute phase. Chronic inflammatory responses in the optic tract and visual dysfunction were assessed by immunostaining for Iba-1 and GFAP and visual evoked potential (VEP), respectively. The effect of GPR110 ligands in vitro was evaluated by the cyclic adenosine monophosphate (cAMP) production in primary microglia isolated from adult WT or KO mouse brains. Results CHIMERA injury acutely upregulated the GPR110 and TNF gene level in mouse brain. Repetitive CHIMERA (rCHIMERA) increased the GFAP and Iba-1 immunostaining of glia cells and silver staining of degenerating axons in the optic tract with significant reduction of N1 amplitude of visual evoked potential at up to 3.5 months after injury. Both GPR110 ligands dose- and GPR110-dependently increased cAMP in cultured primary microglia with A8, a ligand with improved stability, being more effective than synaptamide. Intraperitoneal injection of A8 at 1 mg/kg or synaptamide at 5 mg/kg significantly reduced the acute expression of TNF mRNA in the brain and ameliorated chronic optic tract microgliosis, astrogliosis, and axonal degeneration as well as visual deficit caused by injury in WT but not in GPR110 KO mice. Conclusion Our data demonstrate that ligand-induced activation of the GPR110/cAMP system upregulated after injury ameliorates the long-term optic tract histopathology and visual impairment caused by rCHIMERA. Based on the anti-inflammatory nature of GPR110 activation, we suggest that GPR110 ligands may have therapeutic potential for chronic visual dysfunction associated with mTBI.
topic Synaptamide
A8 (4Z,7Z,10Z,13Z,16Z,19Z)-N-(2-hydroxy-2-methylpropyl) docosa-4,7,10,13,16,19-hexaenamide (dimethylsynaptamide)
repetitive CHIMERA
GPR110
Histopathology
Optic tract
url https://doi.org/10.1186/s12974-021-02195-y
work_keys_str_mv AT huazhenchen gpr110ligandsreducechronicoptictractgliosisandvisualdeficitfollowingrepetitivemildtraumaticbraininjuryinmice
AT karlkevala gpr110ligandsreducechronicoptictractgliosisandvisualdeficitfollowingrepetitivemildtraumaticbraininjuryinmice
AT elmaaflaki gpr110ligandsreducechronicoptictractgliosisandvisualdeficitfollowingrepetitivemildtraumaticbraininjuryinmice
AT juanmarugan gpr110ligandsreducechronicoptictractgliosisandvisualdeficitfollowingrepetitivemildtraumaticbraininjuryinmice
AT heeyongkim gpr110ligandsreducechronicoptictractgliosisandvisualdeficitfollowingrepetitivemildtraumaticbraininjuryinmice
_version_ 1721295748449959936