458
Views
11
CrossRef citations to date
0
Altmetric
Review

The second phase of brain trauma can be controlled by nutraceuticals that suppress DAMP-mediated microglial activation

& ORCID Icon
Pages 559-570 | Received 06 Jan 2021, Accepted 19 Mar 2021, Published online: 05 Apr 2021

References

  • Georges A, Das J M. Traumatic brain injury. 2021 Jan 16. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2021. Jan. PMID: 29083790. Bookshelf ID:NBK459300
  • Capizzi A, Woo J, Verduzco-Gutierrez M. Traumatic brain injury: an overview of epidemiology, pathophysiology, and medical management. Med Clin North Am. 2020;104:213–238.
  • Hugon J, Hourregue C, Cognat E, et al. CHRONIC TRAUMATIC ENCEPHALOPATHY. Neurochirurgie. 2021 Feb 20:S0028-3770(21)00042–4. Online ahead of print. DOI: 10.1016/j.neuchi.2021.02.003
  • Liu T, Zhao DX, Cui H, et al. Therapeutic hypothermia attenuates tissue damage and cytokine expression after traumatic brain injury by inhibiting necroptosis in the rat. Sci Rep. 2016;6:24547.
  • Okuma Y, Liu K, Wake H, et al. Anti-high mobility group box-1 antibody therapy for traumatic brain injury. Ann Neurol. 2012;72:373–384.
  • Park JS, Svetkauskaite D, He Q, et al. Involvement of toll-like receptors 2 and 4 in cellular activation by high mobility group box 1 protein. J Biol Chem. 2004;279:7370–7377.
  • Sims GP, Rowe DC, Rietdijk ST, et al. HMGB1 and RAGE in inflammation and cancer. Annu Rev Immunol. 2010;28:367–388.
  • Paudel YN, Angelopoulou E, Piperi C, et al. Enlightening the role of high mobility group box 1 (HMGB1) in inflammation: updates on receptor signalling. Eur J Pharmacol. 2019;858:172487.
  • Zhang QG, Laird MD, Han D, et al. Critical role of NADPH oxidase in neuronal oxidative damage and microglia activation following traumatic brain injury. PLoS One. 2012;7:e34504.
  • Cooney SJ, Bermudez-Sabogal SL, Byrnes KR. Cellular and temporal expression of NADPH oxidase (NOX) isotypes after brain injury. J Neuroinflammation. 2013;10:155.
  • Shultz SR, Bao F, Weaver LC, et al. Treatment with an anti-CD11d integrin antibody reduces neuroinflammation and improves outcome in a rat model of repeated concussion. J Neuroinflammation. 2013;10:26.
  • Gyoneva S, Ransohoff RM. Inflammatory reaction after traumatic brain injury: therapeutic potential of targeting cell-cell communication by chemokines. Trends Pharmacol Sci. 2015;36:471–480.
  • Utagawa A, Bramlett HM, Daniels L, et al. Transient blockage of the CD11d/CD18 integrin reduces contusion volume and macrophage infiltration after traumatic brain injury in rats. Brain Res. 2008;1207:155–163.
  • Semple BD, Bye N, Rancan M, et al. Role of CCL2 (MCP-1) in traumatic brain injury (TBI): evidence from severe TBI patients and CCL2-/- mice. J Cereb Blood Flow Metab. 2010;30:769–782.
  • Hsieh CL, Niemi EC, Wang SH, et al. CCR2 deficiency impairs macrophage infiltration and improves cognitive function after traumatic brain injury. J Neurotrauma. 2014;31:1677–1688.
  • Liddelow SA, Guttenplan KA, Clarke LE, et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature. 2017;541(7638):481–487. .
  • Loane DJ, Kumar A. Microglia in the TBI brain: the good, the bad, and the dysregulated. Exp Neurol. 2016;275(Pt 3):316–327.
  • Morgan SC, Taylor DL, Pocock JM. Microglia release activators of neuronal proliferation mediated by activation of mitogen-activated protein kinase, phosphatidylinositol-3-kinase/Akt and delta-Notch signalling cascades. J Neurochem. 2004;90:89–101.
  • Butovsky O, Ziv Y, Schwartz A, et al. Microglia activated by IL-4 or IFN-gamma differentially induce neurogenesis and oligodendrogenesis from adult stem/progenitor cells. Mol Cell Neurosci. 2006;31:149–160.
  • Jetten N, Verbruggen S, Gijbels MJ, et al. Anti-inflammatory M2, but not pro-inflammatory M1 macrophages promote angiogenesis in vivo. Angiogenesis. 2014;17:109–118.
  • Miron VE, Boyd A, Zhao JW, et al. M2 microglia and macrophages drive oligodendrocyte differentiation during CNS remyelination. Nat Neurosci. 2013;16:1211–1218.
  • Chang EY, Guo B, Doyle SE, et al. Cutting edge: involvement of the type I IFN production and signaling pathway in lipopolysaccharide-induced IL-10 production. J Immunol. 2007;178:6705–6709.
  • Cao S, Liu J, Song L, et al. The protooncogene c-Maf is an essential transcription factor for IL-10 gene expression in macrophages. J Immunol. 2005;174:3484–3492.
