Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2018

Open Access 01-12-2018 | Research

Omega-3 polyunsaturated fatty acid attenuates the inflammatory response by modulating microglia polarization through SIRT1-mediated deacetylation of the HMGB1/NF-κB pathway following experimental traumatic brain injury

Authors: Xiangrong Chen, Chunnuan Chen, Sining Fan, Shukai Wu, Fuxing Yang, Zhongning Fang, Huangde Fu, Yasong Li

Published in: Journal of Neuroinflammation | Issue 1/2018

Login to get access

Abstract

Background

Microglial polarization and the subsequent neuroinflammatory response are contributing factors for traumatic brain injury (TBI)-induced secondary injury. High mobile group box 1 (HMGB1) mediates the activation of the NF-κB pathway, and it is considered to be pivotal in the late neuroinflammatory response. Activation of the HMGB1/NF-κB pathway is closely related to HMGB1 acetylation, which is regulated by the sirtuin (SIRT) family of proteins. Omega-3 polyunsaturated fatty acids (ω-3 PUFA) are known to have antioxidative and anti-inflammatory effects. We previously demonstrated that ω-3 PUFA inhibited TBI-induced microglial activation and the subsequent neuroinflammatory response by regulating the HMGB1/NF-κB signaling pathway. However, no studies have elucidated if ω-3 PUFA affects the HMGB1/NF-κB pathway in a HMGB1 deacetylation of dependent SIRT1 manner, thus regulating microglial polarization and the subsequent neuroinflammatory response.

Methods

The Feeney DM TBI model was adopted to induce brain injury in rats. Modified neurological severity scores, rotarod test, brain water content, and Nissl staining were employed to determine the neuroprotective effects of ω-3 PUFA supplementation. Assessment of microglia polarization and pro-inflammatory markers, such as tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, and HMGB1, were used to evaluate the neuroinflammatory responses and the anti-inflammatory effects of ω-3 PUFA supplementation. Immunofluorescent staining and western blot analysis were used to detect HMGB1 nuclear translocation, secretion, and HMGB1/NF-κB signaling pathway activation to evaluate the effects of ω-3 PUFA supplementation. The impact of SIRT1 deacetylase activity on HMGB1 acetylation and the interaction between HMGB1 and SIRT1 were assessed to evaluate anti-inflammation effects of ω-3 PUFAs, and also, whether these effects were dependent on a SIRT1-HMGB1/NF-κB axis to gain further insight into the mechanisms underlying the development of the neuroinflammatory response after TBI.

Results

The results of our study showed that ω-3 PUFA supplementation promoted a shift from the M1 microglial phenotype to the M2 microglial phenotype and inhibited microglial activation, thus reducing TBI-induced inflammatory factors. In addition, ω-3 PUFA-mediated downregulation of HMGB1 acetylation and its extracellular secretion was found to be likely due to increased SIRT1 activity. We also found that treatment with ω-3 PUFA inhibited HMGB1 acetylation and induced direct interactions between SIRT1 and HMGB1 by elevating SIRT1 activity following TBI. These events lead to inhibition of HMGB1 nucleocytoplasmic translocation/extracellular secretion and alleviated HMGB1-mediated activation of the NF-κB pathway following TBI-induced microglial activation, thus inhibiting the subsequent inflammatory response.