  • Braun DA, Fribourg M, Sealfon SC. Cytokine response is determined by duration of receptor and signal transducers and activators of transcription 3 (STAT3) activation. J Biol Chem. 2013;288:2986–2993. .
  • Yasukawa H, Ohishi M, Mori H, et al. IL-6 induces an anti-inflammatory response in the absence of SOCS3 in macrophages. Nat Immunol. 2003;4:551–556.
  • Pawate S, Shen Q, Fan F, et al. Redox regulation of glial inflammatory response to lipopolysaccharide and interferongamma. J Neurosci Res. 2004;77:540–551.
  • Li Q, Harraz MM, Zhou W, et al. Nox2 and Rac1 regulate H2O2-dependent recruitment of TRAF6 to endosomal interleukin-1 receptor complexes. Mol Cell Biol. 2006;26:140–154.
  • Li Q, Spencer NY, Oakley FD, et al. Endosomal Nox2 facilitates redox-dependent induction of NF-kappaB by TNF-alpha. Antioxid Redox Signal. 2009;11:1249–1263.
  • Yeung F, Hoberg JE, Ramsey CS, et al. Modulation of NF-kappaB-dependent transcription and cell survival by the SIRT1 deacetylase. Embo J. 2004;23:2369–2380.
  • Zhang WB, Yang F, Wang Y, et al. Inhibition of HDAC6 attenuates LPS-induced inflammation in macrophages by regulating oxidative stress and suppressing the TLR4-MAPK/NF-κB pathways. Biomed Pharmacother. 2019;117:109166.
  • Liu J, Chang G, Huang J, et al. Sodium Butyrate Inhibits the Inflammation of Lipopolysaccharide-Induced Acute Lung Injury in Mice by Regulating the Toll-Like Receptor 4/Nuclear Factor κB Signaling Pathway. J Agric Food Chem. 2019;67:1674–1682.
  • Jiang L, Wang J, Liu Z, et al. Sodium Butyrate Alleviates Lipopolysaccharide-Induced Inflammatory Responses by Down-Regulation of NF-κB, NLRP3 Signaling Pathway, and Activating Histone Acetylation in Bovine Macrophages. Front Vet Sci. 2020;7:579674.
  • Lee C, Kim BG, Kim JH, et al. Sodium butyrate inhibits the NF-kappa B signaling pathway and histone deacetylation, and attenuates experimental colitis in an IL-10 independent manner. Int Immunopharmacol. 2017;51:47–56.
  • Zhang B, Wang B, Cao S, et al. Epigallocatechin-3-Gallate (EGCG) Attenuates Traumatic Brain Injury by Inhibition of Edema Formation and Oxidative Stress. Korean J Physiol Pharmacol. 2015;19:491–497.
  • Itoh T, Tabuchi M, Mizuguchi N, et al. Neuroprotective effect of (-)-epigallocatechin-3-gallate in rats when administered pre- or post-traumatic brain injury. J Neural Transm (Vienna). 2013;120:767–783.
  • Itoh T, Imano M, Nishida S, et al. (-)-Epigallocatechin-3-gallate protects against neuronal cell death and improves cerebral function after traumatic brain injury in rats. Neuromolecular Med. 2011;13:300–309.
  • Erbil G, Sacik U, Yilmaz F, et al. The effect of ferulic acid on experimental traumatic brain damage in rats. Bratisl Lek Listy. 2019;120:372–379.
  • Fetoni AR, Eramo S, Troiani D, et al. Therapeutic window for ferulic acid protection against noise-induced hearing loss in the guinea pig. Acta Otolaryngol. 2011;131:419–427.
  • Chen X, Wu S, Chen C, et al. Omega-3 polyunsaturated fatty acid supplementation attenuates microglial-induced inflammation by inhibiting the HMGB1/TLR4/NF-kappaB pathway following experimental traumatic brain injury. J Neuroinflammation. 2017;14:143.
  • Tang R, Lin YM, Liu HX, et al. Neuroprotective effect of docosahexaenoic acid in rat traumatic brain injury model via regulation of TLR4/NF-Kappa B signaling pathway. Int J Biochem Cell Biol. 2018;99:64–71.
  • Chen X, Chen C, Fan S, et al. Omega-3 polyunsaturated fatty acid attenuates the inflammatory response by modulating microglia polarization through SIRT1-mediated deacetylation of the HMGB1/NF-kappaB pathway following experimental traumatic brain injury. J Neuroinflammation. 2018;15:116.
  • Lewis MD. Concussions, Traumatic Brain Injury, and the Innovative Use of Omega-3s. J Am Coll Nutr. 2016;35:469–475.
  • Soltani Z, Khaksari M, Jafari E, et al. Is genistein neuroprotective in traumatic brain injury? Physiol Behav. 2015;152:26–31.
  • Ding K, Wang H, Xu J, et al. Melatonin reduced microglial activation and alleviated neuroinflammation induced neuron degeneration in experimental traumatic brain injury: possible involvement of mTOR pathway. Neurochem Int. 2014;76:23–31.
  • Wang J, Jiang C, Zhang K, et al. Melatonin receptor activation provides cerebral protection after traumatic brain injury by mitigating oxidative stress and inflammation via the Nrf2 signaling pathway. Free Radic Biol Med. 2019;131:345–355.
  • Lin XJ, Liu R, Li C, et al. Melatonin ameliorates spatial memory and motor deficits via preserving the integrity of cortical and hippocampal dendritic spine morphology in mice with neurotrauma. Inflammopharmacology. 2020;28:1553–1566.
  • Wu L, Mo W, Feng J, et al. Astaxanthin attenuates hepatic damage and mitochondrial dysfunction in non-alcoholic fatty liver disease by up-regulating the FGF21/PGC-1α pathway. Br J Pharmacol. 2020;177:3760–3777.
  • Hong BE, Fujimura Y, Yamada K, et al. TLR4 signaling inhibitory pathway induced by green tea polyphenol epigallocatechin-3-gallate through 67-kDa laminin receptor. J Immunol. 2010;185:33–45.
  • Byun EH, Omura T, Yamada K, et al. Green tea polyphenol epigallocatechin-3-gallate inhibits TLR2 signaling induced by peptidoglycan through the polyphenol sensing molecule 67-kDa laminin receptor. FEBS Lett. 2011;585:814–820.
  • Byun EB, Choi HG, Sung NY, et al. Green tea polyphenol epigallocatechin-3-gallate inhibits TLR4 signaling through the 67-kDa laminin receptor on lipopolysaccharide-stimulated dendritic cells. Biochem Biophys Res Commun. 2012;426:480–485.
  • Kumazoe M, Yamashita M, Nakamura Y, et al. Green Tea Polyphenol EGCG Upregulates Tollip Expression by Suppressing Elf-1 Expression. J Immunol. 2017;199:3261–3269.
  • Byun EB, Kim WS, Sung NY, et al. Epigallocatechin-3-Gallate Regulates Anti-Inflammatory Action Through 67-kDa Laminin Receptor-Mediated Tollip Signaling Induction in Lipopolysaccharide-Stimulated Human Intestinal Epithelial Cells. Cell Physiol Biochem. 2018;46:2072–2081.
  • Ren Z, Zhang R, Li Y, et al. Ferulic acid exerts neuroprotective effects against cerebral ischemia/reperfusion-induced injury via antioxidant and anti-apoptotic mechanisms in vitro and in vivo. Int J Mol Med. 2017;40:1444–1456.
  • McCarty MF, Assanga SBI. Ferulic acid may target MyD88-mediated pro-inflammatory signaling - Implications for the health protection afforded by whole grains, anthocyanins, and coffee. Med Hypotheses. 2018;118:114–120.
  • Ma ZC, Hong Q, Wang YG, et al. Ferulic acid induces heme oxygenase-1 via activation of ERK and Nrf2. Drug Discov Ther. 2011;5:299–305.
  • Catino S, Paciello F, Miceli F, et al. Ferulic Acid Regulates the Nrf2/Heme Oxygenase-1 System and Counteracts Trimethyltin-Induced Neuronal Damage in the Human Neuroblastoma Cell Line SH-SY5Y. Front Pharmacol. 2015;6:305.
  • Li B, Lee DS, Jeong GS, et al. Involvement of heme oxygenase-1 induction in the cytoprotective and immunomodulatory activities of 6,4ʹ-dihydroxy-7-methoxyflavanone in murine hippocampal and microglia cells. Eur J Pharmacol. 2012;674:153–162.
  • Kim KW, Kim HJ, Sohn JH, et al. Anti-neuroinflammatory effect of 6,8,1ʹ-tri-O-methylaverantin, a metabolite from a marine-derived fungal strain Aspergillus sp., via upregulation of heme oxygenase-1 in lipopolysaccharide-activated microglia. Neurochem Int. 2018;113:8–22.
  • Lee DS, Ko W, Yoon CS, et al. KCHO-1, a Novel Antineuroinflammatory Agent, Inhibits Lipopolysaccharide-Induced Neuroinflammatory Responses through Nrf2-Mediated Heme Oxygenase-1 Expression in Mouse BV2 Microglia Cells. Evid Based Complement Alternat Med. 2014;2014:357154.
  • Ren Z, Chen L, Wang Y, et al. Activation of the Omega-3 Fatty Acid Receptor GPR120 Protects against Focal Cerebral Ischemic Injury by Preventing Inflammation and Apoptosis in Mice. J Immunol. 2019;202:747–759.
  • Yin J, Li H, Meng C, et al. Inhibitory effects of omega-3 fatty acids on early brain injury after subarachnoid hemorrhage in rats: possible involvement of G protein-coupled receptor 120/beta-arrestin2/TGF-beta activated kinase-1 binding protein-1 signaling pathway. Int J Biochem Cell Biol. 2016;75:11–22.
  • Karakula-Juchnowicz H, Rog J, Juchnowicz D, et al. GPR120: mechanism of action, role and potential for medical applications. Postepy Hig Med Dosw. 2017;71:942–953.
  • McCarty MF. Isoflavones made simple - genistein’s agonist activity for the beta-type estrogen receptor mediates their health benefits. Med Hypotheses. 2006;66:1093–1114.
  • Asl SZ, Khaksari M, Khachki AS, et al. Contribution of estrogen receptors alpha and beta in the brain response to traumatic brain injury. J Neurosurg. 2013;119:353–361.