Conclusions

The results of this study suggest that ω-3 PUFA supplementation attenuates the inflammatory response by modulating microglial polarization through SIRT1-mediated deacetylation of the HMGB1/NF-κB pathway, leading to neuroprotective effects following experimental traumatic brain injury.
Literature
1.
go back to reference Xu H, Wang Z, Li J, Wu H, Peng Y, Fan L, Chen J, Gu C, Yan F, Wang L, et al. The polarization states of microglia in TBI: a new paradigm for pharmacological intervention. Neural Plast. 2017;2017:5405104.PubMedPubMedCentral Xu H, Wang Z, Li J, Wu H, Peng Y, Fan L, Chen J, Gu C, Yan F, Wang L, et al. The polarization states of microglia in TBI: a new paradigm for pharmacological intervention. Neural Plast. 2017;2017:5405104.PubMedPubMedCentral
2.
go back to reference Kumar A, Alvarez-Croda DM, Stoica BA, Faden AI, Loane DJ. Microglial/macrophage polarization dynamics following traumatic brain injury. J Neurotrauma. 2016;33:1732–50.CrossRefPubMedPubMedCentral Kumar A, Alvarez-Croda DM, Stoica BA, Faden AI, Loane DJ. Microglial/macrophage polarization dynamics following traumatic brain injury. J Neurotrauma. 2016;33:1732–50.CrossRefPubMedPubMedCentral
4.
go back to reference Chen X, Wu S, Chen C, Xie B, Fang Z, Hu W, Chen J, Fu H, He H. Omega-3 polyunsaturated fatty acid supplementation attenuates microglial-induced inflammation by inhibiting the HMGB1/TLR4/NF-κB pathway following experimental traumatic brain injury. J Neuroinflammation. 2017;14:143–55.CrossRefPubMedPubMedCentral Chen X, Wu S, Chen C, Xie B, Fang Z, Hu W, Chen J, Fu H, He H. Omega-3 polyunsaturated fatty acid supplementation attenuates microglial-induced inflammation by inhibiting the HMGB1/TLR4/NF-κB pathway following experimental traumatic brain injury. J Neuroinflammation. 2017;14:143–55.CrossRefPubMedPubMedCentral
5.
go back to reference Ma Y, Matsuwaki T, Yamanouchi K, Nishihara M. Progranulin protects hippocampal neurogenesis via suppression of neuroinflammatory responses under acute immune stress. Mol Neurobiol. 2017;54:3717–28.CrossRefPubMed Ma Y, Matsuwaki T, Yamanouchi K, Nishihara M. Progranulin protects hippocampal neurogenesis via suppression of neuroinflammatory responses under acute immune stress. Mol Neurobiol. 2017;54:3717–28.CrossRefPubMed
6.
go back to reference Wang G, Shi Y, Jiang X, Leak RK, Hu X, Wu Y, Pu H, Li W, Tang B, Wang Y, et al. HDAC inhibition prevents white matter injury by modulating microglia/macrophage polarization through the GSK3β/PTEN/Akt axis. Proc Natl Acad Sci. 2015;112:2853–8.CrossRefPubMedPubMedCentral Wang G, Shi Y, Jiang X, Leak RK, Hu X, Wu Y, Pu H, Li W, Tang B, Wang Y, et al. HDAC inhibition prevents white matter injury by modulating microglia/macrophage polarization through the GSK3β/PTEN/Akt axis. Proc Natl Acad Sci. 2015;112:2853–8.CrossRefPubMedPubMedCentral
7.
go back to reference Yao X, Liu S, Ding W, Yue P, Jiang Q, Zhao M, Hu F, Zhang H. TLR4 signal ablation attenuated neurological deficits by regulating microglial M1/M2 phenotype after traumatic brain injury in mice. J Neuroimmunol. 2017;310:38–45.CrossRefPubMed Yao X, Liu S, Ding W, Yue P, Jiang Q, Zhao M, Hu F, Zhang H. TLR4 signal ablation attenuated neurological deficits by regulating microglial M1/M2 phenotype after traumatic brain injury in mice. J Neuroimmunol. 2017;310:38–45.CrossRefPubMed