  • Khaksari M, Abbasloo E, Dehghan F, et al. The brain cytokine levels are modulated by estrogen following traumatic brain injury: which estrogen receptor serves as modulator? Int Immunopharmacol. 2015;28:279–287.
  • Emerson CS, Headrick JP, Vink R. Estrogen improves biochemical and neurologic outcome following traumatic brain injury in male rats, but not in females. Brain Res. 1993;608:95–100.
  • Meltser I, Tahera Y, Simpson E, et al. Estrogen receptor beta protects against acoustic trauma in mice. J Clin Invest. 2008;118:1563–1570.
  • Garcã-a JA, Volt H, Venegas C, et al. Disruption of the NF-κB/NLRP3 connection by melatonin requires retinoid-related orphan receptor-α and blocks the septic response in mice. Faseb J. 2015;29:3863–3875.
  • Early JO, Menon D, Wyse CA, et al. Circadian clock protein BMAL1 regulates IL-1Î2 in macrophages via NRF2. Proc Natl Acad Sci U S A. 2018;115:E8460–E8468.
  • Chhunchha B, Kubo E, Singh DP. Clock Protein Bmal1 and Nrf2 Cooperatively Control Aging or Oxidative Response and Redox Homeostasis by Regulating Rhythmic Expression of Prdx6. Cells. 2020;9:1861.
  • Zhou B, Zhang Y, Zhang F, et al. CLOCK/BMAL1 regulates circadian change of mouse hepatic insulin sensitivity by SIRT1. Hepatology. 2014;59:2196–2206.
  • Dohi K, Ohtaki H, Nakamachi T, et al. Gp91phox (NOX2) in classically activated microglia exacerbates traumatic brain injury. J Neuroinflammation. 2010;7:41.
  • Lo W, Bravo T, Jadhav V, et al. NADPH oxidase inhibition improves neurological outcomes in surgically-induced brain injury. Neurosci Lett. 2007;414:228–232.
  • Choi BY, Jang BG, Kim JH, et al. Prevention of traumatic brain injury-induced neuronal death by inhibition of NADPH oxidase activation. Brain Res. 2012;1481:49–58.
  • Loane DJ, Stoica BA, Byrnes KR, et al. Activation of mGluR5 and inhibition of NADPH oxidase improves functional recovery after traumatic brain injury. J Neurotrauma. 2013;30:403–412.
  • Ferreira AP, Rodrigues FS, Della-Pace ID, et al. The effect of NADPH-oxidase inhibitor apocynin on cognitive impairment induced by moderate lateral fluid percussion injury: role of inflammatory and oxidative brain damage. Neurochem Int. 2013;63:583–593.
  • Lu XY, Wang HD, Xu JG, et al. NADPH oxidase inhibition improves neurological outcome in experimental traumatic brain injury. Neurochem Int. 2014;69:14–19.
  • Zhang L, Li Z, Feng D, et al. Involvement of Nox2 and Nox4 NADPH oxidases in early brain injury after subarachnoid hemorrhage. Free Radic Res. 2017;51:316–328.
  • Feng Y, Cui C, Liu X, et al. Protective Role of Apocynin via Suppression of Neuronal Autophagy and TLR4/NF-kappaB Signaling Pathway in a Rat Model of Traumatic Brain Injury. Neurochem Res. 2017;42:3296–3309.
  • Ma MW, Wang J, Dhandapani KM, et al. NADPH oxidases in traumatic brain injury - Promising therapeutic targets? Redox Biol. 2018;16:285–293.
  • Kumar A, Barrett JP, Alvarez-Croda DM, et al. NOX2 drives M1-like microglial/macrophage activation and neurodegeneration following experimental traumatic brain injury. Brain Behav Immun. 2016;58:291–309.
  • Barrett JP, Henry RJ, Villapol S, et al. NOX2 deficiency alters macrophage phenotype through an IL-10/STAT3 dependent mechanism: implications for traumatic brain injury. J Neuroinflammation. 2017;14:65.
  • Wang J, Ma MW, Dhandapani KM, et al. Regulatory role of NADPH oxidase 2 in the polarization dynamics and neurotoxicity of microglia/macrophages after traumatic brain injury. Free Radic Biol Med. 2017;113:119–131.
  • Wang J, Ma MW, Dhandapani KM, et al. NADPH oxidase 2 deletion enhances neurogenesis following traumatic brain injury. Free Radic Biol Med. 2018;123:62–71.
  • Miao H, Li R, Han C, et al. Minocycline promotes posthemorrhagic neurogenesis via M2 microglia polarization via upregulation of the TrkB/BDNF pathway in rats. J Neurophysiol. 2018;120:1307–1317.
  • Zhang XJ, Lv MM, Zhu XQ, et al. Microglia M1/M2 polarization contributes to electromagnetic pulse-induced brain injury. J Biol Regul Homeost Agents. 2019;33:1051–1062.
  • Choi SH, Aid S, Kim HW, et al. Inhibition of NADPH oxidase promotes alternative and anti-inflammatory microglial activation during neuroinflammation. J Neurochem. 2012;120:292–301.
  • McCarty MF. Clinical potential of Spirulina as a source of phycocyanobilin. J Med Food. 2007;10:566–570.
  • Zheng J, Inoguchi T, Sasaki S, et al. Phycocyanin and phycocyanobilin from Spirulina platensis protect against diabetic nephropathy by inhibiting oxidative stress. Am J Physiol Regul Integr Comp Physiol. 2013;304:R110–R120.
  • Lanone S, Bloc S, Foresti R, et al. Bilirubin decreases nos2 expression via inhibition of NAD(P)H oxidase: implications for protection against endotoxic shock in rats. Faseb J. 2005;19:1890–1892.
  • Matsumoto H, Ishikawa K, Itabe H, et al. Carbon monoxide and bilirubin from heme oxygenase-1 suppresses reactive oxygen species generation and plasminogen activator inhibitor-1 induction. Mol Cell Biochem. 2006;291:21–28.
  • Jiang F, Roberts SJ, Datla S, et al. NO modulates NADPH oxidase function via heme oxygenase-1 in human endothelial cells. Hypertension. 2006;48:950–957.
  • Datla SR, Dusting GJ, Mori TA, et al. Induction of heme oxygenase-1 in vivo suppresses NADPH oxidase derived oxidative stress. Hypertension. 2007;50:636–642.
  • Luo M, Tian R, Lu N. Nitric oxide protected against NADPH oxidase-derived superoxide generation in vascular endothelium: critical role for heme oxygenase-1. Int J Biol Macromol. 2019;126:549–554.
  • Marin-Prida J, Pavon-Fuentes N, Llopiz-Arzuaga A, et al. Phycocyanobilin promotes PC12 cell survival and modulates immune and inflammatory genes and oxidative stress markers in acute cerebral hypoperfusion in rats. Toxicol Appl Pharmacol. 2013;272:49–60.
  • Penton-Rol G, Marin-Prida J, Falcon-Cama V. C-Phycocyanin and Phycocyanobilin as Remyelination Therapies for Enhancing Recovery in Multiple Sclerosis and Ischemic Stroke: a Preclinical Perspective. Behav Sci. 2018;8:15.
  • Pavon-Fuentes N, Marin-Prida J, Llopiz-Arzuaga A, et al. Phycocyanobilin reduces brain injury after endothelin-1- induced focal cerebral ischemia. Clin Exp Pharmacol Physiol. 2020;47:383–392.
  • Thaakur S, Sravanthi R. Neuroprotective effect of Spirulina in cerebral ischemia-reperfusion injury in rats. J Neural Transm. 2010;117: 1083–1091.
  • Liu Q, Li W, Qin S. Therapeutic effect of phycocyanin on acute liver oxidative damage caused by X-ray. Biomed Pharmacother. 2020;130:110553.
  • Strasky Z, Zemankova L, Nemeckova I, et al. Spirulina platensis and phycocyanobilin activate atheroprotective heme oxygenase-1: a possible implication for atherogenesis. Food Funct. 2013;4:1586–1594.
  • Vã-tek L. Bilirubin as a signaling molecule. Med Res Rev. 2020;40:1335–1351.
  • Miao W, Hu L, Scrivens PJ, et al. Transcriptional regulation of NF-E2 p45-related factor (NRF2) expression by the aryl hydrocarbon receptor-xenobiotic response element signaling pathway: direct cross-talk between phase I and II drug-metabolizing enzymes. J Biol Chem. 2005;280:20340–20348.
  • Liao B, Zhao W, Beers DR, et al. Transformation from a neuroprotective to a neurotoxic microglial phenotype in a mouse model of ALS. Exp Neurol. 2012;237:147–152.
  • Li YH, Yu JZ, Xin YL, et al. Protective effect of a novel Rho kinase inhibitor WAR-5 in experimental autoimmune encephalomyelitis by modulating inflammatory response and neurotrophic factors. Exp Mol Pathol. 2015;99:220–228.
  • Kahn MA, Kumar S, Liebl D, et al. Mice lacking NT-3, and its receptor TrkC, exhibit profound deficiencies in CNS glial cells. Glia. 1999 April;26(2):153–165. .
  • Chen S, Zhang H, Pu H, et al. n-3 PUFA supplementation benefits microglial responses to myelin pathology. Sci Rep. 2014;4:7458.
  • Cai W, Liu S, Hu M, et al. Post-stroke DHA Treatment Protects Against Acute Ischemic Brain Injury by Skewing Macrophage Polarity Toward the M2 Phenotype. Transl Stroke Res. 2018;9:669–680.
  • Popiolek-Barczyk K, Kolosowska N, Piotrowska A, et al. Parthenolide Relieves Pain and Promotes M2 Microglia/Macrophage Polarization in Rat Model of Neuropathy. Neural Plast. 2015;2015:676473.
  • Zhou X, Cao Y, Ao G, et al. CaMKKbeta-dependent activation of AMP-activated protein kinase is critical to suppressive effects of hydrogen sulfide on neuroinflammation. Antioxid Redox Signal. 2014;21:1741–1758.
  • Zhang M, Wu X, Xu Y, et al. The cystathionine beta-synthase/hydrogen sulfide pathway contributes to microglia-mediated neuroinflammation following cerebral ischemia. Brain Behav Immun. 2017;66:332–346.