8.
go back to reference Aryanpour R, Pasbakhsh P, Zibara K, Namjoo Z, Beigi BF, Shahbeigi S, Kashani IR, Beyer C, Zendehdel A. Progesterone therapy induces an M1 to M2 switch in microglia phenotype and suppresses NLRP3 inflammasome in a cuprizone-induced demyelination mouse model. Int Immunopharmacol. 2017;51:131–9.CrossRefPubMed Aryanpour R, Pasbakhsh P, Zibara K, Namjoo Z, Beigi BF, Shahbeigi S, Kashani IR, Beyer C, Zendehdel A. Progesterone therapy induces an M1 to M2 switch in microglia phenotype and suppresses NLRP3 inflammasome in a cuprizone-induced demyelination mouse model. Int Immunopharmacol. 2017;51:131–9.CrossRefPubMed
9.
go back to reference Kumar A, Stoica BA, Loane DJ, Yang M, Abulwerdi G, Khan N, Kumar A, Thom SR, Faden AI. Microglial-derived microparticles mediate neuroinflammation after traumatic brain injury. J Neuroinflammation. 2017;14:47.CrossRefPubMedPubMedCentral Kumar A, Stoica BA, Loane DJ, Yang M, Abulwerdi G, Khan N, Kumar A, Thom SR, Faden AI. Microglial-derived microparticles mediate neuroinflammation after traumatic brain injury. J Neuroinflammation. 2017;14:47.CrossRefPubMedPubMedCentral
10.
go back to reference Braun M, Vaibhav K, Saad NM, Fatima S, Vender JR, Baban B, Hoda MN, Dhandapani KM. White matter damage after traumatic brain injury: a role for damage associated molecular patterns. Biochim Biophys Acta. 1863;2017:2614–26. Braun M, Vaibhav K, Saad NM, Fatima S, Vender JR, Baban B, Hoda MN, Dhandapani KM. White matter damage after traumatic brain injury: a role for damage associated molecular patterns. Biochim Biophys Acta. 1863;2017:2614–26.
11.
go back to reference Fang P, Schachner M, Shen YQ. HMGB1 in development and diseases of the central nervous system. Mol Neurobiol. 2012;45:499–506.CrossRefPubMed Fang P, Schachner M, Shen YQ. HMGB1 in development and diseases of the central nervous system. Mol Neurobiol. 2012;45:499–506.CrossRefPubMed
12.
go back to reference Wang D, Liu K, Wake H, Teshigawara K, Mori S, Nishibori M. Anti-high mobility group box-1 (HMGB1) antibody inhibits hemorrhage-induced brain injury and improved neurological deficits in rats. Sci Rep. 2017;7:46243.CrossRefPubMedPubMedCentral Wang D, Liu K, Wake H, Teshigawara K, Mori S, Nishibori M. Anti-high mobility group box-1 (HMGB1) antibody inhibits hemorrhage-induced brain injury and improved neurological deficits in rats. Sci Rep. 2017;7:46243.CrossRefPubMedPubMedCentral
13.
go back to reference Ding J, Cui X, Liu Q. Emerging role of HMGB1 in lung diseases: friend or foe. J Cell Mol Med. 2017;21:1046–57.CrossRefPubMed Ding J, Cui X, Liu Q. Emerging role of HMGB1 in lung diseases: friend or foe. J Cell Mol Med. 2017;21:1046–57.CrossRefPubMed
14.
go back to reference Qiu Y, Chen Y, Zeng T, Guo W, Zhou W, Yang X. High-mobility group box-B1 (HMGB1) mediates the hypoxia-induced mesenchymal transition of osteoblast cells via activating ERK/JNK signaling. Cell Biol Int. 2016;40:1152–61.CrossRefPubMed Qiu Y, Chen Y, Zeng T, Guo W, Zhou W, Yang X. High-mobility group box-B1 (HMGB1) mediates the hypoxia-induced mesenchymal transition of osteoblast cells via activating ERK/JNK signaling. Cell Biol Int. 2016;40:1152–61.CrossRefPubMed