  • Zhou X, Chu X, Xin D, et al. L-Cysteine-Derived H2S Promotes Microglia M2 Polarization via Activation of the AMPK Pathway in Hypoxia-Ischemic Neonatal Mice. Front Mol Neurosci. 2019;12:58.
  • Meares GP, Qin H, Liu Y, et al. AMP-activated protein kinase restricts IFN-gamma signaling. J Immunol. 2013;190:372–380.
  • Lee YY, Park JS, Lee EJ, et al. Anti-inflammatory mechanism of ginseng saponin metabolite Rh3 in lipopolysaccharide-stimulated microglia: critical role of 5ʹ-adenosine monophosphate-activated protein kinase signaling pathway. J Agric Food Chem. 2015;63:3472–3480.
  • He C, Li H, Viollet B, et al. AMPK Suppresses Vascular Inflammation In Vivo by Inhibiting Signal Transducer and Activator of Transcription-1. Diabetes. 2015;64:4285–4297.
  • McCarty MF, O’Keefe JH, DiNicolantonio JJ. A diet rich in taurine, cysteine, folate, B12 and betaine may lessen risk for Alzheimer’s disease by boosting brain synthesis of hydrogen sulfide. Med Hypotheses. 2019;132:109356.
  • Zhao H, Qu J, Li Q, et al. Taurine supplementation reduces neuroinflammation and protects against white matter injury after intracerebral hemorrhage in rats. Amino Acids. 2018;50:439–451.
  • Su Y, Fan W, Ma Z, et al. Taurine improves functional and histological outcomes and reduces inflammation in traumatic brain injury. Neuroscience. 2014;266:56–65.
  • Wang Q, Fan W, Cai Y, et al. Protective effects of taurine in traumatic brain injury via mitochondria and cerebral blood flow. Amino Acids. 2016;48:2169–2177.
  • Gupte R, Christian S, Keselman P, et al. Evaluation of taurine neuroprotection in aged rats with traumatic brain injury. Brain Imaging Behav. 2019;13:461–471.
  • Jin Q, Cheng J, Liu Y, et al. Improvement of functional recovery by chronic metformin treatment is associated with enhanced alternative activation of microglia/macrophages and increased angiogenesis and neurogenesis following experimental stroke. Brain Behav Immun. 2014;40:131–142.
  • Zhu J, Cao D, Guo C, et al. Berberine Facilitates Angiogenesis Against Ischemic Stroke Through Modulating Microglial Polarization via AMPK Signaling. Cell Mol Neurobiol. 2019;39:751–768.
  • Ma X, Li H, He Y, et al. The emerging link between O-GlcNAcylation and neurological disorders. Cell Mol Life Sci. 2017;74:3667–3686.
  • Yi HA, Yi SD, Jang BC, et al. Inhibitory effects of glucosamine on lipopolysaccharide-induced activation in microglial cells. Clin Exp Pharmacol Physiol. 2005;32:1097–1103.
  • Hwang SY, Shin JH, Hwang JS, et al. Glucosamine exerts a neuroprotective effect via suppression of inflammation in rat brain ischemia/reperfusion injury. Glia. 2010;58:1881–1892.
  • Hwang SY, Hwang JS, Kim SY, et al. Glucosamine inhibits lipopolysaccharide-stimulated inducible nitric oxide synthase induction by inhibiting expression of NF-kappaB/Rel proteins at the mRNA and protein levels. Nitric Oxide. 2013;31:1–8.
  • He Y, Ma X, Li D, et al. Thiamet G mediates neuroprotection in experimental stroke by modulating microglia/macrophage polarization and inhibiting NF-kappaB p65 signaling. J Cereb Blood Flow Metab. 2017;37:2938–2951.
  • Yun Z, Peng HZ, Wang W, et al. A20 inhibits the release of inflammatory cytokines by suppressing the activation of the nuclear factor-kappa B pathway in osteoarthritic fibroblast-like synoviocytes. Biochem Biophys Res Commun. 2019;508:877–881.
  • Yao D, Xu L, Xu O, et al. O-Linked beta-N-Acetylglucosamine Modification of A20 Enhances the Inhibition of NF-kappaB (Nuclear Factor-kappaB) Activation and Elicits Vascular Protection After Acute Endoluminal Arterial Injury. Arterioscler Thromb Vasc Biol. 2018;38:1309–1320.
  • Ma H, Li X, Zhou T, et al. Glucosamine Use, Inflammation, and Genetic Susceptibility, and Incidence of Type 2 Diabetes: a Prospective Study in UK Biobank. Diabetes Care. 2020;43:719–725.
  • King DE, Xiang J. Glucosamine/Chondroitin and Mortality in a US NHANES Cohort. J Am Board Fam Med. 2020;33:842–847.
  • Li ZH, Gao X, Chung VC, et al. Associations of regular glucosamine use with all-cause and cause-specific mortality: a large prospective cohort study. Ann Rheum Dis. 2020;79:829–836.
  • Navarro SL, White E, Kantor ED, et al. Randomized trial of glucosamine and chondroitin supplementation on inflammation and oxidative stress biomarkers and plasma proteomics profiles in healthy humans. PLoS One. 2015;10:e0117534.
  • Katoh A, Kai H, Harada H, et al. Oral Administration of Glucosamine Improves Vascular Endothelial Function by Modulating Intracellular Redox State. Int Heart J. 2017;58:926–932.
  • Weimer S, Priebs J, Kuhlow D, et al. D-Glucosamine supplementation extends life span of nematodes and of ageing mice. Nat Commun. 2014;5:3563.
  • Herrero-Beaumont G, Largo R. Glucosamine and O-GlcNAcylation: a novel immunometabolic therapeutic target for OA and chronic, low-grade systemic inflammation? Ann Rheum Dis. 2020;79:1261–1263.
  • Boontanrart M, Hall SD, Spanier JA, et al. Vitamin D3 alters microglia immune activation by an IL-10 dependent SOCS3 mechanism. J Neuroimmunol. 2016;292:126–136.
  • Zhang Y, Leung DY, Richers BN, et al. Vitamin D inhibits monocyte/macrophage proinflammatory cytokine production by targeting MAPK phosphatase-1. J Immunol. 2012;188:2127–2135.
  • Pemmari A, Paukkeri EL, Hämäläinen M, et al. MKP-1 promotes anti-inflammatory M(IL-4/IL-13) macrophage phenotype and mediates the anti-inflammatory effects of glucocorticoids. Basic Clin Pharmacol Toxicol. 2019;124:404–415.
  • Lawrence DW, Sharma B. A review of the neuroprotective role of vitamin D in traumatic brain injury with implications for supplementation post-concussion. Brain Inj. 2016;30:960–968.
  • Cui C, Song S, Cui J, et al. Vitamin D Receptor Activation Influences NADPH Oxidase (NOX(2)) Activity and Protects against Neurological Deficits and Apoptosis in a Rat Model of Traumatic Brain Injury. Oxid Med Cell Longev. 2017;2017:9245702.
  • Lee JM, Jeong SW, Kim MY, et al. The Effect of Vitamin D Supplementation in Patients with Acute Traumatic Brain Injury. World Neurosurg. 2019;126:e1421–e1426.
  • Shein NA, Grigoriadis N, Alexandrovich AG, et al. Histone deacetylase inhibitor ITF2357 is neuroprotective, improves functional recovery, and induces glial apoptosis following experimental traumatic brain injury. Faseb J. 2009;23:4266–4275.
  • Zhang B, West EJ, Van KC, et al. HDAC inhibitor increases histone H3 acetylation and reduces microglia inflammatory response following traumatic brain injury in rats. Brain Res. 2008;1226:181–191.
  • Shein NA, Shohami E. Histone deacetylase inhibitors as therapeutic agents for acute central nervous system injuries. Mol Med. 2011;17:448–456.
  • Abdanipour A, Schluesener HJ, Tiraihi T. Effects of valproic acid, a histone deacetylase inhibitor, on improvement of locomotor function in rat spinal cord injury based on epigenetic science. Iran Biomed J. 2012;16:90–100.
  • Li S, Lu X, Shao Q, et al. Early Histone Deacetylase Inhibition Mitigates Ischemia/Reperfusion Brain Injury by Reducing Microglia Activation and Modulating Their Phenotype. Front Neurol. 2019;10:893.
  • Patnala R, Arumugam TV, Gupta N, et al. HDAC Inhibitor Sodium Butyrate-Mediated Epigenetic Regulation Enhances Neuroprotective Function of Microglia During Ischemic Stroke. Mol Neurobiol. 2017;54:6391–6411.
  • McDougall A, Bayley M, Munce SE. The ketogenic diet as a treatment for traumatic brain injury: a scoping review. Brain Inj. 2018;32:416–422.
  • Hu ZG, Wang HD, Qiao L, et al. The protective effect of the ketogenic diet on traumatic brain injury-induced cell death in juvenile rats. Brain Inj. 2009;23:459–465.
  • Davis LM, Pauly JR, Readnower RD, et al. Fasting is neuroprotective following traumatic brain injury. J Neurosci Res. 2008;86:1812–1822.
  • McCarty MF, DiNicolantonio JJ, O’Keefe JH. Ketosis may promote brain macroautophagy by activating Sirt1 and hypoxia-inducible factor-1. Med Hypotheses. 2015;85:631–639.
  • McCarty MF, Lerner A. Nutraceutical Induction and Mimicry of Heme Oxygenase Activity as a Strategy for Controlling Excitotoxicity in Brain Trauma and Ischemic Stroke: focus on Oxidative Stress. Expert Rev Neurother. 2021;21:157–168. .
  • Brennan AM, Suh SW, Won SJ, et al. NADPH oxidase is the primary source of superoxide induced by NMDA receptor activation. Nat Neurosci. 2009;12:857–863.
  • Reyes RC, Brennan AM, Shen Y, et al. Activation of neuronal NMDA receptors induces superoxide-mediated oxidative stress in neighboring neurons and astrocytes. J Neurosci. 2012;32:12973–12978.
  • Lam TI, Brennan-Minnella AM, Won SJ, et al. Intracellular pH reduction prevents excitotoxic and ischemic neuronal death by inhibiting NADPH oxidase. Proc Natl Acad Sci U S A. 2013;110:E4362–E4368.
  • Brennan-Minnella AM, Shen Y, El-Benna J, et al. Phosphoinositide 3-kinase couples NMDA receptors to superoxide release in excitotoxic neuronal death. Cell Death Dis. 2013;4:e580.