15.
go back to reference Laird MD, Shields JS, Sukumari-Ramesh S, Kimbler DE, Fessler RD, Shakir B, Youssef P, Yanasak N, Vender JR, Dhandapani KM. High mobility group box protein-1 promotes cerebral edema after traumatic brain injury via activation of toll-like receptor 4. Glia. 2014;62:26–38.CrossRefPubMed Laird MD, Shields JS, Sukumari-Ramesh S, Kimbler DE, Fessler RD, Shakir B, Youssef P, Yanasak N, Vender JR, Dhandapani KM. High mobility group box protein-1 promotes cerebral edema after traumatic brain injury via activation of toll-like receptor 4. Glia. 2014;62:26–38.CrossRefPubMed
16.
go back to reference Wang TH, Xiong LL, Yang SF, You C, Xia QJ, Xu Y, Zhang P, Wang SF, Liu J. LPS pretreatment provides neuroprotective roles in rats with subarachnoid hemorrhage by downregulating MMP9 and Caspase3 associated with TLR4 signaling activation. Mol Neurobiol. 2017;54:7746–60.CrossRefPubMed Wang TH, Xiong LL, Yang SF, You C, Xia QJ, Xu Y, Zhang P, Wang SF, Liu J. LPS pretreatment provides neuroprotective roles in rats with subarachnoid hemorrhage by downregulating MMP9 and Caspase3 associated with TLR4 signaling activation. Mol Neurobiol. 2017;54:7746–60.CrossRefPubMed
17.
go back to reference Leus NG, Zwinderman MR, Dekker FJ. Histone deacetylase 3 (HDAC 3) as emerging drug target in NF-kappaB-mediated inflammation. Curr Opin Chem Biol. 2016;33:160–8.CrossRefPubMedPubMedCentral Leus NG, Zwinderman MR, Dekker FJ. Histone deacetylase 3 (HDAC 3) as emerging drug target in NF-kappaB-mediated inflammation. Curr Opin Chem Biol. 2016;33:160–8.CrossRefPubMedPubMedCentral
18.
go back to reference Schuliga M. NF-kappaB signaling in chronic inflammatory airway disease. Biomol Ther. 2015;5:1266–83. Schuliga M. NF-kappaB signaling in chronic inflammatory airway disease. Biomol Ther. 2015;5:1266–83.
19.
go back to reference Chi J, Seo GS, Cheon JH, Lee SH. Isoliquiritigenin inhibits TNF-α-induced release of high-mobility group box 1 through activation of HDAC in human intestinal epithelial HT-29 cells. Eur J Pharmacol. 2017;796:101–9.CrossRefPubMed Chi J, Seo GS, Cheon JH, Lee SH. Isoliquiritigenin inhibits TNF-α-induced release of high-mobility group box 1 through activation of HDAC in human intestinal epithelial HT-29 cells. Eur J Pharmacol. 2017;796:101–9.CrossRefPubMed
20.
21.
go back to reference Lan K, Chao S, Wu H, Chiang C, Wang C, Liu S, Weng T. Salidroside ameliorates sepsis-induced acute lung injury and mortality via downregulating NF-κB and HMGB1 pathways through the upregulation of SIRT1. Sci Rep-Uk. 2017;7:12026.CrossRef Lan K, Chao S, Wu H, Chiang C, Wang C, Liu S, Weng T. Salidroside ameliorates sepsis-induced acute lung injury and mortality via downregulating NF-κB and HMGB1 pathways through the upregulation of SIRT1. Sci Rep-Uk. 2017;7:12026.CrossRef
22.
go back to reference Zhang X, Wu Q, Wu L, Ye Z, Jiang T, Li W, Zhuang Z, Zhou M, Zhang X, Hang C. Sirtuin 1 activation protects against early brain injury after experimental subarachnoid hemorrhage in rats. Cell Death and Disease. 2016;7:e2416.CrossRefPubMedPubMedCentral Zhang X, Wu Q, Wu L, Ye Z, Jiang T, Li W, Zhuang Z, Zhou M, Zhang X, Hang C. Sirtuin 1 activation protects against early brain injury after experimental subarachnoid hemorrhage in rats. Cell Death and Disease. 2016;7:e2416.CrossRefPubMedPubMedCentral
23.
go back to reference Xie XQ, Zhang P, Tian B, Chen XQ. Downregulation of NAD-dependent deacetylase SIRT2 protects mouse brain against ischemic stroke. Mol Neurobiol. 2016;54:7251–61.CrossRefPubMed Xie XQ, Zhang P, Tian B, Chen XQ. Downregulation of NAD-dependent deacetylase SIRT2 protects mouse brain against ischemic stroke. Mol Neurobiol. 2016;54:7251–61.CrossRefPubMed
24.
go back to reference Wang T, Yang B, Ji R, Xu W, Mai K, Ai Q. Omega-3 polyunsaturated fatty acids alleviate hepatic steatosis-induced inflammation through Sirt1-mediated nuclear translocation of NF-kappaB p65 subunit in hepatocytes of large yellow croaker (Larmichthys crocea). Fish Shellfish Immunol. 2017;71:76–82.CrossRefPubMed Wang T, Yang B, Ji R, Xu W, Mai K, Ai Q. Omega-3 polyunsaturated fatty acids alleviate hepatic steatosis-induced inflammation through Sirt1-mediated nuclear translocation of NF-kappaB p65 subunit in hepatocytes of large yellow croaker (Larmichthys crocea). Fish Shellfish Immunol. 2017;71:76–82.CrossRefPubMed
25.
go back to reference Cai Y, Xu L, Xu H, Fan X. SIRT1 and neural cell fate determination. Mol Neurobiol. 2016;53:2815–25.CrossRefPubMed Cai Y, Xu L, Xu H, Fan X. SIRT1 and neural cell fate determination. Mol Neurobiol. 2016;53:2815–25.CrossRefPubMed
26.
go back to reference Godoy JA, Zolezzi JM, Braidy N, Inestrosa NC. Role of Sirt1 during the ageing process: relevance to protection of synapses in the brain. Mol Neurobiol. 2014;50:744–56.CrossRefPubMed Godoy JA, Zolezzi JM, Braidy N, Inestrosa NC. Role of Sirt1 during the ageing process: relevance to protection of synapses in the brain. Mol Neurobiol. 2014;50:744–56.CrossRefPubMed
27.
go back to reference Kim YM, Park EJ, Kim JH, Park SW, Kim HJ, Chang KC. Ethyl pyruvate inhibits the acetylation and release of HMGB1 via effects on SIRT1/STAT signaling in LPS-activated RAW264.7 cells and peritoneal macrophages. Int Immunopharmacol. 2016;41:98–105.CrossRefPubMed Kim YM, Park EJ, Kim JH, Park SW, Kim HJ, Chang KC. Ethyl pyruvate inhibits the acetylation and release of HMGB1 via effects on SIRT1/STAT signaling in LPS-activated RAW264.7 cells and peritoneal macrophages. Int Immunopharmacol. 2016;41:98–105.CrossRefPubMed
28.
go back to reference Serini S, Calviello G. Reduction of oxidative/Nitrosative stress in brain and its involvement in the neuroprotective effect of n-3 PUFA in Alzheimer’s disease. Curr Alzheimer Res. 2016;13:123–34.CrossRefPubMed Serini S, Calviello G. Reduction of oxidative/Nitrosative stress in brain and its involvement in the neuroprotective effect of n-3 PUFA in Alzheimer’s disease. Curr Alzheimer Res. 2016;13:123–34.CrossRefPubMed
29.