  • Minnella AM, Zhao JX, Jiang X, et al. Excitotoxic superoxide production and neuronal death require both ionotropic and non-ionotropic NMDA receptor signaling. Sci Rep. 2018;8:17522.
  • Tolias KF, Bikoff JB, Burette A, et al. The Rac1-GEF Tiam1 couples the NMDA receptor to the activity-dependent development of dendritic arbors and spines. Neuron. 2005;45:525–538.
  • Cao J, Viholainen JI, Dart C, et al. The PSD95-nNOS interface: a target for inhibition of excitotoxic p38 stress-activated protein kinase activation and cell death. J Cell Biol. 2005;168:117–126.
  • Khan M, Dhammu TS, Matsuda F, et al. Targeting the nNOS/peroxynitrite/calpain system to confer neuroprotection and aid functional recovery in a mouse model of TBI. Brain Res. 2016;1630:159–170.
  • Penton-Rol G, Marin-Prida J, Pardo-Andreu G, et al. C-Phycocyanin is neuroprotective against global cerebral ischemia/reperfusion injury in gerbils. Brain Res Bull. 2011;86:42–52.
  • Marin-Prida J, Penton-Rol G, Rodrigues FP, et al. C-Phycocyanin protects SH-SY5Y cells from oxidative injury, rat retina from transient ischemia and rat brain mitochondria from Ca2+/phosphate-induced impairment. Brain Res Bull. 2012;89:159–167.
  • Oritani Y, Setoguchi Y, Ito R, et al. Comparison of (-)-epigallocatechin-3-O-gallate (EGCG) and O-methyl EGCG bioavailability in rats. Biol Pharm Bull. 2013;36:1577–1582.
  • Fujimura Y, Umeda D, Yano S, et al. The 67kDa laminin receptor as a primary determinant of anti-allergic effects of O-methylated EGCG. Biochem Biophys Res Commun. 2007;364:79–85.
  • Maeda-Yamamoto M, Ema K, Shibuichi I. In vitro and in vivo anti-allergic effects of ‘benifuuki’ green tea containing O-methylated catechin and ginger extract enhancement. Cytotechnology. 2007;55:135–142.
  • Demock M, Kornguth S. A Mechanism for the Development of Chronic Traumatic Encephalopathy From Persistent Traumatic Brain Injury. J Exp Neurosci. 2019;13:1179069519849935.
  • Ghosh S, Wu MD, Shaftel SS, et al. Sustained interleukin-1beta overexpression exacerbates tau pathology despite reduced amyloid burden in an Alzheimer’s mouse model. J Neurosci. 2013;33:5053–5064.
  • Quintanilla RA, Orellana DI, Gonzalez-Billault C, et al. Interleukin-6 induces Alzheimer-type phosphorylation of tau protein by deregulating the cdk5/p35 pathway. Exp Cell Res. 2004;295:245–257.
  • Makinde HM, Just TB, Cuda CM, et al. The Role of Microglia in the Etiology and Evolution of Chronic Traumatic Encephalopathy. Shock. 2017;48:276–283.
  • Ojo JO, Mouzon B, Greenberg MB, et al. Repetitive mild traumatic brain injury augments tau pathology and glial activation in aged hTau mice. J Neuropathol Exp Neurol. 2013;72:137–151.
  • Mckee AC, Abdolmohammadi B, Stein TD. The neuropathology of chronic traumatic encephalopathy. Handb Clin Neurol. 2018;158:297–307.
  • Madathil SK, Wilfred BS, Urankar SE, et al. Early Microglial Activation Following Closed-Head Concussive Injury Is Dominated by Pro-Inflammatory M-1 Type. Front Neurol. 2018;9:964.
  • Yuzwa SA, Shan X, Macauley MS, et al. Increasing O-GlcNAc slows neurodegeneration and stabilizes tau against aggregation. Nat Chem Biol. 2012;8:393–399.
  • Gong CX, Liu F, Iqbal K. O-GlcNAcylation: a regulator of tau pathology and neurodegeneration. Alzheimers Dement. 2016;12:1078–1089.
  • Yuzwa SA, Cheung AH, Okon M, et al. O-GlcNAc modification of tau directly inhibits its aggregation without perturbing the conformational properties of tau monomers. J Mol Biol. 2014;426:1736–1752.
  • Borghgraef P, Menuet C, Theunis C, et al. Increasing brain protein O-GlcNAc-ylation mitigates breathing defects and mortality of Tau.P301L mice. PLoS One. 2013;8:e84442.
  • Gao HM, Zhou H, Zhang F, et al. HMGB1 acts on microglia Mac1 to mediate chronic neuroinflammation that drives progressive neurodegeneration. J Neurosci. 2011;31:1081–1092.
  • Gao HM, Zhang F, Zhou H, et al. Neuroinflammation and α-synuclein dysfunction potentiate each other, driving chronic progression of neurodegeneration in a mouse model of Parkinson’s disease. Environ Health Perspect. 2011;119:807–814.
  • McCarty MF, Lerner A. Nutraceuticals Targeting Generation and Oxidant Activity of Peroxynitrite May Aid Prevention and Control of Parkinson’s Disease. Int J Mol Sci. 2020;21:3624.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.