go back to reference Wang B, Wu X, Guo M, Li M, Xu X, Jin X, Zhang X. Effects of ω-3 fatty acids on toll-like receptor 4 and nuclear factor-κB p56 in lungs of rats with severe acute pancreatitis. World J Gastroentero. 2016;22:9784–93.CrossRef Wang B, Wu X, Guo M, Li M, Xu X, Jin X, Zhang X. Effects of ω-3 fatty acids on toll-like receptor 4 and nuclear factor-κB p56 in lungs of rats with severe acute pancreatitis. World J Gastroentero. 2016;22:9784–93.CrossRef
30.
go back to reference Delattre AM, Carabelli B, Mori MA, Kempe PG, Rizzo DSL, Zanata SM, Machado RB, Suchecki D, Andrade DCB, Lima M, et al. Maternal omega-3 supplement improves dopaminergic system in pre- and postnatal inflammation-induced neurotoxicity in Parkinson’s disease model. Mol Neurobiol. 2017;54:2090–106.CrossRefPubMed Delattre AM, Carabelli B, Mori MA, Kempe PG, Rizzo DSL, Zanata SM, Machado RB, Suchecki D, Andrade DCB, Lima M, et al. Maternal omega-3 supplement improves dopaminergic system in pre- and postnatal inflammation-induced neurotoxicity in Parkinson’s disease model. Mol Neurobiol. 2017;54:2090–106.CrossRefPubMed
31.
go back to reference Chang CY, Kuan YH, Li JR, Chen WY, Ou YC, Pan HC, Liao SL, Raung SL, Chang CJ, Chen CJ. Docosahexaenoic acid reduces cellular inflammatory response following permanent focal cerebral ischemia in rats. J Nutr Biochem. 2013;24:2127–37.CrossRefPubMed Chang CY, Kuan YH, Li JR, Chen WY, Ou YC, Pan HC, Liao SL, Raung SL, Chang CJ, Chen CJ. Docosahexaenoic acid reduces cellular inflammatory response following permanent focal cerebral ischemia in rats. J Nutr Biochem. 2013;24:2127–37.CrossRefPubMed
32.
go back to reference Harvey LD, Yin Y, Attarwala IY, Begum G, Deng J, Yan HQ, Dixon CE, Sun D. Administration of DHA reduces endoplasmic reticulum stress-associated inflammation and alters microglial or macrophage activation in traumatic brain injury. Asn Neuro. 2015;7:1759091415618969.CrossRefPubMedPubMedCentral Harvey LD, Yin Y, Attarwala IY, Begum G, Deng J, Yan HQ, Dixon CE, Sun D. Administration of DHA reduces endoplasmic reticulum stress-associated inflammation and alters microglial or macrophage activation in traumatic brain injury. Asn Neuro. 2015;7:1759091415618969.CrossRefPubMedPubMedCentral
33.
go back to reference Kurtys E, Eisel UL, Verkuyl JM, Broersen LM, Dierckx RA, de Vries EF. The combination of vitamins and omega-3 fatty acids has an enhanced anti-inflammatory effect on microglia. Neurochem Int. 2016;99:206–14.CrossRefPubMed Kurtys E, Eisel UL, Verkuyl JM, Broersen LM, Dierckx RA, de Vries EF. The combination of vitamins and omega-3 fatty acids has an enhanced anti-inflammatory effect on microglia. Neurochem Int. 2016;99:206–14.CrossRefPubMed
34.
go back to reference Pu H, Guo Y, Zhang W, Huang L, Wang G, Liou AK, Zhang J, Zhang P, Leak RK, Wang Y, et al. Omega-3 polyunsaturated fatty acid supplementation improves neurologic recovery and attenuates white matter injury after experimental traumatic brain injury. J Cereb Blood Flow Metab. 2013;33:1474–84.CrossRefPubMedPubMedCentral Pu H, Guo Y, Zhang W, Huang L, Wang G, Liou AK, Zhang J, Zhang P, Leak RK, Wang Y, et al. Omega-3 polyunsaturated fatty acid supplementation improves neurologic recovery and attenuates white matter injury after experimental traumatic brain injury. J Cereb Blood Flow Metab. 2013;33:1474–84.CrossRefPubMedPubMedCentral
35.
go back to reference Yang X, Wu Q, Zhang L, Feng L. Inhibition of histone deacetylase 3 (HDAC3) mediates ischemic preconditioning and protects cortical neurons against ischemia in rats. Front Mol Neurosci. 2016;9:131.PubMedPubMedCentral Yang X, Wu Q, Zhang L, Feng L. Inhibition of histone deacetylase 3 (HDAC3) mediates ischemic preconditioning and protects cortical neurons against ischemia in rats. Front Mol Neurosci. 2016;9:131.PubMedPubMedCentral
36.
go back to reference Hopp S, Nolte MW, Stetter C, Kleinschnitz C, Sirén A, Albert-Weissenberger C. Alleviation of secondary brain injury, posttraumatic inflammation, and brain edema formation by inhibition of factor XIIa. J Neuroinflamm. 2017;14:39.CrossRef Hopp S, Nolte MW, Stetter C, Kleinschnitz C, Sirén A, Albert-Weissenberger C. Alleviation of secondary brain injury, posttraumatic inflammation, and brain edema formation by inhibition of factor XIIa. J Neuroinflamm. 2017;14:39.CrossRef
37.
go back to reference Parker TM, Nguyen AH, Rabang JR, Patil AA, Agrawal DK. The danger zone: systematic review of the role of HMGB1 danger signalling in traumatic brain injury. Brain Inj. 2017;31:2–8.CrossRefPubMed Parker TM, Nguyen AH, Rabang JR, Patil AA, Agrawal DK. The danger zone: systematic review of the role of HMGB1 danger signalling in traumatic brain injury. Brain Inj. 2017;31:2–8.CrossRefPubMed
38.
go back to reference Ma Y, Nie H, Chen H, Li J, Hong Y, Wang B, Wang C, Zhang J, Cao W, Zhang M, et al. NAD(+)/NADH metabolism and NAD(+)-dependent enzymes in cell death and ischemic brain injury: current advances and therapeutic implications. Curr Med Chem. 2015;22:1239–47.CrossRefPubMed Ma Y, Nie H, Chen H, Li J, Hong Y, Wang B, Wang C, Zhang J, Cao W, Zhang M, et al. NAD(+)/NADH metabolism and NAD(+)-dependent enzymes in cell death and ischemic brain injury: current advances and therapeutic implications. Curr Med Chem. 2015;22:1239–47.CrossRefPubMed
39.
go back to reference Logsdon AF, Lucke-Wold BP, Turner RC, Huber JD, Rosen CL, Simpkins JW. Role of microvascular disruption in brain damage from traumatic brain injury. Compr Physiol. 2015;5:1147–60.CrossRefPubMedPubMedCentral Logsdon AF, Lucke-Wold BP, Turner RC, Huber JD, Rosen CL, Simpkins JW. Role of microvascular disruption in brain damage from traumatic brain injury. Compr Physiol. 2015;5:1147–60.CrossRefPubMedPubMedCentral
40.
go back to reference Hamblin MR. Photobiomodulation for traumatic brain injury and stroke. J Neurosci Res. 2018;96:731–43.CrossRefPubMed Hamblin MR. Photobiomodulation for traumatic brain injury and stroke. J Neurosci Res. 2018;96:731–43.CrossRefPubMed
41.
go back to reference Foglio E, Puddighinu G, Germani A, Russo MA, Limana F. HMGB1 inhibits apoptosis following MI and induces autophagy via mTORC1 inhibition. J Cell Physiol. 2017;232:1135–43.CrossRefPubMed Foglio E, Puddighinu G, Germani A, Russo MA, Limana F. HMGB1 inhibits apoptosis following MI and induces autophagy via mTORC1 inhibition. J Cell Physiol. 2017;232:1135–43.CrossRefPubMed
42.
go back to reference Takizawa T, Shibata M, Kayama Y, Shimizu T, Toriumi H, Ebine T, Unekawa M, Koh A, Yoshimura A, Suzuki N. High-mobility group box 1 is an important mediator of microglial activation induced by cortical spreading depression. J Cereb Blood Flow Metab. 2017;37:890–901.CrossRefPubMed Takizawa T, Shibata M, Kayama Y, Shimizu T, Toriumi H, Ebine T, Unekawa M, Koh A, Yoshimura A, Suzuki N. High-mobility group box 1 is an important mediator of microglial activation induced by cortical spreading depression. J Cereb Blood Flow Metab. 2017;37:890–901.CrossRefPubMed
43.
go back to reference Polito F, Cicciu M, Aguennouz M, Cucinotta M, Cristani M, Lauritano F, Sindoni A, Gioffre-Florio M, Fama F. Prognostic value of HMGB1 and oxidative stress markers in multiple trauma patients: a single-centre prospective study. Int J Immunopath Ph. 2016;29:504–9.CrossRef Polito F, Cicciu M, Aguennouz M, Cucinotta M, Cristani M, Lauritano F, Sindoni A, Gioffre-Florio M, Fama F. Prognostic value of HMGB1 and oxidative stress markers in multiple trauma patients: a single-centre prospective study. Int J Immunopath Ph. 2016;29:504–9.CrossRef
44.
go back to reference Kang KA, Piao MJ, Ryu YS, Kang HK, Chang WY, Keum YS, Hyun JW. Interaction of DNA demethylase and histone methyltransferase upregulates Nrf2 in 5-fluorouracil-resistant colon cancer cells. Oncotarget. 2016;7:40594–620.PubMedPubMedCentral Kang KA, Piao MJ, Ryu YS, Kang HK, Chang WY, Keum YS, Hyun JW. Interaction of DNA demethylase and histone methyltransferase upregulates Nrf2 in 5-fluorouracil-resistant colon cancer cells. Oncotarget. 2016;7:40594–620.PubMedPubMedCentral
45.
go back to reference Bai Y, Du S, Li F, Huang F, Deng R, Zhou J, Chen D. Histone deacetylase-high mobility group box-1 pathway targeted by hypaconitine suppresses the apoptosis of endothelial cells. Exp Biol Med (Maywood). 2017;242:527–35.CrossRef Bai Y, Du S, Li F, Huang F, Deng R, Zhou J, Chen D. Histone deacetylase-high mobility group box-1 pathway targeted by hypaconitine suppresses the apoptosis of endothelial cells. Exp Biol Med (Maywood). 2017;242:527–35.CrossRef
46.
go back to reference Garmpis N, Damaskos C, Garmpi A, Dimitroulis D, Spartalis E, Margonis GA, Schizas D, Deskou I, Doula C, Magkouti E. Targeting histone deacetylases in malignant melanoma: a future therapeutic agent or just great expectations? Anticancer Res. 2017;37:5355–62.PubMed Garmpis N, Damaskos C, Garmpi A, Dimitroulis D, Spartalis E, Margonis GA, Schizas D, Deskou I, Doula C, Magkouti E. Targeting histone deacetylases in malignant melanoma: a future therapeutic agent or just great expectations? Anticancer Res. 2017;37:5355–62.PubMed
Metadata
Title
Omega-3 polyunsaturated fatty acid attenuates the inflammatory response by modulating microglia polarization through SIRT1-mediated deacetylation of the HMGB1/NF-κB pathway following experimental traumatic brain injury
Authors
Xiangrong Chen
Chunnuan Chen
Sining Fan
Shukai Wu
Fuxing Yang
Zhongning Fang
Huangde Fu
Yasong Li
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2018
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-018-1151-3

Other articles of this Issue 1/2018

Journal of Neuroinflammation 1/2018 Go to the issue